[go: up one dir, main page]

Next Issue
Volume 14, March
Previous Issue
Volume 14, January
 
 

Pharmaceuticals, Volume 14, Issue 2 (February 2021) – 100 articles

Cover Story (view full-size image): We have demonstrated that the electrochemical method gives rise to stoichiometric cobalt ferrite nanoparticles in an easy and reproducible way, with good magnetic properties. The nanoparticles (NPs) were vectorized with folic acid to render a high uptake dose in the HeLa cancer cell line without a decrease in cell survival, up to 3 mM of Co + Fe concentration. In addition, the cytoskeleton retains its structure and morphology when nanoparticles are internalized at high doses. Relaxivity measurements of the NP-FA colloidal solution up to concentrations of 1 mM result in a high r2 value of 479 Fe+Co mM−1s−1. Finally, in vitro analysis with the HeLa cell line of phantom T2-weighted images present a progressive negative enhancement with the increase of concentration dose, in accordance with the high r2 value obtained. View this paper
  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Section
Select all
Export citation of selected articles as:
15 pages, 4425 KiB  
Article
Discovery of Substituted (2-Aminooxazol-4-yl)Isoxazole-3-carboxylic Acids as Inhibitors of Bacterial Serine Acetyltransferase in the Quest for Novel Potential Antibacterial Adjuvants
by Joana Magalhães, Nina Franko, Samanta Raboni, Giannamaria Annunziato, Päivi Tammela, Agostino Bruno, Stefano Bettati, Stefano Armao, Costanza Spadini, Clotilde Silvia Cabassi, Andrea Mozzarelli, Marco Pieroni, Barbara Campanini and Gabriele Costantino
Pharmaceuticals 2021, 14(2), 174; https://doi.org/10.3390/ph14020174 - 23 Feb 2021
Cited by 7 | Viewed by 2933
Abstract
Many bacteria and actinomycetales use L-cysteine biosynthesis to increase their tolerance to antibacterial treatment and establish a long-lasting infection. In turn, this might lead to the onset of antimicrobial resistance that currently represents one of the most menacing threats to public health worldwide. [...] Read more.
Many bacteria and actinomycetales use L-cysteine biosynthesis to increase their tolerance to antibacterial treatment and establish a long-lasting infection. In turn, this might lead to the onset of antimicrobial resistance that currently represents one of the most menacing threats to public health worldwide. The biosynthetic machinery required to synthesise L-cysteine is absent in mammals; therefore, its exploitation as a drug target is particularly promising. In this article, we report a series of inhibitors of Salmonella thyphimurium serine acetyltransferase (SAT), the enzyme that catalyzes the rate-limiting step of L-cysteine biosynthesis. The development of such inhibitors started with the virtual screening of an in-house library of compounds that led to the selection of seven structurally unrelated hit derivatives. A set of molecules structurally related to hit compound 5, coming either from the original library or from medicinal chemistry efforts, were tested to determine a preliminary structure–activity relationship and, especially, to improve the inhibitory potency of the derivatives, that was indeed ameliorated by several folds compared to hit compound 5 Despite these progresses, at this stage, the most promising compound failed to interfere with bacterial growth when tested on a Gram-negative model organism, anticipating the need for further research efforts. Full article
(This article belongs to the Special Issue Small Molecules as Antimicrobials)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the reductive sulfate assimilation pathway in Salmonella. Inorganic sulfate is transported inside the cell where it is reduced to bisulfide by a multi-step process. L-serine is activated by <span class="html-italic">O</span>-acetylation by SAT to give the product OAS, which spontaneously tautomerizes to NAS, the inducer of the cysteine regulon. A β-substitution reaction between OAS and HS<sup>-</sup>, catalyzed by OASS, leads to L-cysteine formation. This amino acid and the pathways of its biosynthesis influence many processes relevant to infection, like resistance to oxidative stress, biofilm formation and antibiotic resistance. UPAR415 is a competitive inhibitor of OASS.</p>
Full article ">Figure 2
<p>Compounds from the virtual screening.</p>
Full article ">Figure 3
<p>Inhibitory activity of compound <b>5</b>. Panel A: dependence of relative initial reaction rate on the concentration of compound <b>5</b>. The line through data points is the fitting to equation 1 with IC<sub>50</sub> = 110 ± 0 μM. Panel B: dependence of the initial reaction rate on the concentration of acetylCoA in the absence of added inhibitors (grey dots) and in the presence of different concentrations of compound <b>5</b> (green dots 30 μM; pink dots 100 μM, red dots 300 μM). The series of lines through data points is the fitting to the linearised form of equation 2 with K<sub>i</sub> = 64 ± 12 μM.</p>
Full article ">Figure 4
<p>(<b>A</b>) docking pose of compound <b>5</b> in AcCoA pocket in EcSAT (alignment of structures with pdb code 1T3D and pdb code 1SSM). (<b>B</b>) Overlay of the docking pose of compound <b>5</b> and AcCoA structure.</p>
Full article ">Scheme 1
<p><span class="html-italic"><sup>a</sup></span> Reagent and conditions: (<span class="html-italic">a</span>) triethylamine, tetrahydrofuran, rt; (<span class="html-italic">b</span>) N-Bromosuccinimide, <span class="html-italic">p</span>-toluenesulfonic acid, acetonitrile, reflux; (<span class="html-italic">c</span>) urea, dimethylformamide, MW (120 °C, 300 W), 3 min, 49%; (<span class="html-italic">d</span>) suitable bromobenzene derivative or 4-bromopyridine (<b>21e</b>), Sodium <span class="html-italic">t</span>-butoxide, X-Phos Pd G2, t-butanol, toluene, MW (130 °C, 300 W), 15 min, 8–35%; (<span class="html-italic">e</span>) LiOH, THF/H<sub>2</sub>O/MeOH 3:1:1, rt, 3 h, quantitative.</p>
Full article ">
16 pages, 2360 KiB  
Article
Preclinical Pharmacokinetics and Biodistribution of Anticancer Dinuclear Palladium(II)-Spermine Complex (Pd2Spm) in Mice
by Martin Vojtek, Salomé Gonçalves-Monteiro, Edgar Pinto, Sára Kalivodová, Agostinho Almeida, Maria P. M. Marques, Ana L. M. Batista de Carvalho, Clara B. Martins, Helder Mota-Filipe, Isabel M. P. L. V. O. Ferreira and Carmen Diniz
Pharmaceuticals 2021, 14(2), 173; https://doi.org/10.3390/ph14020173 - 23 Feb 2021
Cited by 14 | Viewed by 3784
Abstract
Palladium-based compounds are regarded as potential analogs to platinum anticancer drugs with improved properties. The present study assessed the pharmacokinetics and biodistribution of a dinuclear palladium(II)-spermine chelate (Pd2Spm), which has previously been shown to possess promising in vitro activity against several [...] Read more.
Palladium-based compounds are regarded as potential analogs to platinum anticancer drugs with improved properties. The present study assessed the pharmacokinetics and biodistribution of a dinuclear palladium(II)-spermine chelate (Pd2Spm), which has previously been shown to possess promising in vitro activity against several therapy-resistant cancers. Using inductively coupled plasma-mass spectrometry, the kinetic profiles of palladium/platinum in serum, serum ultrafiltrate and tissues (kidney, liver, brain, heart, lungs, ovaries, adipose tissue and mammary glands) were studied in healthy female Balb/c mice after a single intraperitoneal bolus injection of Pd2Spm (3 mg/kg bw) or cisplatin (3.5 mg/kg bw) between 0.5 and 48 h post-injection. Palladium in serum exhibited biphasic kinetics with a terminal half-life of 20.7 h, while the free palladium in serum ultrafiltrate showed a higher terminal half-life than platinum (35.5 versus 31.5 h). Palladium was distributed throughout most of the tissues except for the brain, with the highest values in the kidney, followed by the liver, lungs, ovaries, adipose tissue and mammary glands. The in vitro cellular accumulation was also evaluated in breast cancer cells, evidencing a passive diffusion as a mechanism of Pd2Spm’s cellular entry. This study reports, for the first time, the favorable pharmacokinetics and biodistribution of Pd2Spm, which may become a promising pharmacological agent for cancer treatment. Full article
(This article belongs to the Special Issue Metal-Based Drugs: Updates and Perspectives)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structure of Pt(II)- and Pd(II)-based agents: Worldwide approved Pt drugs—(<b>a</b>) cisplatin, (<b>b</b>) carboplatin and (<b>c</b>) oxaliplatin. (<b>d</b>) Novel palladium(II)-based complex Pd<sub>2</sub>Spm.</p>
Full article ">Figure 2
<p>Kinetic profiles of Pd (from Pd<sub>2</sub>Spm, 3 mg/kg bw) and Pt (from cisplatin, 3.5 mg/kg bw) in serum and serum ultrafiltrate after single intraperitoneal bolus injection in Balb/c mice. Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 5 animals per time point). (<b>a</b>) Concentration–time plot of mean Pd or Pt levels in serum and serum ultrafiltrate. (<b>b</b>) Concentration–time plot of mean Pd or Pt levels in serum and serum ultrafiltrate normalized to administered metal dose.</p>
Full article ">Figure 3
<p>Drug binding to plasma proteins versus time, for cisplatin and Pd<sub>2</sub>Spm. Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 5 animals per time point) and were compared with Student’s <span class="html-italic">t</span>-test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Biodistribution of total Pd (from Pd<sub>2</sub>Spm, 3 mg/kg bw) and Pt (from cisplatin, 3.5 mg/kg bw) in tissues after single intraperitoneal bolus injection in Balb/c mice. Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 5 animals per time point) and were compared with Student’s <span class="html-italic">t</span>-test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>a</b>) AUC<sub>0-48h</sub> (area under the Pd/Pt concentration–time curve). Units: h × ng/g of tissue; (<b>b</b>) dose-normalized AUC<sub>0–48 h</sub> (area under the Pd/Pt concentration–time curve). Units: h × ng/g per µg metal dose.</p>
Full article ">Figure 5
<p>Cellular accumulation of Pd<sub>2</sub>Spm and cisplatin in (<b>a</b>) MDA-MB-231 and (<b>b</b>) HCC-1143 triple-negative breast cancer cells. Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 4 independent experiments) and were compared with Student’s <span class="html-italic">t</span>-test. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure A1
<p>Kinetic profiles of Pd (from Pd2Spm, 3 mg/kg bw) and Pt (from cisplatin, 3.5 mg/kg bw) in tissues after single intraperitoneal bolus injection in Balb/c mice. Data are expressed as mean ± SEM (n = 5 animals per time point). Units: h.ng/g of tissue (<b>a</b>) Concentration–time plot of mean Pd levels in tissues. (<b>b</b>) Concen-tration–time plot of mean Pt levels in tissues.</p>
Full article ">Figure A2
<p>Metal dose-normalized kinetic profiles of Pd (from Pd<sub>2</sub>Spm, 3 mg/kg bw) and Pt (from cisplatin, 3.5 mg/kg bw) in tissues after single intraperitoneal bolus injection in Balb/c mice. Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 5 animals per time point). Units: h.ng/g per µg metal dose (<b>a</b>) Concentration–time plot of mean Pd levels (normalized to administered metal dose) in tissues. (<b>b</b>) Concentration–time plot of mean Pt levels (normalized to administered metal dose) in tissues.</p>
Full article ">
12 pages, 2587 KiB  
Article
Up-Regulated Vitamin D Receptor by Pelargonium sidoides Extract EPs® 7630 Contributes to Rhinovirus Defense in Bronchial Epithelial Cells
by Michael Roth, Qingzhu Sun and Michael Tamm
Pharmaceuticals 2021, 14(2), 172; https://doi.org/10.3390/ph14020172 - 22 Feb 2021
Cited by 13 | Viewed by 3375
Abstract
EPs®7630, extracted from Pelargonium sidoides, reduces the severity of viral upper respiratory tract infections. Vitamin D also improves anti-viral host defense through similar signaling pathways. This study assessed if EPs®7630 modifies vitamin D receptor (VDR) expression and function [...] Read more.
EPs®7630, extracted from Pelargonium sidoides, reduces the severity of viral upper respiratory tract infections. Vitamin D also improves anti-viral host defense through similar signaling pathways. This study assessed if EPs®7630 modifies vitamin D receptor (VDR) expression and function by human bronchial epithelial cells. Bronchial epithelial cells were incubated with EPs®7630 over 48 h before calcitriol stimulation and/or infection with Rhinovirus (RV)-16. Protein expression was determined by Western-blotting. Intracellular signaling of mitogen activated protein kinases (MAPK) was studied by chemical inhibitors. The anti-viral effect was assessed by immunofluorescence for RV-16 protein. EPs®7630 upregulated VDR expression through Erk1/2 MAPK and thereby increased the cell’s sensitivity to calcitriol. Compared ton untreated cells, the shift of the VDR into the nucleus at 5.3 times lower calcitriol concentration. EPs®7630 increased Erk1/2 MAPK signaling, but reduced p38 phosphorylation, and had no effect on Jun N-terminal kinase (JNK). EPs®7630 improved the anti-viral effect of vitamin D on RV-16 infection by 2.1 folds compared to vitamin D alone or to untreated cells. Furthermore, EPs®7630 improved the differentiation of epithelial cells by upregulating E-cadherin expression through Erk1/2. In conclusion, EPs®7630 increased host defense against Rhinovirus infection by upregulating the VDR and the differentiation of epithelial cells. Full article
(This article belongs to the Special Issue Natural Pharmacons: Biologically Active Plant Based Pharmaceuticals)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>High pressure liquid chromatography (HPLC) fingerprint detected by UV at 280 nm. The major peaks were assigned by analysis of high resolution mass spectrometry (HRMS) data and are given as follows: (<b>a</b>) adenosine 3′,5′-cyclic monophosphate, (<b>b</b>) guanosine 3′,5′-cyclic monophosphate, (<b>c</b>) 1-methylguanosine 3′,5′-cyclic monophosphate, and (<b>d</b>) benzopyranones. The assigned structures are given in <a href="#pharmaceuticals-14-00172-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 2
<p>EPs<sup>®</sup> 7630 modifies vitamin D receptor (VDR) expression. (<b>A</b>) Representative Western-blots and image analyses of VDR expression in primary bronchial epithelial cells (<span class="html-italic">n</span> = 4) stimulated with increasing concentrations of EPs<sup>®</sup> 7630 over 24 h. Bars represent mean ± SEM. (<b>B</b>) Kinetic of EPs<sup>®</sup> 7630 (10 μg/mL) induced VDR expression over 48 h (<span class="html-italic">n</span> = 4). (<b>C</b>) The effect of EPs<sup>®</sup> 7630 on the phosphorylation of Erk1/2 mitogen activated protein kinases (MAPK) over 120 min. (<b>D</b>) The effect of inhibitors for Jun N-terminal kinase (JNK), p38, and Erk1/2 on EPs<sup>®</sup> 7630 induced VDR expression (<span class="html-italic">n</span> = 4). Representative Western-blots are depicted above the corresponding bars. Similar results were obtained in BEAS-2B cells.</p>
Full article ">Figure 3
<p>EPs<sup>®</sup> 7630 and calcitriol activate the VDR. (<b>A</b>) Concentration dependent increase of nuclear VDR by EPs<sup>®</sup> 7630 over 24 h in primary human bronchial epithelial cells (<span class="html-italic">n</span> = 4). (<b>B</b>) Concentration dependent increase of nuclear VDR by calcitriol over 24 h (<span class="html-italic">n</span> = 4). (<b>C</b>) The effect of combined EPs<sup>®</sup> 7630 with calcitriol on the ratio of cytosolic versus nuclear VDR accumulation (<span class="html-italic">n</span> = 4) over 24 h. (<b>D</b>) The effect of MAPK inhibitors for JNK (SP600125), p38 (SB203580), and Erk1/2 (PD98059) on EPs<sup>®</sup> 7630 and calcitriol induced VDR expression (<span class="html-italic">n</span> = 3). Bars represent mean ± SEM. Similar results were obtained in BEAS-2B cells.</p>
Full article ">Figure 4
<p>The effect of EPs<sup>®</sup> 7630 and calcitriol on RV-16 infection in BEAS-2B cells. (<b>A</b>) Concentration dependent effect of EPs<sup>®</sup> 7630 on RV-16 staining in BEAS-2B cells. (<b>B</b>) Concentration dependent effect of calcitriol on RV-16 staining in BEAS-2B cells. (<b>C</b>) The effect of combined EPs<sup>®</sup> 7630 (fixed concentration) with calcitriol (increasing concentrations) on RV-16 staining on BEAS-2B cells. Bars represent mean ± SEM of triplicate experiments. (<b>D</b>) Representative immunofluorescence photographs of RV-16 positive primary epithelial cells in the presence and absence of EPs<sup>®</sup> 7630 (1 µg/mL), or calcitriol (1 µM), or the combination of both (EPs<sup>®</sup> 7630 1 µg/mL + calcitriol 1 µM).</p>
Full article ">Figure 5
<p>EPs<sup>®</sup> 7630 increases E-cadherin expression through Erk1/2 MAPK in BEAS-2B cells. (<b>A</b>) Concentration effect of EPs<sup>®</sup> 7630 on the expression of E-cadherin over 24 h in BEAS-2B cells. (<b>B</b>) The effect of inhibitors for JNK, p38, and Erk1/2 on EPs<sup>®</sup> 7630 induced E-cadherin expression. Bars represent mean ± SEM of triplicate experiments.</p>
Full article ">Figure 6
<p>Cell characterization and treatment. (<b>A</b>) Cell type characterization by phase contrast microscopy and positive immunofluorescence pan-cytokeratin, E-cadherin, and fibronectin (negative control). (<b>B</b>) Treatment scheme of cells. Confluent epithelial cells were pre-treated with either EPs<sup>®</sup> 7630 for 24 h before calcitriol was added for another 24 h and prior to infection with 1 MOI RV-16. RNA and protein were isolated over 4 days. Non-infected cells were used to calculate changes of protein expression at the corresponding time points.</p>
Full article ">
13 pages, 949 KiB  
Article
Cannabis-Based Oral Formulations for Medical Purposes: Preparation, Quality and Stability
by Francesca Baratta, Marco Simiele, Irene Pignata, Lorenzo Ravetto Enri, Antonio D’Avolio, Riccardo Torta, Anna De Luca, Massimo Collino and Paola Brusa
Pharmaceuticals 2021, 14(2), 171; https://doi.org/10.3390/ph14020171 - 22 Feb 2021
Cited by 13 | Viewed by 5969
Abstract
Current legislation in Italy provides that medical Cannabis may be administered orally or by inhalation. One of the fundamental criteria for the administration of oral formulations is that they deliver a known consistent quantity of the active ingredients to ensure uniform therapies leading [...] Read more.
Current legislation in Italy provides that medical Cannabis may be administered orally or by inhalation. One of the fundamental criteria for the administration of oral formulations is that they deliver a known consistent quantity of the active ingredients to ensure uniform therapies leading to the optimisation of the risks/benefits. In 2018, our group developed an improved Cannabis oil extraction technique. The objective of the present work was to carry out a stability study for the oil extracts obtained by this method. Furthermore, in order to facilitate the consumption of the prescribed medical Cannabis therapy by patients, a standard procedure was defined for the preparation of a single-dose preparation for oral use (hard capsules) containing the oil extract; thereafter, the quality and stability were evaluated. The hard capsules loaded with the oil extract were analysed and found to be uniform in content. The encapsulation process did not alter the quantity of the active molecule present in the oil. The stability tests yielded excellent results. Since the capsule dosage form is easily transported and administered, has pleasant organoleptic properties and is stable at room temperature for extended periods of time, this would facilitate the adherence to therapy by patients in treatment. Full article
(This article belongs to the Special Issue Clinical and Forensic Toxicology: The Latest Updates)
Show Figures

Figure 1

Figure 1
<p>Oils stability: average concentration of active constituents compared to T0. (<b>a</b>) Oils stability in fridge; (<b>b</b>) oils stability at room temperature. T0: initial conditions; T 15: 15 days; T 30: 30 days; T 60: 60 days; T 90: 90 days; T 120: 120 days; T 150: 150 days; T 180: 180 days. Standard deviation for oils has been detailed in <a href="#pharmaceuticals-14-00171-t002" class="html-table">Table 2</a>.</p>
Full article ">Figure 2
<p>Capsules stability: average concentration of active constituents compared to T0. (<b>a</b>) Stability of capsules containing 156 µL of oil in fridge and at room temperature; (<b>b</b>) Stability of capsules containing 312 µL of oil in fridge and at room temperature. T0: initial conditions; T 90: 90 days; T 180: 180 days. Standard deviation for oils has been detailed in <a href="#pharmaceuticals-14-00171-t003" class="html-table">Table 3</a>.</p>
Full article ">
15 pages, 1773 KiB  
Communication
PARP Traps Rescue the Pro-Inflammatory Response of Human Macrophages in the In Vitro Model of LPS-Induced Tolerance
by Julita Pietrzak, Karolina Gronkowska and Agnieszka Robaszkiewicz
Pharmaceuticals 2021, 14(2), 170; https://doi.org/10.3390/ph14020170 - 22 Feb 2021
Cited by 3 | Viewed by 3178
Abstract
Secondary infections cause sepsis that lead to patient disability or death. Contact of macrophages with bacterial components (such as lipopolysaccharide—LPS) activates the intracellular signaling pathway downstream of Toll-like receptors (TLR), which initiate an immune proinflammatory response. However, the expression of nuclear factor-kappa B [...] Read more.
Secondary infections cause sepsis that lead to patient disability or death. Contact of macrophages with bacterial components (such as lipopolysaccharide—LPS) activates the intracellular signaling pathway downstream of Toll-like receptors (TLR), which initiate an immune proinflammatory response. However, the expression of nuclear factor-kappa B (NF-κB)-dependent proinflammatory cytokines significantly decreases after single high or multiple LPS stimulations. Knowing that poly(ADP-ribose) polymerase-1 (PARP1) serves as a cofactor of NF-κB, we aimed to verify a hypothesis of the possible contribution of PARP1 to the development of LPS-induced tolerance in human macrophages. Using TNF-α mRNA expression as a readout, we demonstrate that PARP1 interaction with the TNF-α promoter, controls macrophage immunoparalysis. We confirm that PARP1 is extruded from the gene promoter, whereas cell pretreatment with Olaparib maintains macrophage responsiveness to another LPS treatment. Furthermore, cell pretreatment with proteasome inhibitor MG132 completely abrogates the effect of Olaparib, suggesting that PARP1 acts with NF-κB in the same regulatory pathway, which controls pro-inflammatory cytokine transcription. Mechanistically, PARP1 trapping allows for the re-rebinding of p65 to the TNF-α promoter in LPS-stimulated cells. In conclusion, PARP traps prevent PARP1 extrusion from the TNF-α promoter upon macrophage stimulation, thereby maintaining chromatin responsiveness of TLR activation, allowing for the re-binding of p65 and TNF-α transcription. Full article
(This article belongs to the Special Issue Epigenetic Drugs)
Show Figures

Figure 1

Figure 1
<p>PARP traps maintain proinflammatory response in human macrophages activated with tolerance-inducing LPS doses. (<b>a</b>) The scheme of tolerance induction presents an experimental approach and cell treatments: the first dose of LPS aims to induce cell paralysis or priming within 24 h, the second serves to check macrophage pro-inflammatory response using mRNA of TNF-α as a readout after cell stimulation for 2 h. To test a possible effect of the key enzymes involved in the ADP-ribosylation metabolism (PARP inhibitors: olaparib, veliparib, niraparib, and PARG inhibitor: ADP–HPD) on the tolerance development, the corresponding compounds were added for 1 h prior to the tolerance-inducing (first) dose of bacterial endotoxin. Expression of TNF-α was quantified by real-time PCR, normalized to median of ACTB and GAPDH, and shown as a fold change with respect to control cells (LPS untreated = 1). (<b>b</b>) Three doses of LPS were tested for macrophage paralysis and the immunomodulatory effect of PARP inhibitor—olaparib (1 µM) was estimated based on the TNF-α transcription. The red rectangular indicates the couple: olaparib-LPS that was chosen for further experiments. (<b>c</b>) The other two PARP inhibitors, which differ in PARP-DNA binding ability—niraparib (MK-4827) and veliparib (ABT-888), as well as (<b>d</b>–<b>e</b>) PARG inhibitor—ADP–HPD (10 µM) were analyzed for their possible effect on the induction of tolerance by LPS. (<b>d</b>) The increased accumulation of ADP-ribosylated proteins caused by ADP–HPD pretreatment of LPS-induced macrophages was confirmed by western blot. H3 was used as a loading control. Full-length western blot images are included in the <a href="#app1-pharmaceuticals-14-00170" class="html-app">Supplementary Figure S3</a>. (<b>e</b>) The possible modulatory role of PARP1 in the paralysis of macrophage pro-inflammatory phenotype was tested also for other cytokine genes, such as IL-1β, IL-6, IL-12, MIP2A, COX2, and iNOS using real-time PCR for the measurement of mRNA levels. Bars in the figures represent mean ± standard error of the mean (SEM). One-way analysis of variance (ANOVA1) was carried out in GraphPad Prism 5 to compare means in several groups. Once the significance was detected, ANOVA1 was followed by the Tukey post-hoc test and significant differences between the two considered means are marked with * when significant at <span class="html-italic">p</span>  &lt;  0.05, ns—non-significant at <span class="html-italic">p</span> &gt; 0.05. Abbreviations: iPARP—poly(ADP-ribose) polymerase (PARP) inhibitor(s), iPARG—poly(ADP-ribose) glycohydrolase (PARG) inhibitor, LPS—lipopolysaccharide, Olap—olaparib, Velip—veliparib, Nirap—niraparib, TNF—tumor necrosis factor, IL1β—interleukin 1 beta, IL6—interleukin 6, IL-12A—interleukin 12 subunit alpha, COX2—cyclooxygenase-2, MIP2A—macrophage inflammatory protein 2 subunit alpha, iNOS—inducible nitric oxide synthase, ACTB—actin beta, GAPDH—glyceraldehyde 3-phosphate dehydrogenase.</p>
Full article ">Figure 2
<p>The PARP1 level drives LPS-induced immune paralysis in THP1 cells. (<b>a</b>) The induction of tolerance in acute leukemia cells was evaluated based on the <span class="html-italic">TNF-α</span> expression that was quantified by real-time PCR, normalized as median of <span class="html-italic">ACTB</span> and <span class="html-italic">GAPDH</span>, and shown as a fold change with respect to untreated cells. (<b>b</b>) The illustration in outlines the human proximal <span class="html-italic">TNF-α</span> promoter and the position of NF-κB (−98) as well as primer (forward: −124, reverse: −73) binding sites with respect to transcriptional start site (TSS). The fragment between the two indicated primers was amplified to quantify the immunoprecipitated DNA by real-time PCR. (<b>c</b>) The impact of the single LPS dose and olaparib on PARP1 occurrence at the <span class="html-italic">TNF-α</span> promoter was estimated with ChIP-qPCR. (<b>d</b>–<b>e</b>) The silencing efficiency of PARP1 in THP1 cells stably transfected with PARP-1 shRNA Plasmid was determined by real-time PCR and western blot, respectively versus corresponding control cells (shCTRL; control shRNA Plasmid-A). <span class="html-italic">PARP1</span> mRNA was normalized to the median of <span class="html-italic">ACTB</span> and <span class="html-italic">GAPDH</span>, and shown as a fold change with respect to control cells with the normal PARP1 expression. In western blot, H3 was used as a loading control. Full-length western blot images are available in the <a href="#app1-pharmaceuticals-14-00170" class="html-app">Supplementary Figure S4</a>. (<b>f</b>) The mRNA level of <span class="html-italic">TNF-α</span>, <span class="html-italic">MIP2a</span>, and <span class="html-italic">COX2</span>, which served to assess the development of immune tolerance in response to LPS, was measured with real-time PCR as described above. (<b>g</b>–<b>h</b>) PARP1 overexpression in pCMV3–PARP1 versus pCMV3–EMPTY-transfected cells was confirmed by real-time PCR and western blot, respectively. Full-length western blot images are included in the <a href="#app1-pharmaceuticals-14-00170" class="html-app">Supplementary Figure S5</a>. mRNA level of PARP1 was normalized to <span class="html-italic">ACTB</span> and <span class="html-italic">GAPDH</span> and is presented as a fold change of PARP1 knock-in with respect to control cells. (<b>i</b>) The effect of PARP poison (olaparib) on the development of endotoxin tolerance in pCMV3–EMPTY and pCMV3–PARP1 cells was estimated on the basis of <span class="html-italic">TNF-α</span> transcription (mRNA), which was set as a readout. Bars in the figures represent mean ± standard error of the mean (SEM). One-way analysis of variance (ANOVA1) was carried out in GraphPad Prism 5 to compare means in several groups. Once the significance was detected, ANOVA1 was followed by the Tukey post-hoc test and significant differences between the two considered means are marked with * when significant at <span class="html-italic">p</span>  <span class="html-italic">&lt;</span>  0.05, ** when significant at <span class="html-italic">p</span>  <span class="html-italic">&lt;</span>  0.01, *** when significant at <span class="html-italic">p</span>  <span class="html-italic">&lt;</span>  0.001, ns—non-significant at <span class="html-italic">p</span> &gt; 0.05. Abbreviations: THP1—human monocytic cell line, LPS—lipopolysaccharide, Olap—Olaparib, TNF—tumor necrosis factor, ACTB—actin beta, GAPDH—glyceraldehyde 3-phosphate dehydrogenase, shCTRL—THP1 cell transfected with Control shRNA Plasmid-A, shPARP1—THP1 cell transfected with PARP-1 shRNA Plasmid, pCMV3–PARP1—THP1 cells transfected with pCMV3–-PARP1 plasmid, pCMV3–EMPTY—THP1 cells transfected with pCMV3-EMPTY plasmid, IgG—immunoglobulin G.</p>
Full article ">Figure 3
<p>PARP1 eviction from the <span class="html-italic">TNF-α</span> promoter prevents p65 re-binding after LPS re-stimulation. (<b>A</b>) The contribution of canonical NF-κB to macrophage response to LPS was estimated by comparing <span class="html-italic">TNF-α</span> transcription in control and cells pre-treated with proteasome inhibitor (MG132; 1 µM). mRNA level of the cytokine was quantified by real-time PCR, normalized to median of <span class="html-italic">ACTB</span> and <span class="html-italic">GAPDH</span> and presented as a fold change with respect to untreated cells. (<b>B</b>) p65 and (<b>C</b>) p50 occurrence at the <span class="html-italic">TNF-α</span> promoter was analyzed by ChIP-real-time PCR. Bars in the figures represent mean ± standard error of the mean (SEM). One-way analysis of variance (ANOVA1) was carried out in GraphPad Prism 5 to compare means in several groups. Once the significance was detected, ANOVA1 was followed by the Tukey post-hoc test and significant differences between the two considered means are marked with * when significant at <span class="html-italic">p</span>  &lt;  0.05, ** when significant at <span class="html-italic">p</span>  <span class="html-italic">&lt;</span>  0.01, ns—non-significant at <span class="html-italic">p</span> &gt; 0.05. Abbreviations: LPS—lipopolysaccharide, Olap—olaparib, TNF—tumor necrosis factor, ACTB—actin beta, GAPDH—glyceraldehyde 3-phosphate dehydrogenase, IgG—immunoglobulin G, p65—transcription factor p65 (RelA), p50—nuclear factor NF-kappa-B p105 subunit.</p>
Full article ">
54 pages, 14187 KiB  
Article
Synthesis and Biological Evaluation of 1-(Diarylmethyl)-1H-1,2,4-triazoles and 1-(Diarylmethyl)-1H-imidazoles as a Novel Class of Anti-Mitotic Agent for Activity in Breast Cancer
by Gloria Ana, Patrick M. Kelly, Azizah M. Malebari, Sara Noorani, Seema M. Nathwani, Brendan Twamley, Darren Fayne, Niamh M. O’Boyle, Daniela M. Zisterer, Elisangela Flavia Pimentel, Denise Coutinho Endringer and Mary J. Meegan
Pharmaceuticals 2021, 14(2), 169; https://doi.org/10.3390/ph14020169 - 22 Feb 2021
Cited by 6 | Viewed by 5240
Abstract
We report the synthesis and biochemical evaluation of compounds that are designed as hybrids of the microtubule targeting benzophenone phenstatin and the aromatase inhibitor letrozole. A preliminary screening in estrogen receptor (ER)-positive MCF-7 breast cancer cells identified 5-((2H-1,2,3-triazol-1-yl)(3,4,5-trimethoxyphenyl)methyl)-2-methoxyphenol 24 as a [...] Read more.
We report the synthesis and biochemical evaluation of compounds that are designed as hybrids of the microtubule targeting benzophenone phenstatin and the aromatase inhibitor letrozole. A preliminary screening in estrogen receptor (ER)-positive MCF-7 breast cancer cells identified 5-((2H-1,2,3-triazol-1-yl)(3,4,5-trimethoxyphenyl)methyl)-2-methoxyphenol 24 as a potent antiproliferative compound with an IC50 value of 52 nM in MCF-7 breast cancer cells (ER+/PR+) and 74 nM in triple-negative MDA-MB-231 breast cancer cells. The compounds demonstrated significant G2/M phase cell cycle arrest and induction of apoptosis in the MCF-7 cell line, inhibited tubulin polymerisation, and were selective for cancer cells when evaluated in non-tumorigenic MCF-10A breast cells. The immunofluorescence staining of MCF-7 cells confirmed that the compounds targeted tubulin and induced multinucleation, which is a recognised sign of mitotic catastrophe. Computational docking studies of compounds 19e, 21l, and 24 in the colchicine binding site of tubulin indicated potential binding conformations for the compounds. Compounds 19e and 21l were also shown to selectively inhibit aromatase. These compounds are promising candidates for development as antiproliferative, aromatase inhibitory, and microtubule-disrupting agents for breast cancer. Full article
(This article belongs to the Special Issue Anticancer Drugs 2021)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Aromatase inhibitors (letrozole, anastrozole, Exemestane), SERMs (Tamoxifen, Endoxifen, Norendoxifen, Raloxifene), kinase inhibitors Lapatinib, Alpelisib, Tucatinib, steroid sulfatase inhibitor Irosustat and mutant p53 inhibitor PRIMA-1, (<b>b</b>) Combretastatins <b>4a–f</b>, Isocombretastatins <b>5</b>,<b>6</b>, phenstatins <b>7a–e,</b> colchicine binding site inhibitors <b>8a</b>,<b>b</b>, <b>9a–c</b> and target structures <b>10</b>.</p>
Full article ">Figure 1 Cont.
<p>(<b>a</b>) Aromatase inhibitors (letrozole, anastrozole, Exemestane), SERMs (Tamoxifen, Endoxifen, Norendoxifen, Raloxifene), kinase inhibitors Lapatinib, Alpelisib, Tucatinib, steroid sulfatase inhibitor Irosustat and mutant p53 inhibitor PRIMA-1, (<b>b</b>) Combretastatins <b>4a–f</b>, Isocombretastatins <b>5</b>,<b>6</b>, phenstatins <b>7a–e,</b> colchicine binding site inhibitors <b>8a</b>,<b>b</b>, <b>9a–c</b> and target structures <b>10</b>.</p>
Full article ">Figure 2
<p>X-ray crystallography structures of <b>16e</b>, <b>16f</b>, and <b>19c</b> with heteroatoms labelled and displacement shown at 50% probability.</p>
Full article ">Figure 2 Cont.
<p>X-ray crystallography structures of <b>16e</b>, <b>16f</b>, and <b>19c</b> with heteroatoms labelled and displacement shown at 50% probability.</p>
Full article ">Figure 3
<p>X-ray crystallography of compounds <b>21i</b> and <b>26a</b> with heteroatoms labelled and displacement shown at 50% probability. <sup>a</sup> Disordered molecular structure of <b>26a</b> showing the major occupied moiety F1a. <sup>b</sup> Disordered molecular structure of <b>26a</b> showing the minor occupied moiety F1B.</p>
Full article ">Figure 4
<p>Preliminary cell viability data for (<b>A</b>) Series 1: 1-(diarylmethyl)-1<span class="html-italic">H</span>-1,2,4-triazoles <b>13b–g</b>, <b>l–o</b> (<b>B</b>) Series 2: 1-(diarylmethyl)-1<span class="html-italic">H</span>-1,2,4-triazoles <b>16a</b>, <b>c–i</b>, <b>19b–e</b>, 1-(diarylmethyl)-2<span class="html-italic">H</span>-1,2,3-triazole <b>24</b> in MCF-7 breast cancer cells. Cell proliferation of MCF-7 cells was determined with an alamarBlue assay (seeding density 2.5 × 104 cells/mL per well for 96-well plates). Compound concentrations of either 1 or 0.1 μM for 72 h were used to treat the cells (in triplicate) with control wells containing vehicle ethanol (1% <span class="html-italic">v</span>/<span class="html-italic">v</span>). The mean value + SEM for three independent experiments is shown. The positive controls used are CA-4 and phenstatin.</p>
Full article ">Figure 5
<p>Preliminary cell viability data for (<b>A</b>) <b>Series 3:</b> 1-(diarylmethyl)-1<span class="html-italic">H</span>-imidazoles <b>20b–h, k, l</b> and (<b>B</b>) <b>Series 4:</b> 1-(diarylmethyl)-1<span class="html-italic">H</span>-imidazoles <b>21a–g, i–l</b> in MCF-7 breast cancer cells. Cell proliferation of MCF-7 cells was determined with an alamarBlue assay (seeding density 2.5 × 10<sup>4</sup> cells/mL per well for 96-well plates). Compound concentrations of either 1 or 0.1 μM for 72 h were used to treat the cells (in triplicate) with control wells containing vehicle ethanol (1% <span class="html-italic">v</span>/<span class="html-italic">v</span>). The mean value ± SEM for three independent experiments is shown. The positive controls used are CA-4 and phenstatin.</p>
Full article ">Figure 6
<p>Preliminary cell viability data for (<b>A</b>) Series 5: 1-(diarylmethyl)pyrrolidines and 1-(diarylmethyl)piperidines <b>25a–g, 26a–c</b> and (<b>B</b>) Series 5: 1-(diarylmethyl)piperazines <b>27c,d,f,i</b>, and <b>h</b> in MCF-7 breast cancer cells. Cell proliferation of MCF-7 cells was determined with an alamarBlue assay (seeding density 2.5 × 10<sup>4</sup> cells/mL per well for 96-well plates). Compound concentrations of either 10, 1, or 0.1 μM for 72 h were used to treat the cells (in triplicate) with control wells containing vehicle ethanol (1% <span class="html-italic">v</span>/<span class="html-italic">v</span>). The mean value ± SEM for three independent experiments is shown. The positive controls used are CA-4 and phenstatin.</p>
Full article ">Figure 7
<p>Screening of phenstatin derivatives <b>19e</b> (<b>A</b>) and <b>21l</b> (<b>B</b>) in MCF-10A cells at 24, 48, 72 h and (<b>C</b>) <b>24</b> in MCF-10A and MCF-7 cells at 72 h. (<b>A</b>,<b>B</b>) Effect of compounds <b>19e</b> and <b>21l</b> on the viability of non-tumorigenic MCF-10A human mammary epithelial cells. Cells were treated with the indicated concentrations for 21, 48, or 72 h. Cell viability was expressed as a percentage of vehicle control (ethanol 1% (<span class="html-italic">v</span>/<span class="html-italic">v</span>)) and was determined by alamarBlue assay (average + SEM of three independent experiments). (<b>C</b>) Effect of compound <b>24</b> on the viability of non-tumorigenic MCF-10A human mammary epithelial cells and MCF-7 breast cancer cells. Cells were treated with the indicated concentrations for 72 h. Cell viability was expressed as a percentage of vehicle control (ethanol 1% (<span class="html-italic">v</span>/<span class="html-italic">v</span>)) and was determined by alamarBlue assay (average ± SEM of three independent experiments). Statistical analysis was performed using two-way ANOVA (***, <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 8
<p>Compound (<b>A</b>) <b>19e</b>, (<b>B</b>) <b>24</b>, and (<b>C</b>) phenstatin (<b>7a</b>) in MCF-7 induced G2/M arrest followed by apoptosis in a time-dependent manner in MCF-7 cells. Cells were treated with either vehicle control (v) (0.1% ethanol (<span class="html-italic">v</span>/<span class="html-italic">v</span>)] or compound <b>19e</b>, <b>24</b>, or phenstatin (<b>7a</b>) (1 μM) for 24, 48, and 72 h). Then, cells were fixed, stained with PI, and analysed by flow cytometry. Cell cycle analysis was performed on histograms of gated counts per DNA area (FL2-A). The number of cells with &lt;2 N (sub-G1), 2 N (G0G1), and 4 N (G2/M) DNA content was determined with CellQuest software. Values represent as the mean ± SEM for three separate experiments. Statistical analysis was performed using two-way ANOVA (**, <span class="html-italic">p</span> &lt; 0.01, ***, <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 9
<p>Compound <b>21l</b> induced cell apoptosis in (<b>A</b>) MCF-7 breast cancer cells and (<b>B</b>) MDA-MB-231 breast cancer cells. MCF-7 breast cancer cells and MDA-MB-231 breast cancer cells were treated with <b>21l</b> or phenstatin (<b>7a</b>) (1 µM) or control vehicle (0.1% ethanol (<span class="html-italic">v</span>/<span class="html-italic">v</span>)), and the percentage of apoptotic cells was determined by staining with Annexin V-FITC and PI. In each panel, the lower left quadrant shows cells that are negative for both PI and Annexin V-FITC, the upper left shows only PI cells that are necrotic. The lower right quadrant shows Annexin-positive cells that are in the early apoptotic stage and the upper right shows both Annexin/PI positive, which are in late apoptosis/necrosis. Control cells and cells treated with phenstatin <b>7a</b> and <b>21l</b> at 48 h are shown, respectively. Values represent the mean of three independent experiments.</p>
Full article ">Figure 10
<p>Effect of compound <b>21l</b> on tubulin polymerisation in vitro. (<b>A</b>) Tubulin polymerisation assay for compound <b>21l</b> at 10 μM. (<b>B</b>) Paclitaxel (10 μM) and phenstatin (<b>7a</b>) (10 μM) were used as references while ethanol (1% <span class="html-italic">v</span>/<span class="html-italic">v</span>) and DMSO (1% <span class="html-italic">v</span>/<span class="html-italic">v</span>) were used as vehicle controls. Purified bovine tubulin and guanosine-5’-triphosphate (GTP) were mixed in a 96-well plate. The polymerisation reaction was initiated by warming the solution from 4 to 37 °C. The effect on tubulin assembly was monitored in a Spectramax 340PC spectrophotometer at 340 nm at 30 s intervals for 60 min at 37 °C. DMSO. Fold inhibition of tubulin polymerisation was calculated using the V<sub>max</sub> value for each reaction. The results represent the mean for three separate experiments.</p>
Full article ">Figure 11
<p>Compounds <b>19e</b> and <b>21l</b> depolymerise the microtubule network of MCF-7 breast cancer cells. Cells were treated with (<b>A</b>) vehicle control (0.1% ethanol (<span class="html-italic">v</span>/<span class="html-italic">v</span>)), (<b>B</b>) paclitaxel (1 μM), (<b>C</b>) phenstatin (<b>7a</b>) (1 μM), (<b>D</b>) compound <b>19e</b> (10 μM), or (<b>E</b>) compound <b>21l</b> (10 μM) for 16 h. Cells were fixed in ice-cold methanol and stained with mouse monoclonal anti-α-tubulin–fluorescein isothiocyante (FITC) antibody (clone DM1A) (green), Alexa Fluor 488 dye, and counterstained with DAPI 4’-6’-diamidino-2-phenylindole (blue). Images obtained with Leica SP8 confocal microscopy, Leica application suite X software. Representative confocal micrographs of three separate experiments are shown. Scale bar indicates 25 μm.</p>
Full article ">Figure 12
<p>Effects of <b>21l</b> and <b>24</b> on expression of PARP (poly ADP-ribose polymerase) and anti-apoptotic proteins Bcl-2 and Mcl-1. MCF-7 cells were treated with either vehicle control (ethanol, 0.1% <span class="html-italic">v</span>/<span class="html-italic">v</span>) or with compounds <b>21l</b> and <b>24</b> (1 μM) for 24 h. After the required time, cells were harvested and separated by SDS-PAGE to detect the level of the apoptosis-related proteins. The membrane was probed with anti-PARP or anti-cleaved PARP antibodies. Results are representative of three separate experiments. To confirm equal protein loading, each membrane was stripped and re-probed with glyceraldehyde 3-phosphate dehydrogenase (GAPDH) antibody.</p>
Full article ">Figure 13
<p>Docking of compounds <b>19e</b>, <b>21l</b>, and <b>24</b> in the colchicine binding site of tubulin. Overlay of the X-ray structure of tubulin co-crystallised with DAMA-colchicine (PDB entry 1SA0) on the best-ranked docked poses of the three <span class="html-italic">S</span> enantiomers (<b>A</b>) <b>21l,</b> (<b>B</b>) <b>24</b>, and (<b>C</b>) <b>19e</b>. Overlay of the X-ray structure of tubulin co-crystallised with DAMA-colchicine (PDB entry 1SA0) on the best ranked docked poses of the three <span class="html-italic">R</span> enantiomers (<b>D</b>) <b>21l</b>, (<b>E</b>) <b>24</b>, (<b>F</b>) <b>19e</b>, and (<b>G</b>) Phenstatin (<b>7a</b>). Ligands are rendered as tube and amino acids as a line. Tubulin amino acids and DAMA-colchicine are coloured by atom type; the three heterocycles are coloured green. The atoms are coloured by element type, carbon = grey, hydrogen = white, oxygen = red, nitrogen = blue, sulphur = yellow. Key amino acid residues are labelled, and multiple residues are hidden to enable a clearer view.</p>
Full article ">Scheme 1
<p>Synthesis of compounds <b>13a–13o</b> (Series 1). Reagents and conditions: (<b>a</b>) NaBH<sub>4,</sub> MeOH, 0 °C, (38–100%); (b) 1,2,4-triazole, <span class="html-italic">p</span>-TSA, toluene, 4 h, 120 <sup>o</sup>C, microwave open vessel, (3–98%); (<b>c</b>) K<sub>2</sub>CO<sub>3</sub>, MeOH, H<sub>2</sub>O, 20 °C, 72 h, (16%); (<b>d</b>) H<sub>2</sub>, Pd(OH)<sub>2</sub>, ethyl acetate, 20 °C, (67%); (<b>e</b>) TBAF, THF, 0 °C, (90%) [TBDMS, <span class="html-italic">tert</span>-butyldimethylsilyl; Bn, CH<sub>2</sub>C<sub>6</sub>H<sub>5</sub>].</p>
Full article ">Scheme 2
<p>Synthesis of letrozole-phenstatin hybrid compounds <b>16a–16i</b> (Series 2a). Reagents and conditions: (<b>a</b>) <span class="html-italic">n</span>-BuLi, THF, −78 °C, 1.5 h, (21–89%); (<b>b</b>) 1,2,4-triazole, <span class="html-italic">p</span>-TSA, toluene, 4 h, 120 °C, microwave open vessel, (34–93%); (<b>c</b>) Pd(OH)<sub>2</sub>, H<sub>2</sub>, ethyl acetate, 20 °C, (49%). [Bn, CH<sub>2</sub>C<sub>6</sub>H<sub>5</sub>].</p>
Full article ">Scheme 3
<p>Synthesis of letrozole-phenstatin hybrid compounds <b>19a-19e</b> (Series 2a) and 19f–19i (Series 2b). Reagents and conditions: (<b>a</b>): <span class="html-italic">n</span>-BuLi, dry THF, −78 °C,1.5 h, (16–88%); (<b>b</b>) 1,2,4-triazole, <span class="html-italic">p</span>-TSA, microwave open vessel, 4h, (64–95%); (<b>c</b>) Pd(OH)<sub>2</sub>, H<sub>2</sub>, (66–82%). [Bn: CH<sub>2</sub>C<sub>6</sub>H<sub>5</sub>].</p>
Full article ">Scheme 4
<p>Synthesis of hybrid imidazole-phenstatin compounds <b>20a–20l</b> (Series 3). Reagents and conditions: (<b>a</b>) CDI, CH<sub>3</sub>CN, reflux, 3 h, (10–64%); (<b>b</b>) K<sub>2</sub>CO<sub>3</sub>, MeOH, H<sub>2</sub>O, 20 °C, 72, (50%). [Bn: CH<sub>2</sub>C<sub>6</sub>H<sub>5</sub>].</p>
Full article ">Scheme 5
<p>Synthesis of hybrid imidazole-phenstatin compounds <b>21a–21l</b>, (Series 4). Reagents and conditions: (<b>a</b>) CDI, CH<sub>3</sub>CN, reflux, 3 h, (30–100%). (<b>b</b>) H<sub>2</sub>, Pd(OH)<sub>2</sub>, ethyl acetate, 20 °C, (93%). [Bn: CH<sub>2</sub>C<sub>6</sub>H<sub>5</sub>].</p>
Full article ">Scheme 6
<p>Synthesis of phenstatin 7a and phenstatin hybrids <b>21b</b>, <b>21c</b>, <b>24</b>, (Series 4). Reagents and conditions: (<b>a</b>) Eaton’s reagent (P<sub>2</sub>O<sub>5</sub>, CH<sub>3</sub>SO<sub>3</sub>H), 60 °C, 3 h, [<b>23a</b> (60%), <b>23b</b> (57%), <b>23c</b> (17%]); (<b>b</b>) sodium acetate, methanol, refux, 2 h, (89%); (<b>c</b>) NaBH<sub>4,</sub> MeOH, 0 <sup>o</sup>C, [<b>15c</b> (50%), <b>15d</b> (89%),<b>15i</b> (96%)]; (<b>d</b>) 1,2,3-triazole, <span class="html-italic">p</span>-TSA, toluene, 4 h, 120 °C, microwave open vessel, (77%); (<b>e</b>) CDI, CH<sub>3</sub>CN, reflux, 3 h, [<b>21b</b> (39%), <b>21c</b> (67%)].</p>
Full article ">Scheme 7
<p>Synthesis of pyrrolidine <b>25a–g</b> and piperidine derivatives <b>26a–c</b>, (Series 5). Reagents and conditions: (<b>a</b>) SOCl<sub>2</sub>,CH<sub>2</sub>Cl<sub>2</sub>, 12 h, 20 <sup>o</sup>C; (<b>b</b>) pyrrolidine, acetonitrile,12 h, reflux (23–93%); (<b>c</b>) piperidine, acetonitrile,12 h, reflux (84–91%).</p>
Full article ">Scheme 8
<p>Synthesis of piperazine-phenstatin compounds <b>27a–g</b>, 28, (Series 5). Reagents and conditions: (<b>a</b>) SOCl<sub>2</sub>, CH<sub>2</sub>Cl<sub>2</sub>,12 h, 20 <sup>o</sup>C; (<b>b</b>) N-phenylpiperazine, N-benzylpiperazine, N-Boc-piperazine or <span class="html-italic">p</span>-methoxyphenylpiperazine, acetonitrile, reflux, 12 h (6–80%) (<b>c</b>) TFA, CH<sub>2</sub>Cl<sub>2</sub>, 30 min, 20 <sup>o</sup>C (42%); (<b>d</b>) H<sub>2</sub>, Pd(OH)<sub>2</sub> (45%); (<b>e</b>): piperazine, ACN, acetonitrile, reflux, 12 h (12%). [Boc:<span class="html-italic">tert</span>-Butoxycarbony; Bn: CH<sub>2</sub>C<sub>6</sub>H<sub>5</sub>].</p>
Full article ">
12 pages, 766 KiB  
Review
Non-Coding RNAs: The “Dark Side Matter” of the CLL Universe
by Marcello Francesco Lingua, Giovanna Carrà, Beatrice Maffeo and Alessandro Morotti
Pharmaceuticals 2021, 14(2), 168; https://doi.org/10.3390/ph14020168 - 21 Feb 2021
Cited by 5 | Viewed by 2246
Abstract
For many years in the field of onco-hematology much attention has been given to mutations in protein-coding genes or to genetic alterations, including large chromosomal losses or rearrangements. Despite this, biological and clinical needs in this sector remain unmet. Therefore, it is not [...] Read more.
For many years in the field of onco-hematology much attention has been given to mutations in protein-coding genes or to genetic alterations, including large chromosomal losses or rearrangements. Despite this, biological and clinical needs in this sector remain unmet. Therefore, it is not surprising that recent studies have shifted from coded to non-coded matter. The discovery of non-coding RNAs (ncRNAs) has influenced several aspects related to the treatment of cancer. In particular, in chronic lymphocytic leukemia (CLL) the knowledge of ncRNAs and their contextualization have led to the identification of new biomarkers used to follow the course of the disease, to the anticipation of mechanisms that support resistance and relapse, and to the selection of novel targeted treatment regimens. In this review, we will summarize the main ncRNAs discovered in CLL and the molecular mechanisms by which they are affected and how they influence the development and the progression of the disease. Full article
(This article belongs to the Special Issue Non-coding RNA in Hematological Cancers)
Show Figures

Figure 1

Figure 1
<p>Non-coding RNAs represent the “dark side matter” in CLL pathogenesis. Indeed, although often underestimated they balance the levels of oncogene and onco-suppressor proteins responsible of CLL development and progression. In <a href="#pharmaceuticals-14-00168-f001" class="html-fig">Figure 1</a>, the CLL cell is symbolized as a “black death” in which ncRNAs (miRNAs, lncRNAs, circRNAs, and tsRNAs) represent the “dark side matter” as opposed to the coding part (“non-dark matter”), both involved in the deregulated pathways responsible for CLL development and progression. The red swords represent a silencing mechanism, whereas green swords represent an inducing mechanism. Arrows indicate activation, whereas bar-headed lines indicate inhibition.</p>
Full article ">
40 pages, 8763 KiB  
Review
Advances in Development of Radiometal Labeled Amino Acid-Based Compounds for Cancer Imaging and Diagnostics
by Mária Bodnár Mikulová and Peter Mikuš
Pharmaceuticals 2021, 14(2), 167; https://doi.org/10.3390/ph14020167 - 21 Feb 2021
Cited by 15 | Viewed by 3433
Abstract
Radiolabeled biomolecules targeted at tumor-specific enzymes, receptors, and transporters in cancer cells represent an intensively investigated and promising class of molecular tools for the cancer diagnosis and therapy. High specificity of such biomolecules is a prerequisite for the treatment with a lower burden [...] Read more.
Radiolabeled biomolecules targeted at tumor-specific enzymes, receptors, and transporters in cancer cells represent an intensively investigated and promising class of molecular tools for the cancer diagnosis and therapy. High specificity of such biomolecules is a prerequisite for the treatment with a lower burden to normal cells and for the effective and targeted imaging and diagnosis. Undoubtedly, early detection is a key factor in efficient dealing with many severe tumor types. This review provides an overview and critical evaluation of novel approaches in the designing of target-specific probes labeled with metal radionuclides for the diagnosis of most common death-causing cancers, published mainly within the last three years. Advances are discussed such traditional peptide radiolabeling approaches, and click and nanoparticle chemistry. The progress of radiolabeled peptide based ligands as potential radiopharmaceuticals is illustrated via novel structure and application studies, showing how the molecular modifications reflect their binding selectivity to significant onco-receptors, toxicity, and, by that, practical utilization. The most impressive outputs in categories of newly developed structures, as well as imaging and diagnosis approaches, and the most intensively studied oncological diseases in this context, are emphasized in order to show future perspectives of radiometal labeled amino acid-based compounds in nuclear medicine. Full article
(This article belongs to the Special Issue Metal-Based Drugs: Updates and Perspectives)
Show Figures

Figure 1

Figure 1
<p>Basic scheme of a potential target-specific radiopharmaceutical.</p>
Full article ">Figure 2
<p>Chemical structures of the most common acyclic chelators as a base of BFCA.</p>
Full article ">Figure 3
<p>Chemical structures of the most common cyclic chelators as a base of BFCA.</p>
Full article ">Figure 4
<p>Scheme of solid phase peptide synthesis (SPPS). PG<sub>1</sub> = temporary protecting group; PG<sub>2</sub> = semi-permanent protecting group; X = N/O.</p>
Full article ">Figure 5
<p>Scheme of the pre-labeling procedure with technetium-99m (adapted according to [<a href="#B3-pharmaceuticals-14-00167" class="html-bibr">3</a>]).</p>
Full article ">Figure 6
<p>Scheme of the post-labeling procedure with technetium-99m (adapted according to [<a href="#B3-pharmaceuticals-14-00167" class="html-bibr">3</a>]).</p>
Full article ">Figure 7
<p>Selected click reactions for the preparation of intermediates used for metal chelating (R<sub>1</sub>, R<sub>2</sub>—proper chelating and peptide moieties). (<b>A</b>) CuAAC, (<b>B</b>) IEDDA, (<b>C</b>) SPAAC, (<b>D</b>) Staudinger ligation.</p>
Full article ">Figure 8
<p>Examples of attractive clickable chelators for radiolabeling of biomolecules with metal radionuclides [<a href="#B106-pharmaceuticals-14-00167" class="html-bibr">106</a>,<a href="#B108-pharmaceuticals-14-00167" class="html-bibr">108</a>,<a href="#B111-pharmaceuticals-14-00167" class="html-bibr">111</a>,<a href="#B112-pharmaceuticals-14-00167" class="html-bibr">112</a>,<a href="#B113-pharmaceuticals-14-00167" class="html-bibr">113</a>].</p>
Full article ">Figure 9
<p>Representative image of PET-SERRS nanoparticles with non-optimized <sup>68</sup>Ga-labeling (<b>A</b>) with visible degradation of silica shells and after the optimization (<b>B</b>) with improved stability of the silica shells [<a href="#B118-pharmaceuticals-14-00167" class="html-bibr">118</a>].</p>
Full article ">Figure 10
<p>Illustrative scheme of [<sup>99m</sup>Tc]Tc-EDDA/HYNIC-GGC conjugated to RGD derivative and gold NP [<a href="#B119-pharmaceuticals-14-00167" class="html-bibr">119</a>].</p>
Full article ">Figure 11
<p>PET/CT images of a patient with ileal neuroendocrine tumors showing bilobar liver metastases (marked with arrows) after application of [<sup>68</sup>Ga]Ga-OPS202 (<b>A</b>) and its transaxial fusion image (<b>B</b>) and [<sup>68</sup>Ga]Ga-DOTATOC (<b>C</b>) and its transaxial fusion image (<b>D</b>) (adapted from [<a href="#B60-pharmaceuticals-14-00167" class="html-bibr">60</a>]).</p>
Full article ">Figure 12
<p>The AA sequence of two biologically active forms of somatostatin.</p>
Full article ">Figure 13
<p>The AA sequence of bombesin peptide.</p>
Full article ">Figure 14
<p>The AA sequence of octapeptide cholecystokinin.</p>
Full article ">Figure 15
<p>The AA sequence of GLP-1.</p>
Full article ">Figure 16
<p>Structure of a peptide containing RGD sequence.</p>
Full article ">Figure 17
<p>AA sequences of other important peptides for tumor imaging. (<b>A</b>) neurotensin; (<b>B</b>) α-MSH; (<b>C</b>) substance P; (<b>D</b>) VIP.</p>
Full article ">Figure 18
<p>Structural motifs of small peptide inhibitors of proteins. (<b>A</b>) EuK motif as a base for PSMA inhibitors, (<b>B</b>) 2-cyanopyrrolidin-quinoline carboxamides as a base for FAP inhibitors.</p>
Full article ">Figure 19
<p>Position of sulfonamide-derived hCA IX inhibitor docked into the active site of hCA IX (<b>A</b>) and its intermolecular interaction diagram (<b>B</b>) [<a href="#B239-pharmaceuticals-14-00167" class="html-bibr">239</a>].</p>
Full article ">
17 pages, 3684 KiB  
Article
Losartan Improves Memory, Neurogenesis and Cell Motility in Transgenic Alzheimer’s Mice
by Henning Johannes Drews, Roman Klein, Ali Lourhmati, Marine Buadze, Elke Schaeffeler, Thomas Lang, Torgom Seferyan, Leah R. Hanson, William H. Frey II, Tom C.G.M. de Vries, Inge A.E.W. Thijssen-van Loosdregt, Christoph H. Gleiter, Matthias Schwab and Lusine Danielyan
Pharmaceuticals 2021, 14(2), 166; https://doi.org/10.3390/ph14020166 - 20 Feb 2021
Cited by 21 | Viewed by 4193
Abstract
Angiotensin receptor blockers (ARBs) have demonstrated multiple neuroprotective benefits in Alzheimer’s disease (AD) models. However, their beneficial effects on memory deficits, cholinergic activity, neurogenesis and Amyloid beta (Aβ) clearance reveal significant interstudy variability. The delivery route can impact not only delivery but also [...] Read more.
Angiotensin receptor blockers (ARBs) have demonstrated multiple neuroprotective benefits in Alzheimer’s disease (AD) models. However, their beneficial effects on memory deficits, cholinergic activity, neurogenesis and Amyloid beta (Aβ) clearance reveal significant interstudy variability. The delivery route can impact not only delivery but also targeting and therapeutic efficacy of ARBs. Our previous findings on the beneficial effects of intranasally delivered losartan in the APP/PS1 model of AD prompted us to explore the influence of the delivery route by employing here the systemic administration of losartan. Consistent with our previous results with intranasal losartan, repeated intraperitoneal administration (10 mg/kg) resulted in a remarkable decrease in Aβ plaques and soluble Aβ42, as well as inflammatory cytokines (IL-2, IL-6 and TNFα). The Aβ reduction can be ascribed to its facilitated degradation by neprilysin and diminished generation by BACE1. Losartan increased neurogenesis in vivo and in vitro and improved migratory properties of astrocytes isolated from adult transgenic AD mice. In summary, this data together with our previous results suggest therapeutic features of losartan which are independent of delivery route. The improvement of cell motility of Aβ-affected astrocytes by losartan deserves further in vivo investigation, which may lead to new strategies for AD treatment. Full article
(This article belongs to the Special Issue New Drugs and Biologics For Treatment of Central Nervous Dysfunction)
Show Figures

Figure 1

Figure 1
<p>Blood pressure (BP) and cognitive performance of losartan and vehicle treated APP/PS1 mice. Seven-month-old APP/PS1 mice (<span class="html-italic">n</span> = 9 in losartan and <span class="html-italic">n</span> = 8 in control group) were assessed for blood pressure and spatial working memory in losartan and vehicle treated (Ctrl.) groups. (<b>a</b>,<b>b</b>) systolic and diastolic blood pressure measured prior (baseline) and during drug (vehicle treatment (test blocks 1–3); (<b>c</b>) Latency to reach the goal arm and (<b>d</b>) number of incorrect choices in forced choice alternation T-maze assessed at the last 5 days of losartan or vehicle treatment. (<b>c</b>,<b>d</b>) Two-way ANOVA with Holm Sidak’s multiple comparisons test <span class="html-italic">p</span> &lt; 0.05 (*), <span class="html-italic">p</span> &lt; 0.01 (**) comparison between losartan and Ctrl. group; (##) comparison between day 1 and day 5 within the losartan-treated group.</p>
Full article ">Figure 2
<p>Cerebral content of inflammatory cytokines and soluble beta Amyloid peptide 42 (Aβ42) in losartan and vehicle treated APP/PS1 mice. Brain homogenates of APP/PS1 mice (<span class="html-italic">n</span> = 5) were assessed for the levels of inflammatory cytokines. IL-2 (<b>a</b>), IL-6 (<b>b</b>), and TNFα (<b>c</b>) in losartan and vehicle treated (Ctrl.) groups. (<b>d</b>) Brain content of soluble Aβ42 in losartan and vehicle treated APP/PS1 mice. <span class="html-italic">t</span>-test, <span class="html-italic">** p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.0005, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Losartan-mediated changes in neurogenesis and neuroprotection markers in APP/PS1 mice. (<b>a</b>,<b>b</b>) Expression of nestin (green) in the hilus and granular cell layer of the denate gyrus in brain sections of losartan vs. vehicle (Ctrl.) treated APP/PS1 mice. Cell nuclei are stained with 4´,6 diamidino-2-phenylindole (DAPI, blue), scale bar shown in (<b>b</b>) corresponds to the images shown in (<b>a</b>) and (<b>b</b>). (<b>c</b>) Cerebral content of Neuronal Differentiation 1 protein (NeuroD1), SRY-related HMG-box 2 (Sox2) and erythropoietin (EPO) assessed in brain homogenates of APP/PS1 mice (<span class="html-italic">n</span> = 4) by Western Blot. (<b>d</b>,<b>e</b>) Enzymatic activity (<span class="html-italic">n</span> = 8) and expression level of glutamine synthetase (GS) in brain homogenates of losartan and vehicle (Ctrl.) treated APP/PS1 mice (<span class="html-italic">n</span> = 4). Statistical analysis in (<b>d</b>): <span class="html-italic">t</span>-test, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Expression of factors regulating cholinergic activity, Aβ clearance and microglial function in losartan-treated APP/PS1 mice. (<b>a</b>) qPCR of β secretase 1 (BACE1) mRNA (<span class="html-italic">n</span> = 7 in control and <span class="html-italic">n</span> = 9 in losartan treated group); (<b>b</b>) Western blot of neprilysin, Acetylcholine esterase (AChE), Choline acetyltransferase (ChAT) and ionized calcium-binding adapter molecule 1 (Iba1), <span class="html-italic">n</span> = 4; (<b>c</b>–<b>e</b>) qPCR of AChE, ChAT and Iba1 mRNA (<span class="html-italic">n</span> = 7 in control and <span class="html-italic">n</span> = 9 in losartan treated group). Statistical analysis in (<b>a</b>) and (<b>c</b>–<b>e</b>): <span class="html-italic">t</span>-test, * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.0005.</p>
Full article ">Figure 5
<p>Losartan-mediated changes in neurogenesis and neuroprotection markers in the hippocampus of APP/PS1 mice. (<b>a</b>,<b>b</b>) Immunofluorescent analysis of ChAT (green) and glial fibrillary acidic protein (GFAP) (red) in losartan vs. vehicle (Ctrl.) treated APP/PS1 mice. (<b>c</b>,<b>d</b>) Expression of AChE (red) and GFAP (green) in losartan/vehicle treated APP/PS1 mice. (<b>e</b>,<b>f</b>) Aβ plaques (red) and GFAP staining of losartan and vehicle (Ctrl.) treated APP/PS1 mouse brain sections. Cell nuclei are stained with DAPI (blue), scale bar in (<b>b</b>) corresponds to the images shown in (<b>a</b>) and (<b>b</b>), scale bar in (<b>f</b>) corresponds to (<b>c</b>–<b>f</b>). (<b>a</b>–<b>f</b>) Representative images out of 10 sections per brain taken from <span class="html-italic">n</span> = 3 mice analyzed per treatment group.</p>
Full article ">Figure 6
<p>Losartan-induced neurogenesis and ACh production in wild type (WT) astroglial primary cultures (APC). (<b>a</b>,<b>b</b>) Immunofluorescent analysis of GFAP (green) and ACh (red) in losartan vs. vehicle (Ctrl.) treated WT APC. (<b>c</b>,<b>d</b>) Expression of β tubulin III (red) and GFAP (green) in losartan/vehicle treated WT APC. Arrow in (<b>d</b>) indicates young β tubulin III positive neuron in losartan treated culture. Cell nuclei are stained with DAPI (blue), scale bar in (<b>d</b>) corresponds to the images shown in (<b>a</b>–<b>d</b>). (<b>a</b>–<b>d</b>) Representative images out of <span class="html-italic">n</span> = 4 cover slips stained with ACh/GFAP or β tubulin III/GFAP in each treatment condition.</p>
Full article ">Figure 7
<p>In vitro effects of losartan on triple transgenic Alzheimer’s disease (AD) mice (3xTg-AD) APC. (<b>a</b>,<b>b</b>) Western Blot of ChAT and BACE1 in losartan vs. vehicle (Ctrl.) treated 3xTg-APC. (<b>b</b>,<b>c</b>) Expression of neprilysin (green) in losartan (3xTg losartan) and vehicle (3xTg Ctrl.) treated 3xTg-APC. Cell nuclei are stained with DAPI (blue), scale bar in (<b>d</b>) corresponds to the images shown in (<b>b</b>,<b>c</b>). (<b>d</b>) Quantification of neprilysin positive cells in 3xTg-APC (<span class="html-italic">n</span> = 5). (<b>e</b>) Velocity of migrated cells in the control (ctrl.) and losartan treated 3xTg-APC. From the cells shown in videos in <a href="#app1-pharmaceuticals-14-00166" class="html-app">Supplementary Figure S1</a>, we identified and analyzed 32 cells per treatment group (<span class="html-italic">n</span> = 32) which were trackable for at least 12h; (<b>f</b>) accumulated distance calculated for cells described in (<b>e</b>); Statistical analysis in (<b>d</b>–<b>f</b>): <span class="html-italic">t</span>-test, * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.0005.</p>
Full article ">Figure 8
<p>Schematic illustration of processes influenced by losartan in an AD-like pathology in vivo and in vitro. (<b>a</b>) In vivo effects of losartan in a transgenic model of AD (APP/PS1 mouse). Intraperitoneal administration of losartan led to a decrease in: Acetylcholine esterase (AchE), TNFα, IL-6, IL-2, β secretase 1 (BACE1), and beta amyloid (Aβ), and to an increase in Choline acetyltransferase (ChAT), glutamine synthetase (GS), neprilysin, SRY-related HMG-box 2 (Sox-2), Neuronal Differentiation 1 protein (NeuroD1) and erythropoietin (EPO). Blue solid arrows connect the processes (neurogenesis, neuroinflammation, cholinergic activity and Aβ/Glutamate (Glu)-toxicity) with their respective markers. Dashed blue arrows show the effect of EPO on GS, TNFα, IL-6 and neurogenesis evidenced by the literature indicated in white circles; (<b>b</b>) in vitro effects of losartan on the astrocytes and neural precursors. Losartan induced the differentiation of neurons from neural precursors, increased the Acetylcholine (ACh) in neurons, decreased the expression of BACE1 and upregulated ChAT and neprilysin in astrocytes. Losartan improved the motility and migration of astrocytes. This schematic drawing was created using art elements from Servier Medical Art Commons Attribution 3.0 Unported License. Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License.</p>
Full article ">
11 pages, 796 KiB  
Article
Α Multicenter Retrospective Study Evaluating Brivaracetam in the Treatment of Epilepsies in Clinical Practice
by Maria Stefanatou, Eirini Vasileiadou Kapetanou, Vasilios K. Kimiskidis, Vasileios Papaliagkas, Panagiotis Polychronopoulos, Sofia Markoula, Kleoniki Charisiou, Dimitrios Kazis, Anastasia Verentzioti, Panayiotis Patrikelis, Athanasia Alexoudi and Stylianos Gatzonis
Pharmaceuticals 2021, 14(2), 165; https://doi.org/10.3390/ph14020165 - 19 Feb 2021
Cited by 4 | Viewed by 3278
Abstract
Brivaracetam (BRV) is the latest approved antiepileptic drug. The aim of the study was to evaluate the efficacy and tolerability of BRV in everyday clinical practice. In this retrospective, observational, multicenter study, data from epilepsy patients receiving BRV from January 2018 to July [...] Read more.
Brivaracetam (BRV) is the latest approved antiepileptic drug. The aim of the study was to evaluate the efficacy and tolerability of BRV in everyday clinical practice. In this retrospective, observational, multicenter study, data from epilepsy patients receiving BRV from January 2018 to July 2019 were analyzed. Patients with age ≥16 suffering from any type of epilepsy and having at least one follow up encounter after dose titration were included. 156 consecutive patients were included in the study. The mean age was 40 (16–84 years) and the mean duration of epilepsy was 21 years. Of the 156 patients, 81% were diagnosed with focal-onset seizures, 16% with generalized seizures, while 3% suffered from unclassified seizures. Nine patients received BRV as monotherapy as a switching therapy. At the first follow up visit, seizure cessation was achieved in 56 (36%) patients and the rate of ≥50% responders was 36%. Twenty four patients (15%) remained unchanged; six patients (4%) were recorded with increased seizure frequency, while the remaining 9% had a response of less than 50%. Twenty-six patients (17%) showed clinically significant adverse events, but none were life threatening. Brivaracetam seems to be an effective, easy to use and safe antiepileptic drug in the clinical setting. Full article
(This article belongs to the Special Issue Therapeutic Agents for Neurological Disorders)
Show Figures

Figure 1

Figure 1
<p>Response rates in drug-resistant epilepsy.</p>
Full article ">Figure 2
<p>Response rates after BRV initiation correlated to epilepsy syndrome.</p>
Full article ">Figure 3
<p>Response rates after BRV treatment in LEV + and LEV− subgroups.</p>
Full article ">Figure 4
<p>Response rates after BRV treatment correlated to BRV dose.</p>
Full article ">
18 pages, 2859 KiB  
Article
In Vitro Characterization of Inhalable Cationic Hybrid Nanoparticles as Potential Vaccine Carriers
by Iman M. Alfagih, Kan Kaneko, Nitesh K. Kunda, Fars Alanazi, Sarah R. Dennison, Hesham M. Tawfeek and Imran Y. Saleem
Pharmaceuticals 2021, 14(2), 164; https://doi.org/10.3390/ph14020164 - 18 Feb 2021
Cited by 12 | Viewed by 3496
Abstract
In this study, PGA-co-PDL nanoparticles (NPs) encapsulating model antigen, bovine serum albumin (BSA), were prepared via double emulsion solvent evaporation. In addition, chitosan hydrochloride (CHL) was incorporated into the external phase of the emulsion solvent method, which resulted in surface adsorption onto the [...] Read more.
In this study, PGA-co-PDL nanoparticles (NPs) encapsulating model antigen, bovine serum albumin (BSA), were prepared via double emulsion solvent evaporation. In addition, chitosan hydrochloride (CHL) was incorporated into the external phase of the emulsion solvent method, which resulted in surface adsorption onto the NPs to form hybrid cationic CHL NPs. The BSA encapsulated CHL NPs were encompassed into nanocomposite microcarriers (NCMPs) composed of l-leucine to produce CHL NPs/NCMPs via spray drying. The CHL NPs/NCMPs were investigated for in vitro aerosolization, release study, cell viability and uptake, and stability of protein structure. Hybrid cationic CHL NPs (CHL: 10 mg/mL) of particle size (480.2 ± 32.2 nm), charge (+14.2 ± 0.72 mV), and BSA loading (7.28 ± 1.3 µg/mg) were produced. The adsorption pattern was determined to follow the Freundlich model. Aerosolization of CHL NPs/NCMPs indicated fine particle fraction (FPF: 46.79 ± 11.21%) and mass median aerodynamic diameter (MMAD: 1.49 ± 0.29 µm). The BSA α-helical structure was maintained, after release from the CHL NPs/NCMPs, as indicated by circular dichroism. Furthermore, dendritic cells (DCs) and A549 cells showed good viability (≥70% at 2.5 mg/mL after 4–24 h exposure, respectively). Confocal microscopy and flow cytometry data showed hybrid cationic CHL NPs were successfully taken up by DCs within 1 h of incubation. The upregulation of CD40, CD86, and MHC-II cell surface markers indicated that the DCs were successfully activated by the hybrid cationic CHL NPs. These results suggest that the CHL NPs/NCMPs technology platform could potentially be used for the delivery of proteins to the lungs for immunostimulatory applications such as vaccines. Full article
(This article belongs to the Special Issue Nano Drug Carriers 2021)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The relationship of CHL concentration to the amount absorbed on PGA-co-PDL NPs (<b>A</b>). Zeta potential and particle size of hybrid cationic CHL NPs (<b>B</b>) (<span class="html-italic">n</span> = 3, <span class="html-italic">p</span> &lt; 0.05 is indicated by *).</p>
Full article ">Figure 2
<p>Linear description of CHL adsorption onto PGA-co-PDL NPs. Ce is the residual CHL concentrations in the suspension at equilibrium (mg/mL) and q is the amount of adsorbed CHL per unit weight of NPs. All models (<b>A</b>) Langmuir, (<b>B</b>) BET, (<b>C</b>) Freundlich (<b>D</b>) Halsey were fit utilizing concentrations of 2–20 mg/mL; however, the Langmuir model was also fit to low concentrations (0–8 mg/mL) (black) (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>Scanning electron microscopy images of cationic CHL NPs/NCMPs: (<b>A</b>) 1 µm scale and (<b>B</b>) 2 µm scale.</p>
Full article ">Figure 4
<p>In vitro release of BSA from cationic CHL NPs/NCMPs in PBS buffer at 37 °C (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 5
<p>SDS-PAGE to evaluate the BSA stability released from cationic CHL NPs/NCMPs (<b>A</b>). Lanes represent, molecular weight (MW) standard markers, BSA (MW 66,000) (<b>1</b>), BSA standard (<b>2</b>), BSA released from hybrid cationic CHL NPs (<b>3</b>), and BSA released from cationic CHL NPs/NCMPs (<b>4</b>). Difference in band intensity was due to different loading. (The CD spectra of BSA released from cationic CHL NPs/NCMPs (grey) and BSA standard (black) (<b>B</b>) (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 6
<p>MTT assay to determine cell viability of A549 cells after 24 h of exposure to hybrid cationic CHL NPs and cationic CHL NPs/NCMPs, and DCs after 4 h of exposure to hybrid cationic CHL NPs (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 7
<p>Confocal microscopy images of hybrid cationic CHL NPs uptake by DCs after 1 h incubation. DCs incubated without hybrid cationic CHL NPs at 20× (<b>A</b>), DCs incubated with hybrid cationic CHL NPs at 63× (<b>B</b>). Red channel for WGA TR: cell membrane, blue channel for DAPI: nucleus, and green channel for FITC-BSA: hybrid cationic CHL NPs incorporating FITC-BSA.</p>
Full article ">Figure 8
<p>The association/uptake of FITC-BSA in cationic CHL NPs, by DCs. DCs were incubated with FITC-BSA incorporated CHL NPs for either 1 or 4 h, at either 4 or 37 °C (<span class="html-italic">n</span> = 4).</p>
Full article ">Figure 9
<p>Upregulation of cell surface CD40 (<b>A</b>), CD86 (<b>B</b>), and MHC-II (<b>C</b>) on DCs, and cell viability according to 7AAD staining (<b>D</b>), after incubation with BSA incorporated hybrid cationic CHL NPs for 24 h (<span class="html-italic">n</span> = 6). The indicated groups exhibited significantly greater MFI (<span class="html-italic">p</span> &lt; 0.05) compared to negative control. (<b>1</b>), BSA (<b>2</b>), CHL NPs (<b>3</b>), BSA CHL NPs (<b>4</b>), LPS (<b>5</b>).</p>
Full article ">
10 pages, 1632 KiB  
Article
Development and Validation of an LC-MS/MS Method for Quantification of the Novel Antibacterial Candidate DA-7010 in Plasma and Application to a Preclinical Pharmacokinetic Study
by Mi Hye Kwon, Dae Young Lee and Hee Eun Kang
Pharmaceuticals 2021, 14(2), 163; https://doi.org/10.3390/ph14020163 - 18 Feb 2021
Cited by 2 | Viewed by 2683
Abstract
DA-7010 is a new candidate for an antibacterial agent that targets Gram-negative pathogens by acting as a leucyl-tRNA synthetase inhibitor. In this study, a simple and rapid liquid chromatography tandem mass spectrometry (LC-MS/MS) method was developed to determine DA-7010 levels in the plasma [...] Read more.
DA-7010 is a new candidate for an antibacterial agent that targets Gram-negative pathogens by acting as a leucyl-tRNA synthetase inhibitor. In this study, a simple and rapid liquid chromatography tandem mass spectrometry (LC-MS/MS) method was developed to determine DA-7010 levels in the plasma from mice, rats, and dogs. Plasma samples were mixed with methanol for protein precipitation. Chromatographic separation was carried out using a reversed-phase C18 column (Agilent Poroshell 120, 50 × 3.0 mm, 2.7 μm). An isocratic elution of the mobile phase consisting of 5 mM formic acid in water and acetonitrile at a ratio of 84:16 (v/v) was applied at a flow rate of 0.3 mL/min. The total chromatographic run time was 3.5 min. Multiple reaction monitoring (MRM) mode was used for mass spectrometric detection using an Agilent 6460 triple quadrupole coupled with an electrospray ionization (ESI) source operated in positive-ion mode. The MRM transitions analyzed were m/z 220.1→162.1 for DA-7010 and m/z 206.1→170.1 for the internal standard (structural analogue of DA-7010). Calibration curves were constructed in the range of 10–10,000 ng/mL. The intra- and interday precision and accuracy were within 11.3%, excluding those for the lower limit of quantification (LLOQ) samples, which were within 17.1%. The developed LC-MS/MS method was successfully validated and applied in preclinical pharmacokinetic studies of DA-7010 in mice, rats, and dogs following single oral administrations. The oral absorption of DA-7010 was rapid, and the systemic exposure was approximately four times higher in the dogs than in the mice or rats. Full article
(This article belongs to the Special Issue Analytical Techniques in the Pharmaceutical Sciences)
Show Figures

Figure 1

Figure 1
<p>Chemical structures of DA-7010 and the internal standard (IS): (<b>a</b>) DA-7010; (<b>b</b>) the IS.</p>
Full article ">Figure 2
<p>Product ion mass spectra of (<b>a</b>) DA-7010 and (<b>b</b>) the IS.</p>
Full article ">Figure 3
<p>Representative multiple reaction monitoring (MRM) chromatograms of DA-7010 (I) and the IS (II) in each mouse, rat, and dog plasma sample: (<b>a</b>) blank plasma; (<b>b</b>) blank plasma spiked with lower limit of quantification (LLOQ) level of DA-7010 (10 ng/mL) or mixed with the IS at working concentrations (10 μg/mL); (<b>c</b>) plasma samples collected 4 h after oral administration of 3 mg/kg DA-7010 to mouse, rat, and dog with determined DA-7010 concentrations of 334, 648, and 1952 ng/mL, respectively.</p>
Full article ">Figure 4
<p>Mean arterial plasma concentration–time profiles of DA-7010 following oral administration of DA-7010 at a dose of 3 mg/kg to mice (<span class="html-italic">n</span> = 6 for each point), rats (<span class="html-italic">n</span> = 7), and dogs (<span class="html-italic">n</span> = 4). Data are expressed as mean ± SD.</p>
Full article ">
13 pages, 1929 KiB  
Article
Limited Sampling Strategy for Determination of Ibrutinib Plasma Exposure: Joint Analyses with Metabolite Data
by Félicien Le Louedec, Fanny Gallais, Fabienne Thomas, Mélanie White-Koning, Ben Allal, Caroline Protin, Loïc Ysebaert, Étienne Chatelut and Florent Puisset
Pharmaceuticals 2021, 14(2), 162; https://doi.org/10.3390/ph14020162 - 18 Feb 2021
Cited by 5 | Viewed by 3814
Abstract
Therapeutic drug monitoring of ibrutinib is based on the area under the curve of concentration vs. time (AUCIBRU) instead of trough concentration (Cmin,ss) because of a limited accumulation in plasma. Our objective was to identify a limited sampling strategy [...] Read more.
Therapeutic drug monitoring of ibrutinib is based on the area under the curve of concentration vs. time (AUCIBRU) instead of trough concentration (Cmin,ss) because of a limited accumulation in plasma. Our objective was to identify a limited sampling strategy (LSS) to estimate AUCIBRU associated with Bayesian estimation. The actual AUCIBRU of 85 patients was determined by the Bayesian analysis of the full pharmacokinetic profile of ibrutinib concentrations (pre-dose T0 and 0.5, 1, 2, 4 and 6 h post-dose) and experimental AUCIBRU were derived considering combinations of one to four sampling times. The T0–1–2–4 design was the most accurate LSS (root-mean-square error RMSE = 11.0%), and three-point strategies removing the 1 h or 2 h points (RMSE = 22.7% and 14.5%, respectively) also showed good accuracy. The correlation between the actual AUCIBRU and Cmin,ss was poor (r2 = 0.25). The joint analysis of dihydrodiol-ibrutinib metabolite concentrations did not improve the predictive performance of AUCIBRU. These results were confirmed in a prospective validation cohort (n = 27 patients). At least three samples, within the pre-dose and 4 h post-dose period, are necessary to estimate ibrutinib exposure accurately. Full article
Show Figures

Figure 1

Figure 1
<p>Ibrutinib actual area under the curve (AUC) vs. observed steady-state trough concentration. Solid line: linear regression (<span class="html-italic">n</span> = 85 patients).</p>
Full article ">Figure 2
<p>Comparison of the estimation of ibrutinib AUC using ibrutinib concentrations only (<b>left</b>) or both ibrutinib and dihydrodiol-ibrutinib (<b>right</b>), as a function of the number of points used for estimation (seven values with a bias &gt; 10% are out of bounds). MPE: mean percentage error; RMSE: Root-Mean-Square Error.</p>
Full article ">Figure 3
<p>Comparison of the performance of several sampling strategies to predict ibrutinib AUC in the development (<b>top</b>) and validation (<b>bottom</b>) dataset. Crosses (+) are out of bound values censored to the maximal bound of the y-axis (i.e., +/−60%).</p>
Full article ">Figure 4
<p>Distribution of the AUC<sub>ANTI-BTK</sub> / free AUC<sub>IBRU</sub> ratio (dashed lines: +/−25% interval around the median).</p>
Full article ">
16 pages, 1833 KiB  
Review
Bacteriophages as Therapeutic and Diagnostic Vehicles in Cancer
by Valentina Foglizzo and Serena Marchiò
Pharmaceuticals 2021, 14(2), 161; https://doi.org/10.3390/ph14020161 - 17 Feb 2021
Cited by 36 | Viewed by 7214
Abstract
Evolution of nanomedicine is the re-design of synthetic and biological carriers to implement novel theranostic platforms. In recent years, bacteriophage research favors this process, which has opened up new roads in drug and gene delivery studies. By displaying antibodies, peptides, or proteins on [...] Read more.
Evolution of nanomedicine is the re-design of synthetic and biological carriers to implement novel theranostic platforms. In recent years, bacteriophage research favors this process, which has opened up new roads in drug and gene delivery studies. By displaying antibodies, peptides, or proteins on the surface of different bacteriophages through the phage display technique, it is now possible to unravel specific molecular determinants of both cancer cells and tumor-associated microenvironmental molecules. Downstream applications are manifold, with peptides being employed most of the times to functionalize drug carriers and improve their therapeutic index. Bacteriophages themselves were proven, in this scenario, to be good carriers for imaging molecules and therapeutics as well. Moreover, manipulation of their genetic material to stably vehiculate suicide genes within cancer cells substantially changed perspectives in gene therapy. In this review, we provide examples of how amenable phages can be used as anticancer agents, especially because their systemic administration is possible. We also provide some insights into how their immunogenic profile can be modulated and exploited in immuno-oncology for vaccine production. Full article
(This article belongs to the Special Issue Bacteriophages as Therapeutic Delivery Vehicles)
Show Figures

Figure 1

Figure 1
<p>M13-based applications. Examples of therapeutic and diagnostic approaches exploiting M13 phage display-derived peptides [<a href="#B40-pharmaceuticals-14-00161" class="html-bibr">40</a>,<a href="#B42-pharmaceuticals-14-00161" class="html-bibr">42</a>], capsid proteins [<a href="#B44-pharmaceuticals-14-00161" class="html-bibr">44</a>,<a href="#B45-pharmaceuticals-14-00161" class="html-bibr">45</a>], and whole phages [<a href="#B46-pharmaceuticals-14-00161" class="html-bibr">46</a>,<a href="#B48-pharmaceuticals-14-00161" class="html-bibr">48</a>,<a href="#B50-pharmaceuticals-14-00161" class="html-bibr">50</a>] as carriers for tumor-targeted delivery of drugs (e.g., Dox, antisense oligonucleotides, photosensitizers) or imaging dyes (e.g., fluorescein, NIR-SWNTs). Figure created with BioRender.com.</p>
Full article ">Figure 2
<p>Theranostic applications of the hybrid AAVP vector. AAPVs can be ligand-targeted to a tumor-specific cell surface molecule such as α<sub>v</sub>β<sub>3</sub> integrin [<a href="#B53-pharmaceuticals-14-00161" class="html-bibr">53</a>] or GRP78 [<a href="#B59-pharmaceuticals-14-00161" class="html-bibr">59</a>], followed by internalization of DNA and production of AAVP-coded proteins. The <span class="html-italic">HSVtk</span> transgene codes for TK, an enzyme that adds phosphate groups to thymidine analogues and converts (i) the prodrug GCV in the cytotoxic drug GCV triphosphate and (ii) [<sup>18</sup>F]FEAU in [<sup>18</sup>F]FEAU phosphate, which is retained intracellularly, allowing detection by PET imaging. Figure created with BioRender.com.</p>
Full article ">Figure 3
<p>Bacteriophage-based platforms to deliver therapeutic and diagnostic agents via RNA technology. (<b>a</b>) Reintroduction of MEG3 lncRNA in cancer cells via an EGFR-targeted MS2 phage—upon binding to EGFR, the phage is internalized by endocytosis and releases MEG3, thus inducing apoptosis and inhibiting proliferation of cancer cells [<a href="#B63-pharmaceuticals-14-00161" class="html-bibr">63</a>]. (<b>b</b>) VNPs derived from Φ29 bacteriophage pRNA are suitable nanocarriers for the delivery of RNAs (aptamer, ribozyme, siRNA), small molecules (drug, dye), or proteins (targeting moiety) to cancer cells [<a href="#B64-pharmaceuticals-14-00161" class="html-bibr">64</a>,<a href="#B65-pharmaceuticals-14-00161" class="html-bibr">65</a>]. Figure created with BioRender.com.</p>
Full article ">
17 pages, 2631 KiB  
Article
Design and Mechanism of Action of a New Prototype of Combi-Molecule “Programed” to Release Bioactive Species at a pH Range Akin to That of the Tumor Microenvironment
by Anne-Laure Larroque-Lombard, Etienne Chatelut, Jean-Pierre Delord, Diane-Charlotte Imbs, Philippe Rochaix, Bertrand Jean-Claude and Ben Allal
Pharmaceuticals 2021, 14(2), 160; https://doi.org/10.3390/ph14020160 - 16 Feb 2021
Cited by 3 | Viewed by 3863
Abstract
The clinical use of cytotoxic agents is plagued by systemic toxicity. We report a novel approach that seeks to design a “combi-molecule” to behave as an alkylating agent on its own and to undergo acid-catalyzed conversion to two bioactive species at a pH [...] Read more.
The clinical use of cytotoxic agents is plagued by systemic toxicity. We report a novel approach that seeks to design a “combi-molecule” to behave as an alkylating agent on its own and to undergo acid-catalyzed conversion to two bioactive species at a pH range akin to that of a tumor microenvironment: an AL530 prototype was synthesized and we studied its ability to release a chlorambucil analogue (CBL-A) plus a potent mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK) inhibitor (PD98059) at different pHs in buffered solutions, plasma and tumors. Its potency was compared in vitro with CBL+PD98059 (SRB assay) and in vivo in a xenograft model. Its target modulation was studied by western blotting and immunohistochemistry. AL530 released PD98059+CBL-A at mild acidic pH and in vitro was fivefold more potent than CBL and three-to-fivefold more potent than CBL+PD98059. In vivo it released high levels of PD98059 in tumors with a tumor/plasma ratio of five. It induced γ-H2AX phosphorylation and blocked pErk1,2, indirectly indicating its ability to damage DNA and modulate MEK. It induced significant tumor delay and less toxicity at unachievable doses for CBL and CBL+PD98059. We demonstrated the feasibility of a pH-labile combi-molecule capable of delivering high MEK inhibitor concentration in tumors, damaging DNA therein, and inducing tumor growth delay. Full article
Show Figures

Figure 1

Figure 1
<p>Multiple cell signaling pathways leading to MEK activation. GF: Growth Factor, RTK: Receptor Tyrosine Kinase.</p>
Full article ">Figure 2
<p>Influence of pH and temperature on AL530 hydrolysis. Graphs (<b>A</b>) and (<b>B</b>) represent the formation rate (constant k) of PD98059 at ■—37 °C, ▲—40 °C and ▼—42 °C versus the pH ranging from 5.5 to 7. The overlaid chromatograms (<b>C</b>) represent the time course effects (from 24 to 96 h) of AL530 hydrolysis versus the formation of PD98059 at 37 °C at pH = 7.</p>
Full article ">Figure 3
<p>Study of Erk1,2 phosphorylation inhibition in CAL33 cells daily treated with AL530 (25 µM) for 24 to 48 h. At the indicated times in the figure, cells were harvested, lysed (RIPA buffer), an equal amount of protein was separated (12.5% SDS PAGE gel), blotted (PVDF membrane) and coated with antibody of interest.</p>
Full article ">Figure 4
<p>(<b>A</b>) In vivo hydrolysis of AL530 in 4T1 mouse mammary tumors (top panel) and human head and neck tumors generated from the CAL33 human xenograft in nude mice (bottom panel). Each chromatogram represents the analysis of plasma or tumor extract from a mouse treated with 100 mg/kg of AL530 (i.p.) after 3 h (4T1 model) and 50 mg/kg (i.p.) after 1 h (CAL33 model). Following mice sacrifice, plasma and tissue were collected and stored at −80 °C until the day of analysis by HPLC as described in the Materials and Methods section. Consistently higher levels of PD98059 were released in tumor tissues than in plasma. (<b>B</b>) Proposed model for the absorption and distribution of AL530 and the localization of its hydrolytic conversion to PD98059 + CBL-A.</p>
Full article ">Figure 5
<p>Immunohistochemistry analysis of the effect of AL530 on tumor tissue. Thick sections of 4 µm formalin fixed and paraffin embedded in tumors from CAL33 cell xenografts in nude mice treated with vehicle or 50 mg/kg of AL530 i.p. for 13 days, were stained with antibody against γ-H2AX (<b>A</b>) and pErk1,2 (<b>B</b>) (magnification, x20).</p>
Full article ">Figure 6
<p>Effect of vehicle, AL530, Chlorambucil (CBL), PD98059 (MEK inhibitor) or equimolar combination CBL + PD98059 treatment on tumors growth (<b>A</b>,<b>C</b>) and weight (<b>B</b>) properties of CAL33 head and neck cancer cell xenograft. Nude mice (<span class="html-italic">n</span> = 7 per group) bearing subcutaneous 200 mm<sup>3</sup> (at the start of the experiment) bilateral CAL33 tumors xenograft received either saline (orange square, <b>A1</b>), 25 mg/kg (<b>A1</b>) or 50 mg/kg (<b>C</b>) of AL530 (green triangle), or 11 mg/kg of CBL (<b>A2</b>, blue diamond), or 9.7 mg/kg of PD98059 (data not shown) or an equimolar combination of CBL + PD98059 (1:1; <b>A3</b>, orange circle) daily by i.p. route/q5d throughout the experiment. Data are expressed as mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.0001 and * <span class="html-italic">p</span> &lt; 0.01, Mann–Whitney posthoc test.</p>
Full article ">Scheme 1
<p>2-Nitroaryl amides proposed as bioreducible prodrugs of anilines based on the intramolecular cyclization of the 2-aminoaryl amides resulting from the reduction of the 2-nitro group.</p>
Full article ">Scheme 2
<p>Proposed mechanism for the pH dependence of the degradation of the lead combi-molecule AL530.</p>
Full article ">Scheme 3
<p>(<b>i</b>) <b>8S</b>, 1 h and THF/CH<sub>2</sub>Cl<sub>2</sub> 1/1, Et<sub>3</sub>N, 0 °C to 50 °C, 3 days, 44%, (<b>ii</b>) LiOH 2N, MeOH, rt, 18 h, 97%, (<b>iii</b>) CDI, aniline mustard (compound 12S), THF/ACN 1/1, 0 °C and rt, 18 h, 47%, (<b>iv</b>) Pd/C 10%, MeOH, H<sub>2</sub>, rt, 3 h, 65%.</p>
Full article ">
9 pages, 585 KiB  
Case Report
Tumor Type Agnostic Therapy Carrying BRAF Mutation: Case Reports and Review of Literature
by Ottavia Bernocchi, Marianna Sirico, Silvia Paola Corona, Carla Strina, Manuela Milani, Maria Rosa Cappelletti, Giuseppina Ferrero, Nicoletta Ziglioli, Valeria Cervoni, Andrea Macchiavelli, Giandomenico Roviello and Daniele Generali
Pharmaceuticals 2021, 14(2), 159; https://doi.org/10.3390/ph14020159 - 16 Feb 2021
Cited by 6 | Viewed by 3603
Abstract
Background: Precision medicine is based on molecular and genotypic patient characterization to define specific target treatment. BRAF mutation is an oncogenic driver, and the Cancer Genome Atlas has identified BRAF mutations in different cancer types. Tumor type agnostic therapy is based on targeting [...] Read more.
Background: Precision medicine is based on molecular and genotypic patient characterization to define specific target treatment. BRAF mutation is an oncogenic driver, and the Cancer Genome Atlas has identified BRAF mutations in different cancer types. Tumor type agnostic therapy is based on targeting genomic alterations, regardless of tumor origin. In this context, novel therapeutic agents including BRAF and MEK inhibitors based on the molecular landscape in solid tumors have been investigated. Case presentation, Case 1: The first case is chemotherapy-refractory, BRAF V600E mutated intrahepaticcholangiocarcinoma treated with vemurafenib and cobimetinib as third line therapy. In this setting the dual BRAF and MEK inhibition resulted in improved progression-free survival and quality of life; Case 2: The second case shows aBRAF G466A mutated Bellini duct carcinoma (BDC), treated with dabrafenib and trametinib in second line therapy. The disease remained under control for 11 months after the first relapse. Discussion: In the literature there is strong evidence that melanoma, colorectal cancer, non small cell lung cancer and anaplastic thyroid cancer with BRAF mutations are good targets for BRAF/MEK pathway inhibitors. The VE-BASKET and ROAR basket trials explored the efficacy of vemurafenib and the combination of dabrafenib/trametinib, respectively, in BRAF V600 mutation-positive cancers other than melanoma, papillary thyroid cancer, colorectal cancer and non small cell lung cancer. Within the concept of tumor type agnostic therapy, we decided to treat our BRAF-mutated tumors with the association of BRAF and MEK inhibitors. Conclusions: Our results confirm the emerging importance of molecular tumor profiling for the successful management of cancer, and the potential of BRAF-targeted therapy in the treatment of rare solid tumors with poor prognosis and no clinical benefit from systemic therapies with. Full article
(This article belongs to the Special Issue Cancer Translational Biomarkers and Targeted Therapies)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) CT scan, March 2019; (<b>b</b>) CT scan, September 2020.</p>
Full article ">Figure 2
<p>FDG-PET after 4 weeks of treatment with dabrafenib and trametinib showed decreased FDG avidity. (<b>a</b>) FDG-PET performed in January; (<b>b</b>) FDG-PET performed in February 2016.</p>
Full article ">
13 pages, 1210 KiB  
Article
Preparation and Bioevaluation of Novel 99mTc-Labeled Complexes with a 2-Nitroimidazole HYNIC Derivative for Imaging Tumor Hypoxia
by Qing Ruan, Qianqian Gan, Xuran Zhang, Si’an Fang and Junbo Zhang
Pharmaceuticals 2021, 14(2), 158; https://doi.org/10.3390/ph14020158 - 15 Feb 2021
Cited by 6 | Viewed by 3189
Abstract
To develop novel 99mTc-labeled single-photon emission computed tomography (SPECT) radiotracers for imaging hypoxia, a novel HYNICNM ligand (6-hydrazinonicotinamide (HYNIC) 2-nitroimidazole derivative) was designed and synthesized. It was radiolabeled with technetium-99m using tricine/trisodium triphenylphosphine-3,3′,3′′-trisulfonate (TPPTS), tricine/sodium triphenylphosphine-3-monosulfonate (TPPMS) and tricine as co-ligands to [...] Read more.
To develop novel 99mTc-labeled single-photon emission computed tomography (SPECT) radiotracers for imaging hypoxia, a novel HYNICNM ligand (6-hydrazinonicotinamide (HYNIC) 2-nitroimidazole derivative) was designed and synthesized. It was radiolabeled with technetium-99m using tricine/trisodium triphenylphosphine-3,3′,3′′-trisulfonate (TPPTS), tricine/sodium triphenylphosphine-3-monosulfonate (TPPMS) and tricine as co-ligands to obtain [99mTc]Tc-tricine-TPPTS-HYNICNM, [99mTc]Tc-tricine-TPPMS-HYNICNM, and [99mTc]Tc-(tricine)2-HYNICNM, respectively. The three technetium-99m complexes were radiolabeled in one step with a high yield (95%) and had good stability in saline and mouse serum. In vitro cellular uptake results showed that these complexes exhibited good hypoxic selectivity. The partition coefficient indicated that they were good hydrophilic complexes, and [99mTc]Tc-tricine-TPPTS-HYNICNM displayed the highest hydrophilicity (−3.02 ± 0.08). The biodistribution in mice bearing S180 tumors showed that [99mTc]Tc-tricine-TPPTS-HYNICNM exhibited higher tumor uptake (1.05 ± 0.27% IA/g); more rapid clearance from the liver, blood, muscle, and other non-target organs; and a higher tumor/non-target ratio, especially for the tumor/liver ratio (1.95), than [99mTc]Tc-tricine-TPPMS-HYNICNM and [99mTc]Tc-(tricine)2-HYNICNM. The results of single-photon emission computed tomography (SPECT) imaging studies of [99mTc]Tc-tricine-TPPTS-HYNICNM were in accordance with the biodistribution results, which suggested that [99mTc]Tc-tricine-TPPTS-HYNICNM is a promising agent for imaging tumor hypoxia. Full article
Show Figures

Figure 1

Figure 1
<p>HPLC patterns of <sup>99m</sup>Tc-complexes: (<b>a</b>) [<sup>99m</sup>Tc]NaTcO<sub>4</sub>; (<b>b</b>) [<sup>99m</sup>Tc]Tc-tricine-TPPTS-HYNICNM; (<b>c</b>) [<sup>99m</sup>Tc]Tc-tricine-TPPMS-HYNICNM; (<b>d</b>) [<sup>99m</sup>Tc]Tc-(tricine)<sub>2</sub>-HYNICNM.</p>
Full article ">Figure 2
<p>Cellular uptake of <sup>99m</sup>Tc-complexes: (<b>a</b>) [<sup>99m</sup>Tc]Tc-tricine-TPPTS-HYNICNM; (<b>b</b>) [<sup>99m</sup>Tc]Tc-tricine-TPPMS-HYNICNM; (<b>c</b>) [<sup>99m</sup>Tc]Tc-(tricine)<sub>2</sub>-HYNICNM.</p>
Full article ">Figure 3
<p>(<b>a</b>) Comparisons of biodistribution of [<sup>99m</sup>Tc]Tc-tricine-TPPTS-HYNICNM, [<sup>99m</sup>Tc]Tc-tricine-TPPMS-HYNICNM, and [<sup>99m</sup>Tc]Tc-(tricine)<sub>2</sub>-HYNICNM at 2 h post-injection (<span class="html-italic">n</span> = 5). (<b>b</b>) Comparisons of the tumor-to-blood, tumor-to-muscle, and tumor-to-liver ratios of [<sup>99m</sup>Tc]Tc-tricine-TPPTS-HYNICNM, [<sup>99m</sup>Tc]Tc-tricine-TPPMS-HYNICNM, and [<sup>99m</sup>Tc]Tc-(tricine)<sub>2</sub>-HYNICNM at 2 h post-injection (<span class="html-italic">n</span> = 5).</p>
Full article ">Figure 4
<p>Single-photon emission computed tomography (SPECT)/CT images of [<sup>99m</sup>Tc]Tc-tricine-TPPTS-HYNICNM in Kunming female mice bearing S180 tumors at 2 h post-injection: (<b>a</b>) visual imaging; (<b>b</b>) coronal section; (<b>c</b>) sagittal section; (<b>d</b>) transverse section.</p>
Full article ">Scheme 1
<p>The synthesis and speculative structures of [<sup>99m</sup>Tc]Tc-tricine-TPPTS-HYNICNM, [<sup>99m</sup>Tc]Tc-tricine-TPPMS-HYNICNM, and [<sup>99m</sup>Tc]Tc-(tricine)<sub>2</sub>-HYNICNM. Reagents and conditions: (<b>a</b>) DMF, TEA, reflux, 4 h; (<b>b</b>) Trisodium triphenylphosphine-3,3′,3′′-trisulfonate (TPPTS), tricine, SnCl<sub>2</sub>·2H<sub>2</sub>O, acetate buffer solution (pH 5, 0.5 M), [<sup>99m</sup>Tc]NaTcO<sub>4</sub>, 100 °C, 0.5 h; (<b>c</b>) Sodium triphenylphosphine-3-monosulfonate (TPPM, tricine, SnCl<sub>2</sub>·2H<sub>2</sub>O, acetate buffer solution (pH 5, 0.5 M), [<sup>99m</sup>Tc]NaTcO<sub>4</sub>, 100 °C, 0.5 h; (<b>d</b>) tricine, SnCl<sub>2</sub>·2H<sub>2</sub>O, acetate buffer solution (pH 5, 0.5 M), [<sup>99m</sup>Tc]NaTcO<sub>4</sub>, 100 °C, 0.5 h.</p>
Full article ">
28 pages, 6335 KiB  
Review
Status and Challenges of Plant-Anticancer Compounds in Cancer Treatment
by Paula Garcia-Oliveira, Paz Otero, Antia Gonzalez Pereira, Franklin Chamorro, Maria Carpena, Javier Echave, Maria Fraga-Corral, Jesus Simal-Gandara and Miguel Angel Prieto
Pharmaceuticals 2021, 14(2), 157; https://doi.org/10.3390/ph14020157 - 14 Feb 2021
Cited by 136 | Viewed by 12813
Abstract
Nowadays, cancer is one of the deadliest diseases in the world, which has been estimated to cause 9.9 million deaths in 2020. Conventional treatments for cancer commonly involve mono-chemotherapy or a combination of radiotherapy and mono-chemotherapy. However, the negative side effects of these [...] Read more.
Nowadays, cancer is one of the deadliest diseases in the world, which has been estimated to cause 9.9 million deaths in 2020. Conventional treatments for cancer commonly involve mono-chemotherapy or a combination of radiotherapy and mono-chemotherapy. However, the negative side effects of these approaches have been extensively reported and have prompted the search of new therapeutic drugs. In this context, scientific community started to look for innovative sources of anticancer compounds in natural sources, including traditional plants. Currently, numerous studies have evaluated the anticancer properties of natural compounds derived from plants, both in vitro and in vivo. In pre-clinical stages, some promising compounds could be mentioned, such as the sulforaphane or different phenolic compounds. On the other hand, some phytochemicals obtained positive results in clinical stages and were further approved for cancer treatment, such as vinca alkaloids or the paclitaxel. Nevertheless, these compounds are not exempt of limitations, such as low solubility, restricted effect on their own, negative side-effects, etc. This review aims to compile the information about the current phytochemicals used for cancer treatment and also promising candidates, main action mechanisms and also reported limitations. In this sense, some strategies to face the limitations have been considered, such as nano-based formulations to improve solubility or chemical modification to reduce toxicity. In conclusion, although more research is still necessary to develop more efficient and safe phytochemical drugs, more of these compounds might be used in future cancer therapies. Full article
(This article belongs to the Special Issue Anticancer Compounds in Medicinal Plants)
Show Figures

Figure 1

Figure 1
<p>Main causes involved in the development of cancer, according to WHO.</p>
Full article ">Figure 2
<p>Schematic process for the development of anticancer drugs based on plant-derived compounds.</p>
Full article ">Figure 3
<p>Main mechanisms of phytochemical compounds employed in cancer therapy.</p>
Full article ">
8 pages, 1294 KiB  
Article
Quetiapine-Induced Place Preference in Mice: Possible Dopaminergic Pathway
by Yusuf S. Althobaiti
Pharmaceuticals 2021, 14(2), 156; https://doi.org/10.3390/ph14020156 - 14 Feb 2021
Cited by 4 | Viewed by 2806
Abstract
Quetiapine, an atypical antipsychotic, is effective in the management of schizophrenia, depression, and anxiety. Although quetiapine overdosage and misuse have been reported, its abuse potential has not been investigated in animals. In this study, the abuse potential of quetiapine was assessed based on [...] Read more.
Quetiapine, an atypical antipsychotic, is effective in the management of schizophrenia, depression, and anxiety. Although quetiapine overdosage and misuse have been reported, its abuse potential has not been investigated in animals. In this study, the abuse potential of quetiapine was assessed based on the conditioned place preference (CPP) paradigm of drug addiction in a mouse model. First, mice received intraperitoneal injections of quetiapine (40, 80, or 120 mg/kg) every other day during the conditioning phase. In the second experiment, mice were pretreated with 0.03 mg/kg SKF-35866, a D1 receptor antagonist, before receiving saline or quetiapine (120 mg/kg) during the conditioning phase. No significant changes in time spent in the quetiapine-paired chamber were observed compared with time spent in the saline-paired chamber in mice treated with 40 or 80 mg/kg. In contrast, the preference to the quetiapine-paired chamber was significantly increased in mice treated with 120 mg/kg quetiapine, and this effect was blocked by SKF-35866 pretreatment. These results demonstrated, for the first time, the abuse potential of quetiapine in an animal model of drug addiction. Interestingly, this CPP-inducing effect was likely mediated by activating D1 receptors. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Time spent in chamber 1 compared with chamber 2 during the pretest and post-test in the control group. (<b>B</b>) Time spent in the quetiapine-paired chamber compared with the saline-paired chamber during the pretest and post-test in the Quet-40 group (treated with quetiapine 40 mg/kg). Values are shown as means ± standard errors of the means.</p>
Full article ">Figure 2
<p>(<b>A</b>) Time spent in the quetiapine-paired chamber compared with the saline-paired chamber during pretest and post-test in the Quet-80 group (treated with quetiapine; 80 mg/kg). (<b>B</b>) Time spent in the quetiapine-paired chamber compared with the saline-paired chamber during pretest and post-test in the Quet-120 group (treated with quetiapine; 120 mg/kg). Values are shown as means ± standard errors of the means. *<span class="html-italic">p</span> &lt; 0.05 compared with the saline-paired chamber.</p>
Full article ">Figure 3
<p>(<b>A</b>) Time spent in the D1 antagonist-paired chamber compared with the saline-paired chamber during pretest and post-test in the SKF-V group (treated with SKF followed by vehicle). (<b>B</b>) Time spent in the quetiapine-paired chamber compared with the saline-paired chamber during pretest and post-test in the SKF-Quet group (treated with SKF followed by quetiapine). Values are shown as means ± standard errors of the means.</p>
Full article ">Figure 4
<p>Experimental schedule of the conditioned place preference (CPP) experiments.</p>
Full article ">
18 pages, 3040 KiB  
Article
Synthesis, Characterization, and Biological Evaluation of New Derivatives Targeting MbtI as Antitubercular Agents
by Matteo Mori, Giovanni Stelitano, Laurent R. Chiarelli, Giulia Cazzaniga, Arianna Gelain, Daniela Barlocco, Elena Pini, Fiorella Meneghetti and Stefania Villa
Pharmaceuticals 2021, 14(2), 155; https://doi.org/10.3390/ph14020155 - 13 Feb 2021
Cited by 24 | Viewed by 4060
Abstract
Tuberculosis (TB) causes millions of deaths every year, ranking as one of the most dangerous infectious diseases worldwide. Because several pathogenic strains of Mycobacterium tuberculosis (Mtb) have developed resistance against most of the established anti-TB drugs, new therapeutic options are urgently needed. An [...] Read more.
Tuberculosis (TB) causes millions of deaths every year, ranking as one of the most dangerous infectious diseases worldwide. Because several pathogenic strains of Mycobacterium tuberculosis (Mtb) have developed resistance against most of the established anti-TB drugs, new therapeutic options are urgently needed. An attractive target for the development of new antitubercular agents is the salicylate synthase MbtI, an essential enzyme for the mycobacterial siderophore biochemical machinery, absent in human cells. A set of analogues of I and II, two of the most potent MbtI inhibitors identified to date, was synthesized, characterized, and tested to elucidate the structural requirements for achieving an efficient MbtI inhibition and a potent antitubercular activity with this class of compounds. The structure-activity relationships (SAR) here discussed evidenced the importance of the furan as part of the pharmacophore and led to the preparation of six new compounds (IVIX), which gave us the opportunity to examine a hitherto unexplored position of the phenyl ring. Among them emerged 5-(3-cyano-5-(trifluoromethyl)phenyl)furan-2-carboxylic acid (IV), endowed with comparable inhibitory properties to the previous leads, but a better antitubercular activity, which is a key issue in MbtI inhibitor research. Therefore, compound IV offers promising prospects for future studies on the development of novel agents against mycobacterial infections. Full article
(This article belongs to the Special Issue Novel Antibacterial Agents)
Show Figures

Figure 1

Figure 1
<p>Reactions catalyzed by MbtI.</p>
Full article ">Figure 2
<p>Chemical structure of the lead compounds <b>I</b>, <b>II</b>, and <b>III</b>.</p>
Full article ">Figure 3
<p>Chemical structure of the heterocyclic cores tested in this study: <b>1</b> (thiophene), <b>2</b> (thiazole), <b>3</b> (oxazole), <b>4</b> (imidazole), <b>5</b> (1,3,4-oxadiazole), <b>6</b> (1,2,3-triazole).</p>
Full article ">Figure 4
<p>Chemical structure of compounds <b>IV</b>–<b>IX</b>.</p>
Full article ">Figure 5
<p>Biological characterization of <b>IV</b>. (<b>A</b>) IC<sub>50</sub> determination of <b>IV</b> against MbtI activity. (<b>B</b>) Global reciprocal plot of data from MbtI steady-state kinetics analysis towards chorismic acid, in the presence of different concentrations of <b>IV</b> (50, 20, 10, 5, 1, and 0 μM). (<b>C</b>) MIC<sup>99</sup> determination of <b>IV</b> against <span class="html-italic">M. bovis</span> BCG growth.</p>
Full article ">Scheme 1
<p>Synthetic procedure for the preparation of <b>1a,b</b>. Reagents and conditions: (<b>a</b>) MeOH, conc. H<sub>2</sub>SO<sub>4</sub>, reflux, overnight; (<b>b</b>) Pd(PPh<sub>3</sub>)<sub>2</sub>Cl<sub>2</sub>, 2 M Na<sub>2</sub>CO<sub>3</sub>, dry 1,4-dioxane, 90 °C, overnight, N<sub>2</sub> atm; (<b>c</b>) <span class="html-italic">1.</span> LiOH, THF-H<sub>2</sub>O 2:1, r.t., 2 h for <b>1a</b>; 1 M NaOH, EtOH-THF 1:1, reflux, 5 h for <b>1b</b>; <span class="html-italic">2.</span> 1 M HCl, 0 °C.</p>
Full article ">Scheme 2
<p>Synthetic procedure for the preparation of <b>2a,b</b>. Reagents and conditions: (<b>a</b>) NBS, <span class="html-italic">p</span>-TsOH, DCM, overnight, r.t, N<sub>2</sub> atm.; (<b>b</b>) <span class="html-italic">1</span>. hexamine, DCM, 8 h, r.t.; <span class="html-italic">2</span>. conc. HCl, EtOH, overnight, r.t.; (<b>c</b>) TEA, EtOAc, 3 h, reflux; (<b>d</b>) Lawesson’s reagent, 1,4-dioxane, 2 h, reflux; (<b>e</b>) NaOH, THF-H<sub>2</sub>O 1:1, 1.5 h, r.t.</p>
Full article ">Scheme 3
<p>Synthetic procedure for the preparation of <b>3a,b</b>. Reagents and conditions: (<b>a</b>) I<sub>2</sub>, DMSO, 3 h, 130 °C; (<b>b</b>) NaOH, THF-H<sub>2</sub>O 1:1, 1.5 h, r.t.</p>
Full article ">Scheme 4
<p>Synthetic procedure for the preparation of <b>4a,b</b>. Reagents and conditions: (<b>a</b>) SeO<sub>2</sub>, 1,4-dioxane/H<sub>2</sub>O, reflux, 7 h, N<sub>2</sub> atm; (<b>b</b>) NH<sub>4</sub>OAc, CH<sub>3</sub>CN, H<sub>2</sub>O, r.t., 2 h; (<b>c</b>) <span class="html-italic">1.</span> LiOH, THF-H<sub>2</sub>O 2:1, r.t., overnight for <b>4a</b>; NaOH, THF-H<sub>2</sub>O 1:1, reflux, 6 h for <b>4b</b>; <span class="html-italic">2.</span> 3 M HCl, 0 °C.</p>
Full article ">Scheme 5
<p>(<b>A</b>) Synthetic procedure for the preparation of <b>5a</b>. Reagents and conditions: (<b>a</b>) dry MeOH, conc. H<sub>2</sub>SO<sub>4</sub>, reflux, 3 h, N<sub>2</sub> atm; (<b>b</b>) NH<sub>2</sub>NH<sub>2</sub>∙H<sub>2</sub>O, MeOH, r.t., overnight; (<b>c</b>) TEA, DCM, r.t., 2 h; (<b>d</b>) TEA, DCM, TsCl, r.t., 2 h; (<b>e</b>) <span class="html-italic">1.</span> NaOH, THF-H<sub>2</sub>O 1:1, r.t., 1 h; <span class="html-italic">2.</span> Amberlite IR120, 0 °C. (<b>B</b>) Synthetic procedure for the preparation of <b>5b</b>. Reagents and conditions: (<b>a</b>) EtOH, conc. H<sub>2</sub>SO<sub>4</sub>, reflux, overnight; (<b>b</b>) NH<sub>2</sub>NH<sub>2</sub>∙H<sub>2</sub>O<sub>,</sub> EtOH, reflux, overnight; (<b>c</b>) 86% PPA, 120-130 °C, 1.5 h; (<b>d</b>) <span class="html-italic">1.</span> LiOH, THF-H<sub>2</sub>O 1:1, r.t., 1 h; <span class="html-italic">2.</span> Amberlite IR120, 0 °C.</p>
Full article ">Scheme 6
<p>Synthetic procedure for the preparation of <b>6a,b</b>. Reagents and conditions: (<b>a</b>) <span class="html-italic">1</span>. NaN<sub>3</sub>, Cu(OAc)<sub>2</sub>, MeOH, 55 °C, 1.5-4 h, N<sub>2</sub> atm; <span class="html-italic">2.</span> ethyl propiolate, (+)-sodium <span class="html-small-caps">l</span>-ascorbate, r.t., overnight-24 h; (<b>b</b>) <span class="html-italic">1</span>. LiOH, THF-H<sub>2</sub>O 2:1, r.t., 1 h for <b>6a</b>; NaOH, THF-H<sub>2</sub>O 1:1, reflux, 5 h for <b>6b</b>; <span class="html-italic">2.</span> 3 M HCl, 0 °C for <b>6a</b>; 1 M HCl, 0 °C for <b>6b</b>.</p>
Full article ">
12 pages, 304 KiB  
Review
The Treatment of Lung Involvement in Systemic Sclerosis
by Barbara Ruaro, Marco Confalonieri, Marco Matucci-Cerinic, Francesco Salton, Paola Confalonieri, Mario Santagiuliana, Gloria Maria Citton, Elisa Baratella and Cosimo Bruni
Pharmaceuticals 2021, 14(2), 154; https://doi.org/10.3390/ph14020154 - 13 Feb 2021
Cited by 17 | Viewed by 3394
Abstract
Systemic sclerosis (SSc) patients are often affected by interstitial lung disease (ILD) and, although there have been recent treatment advances, it remains the leading cause of death among SSc, with a 10-year mortality up to 40%. African Americans and subjects with diffuse cutaneous [...] Read more.
Systemic sclerosis (SSc) patients are often affected by interstitial lung disease (ILD) and, although there have been recent treatment advances, it remains the leading cause of death among SSc, with a 10-year mortality up to 40%. African Americans and subjects with diffuse cutaneous SSc or anti-topoisomerase 1 antibodies are most commonly affected. Currently, early ILD diagnosis can be made, and it is pivotal to improve the prognosis. The diagnostic mainstay test for SSc-ILD is high-resolution computed tomography for the morphology and pulmonary function tests for the functional aspects. Treatment planning and intensity are guided by the disease severity and risk of progression. Traditionally, therapy has depended on combinations of immunosuppressants, particularly cyclophosphamide and mycophenolate mofetil, which can be supplemented by targeted biological and antifibrotic therapies. Benefits have been observed in trials on hematopoietic autologous stem cell transplantation for patients with progressive SSc, whilst lung transplantation is reserved for refractory SSc-ILD cases. Herein, recent advances in SSc-ILD treatment will be explored. Full article
17 pages, 3619 KiB  
Article
Pharyngeal Pumping and Tissue-Specific Transgenic P-Glycoprotein Expression Influence Macrocyclic Lactone Susceptibility in Caenorhabditis elegans
by Alexander P. Gerhard, Jürgen Krücken, Cedric Neveu, Claude L. Charvet, Abdallah Harmache and Georg von Samson-Himmelstjerna
Pharmaceuticals 2021, 14(2), 153; https://doi.org/10.3390/ph14020153 - 13 Feb 2021
Cited by 13 | Viewed by 3522
Abstract
Macrocyclic lactones (MLs) are widely used drugs to treat and prevent parasitic nematode infections. In many nematode species including a major pathogen of foals, Parascaris univalens, resistance against MLs is widespread, but the underlying resistance mechanisms and ML penetration routes into nematodes [...] Read more.
Macrocyclic lactones (MLs) are widely used drugs to treat and prevent parasitic nematode infections. In many nematode species including a major pathogen of foals, Parascaris univalens, resistance against MLs is widespread, but the underlying resistance mechanisms and ML penetration routes into nematodes remain unknown. Here, we examined how the P-glycoprotein efflux pumps, candidate genes for ML resistance, can modulate drug susceptibility and investigated the role of active drug ingestion for ML susceptibility in the model nematode Caenorhabditis elegans. Wildtype or transgenic worms, modified to overexpress P. univalens PGP-9 (Pun-PGP-9) at the intestine or epidermis, were incubated with ivermectin or moxidectin in the presence (bacteria or serotonin) or absence (no specific stimulus) of pharyngeal pumping (PP). Active drug ingestion by PP was identified as an important factor for ivermectin susceptibility, while moxidectin susceptibility was only moderately affected. Intestinal Pun-PGP-9 expression elicited a protective effect against ivermectin and moxidectin only in the presence of PP stimulation. Conversely, epidermal Pun-PGP-9 expression protected against moxidectin regardless of PP and against ivermectin only in the absence of active drug ingestion. Our results demonstrate the role of active drug ingestion by nematodes for susceptibility and provide functional evidence for the contribution of P-glycoproteins to ML resistance in a tissue-specific manner. Full article
(This article belongs to the Special Issue Antiparasitics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Tissue-specific expression of <span class="html-italic">Pun</span>-PGP-9 in <span class="html-italic">Cel-pgp-9</span> loss-of-function strain, strain tm830. Verification of transcription and tissue-specific expression. (<b>a</b>) Visible bands for all reverse-transcriptase (RT) PCRs on cDNA templates made from whole-worm total RNA, and no bands for no RT controls and the control strain. (<b>b</b>–<b>e</b>) Confocal microscope images of tissue-specific expression patterns of fixed, freeze cracked, and immunofluorescence-stained transgenic adult <span class="html-italic">Caenorhabditis elegans</span> (blue lookup table (LUT), 405 nM excitation), pharyngeal GFP expression (green LUT, 488 nm excitation) and merged images (DIC +405 nM + 488 nM). Primary antibodies target the FLAG-tag fused to <span class="html-italic">Pun</span>-PGP-9 and the secondary antibody is conjugated with DyLight405. Images were acquired with a confocal Eclipse Ti-U inverted research microscope and processed and merged with ImageJ [<a href="#B40-pharmaceuticals-14-00153" class="html-bibr">40</a>]. All scalebars are 100 µm. (<b>b</b>) Immunostaining in the intestine-Pgp-9 line 1 (white arrows indicate the intestine) and GFP expression at the pharynx. (<b>c</b>) Immunostaining of the epidermis-Pgp-9 strain (white arrows indicate the epidermal syncytium) and pharyngeal GFP expression. (<b>d</b>) Immunostaining in the control strain and pharyngeal GFP expression. (<b>e</b>) Higher magnification of the intestine-Pgp-9 strain (white arrow indicates the apical membrane), <span class="html-italic">Pun</span>-PGP-9: <span class="html-italic">Parascaris univalens</span> P-glycoprotein-9, GFP: green fluorescence protein, DIC: differential interference contrast, int: intestine, vul: vulva, hyp7: hyp7 syncytium, N2∆<span class="html-italic">Cel</span>Pgp<span class="html-italic">-9</span> is tm830 (NBRP) [<span class="html-italic">Cel-pgp-9(-)</span>]; Transgenic strains genotypes: Epidermis-Pgp-9 <span class="html-italic">EpiPgp-9Ex1</span> [<span class="html-italic">Cel-pgp-9(-); Cel-col-19p::Pun-pgp-9::FLAG::Cel-unc-54_3′-UTR; Cel-myo-2p::gfp:: Cel-unc-54_3′UTR</span>]; Intestine-Pgp-9 Line 1 <span class="html-italic">IntPgp-9Ex1</span> [<span class="html-italic">Cel-pgp-9(-); Cel-ges-1p::Pun-pgp-9::FLAG::Celunc-54_3′-UTR; Cel-myo-2p::gfp::Cel-unc-54_3′UTR</span>]; Control strain <span class="html-italic">CtrlEx1</span> [<span class="html-italic">Cel-pgp-9(-); Cel-myo-2p::gfp:: Cel-unc-54_3′-UTR</span>].</p>
Full article ">Figure 2
<p>Pharyngeal pumping increases ivermectin and moxidectin susceptibility. Effect of pharyngeal pumping (PP) stimulation by OP50 food bacteria or serotonin in <span class="html-italic">Caenorhabditis elegans</span> on ivermectin and moxidectin susceptibility. (<b>a</b>) Schematic illustration of the experimental setup with active ingestion and intestine drug exposure by PP stimulation. (<b>b</b>) Mean thrashes/minute ± standard error of the mean (SEM) in the negative control (no drug, 1% DMSO) between strains and conditions. Each strain/condition combination was compared to WT OP50<sup>−</sup> with a Kruskal–Wallis test with Dunn’s post hoc, and <span class="html-italic">p</span> &gt; 0.05 was considered not significant. (<b>c</b>,<b>d</b>) Ivermectin and moxidectin concentration–response curves calculated with GraphPad v8.3.0 based on thrashes/minute in the WT strain with n = 36 per concentration spread equally on three separate days. PP stimulation by OP50 bacteria (OP50<sup>+</sup>) (black), 4 mM 5-HT (red), or in the absence of a PP stimulus (OP50<sup>−</sup>) (blue). Significant differences between half-maximal effective concentration (EC<sub>50</sub>) were calculated using the extra-sum-of-squares-F test and Bonferroni correction. (<b>e</b>) Comparison of the effect of PP stimulation by OP50 <span class="html-italic">Escherichia coli</span> food bacteria (red) or 5-HT (black) in different transgenic and wildtype strains. (<b>c</b>–<b>e</b>) All calculated four parameter non-linear regression models and SEM at each concentration correspond to <a href="#app1-pharmaceuticals-14-00153" class="html-app">Supplementary Tables S1 and S2</a>. Prior to the calculation, the no-drug negative control was set to 0.1 nM and all concentrations were log<sub>10</sub> transformed. On the x-axis, the negative control was visualised as “0 M (no drug)” and separated by a break in the axis. <span class="html-italic">P</span>-values &lt; 0.05 were considered significant and are indicated with an asterisk, while corresponding fold-changes are indicated with arrows. 5-HT: serotonin/5-hydroxytryptamine; WT: <span class="html-italic">C. elegans</span> N2 Bristol; Control strain genotype <span class="html-italic">CtrlEx1</span> [<span class="html-italic">Cel-pgp-9(-); Cel-myo-2p::gfp:: Cel-unc-54_3′-UTR</span>].</p>
Full article ">Figure 3
<p>Modulation of ivermectin and moxidectin susceptibility in <span class="html-italic">C. elegans</span> by transgenic tissue-specific <span class="html-italic">Pun</span>-PGP-9 expression. (<b>a</b>–<b>d</b>) Concentration–response curves corresponding to <a href="#app1-pharmaceuticals-14-00153" class="html-app">Tables S1 and S2</a> for ivermectin (<b>a</b>/<b>c</b>) and moxidectin (<b>b</b>/<b>d</b>) in the absence of a PP stimulus (OP50<sup>−</sup>) or PP stimulation by serotonin (5-HT<sup>+</sup>/OP50<sup>-</sup>) in liquid S-medium. (<b>a</b>–<b>d</b>) show the control strain’s response for 5-HT<sup>+</sup> (black dashed line with triangles) or OP50<sup>−</sup> (grey dashed line with circles), (<b>a</b>,<b>b</b>) show the epidermis-Pgp-9 strain for 5-HT<sup>+</sup> (dark blue with open squares) or OP50<sup>−</sup> (orange with rectangles) and (<b>c</b>,<b>d</b>) show the intestine-Pgp-9 line 1 for 5-HT<sup>+</sup> (light blue with open squares) or OP50<sup>−</sup> (red with rectangles). All concentration–response curves are based on four-parameter non-linear regression models calculated from the motility response (thrashes/minute) of 36 synchronised 1-day adults per concentration, and error bars represent standard error of the mean. Prior to calculation, concentrations were log<sub>10</sub> transformed, and the no-drug negative control was set to 0.1 nM. On the x-axis, the negative control was visualised as “0 M (no drug)” and separated by a break in the axis. Significant differences in EC<sub>50</sub> values were compared using the extra-sum-of-squares-F test and Bonferroni correction; <span class="html-italic">p</span>-values &lt; 0.05 were considered significant and indicated with an asterisk (*), while corresponding fold-changes are indicated with arrows. <span class="html-italic">Pun</span>-PGP-9: <span class="html-italic">Parascaris univalens</span> P-glycoprotein-9; N2∆<span class="html-italic">Cel</span>Pgp-<span class="html-italic">9</span> is tm830 (NBRP) [<span class="html-italic">Cel-pgp-9(-)</span>]; Transgenic strains genotypes: Epidermis-Pgp-9 <span class="html-italic">EpiPgp-9Ex1</span> [<span class="html-italic">Cel-pgp-9(-); Cel-col-19p::Pun-pgp-9::FLAG::Cel-unc-54_3′-UTR; Cel-myo-2p::gfp:: Cel-unc-54_3′UTR</span>]; Intestine-Pgp-9 Line 1 <span class="html-italic">IntPgp-9Ex1</span> [<span class="html-italic">Cel-pgp-9(-); Cel-ges-1p::Pun-pgp-9::FLAG::Celunc-54_3′-UTR; Cel-myo-2p::gfp::Cel-unc-54_3′UTR</span>]; Control strain <span class="html-italic">CtrlEx1</span> [<span class="html-italic">Cel-pgp-9(-); Cel-myo-2p::gfp:: Cel-unc-54_3′-UTR</span>].</p>
Full article ">Figure 4
<p>Comparison of moxidectin and ivermectin in wildtype and transgenic <span class="html-italic">Caenorhabditis elegans</span> strains. Forrest plot visualising the EC<sub>50</sub> and corresponding 95% confidence intervals for ivermectin (red) and moxidectin (turquoise) in transgenic and wildtype <span class="html-italic">Caenorhabditis elegans</span> strains in the presence of pharyngeal pumping (PP) stimulation by OP50 food bacteria (OP50<sup>+</sup>) or 5-hydroxytryptamine (5-HT<sup>+</sup>) or no PP stimulation (OP50<sup>−</sup>) were visualised using ggplot2 [<a href="#B42-pharmaceuticals-14-00153" class="html-bibr">42</a>] in R v4.0.3 [<a href="#B43-pharmaceuticals-14-00153" class="html-bibr">43</a>]. EC<sub>50</sub> values were inferred from four-parameter linear regression models calculated from 36 synchronised 1-day adult worms per concentration, strain, and condition using GraphPad v8.3.0. Worms were incubated for 24 h in S-medium containing a concentration series of ivermectin or moxidectin in a final DMSO concentration of 1%. Epidermis-Pgp-9 genotype is <span class="html-italic">EpiPgp-9Ex1</span> [<span class="html-italic">Cel-pgp-9(-); Cel-col-19p::Pun-pgp-9::FLAG::Cel-unc-54_3′-UTR; Cel-myo-2p::gfp:: Cel-unc-54_3′UTR</span>];. Intestine Pgp-9 Line 1 (L1) and Line 2 (L2) genotype is <span class="html-italic">IntPgp-9Ex1 or 2</span> [<span class="html-italic">Cel-pgp-9(-); Cel-ges-1p::Pun-pgp-9::FLAG::Celunc-54_3′-UTR; Cel-myo-2p::gfp::Cel-unc-54_3′UTR</span>];. Control strain genotype is <span class="html-italic">CtrlEx1</span> [<span class="html-italic">Cel-pgp-9(-); Cel-myo-2p::gfp::Cel-unc-54_3′-UTR</span>]; WT is N2 Bristol. Pgp: P-glycoprotein; <span class="html-italic">Pun</span>-PGP-9: <span class="html-italic">Parascaris univalens</span> P-glycoprotein-9; IVM: ivermectin; MOX: moxidectin; PP: pharyngeal pumping; WT: wildtype.</p>
Full article ">Figure 5
<p>Schematic illustration of P-glycoprotein-mediated barrier function. Hypothetical schematic illustration of Pgp-mediated barrier function in a <span class="html-italic">Caenorhabditis elegans</span> adult. Expression of P-glycoproteins in specific barrier tissues, i.e., the epidermis and the intestine prohibit MLs from reaching target tissues, thereby preventing an ML-induced hyperpolarisation of the neurons and muscle paralysis.</p>
Full article ">
14 pages, 824 KiB  
Review
Therapeutic Applications of Type 2 Diabetes Mellitus Drug Metformin in Patients with Osteoarthritis
by Parkyong Song, Ji Sun Hwang, Hyean Cheal Park, Keun Ki Kim, Hong-Joo Son, Yu-Jin Kim and Kwang Min Lee
Pharmaceuticals 2021, 14(2), 152; https://doi.org/10.3390/ph14020152 - 13 Feb 2021
Cited by 13 | Viewed by 3657
Abstract
Type 2 diabetes mellitus (T2DM) and osteoarthritis (OA) are common chronic diseases that frequently co-exist. The link between OA and T2DM is attributed to common risk factors, including age and obesity. Several reports suggest that hyperglycemia and accumulated advanced glycosylation end-products might regulate [...] Read more.
Type 2 diabetes mellitus (T2DM) and osteoarthritis (OA) are common chronic diseases that frequently co-exist. The link between OA and T2DM is attributed to common risk factors, including age and obesity. Several reports suggest that hyperglycemia and accumulated advanced glycosylation end-products might regulate cartilage homeostasis and contribute to the development and progression of OA. Metformin is used widely as the first-line treatment for T2DM. The drug acts by regulating glucose levels and improving insulin sensitivity. The anti-diabetic effects of metformin are mediated mainly via activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK), which is an energy sensing enzyme activated directly by an increase in the AMP/ATP ratio under conditions of metabolic stress. Dysregulation of AMPK is strongly associated with development of T2DM and metabolic syndrome. In this review, we discuss common risk factors, the association between OA and T2DM, and the role of AMPK. We also address the adaptive use of metformin, a known AMPK activator, as a new drug for treatment of patients with OA and T2DM. Full article
(This article belongs to the Special Issue Drug and Therapy for Osteoarthritis (OA))
Show Figures

Figure 1

Figure 1
<p>A schematic of common risk factors for osteoarthritis (OA) and type 2 diabetes mellitus (T2DM).</p>
Full article ">Figure 2
<p>Structure of metformin.</p>
Full article ">Figure 3
<p>Simplified schematic of therapeutic applications of metformin for OA and T2DM.</p>
Full article ">
18 pages, 982 KiB  
Review
Immune Checkpoint Inhibition in Oesophago-Gastric Carcinoma
by Anica Högner and Peter Thuss-Patience
Pharmaceuticals 2021, 14(2), 151; https://doi.org/10.3390/ph14020151 - 12 Feb 2021
Cited by 18 | Viewed by 5126
Abstract
Immune checkpoint inhibitors enrich the therapeutic landscape in oesophago-gastric carcinoma. With regard to oesophageal squamous cell carcinoma (ESCC), the selective PD-1 (programmed cell death receptor 1)-inhibitor nivolumab improves disease-free survival in the adjuvant therapy setting (CHECKMATE-577). In first-line treatment, ESCC patients (pts) benefit [...] Read more.
Immune checkpoint inhibitors enrich the therapeutic landscape in oesophago-gastric carcinoma. With regard to oesophageal squamous cell carcinoma (ESCC), the selective PD-1 (programmed cell death receptor 1)-inhibitor nivolumab improves disease-free survival in the adjuvant therapy setting (CHECKMATE-577). In first-line treatment, ESCC patients (pts) benefit in overall survival (OS) from the PD-1-inhibitor pembrolizumab in combination with chemotherapy (KEYNOTE-590). In the second-line setting, nivolumab (ATTRACTION-03) and pembrolizumab (KEYNOTE-181) demonstrate a benefit in OS compared with chemotherapy. These data resulted in the approval of nivolumab for the second-line treatment of advanced ESCC pts regardless of PD-L1 (programmed cell death ligand 1) status in Europe, Asia, and the USA, and pembrolizumab for pts with PD-L1 CPS (combined positivity score) ≥ 10 in Asia and the USA. Further approvals can be expected. In gastro-oesophageal junction and gastric cancer, the addition of nivolumab to chemotherapy in first-line treatment improves OS in pts with advanced disease with PD-L1 CPS ≥ 5 (CHECKMATE-649). Additionally, pembrolizumab was non-inferior to chemotherapy for OS in PD-L1 CPS ≥ 1 pts (KEYNOTE-062). In third-line treatment, nivolumab shows benefits in OS regardless of PD-L1 expression (ATTRACTION-02) with approval in Asia, and pembrolizumab prolonged the duration of response in PD-L1 positive pts (KEYNOTE-059) with approval in the USA. We discuss the recent results of the completed phase II and III clinical trials. Full article
(This article belongs to the Special Issue Immune Checkpoint Inhibitor Therapy)
Show Figures

Figure 1

Figure 1
<p>Functional interaction of immune system and tumour cells in checkpoint inhibition (adapted from Taieb et al., 2018 [<a href="#B13-pharmaceuticals-14-00151" class="html-bibr">13</a>].) Blocking checkpoint proteins, PD-1 (programmed death receptor-1), PD-L1 (programmed death receptor ligand-1), CTLA-4 (anti-cytotoxic T-lymphocyte-associated antigen 4), amplify T-cell immune response against tumour cells by blocking inhibitory signals of tumour cells. NK—natural killer, FcyR—Fc receptors for IgG, ADCC—antibody-dependent cellular cytotoxicity, MHC I/II—major histocompatibility complex, TCR—T-cell receptor.</p>
Full article ">Figure 2
<p>Overview of curative therapy regimes of oesophageal and gastric cancer.</p>
Full article ">Figure 3
<p>Overview of a possible algorithm for palliative therapy regimes in oesophageal cancer.</p>
Full article ">Figure 4
<p>Overview of a possible algorithm for palliative therapy regimes in gastric cancer.</p>
Full article ">
13 pages, 379 KiB  
Article
Assessment of Novel Inhaler Technique Reminder Labels in Image Format on the Correct Demonstration of Inhaler Technique Skills in Asthma: A Single-Blinded Randomized Controlled Trial
by Iman Basheti, Bassam Mahboub, Laila Salameh, Mena Al-Ani, Ammar Abdulrahman Jairoun, Basema Saddik and Eman Abu-Gharbieh
Pharmaceuticals 2021, 14(2), 150; https://doi.org/10.3390/ph14020150 - 12 Feb 2021
Cited by 4 | Viewed by 2268
Abstract
Background: Prevalence of asthma in the United Arab Emirates (UAE) is high, and training patients on correct inhaler technique is vital. Objectives: To assess the effectiveness of inhaler technique labels incorporating the individual technique steps in image format on the retention of correct [...] Read more.
Background: Prevalence of asthma in the United Arab Emirates (UAE) is high, and training patients on correct inhaler technique is vital. Objectives: To assess the effectiveness of inhaler technique labels incorporating the individual technique steps in image format on the retention of correct inhaler technique for patients with asthma living in the UAE and following inhaler training; secondly to investigate the effect of inhaler technique education using self-check pictorial labels on patients’ overall asthma control. Methods: This single-blinded randomized controlled study was conducted in 2019 and followed consecutive recruitment of asthma patients visiting respiratory clinics at Rashid Hospital in Dubai. Patients were using a controller inhaler (Turbuhaler (TH), Accuhaler (ACC), or pressurized metered-dose inhaler (pMDI)). Following recruitment, patients were randomized into active group receiving educational intervention plus the inhaler label, and control group receiving educational intervention without the label. Patients were assessed at baseline and at one-month on their inhaler technique and asthma control. Results: Participants (n = 245; 93 = TH, 70 = ACC, 82 = pMDI) showed a significant difference between the groups at one-month for inhaler technique scores for TH (active 5.29 ± 1.86 vs. control = 24.4 ± 21.28), ACC (active = 3.99 ± 1.43 vs. control = 25.45 ± 22.57), and pMDI (active = 4.59 ± 0.10 vs. control = 120.55 ± 17.2), p < 0.001 for all. Asthma control for active group indicated significant improvements compared to control for TH and pMDI (p < 0.001 for both), but not ACC group (p = 0.087). Conclusions: Retention of correct inhaler technique and improved asthma control can be enhanced by using a specialized inhaler technique label in image format. Full article
Show Figures

Figure 1

Figure 1
<p>Consort diagram showing participants’ recruitment and retention during the study period.</p>
Full article ">
23 pages, 3860 KiB  
Review
The Role of miRNA-7 in the Biology of Cancer and Modulation of Drug Resistance
by Ewa Gajda, Małgorzata Grzanka, Marlena Godlewska and Damian Gawel
Pharmaceuticals 2021, 14(2), 149; https://doi.org/10.3390/ph14020149 - 12 Feb 2021
Cited by 16 | Viewed by 3409
Abstract
MicroRNAs (miRNAs, miRs) are small non-coding RNA (ncRNA) molecules capable of regulating post-transcriptional gene expression. Imbalances in the miRNA network have been associated with the development of many pathological conditions and diseases, including cancer. Recently, miRNAs have also been linked to the phenomenon [...] Read more.
MicroRNAs (miRNAs, miRs) are small non-coding RNA (ncRNA) molecules capable of regulating post-transcriptional gene expression. Imbalances in the miRNA network have been associated with the development of many pathological conditions and diseases, including cancer. Recently, miRNAs have also been linked to the phenomenon of multidrug resistance (MDR). MiR-7 is one of the extensively studied miRNAs and its role in cancer progression and MDR modulation has been highlighted. MiR-7 is engaged in multiple cellular pathways and acts as a tumor suppressor in the majority of human neoplasia. Its depletion limits the effectiveness of anti-cancer therapies, while its restoration sensitizes cells to the administered drugs. Therefore, miR-7 might be considered as a potential adjuvant agent, which can increase the efficiency of standard chemotherapeutics. Full article
(This article belongs to the Special Issue MiRNA-Based Therapeutics in Cancer)
Show Figures

Figure 1

Figure 1
<p>A graphical representation of the canonical pathway of miRNA biogenesis in animals. The <span class="html-italic">MIR</span> gene encodes pri-miRNA. Modification of the pri-miRNA hairpin with two free ends takes place in the cell nucleus. The DGCR8 (DiGeorge critical region 8) and Drosha enzymes cut off the free strands from the hairpin giving pre-miRNA. Then, pre-miRNA is transported into the cytoplasm by Exportin 5 (Exp5), where the Dicer and TRBP (TAR double-stranded RNA binding protein) complex removes the hairpin’s loop and cleaves the molecule into the miRNA duplex. One of the duplex strands, along with the RNA-induced silencing complex (RISC), is involved in mRNA targeting. The second one is degraded.</p>
Full article ">Figure 2
<p>Biogenesis of miR-7. (<b>A</b>) miR-7 is the result of transcription of the <span class="html-italic">MIR7-1</span>, <span class="html-italic">MIR7-2</span>, and <span class="html-italic">MIR7-3</span> genes located on chromosomes 9, 15, and 19, respectively. The generated pri-miRs are transformed into pre-miR-7. Exp5 transports them through the nuclear pores to the cytoplasm (<b>B</b>), where pre-miR-7-1, pre-miR-7-2, and pre-miR-7-3 (<b>C</b>) undergo further modifications. The Dicer complex, cleaves double-stranded RNA (dsRNA) into shorter nucleotide duplexes. Sequences marked in pink are involved in the regulation of mRNA expression.</p>
Full article ">Figure 3
<p>Tumor-related signaling pathways and proteins associated with depletion of miR-7.</p>
Full article ">
46 pages, 6710 KiB  
Review
Modulation of the Serotonergic Receptosome in the Treatment of Anxiety and Depression: A Narrative Review of the Experimental Evidence
by Gustavo R. Villas-Boas, Stefânia N. Lavorato, Marina M. Paes, Pablinny M. G. de Carvalho, Vanessa C. Rescia, Mila S. Cunha, Manoel F. de Magalhães-Filho, Luis F. Ponsoni, Adryano Augustto Valladao de Carvalho, Roseli B. de Lacerda, Lais da S. Leite, Matheus da S. Tavares-Henriques, Luiz A. F. Lopes, Luiz G. R. Oliveira, Saulo E. Silva-Filho, Ana P. S. da Silveira, Roberto K. N. Cuman, Francielli M. de S. Silva-Comar, Jurandir F. Comar, Luana do A. Brasileiro, Jussileide N. dos Santos, William R. de Freitas, Katyuscya V. Leão, Jonatas G. da Silva, Raphael C. Klein, Mary H. F. Klein, Bruno H. da S. Ramos, Cristiane K. C. Fernandes, Dayane G. de L. Ribas and Silvia A. Oesterreichadd Show full author list remove Hide full author list
Pharmaceuticals 2021, 14(2), 148; https://doi.org/10.3390/ph14020148 - 12 Feb 2021
Cited by 20 | Viewed by 6592
Abstract
Serotonin (5-HT) receptors are found throughout central and peripheral nervous systems, mainly in brain regions involved in the neurobiology of anxiety and depression. 5-HT receptors are currently promising targets for discovering new drugs for treating disorders ranging from migraine to neuropsychiatric upsets, such [...] Read more.
Serotonin (5-HT) receptors are found throughout central and peripheral nervous systems, mainly in brain regions involved in the neurobiology of anxiety and depression. 5-HT receptors are currently promising targets for discovering new drugs for treating disorders ranging from migraine to neuropsychiatric upsets, such as anxiety and depression. It is well described in the current literature that the brain expresses seven types of 5-HT receptors comprising eighteen distinct subtypes. In this article, we comprehensively reviewed 5-HT1-7 receptors. Of the eighteen 5-HT receptors known today, thirteen are G protein-coupled receptors (GPCRs) and represent targets for approximately 40% of drugs used in humans. Signaling pathways related to these receptors play a crucial role in neurodevelopment and can be modulated to develop effective therapies to treat anxiety and depression. This review presents the experimental evidence of the modulation of the “serotonergic receptosome” in the treatment of anxiety and depression, as well as demonstrating state-of-the-art research related to phytochemicals and these disorders. In addition, detailed aspects of the pharmacological mechanism of action of all currently known 5-HT receptor families were reviewed. From this review, it will be possible to direct the rational design of drugs towards new therapies that involve signaling via 5-HT receptors. Full article
(This article belongs to the Special Issue Molecular Pharmacology of 5-HT Receptors)
Show Figures

Figure 1

Figure 1
<p>Serotonin (5-HT<sub>1A</sub>) receptor signaling pathways: (<b>a</b>) The 5-HT<sub>1A</sub> receptor is coupled to the Gα<sub>i/0</sub> protein. Its activation blocks the adenylate cyclase activity (AC), reducing the conversion of adenosine triphosphate (ATP) into cyclic adenosine monophosphate (cAMP), which is responsible for the activation of protein kinase A (PKA). The activation of these receptors decreases the release of neurotransmitters in neurons through opposite changes in K<sup>+</sup> (increase) and Ca<sup>2+</sup> (decrease) conductances. In addition, stimulation of the 5-HT<sub>1A</sub> receptor regulates the phosphorylation of the extracellular signal-regulated kinase (ERK). The “atypical” coupling is represented by the orange circle and demonstrates the overexpressing cell Jurkat T-like cell line. The yellow rectangle represents a specific effect on the rat hippocampus, where stimulation of the 5-HT1A receptor reduced MEK1/2 and ERK and Elk-1 phosphorylation. This change in pERK levels was not seen in the cortex [<a href="#B12-pharmaceuticals-14-00148" class="html-bibr">12</a>]; (<b>b</b>) the activation of the 5-HT<sub>1A</sub> receptor also induces stimulation of the Janus kinase 2 protein (JAK2), which phosphorylates the calmodulin protein, which in turn is associated with the sodium and hydrogen 1 (NHE-1) transporter, activating it. The massive outlet of protons caused by the 5-HT<sub>1A</sub> activation culminates in an increase in intracellular pH. The numbers represent the amino acids (approximate region) of the cytoplasmic domain of the NHE1 transporter in which the other proteins interact.</p>
Full article ">Figure 2
<p>5-HT<sub>1B</sub> receptor signaling pathways. The 5-HT<sub>1B</sub> receptor is coupled to the Gα<sub>i/0</sub> protein. Its activation blocks the activity of adenylate cyclase (AC), reducing the conversion of adenosine triphosphate (ATP) into cyclic adenosine monophosphate (cAMP), which is responsible for the activation of protein kinase A (PKA). The activation of these receptors decreases the release of neurotransmitters in neurons through opposite changes in K<sup>+</sup> (increase) and Ca<sup>2+</sup> (decrease) conductances. After 5-HT<sub>1B</sub> activation, a cascade of kinases regulates the translocation of extracellular signal regulated kinase (ERK) (transcription activation) [<a href="#B12-pharmaceuticals-14-00148" class="html-bibr">12</a>]. In addition, kinase B protein (AKT) is stimulated with consequent activation of glycogen synthase kinase 3 (GSK3), which is involved with phosphorylation and regulation of the 5-HT<sub>1B</sub> receptor activity [<a href="#B16-pharmaceuticals-14-00148" class="html-bibr">16</a>].</p>
Full article ">Figure 3
<p>Anxiolytic mechanism of tandospirone. 5-HT<sub>1A</sub>: (<b>a</b>) Biological factors have been implicated in the pathogenesis of anxiety. Neurochemical evidence suggests that the dysregulation of serotonin, norepinephrine, gamma-aminobutyric acid (GABA), glutamate, and peptides (corticotropin releasing factor, cholecystokinin, neuropeptide Y) transmission systems are strongly correlated with the pathophysiology of anxiety. In the classic concept of stress, homeostasis is threatened by physical and psychological events, known as stressors. Behavior is aimed at assessing the destabilizing potential of the stressor. If the stressor event does not correspond to any cognitive representation based on previous subjective experiences, there is an increase in alertness, hypervigilance, focused attention, and cognitive processing. The interface between sensory information received and the evaluation process is formed in the limbic region of the brain, which comprises structures such as the hippocampus, the amygdala, and the medial prefrontal cortex (mPFC). The components of the human fear circuit, which can be of particular relevance for destabilization produced by the stressor and, consequently, induction of anxiety, include mPFC (subservient fear extinction processes), the amygdala (risk assessment processing), and the hippocampus. The ascending serotonergic pathway that projects from the DRN into the hippocampus may be implicated in the genesis of behavioral inhibition observed in dangerous situations. During episodes of anxiety crisis, it is postulated that these serotonergic inputs in the hippocampus activate 5-HT<sub>1A</sub> receptors in GABAergic interneurons. In this case, 5-HT<sub>1A</sub> receptors coupled to the Gαi/0 protein block adenylate cyclase (AC), prevent the formation of cyclic adenosine monophosphate (cAMP) and reduce the activation of protein kinase A (PKA) which, once reduced, decrease the permeability of calcium (Ca<sup>2+</sup>) to specific channels located in the cell membrane. This reduction in the calcium influx into the GABAergic interneuron culminates in the blocking the exocytosis of the GABA neurotransmitter. The reduction of the hippocampal GABAergic transmission causes an increase in the number of post-synaptic pyramidal glutamatergic neurons fires, stimulating anxiety-related regions such as the amygdala and the DRN, which feeds back and over-stimulates the anxiety-related circuit; (<b>b</b>,<b>c</b>); tandospirone has a promising anxiolytic effect, which has been shown in animal models, especially of the generalized anxiety disorder (GAD). Tandospirone acts as an anxiolytic by activating the post-synaptic 5-HT<sub>1A</sub> receptor coupled to the Gαi/0protein, resulting in reduced cAMP formation and PKA inhibition. On the other hand, it activates the G protein-controlled internal rectifying potassium channels (GIRK) by releasing Gβγ subunits, leading to intracellular potassium (K<sup>+</sup>) efflux, hyperpolarization of target neurons and, finally, inhibition of the local neuronal activity [<a href="#B24-pharmaceuticals-14-00148" class="html-bibr">24</a>]; (<b>d</b>) another mechanism by which tandospirone exerts its anxiolytic effect is by increasing the release of dopamine (DA) in the VTA. In this case, tandospirone activates the 5-HT<sub>1A</sub> receptor in DRN or mPFC, directly or indirectly, stimulating dopaminergic transmission in VTA [<a href="#B24-pharmaceuticals-14-00148" class="html-bibr">24</a>].</p>
Full article ">Figure 4
<p>Hypothetical scheme of serotonergic projections resulting from the raphe and innervation of the dorsal horn spinal neurons (DHS) and limbic regions. In the dorsal raphe nucleus (DRN), 5-HT<sub>1A</sub> receptors activate G protein-controlled internal rectifying potassium channels (GIRK) by releasing Gβγ subunits of the Gα<sub>i/0</sub> protein, leading to intracellular potassium (K<sup>+</sup>) efflux, hyperpolarization of target neurons, and, finally, inhibition of the local neuronal activity. Hippocampus and cortex heteroreceptors are also coupled to GIRK channels. Thus, activation of both 5-HT<sub>1A</sub> autoreceptors and the hippocampus and cortex heteroreceptors increases the GIRK current, leading to neuronal hyperpolarization. As shown in the figure, the activation of 5-HT<sub>1A</sub> autoreceptors reduces the release of serotonin (5-HT) in limbic regions or in DHS. However, the activation of 5-HT<sub>1A</sub> heteroreceptors located in the DHS reduces the release of local 5-HT, consequently decreasing the release of nociceptive neurotransmitters and reducing pain signals (antinociceptive effect). In limbic regions, the stimulation of these receptors activates GIRK in GABAergic interneurons, hyperpolarizing them and consequently decreasing the influence of GABA on local glutamatergic neurons. As a consequence, there is an increase in the excitatory glutamatergic influence on underlying dopaminergic neurons [<a href="#B37-pharmaceuticals-14-00148" class="html-bibr">37</a>].</p>
Full article ">Figure 5
<p>5-HT<sub>2A</sub> receptor signaling pathways: (<b>a</b>) In blood vessels (and other smooth muscles), stimulation of the 5-HT<sub>2A</sub> by 5-HT receptor activates several signal transduction pathways through the Gα<sub>q/11</sub> protein (phospholipase C (PLC)/Diacylglycerol (DAG)/protein kinase C (PKC)/calcium (Ca<sup>2+</sup>) and kinase phosphorylation regulated by extracellular signal (ERK), leading to vascular smooth muscle contraction. The “atypical” coupling represented by the green circle demonstrates the specific signaling of the tracheal tissue mediated by the 5-HT<sub>2A</sub> receptor, corroborating the hypothesis that the activity of these receptors, as well as the integration of their multiple pathways, varies from one tissue to another. In the trachea, activation of the 5-HT<sub>2A</sub> receptor produces its downstream effects primarily through the mammalian target of rapamycin (mTOR)/p70 ribosomal protein S6 kinase (S6K1) pathway [<a href="#B12-pharmaceuticals-14-00148" class="html-bibr">12</a>]. (<b>b</b>) In glutamatergic neurons of the medial prefrontal cortex (mPFC), the coupling of 5-HT to the 5-HT<sub>2A</sub> receptor stimulates the Gα<sub>q/11</sub> protein that activates several signal transduction pathways through PLC/DAG/AC/PKC. These, in turn, produce inhibition of Ca<sup>2+</sup> and Na<sup>+</sup> conductances. The “atypical” coupling represented by the blue circle demonstrates the activation of phospholipase A2 (PLA<sub>2</sub>) mediated by the 5-HT<sub>2A</sub> receptor and the subsequent release of arachidonic acid, described as a result of Gα<sub>12/13</sub>-coupled, Rho-mediated, p38 activation in NIH 3T3 cells [<a href="#B48-pharmaceuticals-14-00148" class="html-bibr">48</a>]. Stimulation of PLA<sub>2</sub> causes the release of arachidonic acid. The pathways of several kinases are involved in the modulation of neuron morphology and plasticity through direct interaction between the C terminal portion of the receptor and specific modulating proteins [<a href="#B12-pharmaceuticals-14-00148" class="html-bibr">12</a>].</p>
Full article ">Figure 6
<p>Scheme of pathways that stimulate extracellular signal-regulated kinase (ERK) and phosphorylate protein kinase B (AKT) after stimulation of the 5-HT<sub>2B</sub> receptor by fluoxetine in astrocytes. Fluoxetine binds and activates 5-HT<sub>2B</sub> receptors, culminating in the stimulation of Gα<sub>q/11</sub> protein, which stimulates enzyme phospholipase C (PLC), which catalyzes the hydrolysis of phosphatidylinositol-4,5-bisphosphate in inositol 1,4,5-triphosphate (IP3) and diacylglycerol (DAG). DAG induces protein kinase C (PKC) activity, while IP3 increases the intracellular calcium (Ca<sup>2+</sup>) concentration due to the release of Ca<sup>2+</sup> from endoplasmic reticulum stocks. Ca<sup>2+</sup> stimulates zinc-dependent metalloproteinases (MMPs) and leads to the release of growth factors. The released epithelial growth factor receptor (EGFR) ligand stimulates EGFR phosphorylation. ERK, the downstream target of EGFR, is phosphorylated via the Ras/Raf/MEK pathway, and AKT is phosphorylated via the phosphoinositide 3-kinase pathway (PI3K). In addition, PIK3 catalyzes the formation of PIP3 from PIP2. After fluoxetine administration, ERK and AKT phosphorylation was blocked when iRNA against the 5-HT<sub>2B</sub> receptor was administered or after administration of inhibitors of this receptor (SB204741) (shown in blue), PKC (GF109293X), intracellular Ca<sup>2+</sup> homeostasis (BAPTA/AM, an intracellular Ca<sup>2+</sup> chelator), zinc-dependent MMPs (GM6001), EGFR (AG1478), ERK phosphorylation (U0126, a mitogen-activated protein Kinase (MEK) inhibitor), or AKT pathway (LY294002, a PI3K inhibitor) [<a href="#B55-pharmaceuticals-14-00148" class="html-bibr">55</a>].</p>
Full article ">Figure 7
<p>5-HT<sub>2C</sub> receptor signaling. After activation of the 5-HT<sub>2C</sub> receptor by 5-HT, the Gα<sub>q/11</sub> protein is activated and subsequently stimulates phospholipase C (PLC) activity. PLC catalyzes the hydrolysis of phosphatidylinositol-4,5-bisphosphate into inositol 1,4,5-triphosphate (IP3) and diacylglycerol (DAG). IP3 diffuses through the cytoplasm and stimulates the release of calcium (Ca<sup>2+</sup>) from the endoplasmic reticulum and DAG activates protein kinase C (PKC), leading to the phosphorylation of various cellular substrates, which in turn activate several intracellular biochemical cascades. The activation of the 5-HT<sub>2C</sub> receptor can also stimulate the activity of enzyme phospholipase A<sub>2</sub> (PLA<sub>2</sub>), which is responsible for the synthesis of arachidonic acid (causing its release in the intracellular space). The “atypical” coupling represented by the orange circle demonstrates that through the coupling of the Gα<sub>12/13</sub> protein, the activation of the 5-HT<sub>2C</sub> receptor stimulates phospholipase D (PLD), whose activity converges to PKC activation, which subsequently activates extracellular signal-regulated kinases 1 and 2 (ERK1/2). Another ERK activation pathway is provided through Gα<sub>i/0</sub>, which induces the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/glycogen synthase kinase-3β (GSK-3β) cascade. Finally, this signal transduction system controls genetic transcription [<a href="#B58-pharmaceuticals-14-00148" class="html-bibr">58</a>]. The β-Arrestin protein interacts with 5-HT<sub>2C</sub> receptors and creates a steric impediment that blocks the coupling of heterotrimeric G proteins to the receptor, preventing the activation of Gα proteins.</p>
Full article ">Figure 8
<p>Structural formulas of phytochemicals cited in <a href="#pharmaceuticals-14-00148-t001" class="html-table">Table 1</a> and <a href="#pharmaceuticals-14-00148-t002" class="html-table">Table 2</a>.</p>
Full article ">
17 pages, 1448 KiB  
Article
Application of RP-18 TLC Retention Data to the Prediction of the Transdermal Absorption of Drugs
by Anna W. Sobańska, Jeremy Robertson and Elżbieta Brzezińska
Pharmaceuticals 2021, 14(2), 147; https://doi.org/10.3390/ph14020147 - 12 Feb 2021
Cited by 13 | Viewed by 2596
Abstract
Several chromatographic parameters (RM0 and S obtained from RP-18 TLC with methanol—pH 7.4 phosphate buffer mobile phases by extrapolation to zero concentration of methanol; Rf and RM obtained from RP-18 TLC with acetonitrile—pH 7.4 phosphate buffer 70:30 v [...] Read more.
Several chromatographic parameters (RM0 and S obtained from RP-18 TLC with methanol—pH 7.4 phosphate buffer mobile phases by extrapolation to zero concentration of methanol; Rf and RM obtained from RP-18 TLC with acetonitrile—pH 7.4 phosphate buffer 70:30 v/v as a mobile phase) and calculated molecular descriptors (molecular weight—MW; molar volume—VM; polar surface area—PSA; total count of nitrogen and oxygen atoms—(N+O); H-bond donor count—HD; H-bond acceptor count—HA; distribution coefficient—log D; total energy—ET; binding energy—Eb; hydration energy—Eh; energy of the highest occupied molecular orbital—EHOMO; energy of the lowest unoccupied orbital—ELUMO; electronic energy—Ee; surface area—Sa; octanol-water partition coefficient—log P; dipole moment—DM; refractivity—R, polarizability—α) and their combinations (Rf/PSA, RM/MW, RM/VM) were tested in order to generate useful models of solutes’ skin permeability coefficient log Kp. It was established that neither RM0 nor S obtained in the conditions used in this study is a good predictor of the skin permeability coefficient. The chromatographic parameters Rf and Rf/PSA were also unsuitable for this purpose. A simple and potentially useful, purely computational model based on (N+O), log D and HD as independent variables and accounting for ca. 83% of total variability was obtained. The evaluation of parameters derived from RM (RM, RM/MW, RM/VM) as independent variables in log Kp models proved that RM/VM is the most suitable descriptor belonging to this group. In a search for a reliable log Kp model based on this descriptor two possibilities were considered: a relatively simple model based on 5 independent variables: (N+O), log D, RM/VM, ET and Eh and a more complex one, involving also Eb, MW and PSA. Full article
Show Figures

Figure 1

Figure 1
<p>Scatterplots of log <b><span class="html-italic">K<sub>p</sub></span></b><sup>EPI</sup> vs. <b><span class="html-italic">S</span></b> and <b><span class="html-italic">R<sub>M</sub></span><sup>0</sup></b> (<span class="html-italic">n</span> = 14).</p>
Full article ">Figure 2
<p>Equation (5)—predicted vs. observed values.</p>
Full article ">Figure 3
<p>Equation (6)—predicted vs. observed values.</p>
Full article ">Figure 4
<p>Equation (8)—predicted vs. observed values.</p>
Full article ">Figure 5
<p>Equation (10), predicted vs. observed values.</p>
Full article ">
26 pages, 7174 KiB  
Article
Zingerone Targets Status Epilepticus by Blocking Hippocampal Neurodegeneration via Regulation of Redox Imbalance, Inflammation and Apoptosis
by Summya Rashid, Adil Farooq Wali, Shahzada Mudasir Rashid, Rana M. Alsaffar, Ajaz Ahmad, Basit L. Jan, Bilal Ahmad Paray, Saeed M. A. Alqahtani, Azher Arafah and Muneeb U. Rehman
Pharmaceuticals 2021, 14(2), 146; https://doi.org/10.3390/ph14020146 - 11 Feb 2021
Cited by 23 | Viewed by 3366
Abstract
Epilepsy is an intricate neurological disease where the neurons are severely affected, leading to the mortality of millions worldwide. Status epilepticus (SE), induced by lithium chloride (LiCl) and pilocarpine, is the most accepted model for epilepsy. The current work aims to unravel the [...] Read more.
Epilepsy is an intricate neurological disease where the neurons are severely affected, leading to the mortality of millions worldwide. Status epilepticus (SE), induced by lithium chloride (LiCl) and pilocarpine, is the most accepted model for epilepsy. The current work aims to unravel the mechanisms underlying the anti-epileptic efficacy of zingerone (an active ingredient of ginger), which has beneficial pharmacological activities on seizure-induced behavioral, histological, neurochemical, and molecular patterns in mice. Zingerone restored cognitive function by diminishing seizure activity, escape latency, and subsequent hippocampal damage manifested in histology. Seizures are associated with local inflammation, redox imbalance, and neural loss, confirmed by the present study of SE, and was attenuated by zingerone treatment. Nuclear factor-kappa B and its downstream signaling molecules (TNF-α, IL-1β, IL-6, NO, MPO) were activated in the LiCl-and-pilocarpine-induced group leading to inflammatory signaling, which was substantially ameliorated by zingerone treatment. The intrinsic apoptotic process was triggered subsequent to SE, as demonstrated by augmentation of cleaved caspase-3, downregulation of Bcl-2. However, zingerone treatment downregulated caspase-3 and upregulated Bcl-2, increasing cell survival and decreasing hippocampal neural death, deciphering involvement of apoptosis in SE. Therefore, zingerone plays an essential role in neuroprotection, probably by precluding oxidative stress, inflammation, and obstructing the mitochondrial pathway of apoptosis. Full article
(This article belongs to the Special Issue Epilepsy and Neurodegeneration: Current Therapeutic Implications 2021)
Show Figures

Figure 1

Figure 1
<p>Effect of zingerone on the cognitive function of LiCl-and-pilocarpine-induced SE in mice by Morris water maze. (<b>A</b>) Effect on escape latency (sec) on pilocarpine-induced status epilepticus. Results are representative of mean ± SE of fourteen mice per group. In group-II, the escape latency was increased significantly (*** <span class="html-italic">p</span> &lt; 0.001) as compared to control group (group I). Treatment with sodium valproate and zingerone (25 and 50 mg/kg b.w.) significantly attenuated escape latency level in group III (<sup>###</sup> <span class="html-italic">p</span> &lt; 0.001), group IV (<sup>##</sup> <span class="html-italic">p</span> &lt; 0.01), and group V (<sup>###</sup> <span class="html-italic">p</span> &lt; 0.001) as compared to group II. Group I: Normal saline (10 mL/kg b.w.), Group II: LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group III: Sodium Valproate (300 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group IV: zingerone (25 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group V: zingerone (50 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.) (<b>B</b>) Effect of zingerone on % time in target quadrant (seconds) on LiCl-and-pilocarpine-induced SE. Results are representative of mean ± SE of eight mice per group. The results that we got are significantly different from pilocarpine group as the main comparison is with pilocarpine group only (<sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001). Group I: Normal saline (10 mL/kg b.w.), Group II: LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group III: Sodium Valproate (300 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group IV: zingerone (25 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group V: zingerone (50 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.).</p>
Full article ">Figure 2
<p>Panel of graphs represents effects of zingerone on AChE (<b>a</b>), MPO (<b>b</b>), ROS (<b>c</b>), and NO (<b>d</b>) levels on LiCl-and-pilocarpine-induced SE. Values are significantly different in LiCl and pilocarpine group (*** <span class="html-italic">p</span> &lt; 0.001) as compared to control group. Results that we got are significantly different from LiCl and pilocarpine group as the main comparison is with LiCl and pilocarpine group only (<sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001). Group I: Normal saline (10 mL/kg b.w.), Group II: LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group III: Sodium Valproate (300 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group IV: zingerone (25 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group V: zingerone (50 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.)</p>
Full article ">Figure 3
<p>Zingerone effects on histomorphological features in LiCl-and-pilocarpine-induced SE. There is normal histology of group I (<b>A</b>,<b>B</b>) with intact and appropriately sized nuclei and neural cells. In group II (<b>C</b>,<b>D</b>) there is distortion of neurons along with neural condensation and necrosis, pyknotic nuclei in pilocarpine-administered rats as compared to negative control mice. Group V (<b>E</b>,<b>F</b>) treatment of zingerone (50 mg/kg) recovered the damage which was evident by retaining of normal histology and decrease in neural distortion and death. Values are expressed as mean ± SEM (<span class="html-italic">n</span> = 6). Photomicrographs of hippocampus depicting Hematoxylin and eosin staining analyses. Below photomicrographs is the panel which shows quantitative evaluation of neural loss. Significant difference was indicated by *** <span class="html-italic">p</span> &lt; 0.001 when compared with group I and (<sup>###</sup> <span class="html-italic">p</span> &lt; 0.001) when compared with group II. Zingerone treatment significantly protected neural loss in group V (<sup>###</sup> <span class="html-italic">p</span> &lt; 0.001) when compared with group II (<b>G</b>). Group I: Normal saline (10 mL/kg b.w.), Group II: LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group V: zingerone (50 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.).</p>
Full article ">Figure 4
<p>Effect of zingerone on choline acetyl transferase (ChAT) in LiCl-and-pilocarpine-induced SE. Photomicrographs of hippocampus depicting immunohistochemical analyses. Below photomicrographs is the panel which shows quantitative evaluation of ChAT. Values are expressed as mean ± SEM (<span class="html-italic">n</span> = 6). Significant differences were indicated by <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 when compared with group II. Brain sections showing immunohistochemical results demonstrate specific immune positive staining of ChAT with brown color. The CA1 section of hippocampus in LiCl and pilocarpine-administered group-II (<b>C</b>,<b>D</b>) has decreased immunopositive staining of ChAT as quantified by brown color in comparison to negative control (<b>A</b>,<b>B</b>). But treatment of zingerone (50 mg/kg b.w.) in group V (<b>E</b>,<b>F</b>) increased ChAT immune-staining compared to group II (### <span class="html-italic">p</span> &lt; 0.001) (<b>G</b>). Group I: Normal saline (10 mL/kg b.w.), Group II: LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group V: zingerone (50 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.); *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Zingerone upregulates Bcl-2 expression in LiCl-and-pilocarpine-induced SE. Photomicrographs of hippocampus depicting immunohistochemical analyses indicating specific immune positive staining of Bcl-2 with brown color. Below photomicrographs is the panel which show quantitative evaluation of Bcl-2. Values are expressed as mean ± SEM (<span class="html-italic">n</span> = 6). Significant differences were indicated by *** <span class="html-italic">p</span> &lt; 0.001 when compared with group I and (### <span class="html-italic">p</span> &lt; 0.001) when compared with group II. The CA1 section of hippocampus in LiCl and pilocarpine administered group-II (<b>C</b>,<b>D</b>) has decreased immuno-positive staining of Bcl-2 as specified by brown color in comparison to negative control (group I) (<b>A</b>,<b>B</b>). However, treatment with zingerone (50 mg/kg b.w.) in group V (<b>E</b>,<b>F</b>) enhanced immune positive staining of Bcl-2 in comparison to positive control (group II) (### <span class="html-italic">p</span> &lt; 0.001) (<b>G</b>). Group I: Normal saline (10 mL/kg b.w.), Group II: LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group V: zingerone (50 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.).</p>
Full article ">Figure 6
<p>Zingerone downregulates caspase-3 expression in LiCl-and-pilocarpine-induced SE. Photomicrographs of hippocampus depicting immunohistochemical analyses. Below photomicrographs is the panel which shows quantitative evaluation of activated caspase-3 indicating specific immune positive staining of activated caspase-3 with brown color. Values are expressed as mean ± SEM (<span class="html-italic">n</span> = 6). Significant differences were indicated by *** <span class="html-italic">p</span> &lt; 0.001 when compared with group I and (<sup>###</sup> <span class="html-italic">p</span> &lt; 0.001) when compared with group II. The CA1 section of hippocampus in LiCl and pilocarpine administered group-II (<b>C</b>,<b>D</b>) has enhanced caspase-3 immuno-positive staining as stipulated by brown color in comparison to negative control group (group I) (<b>A</b>,<b>B</b>). But zingerone treatment (50 mg/kg b.w.) in group V (<b>E</b>,<b>F</b>) decreased activated caspase-3 in comparison to positive control (group I) (<sup>###</sup> <span class="html-italic">p</span> &lt; 0.001) (<b>G</b>). Group I: Normal saline (10 mL/kg b.w.), Group II: LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.), Group V: zingerone (50 mg/kg b.w.) + LiCl (3 mEq/kg b.w.) + pilocarpine (30 mg/kg b.w.).</p>
Full article ">Figure 7
<p>A schematic of status epilepticus induced by pilocarpine treatment protocol.</p>
Full article ">
14 pages, 1726 KiB  
Review
2020 FDA TIDES (Peptides and Oligonucleotides) Harvest
by Othman Al Musaimi, Danah Al Shaer, Fernando Albericio and Beatriz G. de la Torre
Pharmaceuticals 2021, 14(2), 145; https://doi.org/10.3390/ph14020145 - 11 Feb 2021
Cited by 56 | Viewed by 5912
Abstract
2020 has been an extremely difficult and challenging year as a result of the coronavirus disease 2019 (COVID-19) pandemic and one in which most efforts have been channeled into tackling the global health crisis. The US Food and Drug Administration (FDA) has approved [...] Read more.
2020 has been an extremely difficult and challenging year as a result of the coronavirus disease 2019 (COVID-19) pandemic and one in which most efforts have been channeled into tackling the global health crisis. The US Food and Drug Administration (FDA) has approved 53 new drug entities, six of which fall in the peptides and oligonucleotides (TIDES) category. The number of authorizations for these kinds of drugs has been similar to that of previous years, thereby reflecting the consolidation of the TIDES market. Here, the TIDES approved in 2020 are analyzed in terms of chemical structure, medical target, mode of action, and adverse effects. Full article
(This article belongs to the Special Issue The Story of Successful Drugs and Recent FDA-Approved Molecules)
Show Figures

Figure 1

Figure 1
<p>A total of 228 new drugs were approved by the Food and Drug Administration (FDA) from 2016 to 2020 [<a href="#B2-pharmaceuticals-14-00145" class="html-bibr">2</a>]. mAbs, monoclonal antibodies; ADCs, antibody drug conjugates; Oligos, oligonucleotides.</p>
Full article ">Figure 2
<p>Structure of viltolarsen (Viltepso<sup>TM</sup>).</p>
Full article ">Figure 3
<p>Chemical structure of lumasiran (Oxlumo<sup>TM</sup>).</p>
Full article ">Figure 4
<p>Chemical structure of setmelanotide (Imcivree<sup>TM</sup>).</p>
Full article ">Figure 5
<p>Chemical structure of <sup>64</sup>Cu -DOTATATE (Detectnet<sup>TM</sup>).</p>
Full article ">Figure 6
<p>Chemical structure of <sup>68</sup>Ga-PMSA-11.</p>
Full article ">Figure 7
<p>Chemical structure of belantamab mafodotin-blmf (Blenrep<sup>TM</sup>).</p>
Full article ">Figure 8
<p>Chemical structure of: (<b>A</b>) natural dolastatin 10; (<b>B</b>) monomethyl auristatin F (MMAF); and (<b>C</b>) monomethyl auristatin E (MMAE). Differences from the natural molecule are shown in red [<a href="#B44-pharmaceuticals-14-00145" class="html-bibr">44</a>].</p>
Full article ">
Previous Issue
Next Issue
Back to TopTop