Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (5)

Search Parameters:
Keywords = onvansertib

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 2388 KiB  
Article
Polo-like Kinase 1 Inhibitors Demonstrate In Vitro and In Vivo Efficacy in Preclinical Models of Small Cell Lung Cancer
by Guojing Zhang, Abbe Pannucci, Andrey A. Ivanov, Jeffrey Switchenko, Shi-Yong Sun, Gabriel L. Sica, Zhentao Liu, Yufei Huang, John C. Schmitz and Taofeek K. Owonikoko
Cancers 2025, 17(3), 446; https://doi.org/10.3390/cancers17030446 - 28 Jan 2025
Viewed by 557
Abstract
Objective: To investigate the preclinical efficacy and identify predictive biomarkers of polo-like kinase 1 (PLK1) inhibitors in small cell lung cancer (SCLC) models. Methods: We tested the cytotoxicity of selective PLK1 inhibitors (rigosertib, volasertib, and onvansertib) in a panel of SCLC cell lines. [...] Read more.
Objective: To investigate the preclinical efficacy and identify predictive biomarkers of polo-like kinase 1 (PLK1) inhibitors in small cell lung cancer (SCLC) models. Methods: We tested the cytotoxicity of selective PLK1 inhibitors (rigosertib, volasertib, and onvansertib) in a panel of SCLC cell lines. We confirmed the therapeutic efficacy of subcutaneous xenografts of representative cell lines and in four patient-derived xenograft models generated from patients with platinum-sensitive and platinum-resistant SCLC. We employed an integrated analysis of genomic and transcriptomic sequencing data to identify potential biomarkers of the activity and mechanisms of resistance in laboratory-derived resistance models. Results: Volasertib, rigosertib, and onvansertib showed strong in vitro cytotoxicity at nanomolar concentrations in human SCLC cell lines. Rigosertib, volasertib, and onvansertib showed equivalent efficacy to that of standard care agents (irinotecan and cisplatin) in vivo with significant growth inhibition superior to cisplatin in PDX models of platinum-sensitive and platinum-resistant SCLC. There was an association between YAP1 expression and disruptive or inactivation TP53 gene mutations, with greater efficacy of PLK1 inhibitors. Comparison of lab-derived onvansertib-resistant H526 cells to parental cells revealed differential gene expression with upregulation of NAP1L3, CYP7B1, AKAP7, and FOXG1 and downregulation of RPS4Y1, KDM5D, USP9Y, and EIF1AY highlighting the potential mechanisms of resistance in the clinical setting. Conclusions: We established the efficacy of PLK1 inhibitors in vitro and in vivo using PDX models of platinum-sensitive and resistant relapsed SCLC. An ongoing phase II trial is currently testing the efficacy of onvansertib in patients with SCLC (NCT05450965). Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

Figure 1
<p>Assessment of in vitro antiproliferative activity of targeted agents in a panel of SCLC cell lines (<b>a</b>); effect of volasertib (<b>b</b>) and onvansertib (<b>c</b>) on proliferation of SCLC cell lines. Cells were treated for 72 h with indicated agents. Cell proliferation was determined using colorimetric or luminescent assays depending on the degree of clustering of SCLC cell lines in culture. Values represent the mean ± S.D. from a minimum of 3 independent experiments. Blue and orange curves define cell lines with non-disruptive and disruptive p53 mutations, respectively. Basal protein expression in SCLC cell lines (<b>d</b>). SCLC subtype based on expression are indicated after each cell line: ASCL1 (A), POU2F3 (P), YAP1 (Y).</p>
Full article ">Figure 2
<p>In vivo efficacy of PLK1 inhibitors in SCLC. Mice bearing H526 xenografts were i.p. administered volasertib (20 mg/kg), irinotecan (25 mg/kg), or cisplatin (3 mg/kg) weekly. Tumor volumes represent the mean ± SEM from groups of 6 mice. *** <span class="html-italic">p</span> ≤ 0.001.</p>
Full article ">Figure 3
<p>Antitumor efficacy of PLK1 inhibitors in SCLC PDXs. Mice bearing platinum-resistant PDXs TKO-002 and TKO-008 (<b>a</b>,<b>b</b>) and platinum-sensitive PDXs TKO-005 and TKO-010 (<b>c</b>,<b>d</b>) were administered cisplatin (3 mg/kg; i.p. weekly), rigosertib (250 mg/kg; i.p. daily), and onvansertib (60 mg/kg; oral × 10 days, 4 days off). Tumor volumes represent the mean ± S.D. from groups of 6 mice per group. *: significant and ns: not significant versus control group. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01.</p>
Full article ">Figure 4
<p>Correlative analysis between <span class="html-italic">TP53</span>, <span class="html-italic">PLK1</span>, and <span class="html-italic">MYC</span> expression (NCBI public database Gene Expression Omnibus GSE55830 [<a href="#B30-cancers-17-00446" class="html-bibr">30</a>]) and cell line sensitivity to PLK1 inhibition (<b>a</b>); YAP1 expression in SCLC-Y cell lines versus other subtypes (<b>b</b>); volcano plot of differentially expressed genes between SCLC-Y and not SCLC-Y cell lines (<b>c</b>); analysis of therapeutic vulnerability based on differential sensitivity of YAP1-positive cell lines showing PLK1 inhibitor as a potential candidate (<b>d</b>); Reactome analysis of active cellular function based on DEG between SCLC-Y and not SCLC-Y lines identified major differences in immune regulation and muscle contraction (<b>e</b>); KEGG analysis of differentially activated signaling pathways between the 2 groups (<b>f</b>).</p>
Full article ">Figure 5
<p>Effect of <span class="html-italic">TP53</span> mutational status on PLK1 inhibitor sensitivity. Comparison of mean IC<sub>50</sub> to <span class="html-italic">TP53</span> gene mutation status (<b>a</b>). <span class="html-italic">TP53</span> gene status in 166 tumor samples in cbioportal.org (<b>b</b>) and 50 SCLC cell lines from publicly available CCLE data (<b>c</b>). Activity of PLK1 inhibitor onvansertib in parental and resistant H526 cells (IC<sub>50</sub> concentration in the resistant vs. parent: 447 nM vs. 51 nM) (<b>d</b>). Gene expression profiles of matched parental and PLK1 inhibitor resistant H526 cells from 3 separate samples (<b>e</b>). Heatmap shows the top differential gene expression (<span class="html-italic">p</span>-adj &lt; 0.5; logFC &gt; 4 cut-off) with red indicating high and blue indicating low natural log-transformed expression.</p>
Full article ">
17 pages, 2681 KiB  
Article
Onvansertib and Navitoclax Combination as a New Therapeutic Option for Mucinous Ovarian Carcinoma
by Serena Petrella, Marika Colombo, Mirko Marabese, Chiara Grasselli, Andrea Panfili, Michela Chiappa, Valentina Sancisi, Ilaria Craparotta, Maria C. Barbera, Giada A. Cassanmagnago, Marco Bolis and Giovanna Damia
Int. J. Mol. Sci. 2025, 26(2), 472; https://doi.org/10.3390/ijms26020472 - 8 Jan 2025
Viewed by 747
Abstract
Mucinous epithelial ovarian cancer (mEOC) is a rare subtype of epithelial ovarian cancer, characterized by poor responses to standard platinum-based chemotherapy. Polo-like kinase 1 (PLK1) is a key regulator of mitosis and cell cycle progression and its inhibition has been recently identified as [...] Read more.
Mucinous epithelial ovarian cancer (mEOC) is a rare subtype of epithelial ovarian cancer, characterized by poor responses to standard platinum-based chemotherapy. Polo-like kinase 1 (PLK1) is a key regulator of mitosis and cell cycle progression and its inhibition has been recently identified as a target in mEOC. In this study, we aimed to identify further therapeutic targets in mEOC using a CRISPR/Cas9 library targeting 3015 genes, with and without treatment with onvansertib, a PLK1 inhibitor. We identified twelve genes associated with cell survival (ZC2HC1C, RPA2, KIN17, TUBG1, SMC2, CDC26, CDC42, HOXA9, TAF10, SENP1, MRPS31, and COPS2) and three genes (JUND, CARD9, and BCL2L2) in synthetic lethality with onvansertib treatment. We validated that SENP1 downregulation is important for the growth of mEOC cells through esiRNA interference and the use of a pharmacological inhibitor Momordin Ic. The downregulation of CARD9 and BCL2L2 combined with subtoxic doses of onvansertib interfered with mEOC cell growth. Interestingly, the combination of navitoclax, an inhibitor of BcL2 family members including BCL2L2, was synergistic in all four of the mEOC cell lines tested and substantially induced cell death through apoptosis. These data support the use of a combination of navitoclax and onvansertib as a new therapeutic strategy for mEOC. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

Figure 1
<p>Screening experiment and Cas9 in mEOC cell lines. (<b>A</b>) Western blot analysis of Cas9 in mEOC cells. (<b>B</b>–<b>D</b>) Flow cytometric analyses of GFP positive (GFP+) and negative (GFP−) cells after lentivirus transduction of GFP plasmid carrying gRNA for GFP in MCAS/Cas9 (<b>B</b>), EFO27/Cas9 (<b>C</b>) and TOV2414/Cas9 (<b>D</b>). (<b>E</b>) Workflow of screening experiment.</p>
Full article ">Figure 2
<p>Genes identified by CRISPR/Cas9 screening in EFO27/Cas9. (<b>A</b>) There were 12 genes with an FDR &lt; 0.05 compared to the not treated groups T1 and T0. These are the genes that, if depleted, lead to cell death. (<b>B</b>) Comparing T1-treated with T1-untreated genes, the selection based on the FDR &lt; 0.1 of individual sgRNAs allowed for the identification of three genes with potential synthetic lethality with onvansertib. For each indicated gene, blue lines represent the gRNAs that were downregulated, while the red lines are the gRNAs that were upregulated. CRISPR-seq data were processed using the MAGeCK pipeline [<a href="#B36-ijms-26-00472" class="html-bibr">36</a>] in the MAGeCKFlute R package [<a href="#B37-ijms-26-00472" class="html-bibr">37</a>].</p>
Full article ">Figure 3
<p>Effects of esiRNA <span class="html-italic">KIN17</span> and <span class="html-italic">SENP1</span> transfection. Cell viability of EFO27 (<b>A</b>,<b>B</b>), OCM.72 (<b>E</b>,<b>F</b>), and TOV2414 (<b>I</b>,<b>J</b>) cells transfected with esiRNA for <span class="html-italic">KIN17</span> and <span class="html-italic">SENP1</span>. Data are the mean ± SD of at least three independent experiments run in triplicate, and are expressed as the percentage on scramble-transfected cells. After 72 h following transfection, RNA was extracted from cells transfected with target and scramble esiRNAs and analyzed to evaluate the expression of targeted genes. The relative-fold change in the selected genes after transfection with the selected esiRNA compared to scramble esiRNA-transfected cells. Mean ± SD of at least three independent experiments (<b>C</b>,<b>D</b>,<b>G</b>,<b>H</b>,<b>K</b>,<b>L</b>). ns: not significant; *: <span class="html-italic">p</span> &lt; 0.05; **: <span class="html-italic">p</span> &lt; 0.01; ****: <span class="html-italic">p</span> &lt; 0.0001</p>
Full article ">Figure 4
<p>Effects of esiRNA <span class="html-italic">JUND</span>, <span class="html-italic">CARD9</span> and <span class="html-italic">BCL2L2</span> transfection in EFO27. Cellular viability of EFO27 cells transfected with the esiRNAs <span class="html-italic">JUND</span> (<b>A</b>), CARD9 (<b>B</b>), and BCL2L2 (<b>C</b>) with or without onvansertib treatment. Data are the mean ± SD of at least two independent experiments and are expressed as percentage of negative control cells. After 72 h following transfection, RNA was extracted from cells transfected with target and scramble esiRNAs and analyzed to evaluate the expression of targeted genes (<b>D</b>–<b>F</b>). Data expressed as relative-fold change in target compared to scramble. Results are the mean ± SD of at least two independent experiments. ONV: onvansertib. ns: not significant; *: <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; ****: <span class="html-italic">p</span> &lt; 0.0001</p>
Full article ">Figure 5
<p>Combination of navitoclax and onvansertib on mEOC cells. Dose–response curve for navitocax in EFO27 (<b>A</b>), OCM.72 (<b>B</b>), TOV2414 (<b>C</b>), and MCAS (<b>D</b>) cells. Combination of onvansertib and navitoclax in EFO27 (<b>E</b>), OCM.72 (<b>F</b>), TOV2414 (<b>G</b>), and MCAS (<b>H</b>) cells; the blue curve is onvansertib alone, and the other curves are the combinations with different concentrations of navitoclax. Data are the mean ± SD of at least two independent experiments and are expressed as the percentage of control untreated cells. (<b>I</b>–<b>L</b>) Drug synergy is indicated by blue squares in the Bliss Synergy Heatmap ONV: onvansertib.</p>
Full article ">Figure 6
<p>Molecular investigation of the combination in EFO27. Cells were treated with onvansertib 75 nM, navitoclax 1.25 μM, or their combination. At the times indicated, cells were analyzed for Annexin V positivity (<b>A</b>), caspase 3/7 activity (<b>B</b>), and cell cycle perturbation (<b>C</b>). Representative histograms show the percentages of Annexin V-positive cells, analyzed by flow cytometry (<b>A</b>), and luminescence signals corresponding to caspase 3/7 activation (<b>B</b>). Bar graphs display the quantification of cells in different cell cycle phases (<b>C</b>). Data are presented as the mean ± SD of three independent experiments.Ctrl: control; Onv: Onvansertib; *: <span class="html-italic">p</span> &lt; 0.05; **: <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
15 pages, 3971 KiB  
Article
PLK1 Inhibitor Onvansertib Enhances the Efficacy of Alpelisib in PIK3CA-Mutated HR-Positive Breast Cancer Resistant to Palbociclib and Endocrine Therapy: Preclinical Insights
by Sreeja Sreekumar, Elodie Montaudon, Davis Klein, Migdalia E. Gonzalez, Pierre Painsec, Héloise Derrien, Laura Sourd, Tod Smeal, Elisabetta Marangoni and Maya Ridinger
Cancers 2024, 16(19), 3259; https://doi.org/10.3390/cancers16193259 - 25 Sep 2024
Viewed by 2280
Abstract
Background: Endocrine therapy (ET) combined with cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) is the preferred first-line treatment for hormone receptor-positive (HR+)/HER2- metastatic breast cancer. Although this is beneficial, acquired resistance leads to disease progression, and patients harboring PIK3CA mutations are treated with targeted therapies [...] Read more.
Background: Endocrine therapy (ET) combined with cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) is the preferred first-line treatment for hormone receptor-positive (HR+)/HER2- metastatic breast cancer. Although this is beneficial, acquired resistance leads to disease progression, and patients harboring PIK3CA mutations are treated with targeted therapies such as the PI3Kα inhibitor, alpelisib, alongside ET. Drug-associated resistance mechanisms limit the efficacy of alpelisib, highlighting the need for better combination therapies. This study aimed to evaluate the efficacy of combining alpelisib with a highly specific PLK1 inhibitor, onvansertib, in PIK3CA-mutant HR+ breast cancer preclinical models. Methods: We assessed the effect of the alpelisib and onvansertib combination on cell viability, PI3K signaling pathway, cell cycle phase distribution and apoptosis in PI3K-activated HR+ breast cancer cell lines. The antitumor activity of the combination was evaluated in three PIK3CA-mutant HR+ breast cancer patient-derived xenograft (PDX) models, resistant to ET and CDK4/6 inhibitor palbociclib. Pharmacodynamics studies were performed using immunohistochemistry and Simple Western analyses in tumor tissues. Results: The combination synergistically inhibited cell viability, suppressed PI3K signaling, induced G2/M arrest and apoptosis in PI3K-activated cell lines. In the three PDX models, the combination demonstrated superior anti-tumor activity compared to the single agents. Pharmacodynamic studies confirmed the inhibition of both PLK1 and PI3K activity and pronounced apoptosis in the combination-treated tumors. Conclusions: Our findings support that targeting PLK1 and PI3Kα with onvansertib and alpelisib, respectively, may be a promising strategy for patients with PIK3CA-mutant HR+ breast cancer failing ET + CDK4/6i therapies and warrant clinical evaluation. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

Figure 1
<p>Combination of alpelisib and onvansertib displays a synergistic effect in HR+ breast cancer cell lines. (<b>A</b>) Cell viability was assessed after 6–7 days of treatment with onvansertib (Onv), alpelisib (Alp), or the combination of Onv + Alp at the indicated concentrations, and synergy was assessed. Heatmaps of the combination responses for onvansertib and alpelisib based on the Bliss synergy model are shown. Blue color indicates synergistic interaction (<span class="html-italic">n</span> = 3). (<b>B</b>) Colony formation assay of MCF7, T-47D, and EFM-19 cells treated with Alp and Onv or their combination (Alp + Onv) at the indicated concentrations. Representative wells are shown in the top panel, and percentages of colonies normalized to the DMSO-treated control (Ctrl) from three independent experiments are shown in the bottom panel. Results presented as mean ± SEM. Asterisks indicate significance by one-way ANOVA (* <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, **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 2
<p>Alpelisib and onvansertib combination induces G2/M arrest and apoptosis in ER+ breast cancer cell lines. (<b>A</b>,<b>B</b>) The cells were treated with alpelisib (Alp), onvansertib (Onv), or the combination (Alp + Onv) for 72–96 h at the indicated concentrations, and cell cycle analysis was performed. (<b>A</b>) Representative DNA histograms showing the population of cells in the G1, S, and G2/M cell cycle phases of MCF7 treated with the drugs. (<b>B</b>) The bar graph showing percentage of cells in the G1, S, and G2/M phases of MCF7, T-47D, EFM-19, ZR-75-1, and MCF7/164R-7 cells after treating with Onv, Alp or Alp + Onv. (<b>C</b>) The cells were treated with Onv, Alp, or the combination at the indicated concentrations, and the percentage of apoptotic cells was analyzed by a TUNEL assay. The percentage of TUNEL+ cells is plotted. (<b>D</b>) MCF7 and EFM-19 cells were treated with DMSO vehicle (Ctrl), Onv (25 nM for MCF7, 15 nM for EFM-19), Alp (200 nM for MCF7, 100 nM for EFM-19), or Onv + Alp for 24 h. Representative Simple Western images of cleaved-PARP (cl-PARP) and total PARP protein expression. The uncropped Simple Western images are shown in <a href="#app1-cancers-16-03259" class="html-app">Figure S6</a>. (<b>E</b>) Densitometric ratio of cleaved-PARP to total PARP expression levels normalized to DMSO control are plotted. All results are the mean of three experiments and are presented as mean ± SEM. One-way ANOVA was used to compare the means. Asterisks indicate significance (* <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, **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 3
<p>The combination of onvansertib and alpelisib shows robust anti-tumor activity <span class="html-italic">in vivo</span>. (<b>A</b>) HBCx-86, (<b>B</b>,<b>C</b>) HBCx-180, and (<b>D</b>) HBCx-134palboR31 PDX models were treated with vehicle (Ctrl), onvansertib (Onv, 45 mg/kg), alpelisib (Alp, 25 mg/kg), or combination of Alp and Onv (<span class="html-italic">n</span> = 6 to 8 per group) for the indicated duration (—). (<b>A</b>,<b>B</b>,<b>D</b>) Mean ± SEM tumor volumes are shown on the left, and individual relative tumor volumes (RTV) are shown on the right. Tumor regression is reported if RTV &lt; 0.5 in at least 1 tumor measurement. An unpaired <span class="html-italic">t</span>-test was used to compare RTV at the last measurement. (<b>C</b>) Kaplan–Meier survival curve for event-free survival (time for RTV = 4) was calculated. Log-rank Mantel–Cox test was used for survival analyses * <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>Onvansertib and alpelisib combination inhibits the PLK1 and PI3K pathways and induces apoptosis <span class="html-italic">in vivo</span>. (<b>A</b>) Protein levels of pAKT-Ser473, AKT, phospho-TCTP, and TCTP in HBCx-134palboR31 tumors treated with vehicle (Veh), onvansertib (Onv), alpelisib (Alp), or the combination (Onv + Alp) for 4 days. (<b>B</b>) The normalized densitometric ratio of the phospho-proteins to total protein levels is plotted. (<b>C</b>) H&amp;E-stained photomicrographs (40×) showing apoptotic cells in HBCx-86 and HBCx-134palboR31 tumors treated with Veh, Onv, Alp, or Alp + Onv for 32 days (HBCx-86) or 4 days (HBCx-134palboR31). (<b>D</b>,<b>E</b>) Apoptotic cells were manually counted and plotted. (<b>F</b>) Cleaved-PARP and total PARP protein expression in HBCx-134palboR31 tumors. (<b>G</b>) Graphs show normalized densitometric ratio of cleaved-PARP to total PARP. The uncropped Simple Western images are shown in <a href="#app1-cancers-16-03259" class="html-app">Figure S6</a>. Data are expressed as mean ± SEM. One-way ANOVA was used to compare the means. Asterisks indicate significance (* <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 ">
20 pages, 8325 KiB  
Article
Pentafuhalol-B, a Phlorotannin from Brown Algae, Strongly Inhibits the PLK-1 Overexpression in Cancer Cells as Revealed by Computational Analysis
by Waseem Ahmad Ansari, Safia Obaidur Rab, Mohammad Saquib, Aqib Sarfraz, Mohd Kamil Hussain, Mohd Sayeed Akhtar, Irfan Ahmad and Mohammad Faheem Khan
Molecules 2023, 28(15), 5853; https://doi.org/10.3390/molecules28155853 - 3 Aug 2023
Cited by 12 | Viewed by 2465
Abstract
Polo-like kinase-1 (PLK-1) is an essential mitotic serine/threonine (Ser/Thr) kinase that belongs to the Polo-like kinase (PLK) family and is overexpressed in non-small cell lung cancer (NSCLC) via promotion of cell division. Therefore, PLK-1 may act as a promising target for the therapeutic [...] Read more.
Polo-like kinase-1 (PLK-1) is an essential mitotic serine/threonine (Ser/Thr) kinase that belongs to the Polo-like kinase (PLK) family and is overexpressed in non-small cell lung cancer (NSCLC) via promotion of cell division. Therefore, PLK-1 may act as a promising target for the therapeutic cure of various cancers. Although a variety of anti-cancer drugs, both synthetic and naturally occurring, such as volasertib, onvansertib, thymoquinone, and quercetin, are available either alone or in combination with other therapies, they have limited efficacy, especially in the advanced stages of cancer. To the best of our knowledge, no anticancer agent has been reported from marine algae or microorganisms to date. Thus, the aim of the present study is a high-throughput virtual screening of phlorotannins, obtained from edible brown algae, using molecular docking and molecular dynamic simulation analysis. Among these, Pentafuhalol-B (PtB) showed the lowest binding energy (best of triplicate runs) against the target protein PLK-1 as compared to the reference drug volasertib. Further, in MD simulation (best of triplicate runs), the PtB-PLK-1 complex displayed stability in an implicit water system through the formation of strong molecular interactions. Additionally, MMGBSA calculation (best of triplicate runs) was also performed to validate the PtB-PLK-1 complex binding affinities and stability. Moreover, the chemical reactivity of PtB towards the PLK-1 target was also optimised using density functional theory (DFT) calculations, which exhibited a lower HOMO-LUMO energy gap. Overall, these studies suggest that PtB binds strongly within the pocket sites of PLK-1 through the formation of a stable complex, and also shows higher chemical reactivity than the reference drug volasertib. The present study demonstrated the inhibitory nature of PtB against the PLK-1 protein, establishing its potential usefulness as a small molecule inhibitor for the treatment of different types of cancer. Full article
(This article belongs to the Special Issue Virtual Screening)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Geometrically optimised structures of phlorotannins (<b>1</b>–<b>15</b>) as well as the control drug (<b>16,</b> volasertib) calculated by the B3LYP hybrid function combined with a 6–31 basis set using the DFT method.</p>
Full article ">Figure 2
<p>(<b>A</b>) Pictorial representation of HOMO orbitals for phlorotannins (<b>1</b>–<b>15</b>) and the control drug (<b>16</b>). (<b>B</b>) Pictorial representation of LUMO orbitals for phlorotannins (<b>1</b>–<b>15</b>) and the control drug (<b>16</b>).</p>
Full article ">Figure 2 Cont.
<p>(<b>A</b>) Pictorial representation of HOMO orbitals for phlorotannins (<b>1</b>–<b>15</b>) and the control drug (<b>16</b>). (<b>B</b>) Pictorial representation of LUMO orbitals for phlorotannins (<b>1</b>–<b>15</b>) and the control drug (<b>16</b>).</p>
Full article ">Figure 3
<p>Density of states (DOS) plots of phlorotannins (<b>1</b>–<b>15</b>) and the control drug (<b>16</b>).</p>
Full article ">Figure 4
<p>Molecular electrostatic potential (MEP) map of phlorotannins (<b>1</b>–<b>15</b>) and control drug (<b>16</b>).</p>
Full article ">Figure 5
<p>Docking poses along with molecular interactions of PtB within the binding pockets of the 2YAC receptor.</p>
Full article ">Figure 6
<p>(<b>A</b>) RMSD graph of the PtB (red) and 2YAC protein (blue); (<b>B</b>) RMSF graph of the 2YAC protein (cyan: β-strand, orange: helices, green: H-bond); (<b>C</b>) PtB interactions with protein residues; (<b>D</b>) histogram of interaction amino acids of the 2YAC protein (green: H- bond, Blue: water bridges, Pink: ionic bond, cyan: hydrophobic interaction); (<b>E</b>) fingerprints of PtB and 2YAC contacts (blue: amino acids contribution, orange: amino acid interactions); and (<b>F</b>) PtB properties, e.g., rGyr (green), MolSA (yellow), SASA (light blue), and PSA (dark red). Plots A and B are the RMSD and RMSF presentations among the triplicate runs (<a href="#app1-molecules-28-05853" class="html-app">Supplementary Information</a>, <a href="#app1-molecules-28-05853" class="html-app">Figure S1</a>).</p>
Full article ">Figure 7
<p>The chemical structures of phlorotannin compounds (<b>1</b>–<b>15</b>) and the control drug, volasertib (<b>16</b>).</p>
Full article ">
14 pages, 1876 KiB  
Article
Identification of PLK1 as a New Therapeutic Target in Mucinous Ovarian Carcinoma
by Roberta Affatato, Laura Carrassa, Rosaria Chilà, Monica Lupi, Valentina Restelli and Giovanna Damia
Cancers 2020, 12(3), 672; https://doi.org/10.3390/cancers12030672 - 13 Mar 2020
Cited by 25 | Viewed by 3777
Abstract
Mucinous epithelial ovarian cancer (mEOC) is a rare subset of epithelial ovarian cancer. When diagnosed at a late stage, its prognosis is very poor, as it is quite chemo-resistant. To find new therapeutic options for mEOC, we performed high-throughput screening using a siRNA [...] Read more.
Mucinous epithelial ovarian cancer (mEOC) is a rare subset of epithelial ovarian cancer. When diagnosed at a late stage, its prognosis is very poor, as it is quite chemo-resistant. To find new therapeutic options for mEOC, we performed high-throughput screening using a siRNA library directed against human protein kinases in a mEOC cell line, and polo-like kinase1 (PLK1) was identified as the kinase whose downregulation interfered with cell proliferation. Both PLK1 siRNA and two specific PLK1 inhibitors (onvansertib and volasertib) were able to inhibit cell growth, induce apoptosis and block cells in the G2/M phase of the cell cycle. We evaluated, in vitro, the combinations of PLK1 inhibitors and different chemotherapeutic drugs currently used in the treatment of mEOC, and we observed a synergistic effect of PLK1 inhibitors and antimitotic drugs. When translated into an in vivo xenograft model, the combination of onvansertib and paclitaxel resulted in stronger tumor regressions and in a longer mice survival than the single treatments. These effects were associated with a higher induction of mitotic block and induction of apoptosis, similarly to what was observed in vitro. These data suggest that the combination onvansertib/paclitaxel could represent a new active therapeutic option in mEOC. Full article
Show Figures

Figure 1

Figure 1
<p>High-throughput siRNA screening in the mucinous epithelial ovarian cancer (mEOC) cell line MCAS. (<b>A</b>) Summary of the effects of downregulation on each member of the 719-in-size target pool of the siRNA library. Data are plotted as ratios of transfected siRNA/negative control siRNA mean absorbance values (T/C) and represent the means of the independent siRNA screenings done. The dashed line selected 17 siRNA with a T/C ≤ 0.6. The arrow underlines the T/C value of PLK1. (<b>B</b>) Validation of siRNA screening. MCAS cell survival after 96 h of transfection using specific esiRNA against positive hits identified in the screening.</p>
Full article ">Figure 2
<p>PLK1 downregulation in MCAS and EFO27 cell lines. (<b>A</b>) Western blot analysis showing PLK1 downregulation after transfection with esiRNA in MCAS and EFO27 cell lines. (<b>B</b>) Percentages of cell viability at 48 and 72 h after esiRNA transfection. Black bar, negative esiRNA; white bar, PLK1 esiRNA. (<b>C</b>) Flow cytometric analysis of DNA content. (<b>D</b>) Caspase-3 activity in MCAS (black bar) and EFO27 (white bar) cell lines.</p>
Full article ">Figure 3
<p>Drug combination of onvansertib and paclitaxel or eribulin in mEOC cell lines. (<b>A</b>–<b>C</b>) MCAS, JHOM1 and EFO27 treated with non-toxic concentrations of onvansertib and increasing concentrations of paclitaxel and eribulin. (<b>D</b>,<b>E</b>) normalized IC50 isobolograms showing the synergistic effects of the combination of onvansertib with paclitaxel and eribulin in MCAS and JHOM1 cell lines and the antagonistic effect in EFO27. Each symbol represents an independent experiment.</p>
Full article ">Figure 4
<p>Effect of the combined treatment of onvansertib/paclitaxel on the MCAS cell line. (<b>A</b>) Cell growth curves of cells untreated (-●-), treated with 15nM of onvansertib (-■-), 2nM of paclitaxel (-▲-) or the combination (-<b>○</b>-). Cell growth was followed from the day of treatment and up to 48 h after treatment. The data are expressed as number of cells ±SD of three replicates. A two-way ANOVA test performed with GraphPad Prism was used for statistical analysis. Statistically significance differences are observed from 24 to 48 h in MCAS cell combined treatment groups vs. control and vs. single agents: **** <span class="html-italic">p</span> &lt; 0.0001. (<b>B</b>) Analysis of DNA content after 8, 24 and 48 h of treatment with the two drugs either single (paclitaxel 2nM, onvansertib 15nM) or combined, and biparametric analysis of pS10-histone H3 and DNA after 24 and 48 h. Cells detected above the horizontal line were considered as pS10-histone H3-positive, and this percentage was reported in each plot. (<b>C</b>) Activation of caspase-3 by enzymatic assay 24 and 48 h after treatment. Data are percentages of untreated cells and represent the means ± SDs. For statistical analysis, a two-way ANOVA test with Bonferroni multiple comparison was used, paclitaxel vs. combination 24 h **** <span class="html-italic">p</span> &lt; 0.0001, paclitaxel vs. combination 48 h **** <span class="html-italic">p</span> &lt; 0.0001. (<b>D</b>) Western blot analysis showing PLK1, γH2AX, actin and beta-tubulin protein levels in MCAS cells protein extracts.</p>
Full article ">Figure 5
<p>Antitumor effect and target modulation of the combined treatment in MCAS xenografts. (<b>A</b>) Tumor growth curves of MCAS xenograft control (-●-) and treatments with onvansertib (-■-), paclitaxel (-▲-) or both (-<b>○</b>-). Data are represented as means ± SEs. For statistical analysis, two-way ANOVA test with Bonferroni multiple comparison was used. On day 35 the significant differences were as follows: control vs. paclitaxel and combination, ****, <span class="html-italic">p</span> &lt; 0.0001; onvansertib vs. paclitaxel and combination, ****, <span class="html-italic">p</span> &lt; 0.0001. One-way ANOVA test for repeated measures with Bonferroni multiple comparison was used for statistical analysis at day 45 and the significant differences were as follows: control vs. combination, **, <span class="html-italic">p</span> &lt; 0.01; onvansertib vs. combination, ****, <span class="html-italic">p</span> &lt; 0.0001; paclitaxel vs. combination, *, <span class="html-italic">p</span> &lt; 0.05. (<b>B</b>) Kaplan–Meier survival curves of mice. Log-rank test (Mantel–Cox) was used to calculate p-values comparing the survival curves. Control vs. paclitaxel and combination ****, <span class="html-italic">p</span> &lt; 0.0001; onvansertib vs. paclitaxel and combination ****, <span class="html-italic">p</span> &lt; 0.0001; paclitaxel vs. combination **, <span class="html-italic">p</span> &lt; 0.01. (<b>C</b>) Caspase 3 activity in tumor tissue extracts from mice treated as described in the methods. Data are percentages of untreated cells and represent the means ± SDs of two independent experiments each. One-way ANOVA test with Tukey’s multiple comparison was used, and the differences were as follows: control vs. combination *, <span class="html-italic">p</span> &lt; 0.05; onvansertib vs. paclitaxel *, <span class="html-italic">p</span> &lt; 0.05; onvansertib vs. combination **, <span class="html-italic">p</span> &lt; 0.01. (<b>D</b>) Western blot analysis showing pS10 Histone H3 and γH2AX protein levels in xenograft tumors protein extracts. Two replicates for each condition were used.</p>
Full article ">
Back to TopTop