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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (10,761)

Search Parameters:
Keywords = lung cancer

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 2676 KiB  
Article
Polysaccharide Fraction Isolated from Saccharina japonica Exhibits Anti-Cancer Effects Through Immunostimulating Activities
by Min Seung Park, Seung-U Son, Tae Eun Kim, Se Hyun Shim, Bong-Keun Jang, Sunyoung Park and Kwang-Soon Shin
Mar. Drugs 2025, 23(1), 38; https://doi.org/10.3390/md23010038 (registering DOI) - 13 Jan 2025
Abstract
The present research aimed to assess the anti-cancer effects of the polysaccharide fraction (SJP) isolated from Saccharina japonica. The release of immune-activating cytokines, including IL-6, IL-12, and TNF-α, was markedly stimulated by the SJP in a concentration-dependent manner within the range of [...] Read more.
The present research aimed to assess the anti-cancer effects of the polysaccharide fraction (SJP) isolated from Saccharina japonica. The release of immune-activating cytokines, including IL-6, IL-12, and TNF-α, was markedly stimulated by the SJP in a concentration-dependent manner within the range of 1 to 100 µg/mL. Furthermore, the prophylactic intravenous (p.i.v.) and per os (p.p.o.) injection of SJP boosted the cytolytic activity mediated by NK cells and CTLs against tumor cells. In a study involving Colon26-M3.1 carcinoma as a lung cancer model, both p.i.v. and p.p.o. exhibited significant anti-lung-cancer effects. Notably, p.i.v. and p.p.o. administration of SJP at a dose of 50 mg/kg reduced tumor colonies by 84% and 40%, respectively, compared to the control. Moreover, the anti-lung-cancer effects of SJP remained substantial, even when NK cell function was inhibited using anti-asialo-GM1. Fractionation with CaCl2 suggested that SJP is a mixture of alginate and fucoidan. The fucoidan fraction stimulated the immune response of macrophages more strongly than the alginate fraction. Consequently, this finding suggested that SJP from S. japonica possesses remarkable anti-cancer effects through the activation of various immunocytes. In addition, this finding indicates that the potent biological activity of SJP may be attributed to fucoidan. Full article
(This article belongs to the Special Issue Marine Natural Products as Anticancer Agents, 4th Edition)
Show Figures

Figure 1

Figure 1
<p>Effects of SJP isolated from <span class="html-italic">Saccharina japonica</span> about cytokine release by macrophages. (<b>a</b>) IL-6, (<b>b</b>) IL-12, and (<b>c</b>) TNF-α concentration of culture supernatant. The medium and LPS (1 μg/mL) represent the NC and PC, respectively. All data are presented as the mean ± standard deviation (<span class="html-italic">n</span> = 3); the different letters (a–e) indicate statistical significance (<span class="html-italic">p</span> &lt; 0.05). Significant differences were evaluated using the one-way analysis of variance followed by Duncan’s test for multiple comparisons. NC, negative control; PC, positive control.</p>
Full article ">Figure 2
<p>Comparison of NK cell and CTL cytotoxic activity against cancer cells by SJP. (<b>a</b>,<b>b</b>) Cytotoxicity of NK cells and CTLs by <span class="html-italic">i.v.</span> administration. (<b>c</b>,<b>d</b>) Cytotoxicity of NK cells and CTLs by oral administration. NK cells and CTLs were co-cultured with YAC-1 lymphoma and Colon26-M3.1 carcinoma, respectively, for 6 h in a 5% CO<sub>2</sub> incubator at 37 °C. Saline and distilled water were administered to the NC groups. SJP was administered at various doses including at 0.5 (SL), 5 (SM), and 50 mg/kg (SH). All data are presented as the mean ± standard deviation (<span class="html-italic">n</span> = 3); the different letters (a–d, A–D, I–IV) indicate statistical significance (<span class="html-italic">p</span> &lt; 0.05). Significant differences were evaluated using the one-way analysis of variance followed by Duncan’s test for multiple comparisons. NC, negative control; SL, SJP-low; SM, SJP-medium; SH, SJP-high.</p>
Full article ">Figure 3
<p>Inhibitory effects of SJP on Colon26-M3.1-caused lung cancer in BALB/c mice. (<b>a</b>) Intravenous administration. (<b>b</b>) Oral administration. Lung-cancer-bearing mice model induced by the <span class="html-italic">i.v.</span> inoculation of Colon26-M3.1 carcinoma. Saline and distilled water were administered to the NC group. Krestin (50 mg/kg), a known reference drug isolated from <span class="html-italic">Coriolus versicolor</span>, was used in the PC group. SJP was intravenously and orally administered at various doses including at 0.5 (SL), 5 (SM), and 50 mg/kg (SH), followed by <span class="html-italic">i.v.</span> inoculation with Colon26-M3.1 carcinoma. All data are presented as the mean ± standard deviation (<span class="html-italic">n</span> = 8); the different letters (a–d) indicate statistical significance (<span class="html-italic">p</span> &lt; 0.05). Significant differences were evaluated using the one-way analysis of variance followed by Duncan’s test for multiple comparisons. NC, negative control; PC, positive control; SL, SJP-low; SM, SJP-medium; SH, SJP-high.</p>
Full article ">Figure 4
<p>Cancer inhibitory effects of SJP on lung cancer in BALB/c mice with impaired NK cell function. Lung-cancer-bearing mice model induced by the <span class="html-italic">i.v.</span> inoculation of Colon26-M3.1 carcinoma. Saline was administered to the NC group. SJP was intravenously administered at 5 mg/kg, followed by <span class="html-italic">i.v.</span> inoculation with Colon26-M3.1 carcinoma. The rabbit anti-asialo-GM1 serum was intravenously injected into the mice, two days before inoculation with Colon26-M3.1 carcinoma, to deplete NK cell function. All data are presented as the mean ± standard deviation (<span class="html-italic">n</span> = 8); the different letters (a–d) indicate statistical significance (<span class="html-italic">p</span> &lt; 0.05). Significant differences were evaluated using the one-way analysis of variance followed by Duncan’s test for multiple comparisons. NC, negative control; SO, SJP-only; AO, anti-asialo-GM1-only; AS, anti-asialo-GM1-SJP.</p>
Full article ">Figure 5
<p>Molecular weight of polysaccharide from <span class="html-italic">S. japonica</span> on RID-HPLC. (<b>a</b>) SJP. (<b>b</b>) SJP-CS. (<b>c</b>) SJP-CP. Elution pattern of three polysaccharide fractions on RID-HPLC equipped with Superdex 75 column, stabilized with 50 mM ammonium formate buffer (pH 5.5).</p>
Full article ">Figure 6
<p>Scheme tree to separate polysaccharide from <span class="html-italic">S. japonica</span>.</p>
Full article ">Figure 7
<p>Effects of polysaccharide fractions isolated from <span class="html-italic">S. japonica</span> on cytokine secretion of murine peritoneal macrophages. (<b>a</b>) IL-6, (<b>b</b>) IL-12, and (<b>c</b>) TNF-α concentration of culture supernatant. The medium and LPS (1 μg/mL) represent the NC and PC, respectively. All data are presented as the mean ± standard deviation (<span class="html-italic">n</span> = 3); the different letters (a–i) indicate statistical significance (<span class="html-italic">p</span> &lt; 0.05). Significant differences were evaluated using the one-way analysis of variance followed by Duncan’s test for multiple comparisons. Among the experimental groups, statistical significance was set at <span class="html-italic">p</span> value &lt; 0.05. NC, negative control; PC, positive control.</p>
Full article ">
16 pages, 1545 KiB  
Article
Circulating Tumor Cell-Free DNA as Prognostic Biomarker in Non-Small Cell Lung Cancer Patients Undergoing Immunotherapy: The CORELAB Experience
by Stefania Gelmini, Adele Calabri, Irene Mancini, Camilla Eva Comin, Valeria Pasini, Marco Banini, Vieri Scotti and Pamela Pinzani
Int. J. Mol. Sci. 2025, 26(2), 611; https://doi.org/10.3390/ijms26020611 - 13 Jan 2025
Abstract
The expression level of Programmed Death-Ligand 1 (PD-L1) determined by the immunohistochemical method is currently approved to test the potential efficacy of immune-checkpoint inhibitors and to candidate patients with Non-Small Cell Lung Cancer (NSCLC) for treatment with immunotherapeutic drugs. As part of the [...] Read more.
The expression level of Programmed Death-Ligand 1 (PD-L1) determined by the immunohistochemical method is currently approved to test the potential efficacy of immune-checkpoint inhibitors and to candidate patients with Non-Small Cell Lung Cancer (NSCLC) for treatment with immunotherapeutic drugs. As part of the CORELAB (New prediCtivebiOmaRkers of activity and Efficacy of immune checkpoint inhibitors in advanced non-small cell Lung cArcinoma) project, aimed at identifying new predictive and prognostic biomarkers in NSCLC patients receiving immunotherapeutic drugs, we investigated the role of circulating tumor DNA (ctDNA) molecular characterization as an additional predictive biomarker. We analyzed plasma ctDNA by targeted Next Generation Sequencing in a subset of 50 patients at different time points. ctDNA content was inversely correlated with the clinical outcome both at a baseline and after 2 months of treatment. OS was significantly higher in patients with ≥50% ctDNA reduction. TP53 and KRAS were the most frequently mutated genes, and patients with KRAS and/or TP53 mutations showed worse outcomes than patients without detectable variants or with mutations in other genes. Fewer common variants were found in BRAF, EGFR, MAP2K1, MET, NRAS, and PIK3CA genes. Our data demonstrated that molecular characterization of ctDNA and also its quantitative evaluation could serve as a dynamic, real-time prognostic, and predictive biomarker, enabling regular molecular monitoring of therapy efficacy in support of other medical examinations. Full article
(This article belongs to the Special Issue Advances in the Diagnosis and Treatment of Non-small Cell Lung Cancer)
Show Figures

Figure 1

Figure 1
<p>ctDNA and radiological variation between T0 (untextured) and T1(textured); patients without detectable variants at both times are not represented. Patients 1–14 had a reduction in ctDNA, patients 15–19 had stable ctDNA, and patients 20–24 had an increase in ctDNA. Bars are represented in green for patients with radiological Stable Disease (SD) per RECIST, blue for Partial Response (PR), and red for Progressive Disease (PD).</p>
Full article ">Figure 2
<p>Distribution of ctDNA values at baseline and after 2 months of therapy based on radiological response. Distribution of ctDNA levels at T0 (<b>A</b>), after two months of immunotherapy, T1 (<b>B</b>) and the variation of ctDNA amount between the two time points ∆ctDNA (ctDNAT1 − ctDNAT0)/ctDNAT0; (<b>C</b>), according to the radiologic response; (Mann–Whitney test).</p>
Full article ">Figure 3
<p>Association of molecular response with survival outcomes (<b>A</b>); association of the pre-treatment genomic status with survival outcomes (<b>B</b>).</p>
Full article ">Figure 4
<p>Longitudinal monitoring of an NSCLC patient with a <span class="html-italic">TP53</span> mutation. ctDNA longitudinal monitoring of a case study patient at T0 (day 0), T1 (day 54), T2 (day 112) and at TP (day 184). ctDNA VAF% (corresponding to a <span class="html-italic">TP53</span> variant) at different follow-ups are shown with associated clinical data.</p>
Full article ">
15 pages, 2220 KiB  
Article
Enhancing Treatment Decisions for Advanced Non-Small Cell Lung Cancer with Epidermal Growth Factor Receptor Mutations: A Reinforcement Learning Approach
by Hakan Şat Bozcuk, Leyla Sert, Muhammet Ali Kaplan, Ali Murat Tatlı, Mustafa Karaca, Harun Muğlu, Ahmet Bilici, Bilge Şah Kılıçtaş, Mehmet Artaç, Pınar Erel, Perran Fulden Yumuk, Burak Bilgin, Mehmet Ali Nahit Şendur, Saadettin Kılıçkap, Hakan Taban, Sevinç Ballı, Ahmet Demirkazık, Fatma Akdağ, İlhan Hacıbekiroğlu, Halil Göksel Güzel, Murat Koçer, Pınar Gürsoy, Bahadır Köylü, Fatih Selçukbiricik, Gökhan Karakaya and Mustafa Serkan Alemdaradd Show full author list remove Hide full author list
Cancers 2025, 17(2), 233; https://doi.org/10.3390/cancers17020233 - 13 Jan 2025
Abstract
Background: Although higher-generation TKIs are associated with improved progression-free survival in advanced NSCLC patients with EGFR mutations, the optimal selection of TKI treatment remains uncertain. To address this gap, we developed a web application powered by a reinforcement learning (RL) algorithm to assist [...] Read more.
Background: Although higher-generation TKIs are associated with improved progression-free survival in advanced NSCLC patients with EGFR mutations, the optimal selection of TKI treatment remains uncertain. To address this gap, we developed a web application powered by a reinforcement learning (RL) algorithm to assist in guiding initial TKI treatment decisions. Methods: Clinical and mutational data from advanced NSCLC patients were retrospectively collected from 14 medical centers. Only patients with complete data and sufficient follow-up were included. Multiple supervised machine learning models were tested, with the Extra Trees Classifier (ETC) identified as the most effective for predicting progression-free survival. Feature importance scores were calculated by the ETC, and features were then integrated into a Deep Q-Network (DQN) RL algorithm. The RL model was designed to select optimal TKI generation and a treatment line for each patient and was embedded into an open-source web application for experimental clinical use. Results: In total, 318 cases of EGFR-mutant advanced NSCLC were analyzed, with a median patient age of 63. A total of 52.2% of patients were female, and 83.3% had ECOG scores of 0 or 1. The top three most influential features identified were neutrophil-to-lymphocyte ratio (log-transformed), age (log-transformed), and the treatment line of TKI administration, as tested by the ETC algorithm, with an area under curve (AUC) value of 0.73, whereas the DQN RL algorithm achieved a higher AUC value of 0.80, assigning distinct Q-values across four TKI treatment categories. This supports the decision-making process in the web-based ‘EGFR Mutant NSCLC Treatment Advisory System’, where clinicians can input patient-specific data to receive tailored recommendations. Conclusions: The RL-based web application shows promise in assisting TKI treatment selection for EGFR-mutant advanced NSCLC patients, underscoring the potential for reinforcement learning to enhance decision-making in oncology care. Full article
(This article belongs to the Section Methods and Technologies Development)
Show Figures

Figure 1

Figure 1
<p>Feature importance plot. Variable importance figures for features in the study. Lognlr, logarithmic transformation of neutrophil-to-lymphocyte ratio; logage, logarithmic transformation of age; line_treatment, line of initial TKI usage; brain_met, presence or absence of brain metastases; ecog_cat, ECOG performance status; mutation_cat, Exon 19 mutation or other mutations; smoking_status, never or ever smoker; mets_number, number of metastases up to three or more; bone_or_liver_met, presence or absence of bone and/or liver metastases.</p>
Full article ">Figure 2
<p>Action frequencies with respect to the line of first TKI treatment. Action frequency figures at first and subsequent lines of first TKI treatment (in chemotherapy naïve and refractory patients). Action definitions: 0; first-line, first-generation TKI, 1; first-line, second- or higher-generation TKI, 2; second- or later-line, first-generation TKI, 3; second- or later-line, second- or higher-generation TKI. (<b>a</b>). Action frequencies at first line (chemotherapy naïve patients). (<b>b</b>). Action frequencies at second or subsequent line (chemotherapy refractory patients).</p>
Full article ">Figure 3
<p>Partial dependence plots. Visualizing action–feature interactions, plotting action frequencies, and calculating summary Q-values for each action within the RL model. (<b>a</b>). Gender and actions. (<b>b</b>). Type of EGFR mutation and actions. (<b>c</b>). Age (logarithmic transformation) and actions. (<b>d</b>). NLR (logarithmic transformation) and actions.</p>
Full article ">Figure 4
<p>Web application interface. The web application, named “EGFR Mutant NSCLC Treatment Advisory System” and its interface, accessible at <a href="https://egfr-recommender.streamlit.app/" target="_blank">https://egfr-recommender.streamlit.app/</a>, accessed on 6 January 2025.</p>
Full article ">
15 pages, 7711 KiB  
Article
Neo-BCV: A Novel Bacterial Liquid Complex Vaccine for Enhancing Dendritic Cell-Mediated Immune Responses Against Lung Cancer
by Zilong Zhu, Zhuze Chu, Fei Fei, Chenxi Wu, Zhengyue Fei, Yuxia Sun, Yun Chen and Peihua Lu
Vaccines 2025, 13(1), 64; https://doi.org/10.3390/vaccines13010064 - 13 Jan 2025
Viewed by 217
Abstract
Background: In the past decade, immunotherapy has become a major choice for the treatment of lung cancer, yet its therapeutic efficacy is still relatively limited due to the various immune escape mechanisms of tumors. Based on this, we introduce Neo-BCV, a novel bacterial [...] Read more.
Background: In the past decade, immunotherapy has become a major choice for the treatment of lung cancer, yet its therapeutic efficacy is still relatively limited due to the various immune escape mechanisms of tumors. Based on this, we introduce Neo-BCV, a novel bacterial composite vaccine designed to enhance immune responses against lung cancer. Methods: We investigated the immune enhancing effect of Neo-BCV through in vivo and in vitro experiments, including flow cytometry, RNA-seq, and Western blot. Results: We have demonstrated that Neo-BCV can promote Dendritic cells (DCs) maturation and induce DCs differentiation into pro-inflammatory subgroups, significantly enhancing cytotoxic T lymphocyte (CTL)-mediated anti-tumor responses. Transcriptome sequencing revealed that Neo-BCV exerts its effects by specifically inhibiting the JAK2-STAT3 signaling pathway, a crucial regulator of cancer progression, metabolism, and inflammation. Moreover, Neo-BCV significantly improved the immune microenvironment in both tumor and spleen tissues without inducing notable toxic effects in major organs. Conclusions: These findings highlight Neo-BCV’s potential as a safe and effective therapeutic strategy, offering a novel avenue for clinical translation in lung cancer immunotherapy. Full article
Show Figures

Figure 1

Figure 1
<p>The effect of Neo-BCV on BMDC maturation in vitro. (<b>A</b>) Morphology of BMDCs under light microscope. (<b>B</b>) Flow cytometry was used to detect the expression levels of CD86 and MHC-II on the surface of DCs, along with a semi-quantitative statistical chart. ** <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>Neo-BCV vaccine induces tumor-specific immune responses in mice. (<b>A</b>) Neo-BCV experimental treatment protocol; (<b>B</b>) Images of mouse tumor tissues after treatment; (<b>C</b>) Tumor growth curves in mice; (<b>D</b>) Tumor tissue weights in mice after treatment. **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Immune response induced by Neo-BCV in the tumor. (<b>A</b>) Flow cytometry analysis of CD8<sup>+</sup> T and CD4<sup>+</sup> T lymphocyte expression in tumor tissues and semi-quantitative statistical charts. (<b>B</b>) Flow cytometry analysis of cDC1, CD103<sup>+</sup> DC, and CD83<sup>+</sup> DC expression in tumor tissues and semi-quantitative statistical charts. (<b>C</b>) Flow cytometry analysis of cytotoxic T lymphocyte perforin and granzyme B expression in tumor tissues and semi-quantitative statistical charts. n = 6, data are presented as mean ± standard deviation ( <math display="inline"><semantics> <mrow> <mover accent="true"> <mrow> <mi mathvariant="normal">x</mi> </mrow> <mo>¯</mo> </mover> </mrow> </semantics></math> ± s). Inter-group comparisons were analyzed using 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; 0.0001; ns indicates no significant difference.</p>
Full article ">Figure 4
<p>Immune markers induced by Neo-BCV in the spleen. (<b>A</b>) Flow cytometry analysis of DC expression in spleen tissues and semi-quantitative statistical charts. (<b>B</b>,<b>C</b>) Expression levels of CCR7, CD44, CD69, CD83, CD86, and Ki-67 in the spleen of the PBS and Neo-BCV-treated groups, along with semi-quantitative statistical charts. n = 6; staining intensity was used as the analysis metric, and statistical comparisons were made using the average OD value of the positive regions. Data are presented as mean ± standard deviation ( <math display="inline"><semantics> <mrow> <mover accent="true"> <mrow> <mi mathvariant="normal">x</mi> </mrow> <mo>¯</mo> </mover> </mrow> </semantics></math> ± s). Inter-group comparisons were analyzed using 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, **** <span class="html-italic">p</span> &lt; 0.0001; ns indicates no significant difference.</p>
Full article ">Figure 5
<p>Tumor tissue transcriptomics.(<b>A</b>,<b>B</b>) Differential Gene Expression. (<b>A</b>) Volcano plot of differential genes, where the x-axis represents the fold change in gene or transcript expression between the two samples, and the y-axis represents the statistical significance of the differential expression, indicated by the <span class="html-italic">p</span>-value. (<b>B</b>) Clustering analysis of differential genes, with the color in the figure representing the expression level of that gene in this group of samples; red indicates high expression, while blue indicates low expression. (<b>C</b>,<b>D</b>) Enrichment Analysis of Differential Genes. (<b>C</b>) GO enrichment analysis, where each node represents a GO term, and the color intensity indicates the enrichment level; darker colors represent higher enrichment levels, with each node displaying the name of the GO term and the <span class="html-italic">p</span>-value from the enrichment analysis. (<b>D</b>) KEGG enrichment analysis, where the x-axis represents the enrichment factor and the y-axis represents the functional pathways enriched in the KEGG pathway. (<b>E</b>,<b>F</b>) Representative Western blot images and semi-quantitative statistical charts of the phosphorylation and total protein levels of STAT3 and JAK2 in tumor tissues. **** <span class="html-italic">p</span> &lt; 0.0001. The original Western blot figures can be found in <a href="#app1-vaccines-13-00064" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 6
<p>Safety considerations of Neo-BCV. (<b>A</b>) Body weight change curves of mice in different treatment groups. (<b>B</b>) H&amp;E staining of major organs (lung, liver, kidney, and heart) after two weeks of different treatments (scale bar = 100 μm). ns indicates no significant difference.</p>
Full article ">Figure 7
<p>Summary diagram. Neo-BCV mediates CTL anti-tumor immune responses by activating /dendritic cells (DCs). Simultaneously, it exerts anti-tumor effects through the inhibition of the JAK2-STAT3 signaling pathway.</p>
Full article ">
20 pages, 6986 KiB  
Article
Rab21-Targeted Nano Drug Delivery System-Based FFPG for Efficient Paclitaxel Delivery to Inhibit Lung Cancer Progression
by Jing Wang, Xueying Yan, Wenfei Wang, Shu Wang, Hongxiang Jiang, Xinhua Zhu, Zhehui Li, Defu Cai and Yonggang Xia
Pharmaceutics 2025, 17(1), 94; https://doi.org/10.3390/pharmaceutics17010094 (registering DOI) - 12 Jan 2025
Viewed by 216
Abstract
Background/Objectives: Platycodon grandiflorus (PG) has been widely researched as a conductant drug for the treatment of lung diseases by ancient and modern traditional Chinese medicine (TCM) practitioners. Inspired by the mechanism and our previous finding about fructans and fructooligosaccharides from Platycodon grandiflorus [...] Read more.
Background/Objectives: Platycodon grandiflorus (PG) has been widely researched as a conductant drug for the treatment of lung diseases by ancient and modern traditional Chinese medicine (TCM) practitioners. Inspired by the mechanism and our previous finding about fructans and fructooligosaccharides from Platycodon grandiflorus (FFPG), we developed a nano drug delivery system (NDDS) targeting lung cancer. The aim was to improve the efficiency of the liposomal delivery of Paclitaxel (PTX) and enhance the anti-tumor efficacy. Methods: The FFPG-Lip-PTX NDDS was prepared by electrostatic adsorption. Dynamic light scattering, zeta potential, and transmission electron microscopy were used for physical characterization. The release behavior of the NDDS was simulated by dialysis. The uptake of the NDDS was observed by confocal microscopy and flow cytometry. Cytotoxicity, apoptosis, migration, and invasion experiments were used to evaluate the anti-tumor ability of the NDDS in vitro. The penetration and inhibition of tumor proliferation were further analyzed via a 3D tumor sphere model. Finally, in vivo biological distribution and pharmacodynamic experiments verified the targeting and anti-tumor ability of the FFPG-Lip-PTX NDDS. Results: FFPG-Lip-PTX possessed a homogeneous particle size distribution, high encapsulation efficiency, and stability. In vitro experiments confirmed that FFPG promoted the uptake of the NNDS by tumor cells and enhanced cytotoxicity. It also increased the anti-tumor effect by promoting cell apoptosis and inhibiting invasion and metastasis. The same conclusion was obtained in 3D tumor spheres. In vivo experiments exhibited that FFPG-lips-PTX showed more significant lung cancer-targeting activity and anti-tumor effects. Conclusions: In this study, a novel lung-targeted NDDS is proposed to enhance the therapeutic effect of chemotherapy drugs on lung cancer. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

Figure 1
<p>Preparation and characterization of the physical and chemical properties of FFPG-Lip-PTX. (<b>a</b>) The representative images of FFPG-Lip-PTX at different FFPG concentrations and effect of FFPG concentration on FFPG-Lip-PTX particle diameter. (<b>b</b>) Effect of FFPG concentration on FFPG-Lip-PTX zeta potential. (<b>c</b>) TEM image of Lip-PTX and FFPG-Lip-PTX. (<b>d</b>) Stability studies of FFPG-Lip-PTX and Lip-PTX after incubation with PBS containing 50% FBS at 37 ℃ for 72 h. (<b>e</b>) The in vitro cumulative release profile of PTX from the Lip-PTX and FFPG-Lip-PTX at 37 ℃ during 48 h. Data are shown as mean ± SD (n = 3).</p>
Full article ">Figure 2
<p>Evaluation of extracapsular efficacy of FFPG-Lip-PTX liposomes on A549 and LLC cells after incubation for 1 h at 37 ℃. The final concentration of Cou6 was 100 ng/mL. Laser scanning confocal microscopy images of A549 (<b>a</b>) and LLC cells (<b>b</b>) incubated with different formulations. Green and blue indicate the fluorescence of Cou6 and Hoechst 33258, respectively. Flow cytometric quantitative determination of Cou 6 uptake on A549 cells (<b>c</b>) and LLC cells (<b>d</b>). Quantitative analysis of Cou6 uptake based on flow cytometric plots on A549 (<b>e</b>) and LLC cells (<b>f</b>). Each bar represents the mean fluorescence intensity ± SD (n = 3), * <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 3
<p>Evaluation of extracapsular efficacy of FFPG-Lip-PTX liposomes. The in vitro cytotoxicity of different PTX preparations to (<b>a</b>) A549 and (<b>b</b>) LLC cells was determined by the SRB method. (<b>c</b>) In vitro apoptosis of different PTX preparations in A549 and LLC cells was evaluated by flow cytometry. (<b>d</b>) In vitro inhibition of A549 cell migration and invasion by different PTX preparations observed using transwell images (30 ng/mL). Quantitative inhibition of A549 cell migration (<b>e</b>) and invasion (<b>f</b>). Data are expressed as mean ± SD (n = 3), * <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>Effects of different Cou6 and PTX preparations (PTX = 40 ng/mL) on the penetration and growth of A549 tumor spheres. (<b>a</b>) Silver light image of penetration intensity of different Cou6 preparations on 3D tumor spheres (sale bar, 50 μm). (<b>b</b>) Semi-quantitative penetration strength of different Cou6 preparations on tumor spheres. (<b>c</b>) Image of a typical tumor sphere under an inverted microscope. (<b>d</b>) Quantified size of tumor spheres at different time points. Data are expressed as mean ± SD (n = 3), * <span class="html-italic">p</span> &lt; <span class="html-italic">0</span>.05,** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Biological distribution of Lip-DiR and FFPG-Lip-DiR in LLC tumor-bearing mice. (<b>a</b>) LLC real-time imaging of the tail vein of tumor-bearing mice within 48 h. (<b>b</b>) Fluorescence images of tumors and organs in vitro. (<b>c</b>) Semi-quantitative analysis of fluorescence intensity at tumor site. Data are expressed as mean ± SD (n = 3), ** <span class="html-italic">p</span> &lt; 0.01. Tumors are represented by red ovals.</p>
Full article ">Figure 6
<p>Anti-tumor effects of different PTX prescriptions (10mg/kg) on LLC tumor-bearing mouse model. (<b>a</b>) Schematic diagram of administration of mouse lung cancer-transplanted tumor model. (<b>b</b>) Tumor volume curve. (<b>c</b>) Photographs of isolated tumor tissue. (<b>d</b>) The weight of the tumor removed. (<b>e</b>). H&amp;E pathological images of tumor tissue were resected, the proliferation images of tumor tissue were evaluated by IHC, and TUNEL fluorescence staining of apoptotic cells in tumor tissue was performed. (<b>f</b>). Weight change curve. The data are mean ± SD (n = 6), * <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 7
<p>The effects of FFPG on Rab21, Golgi apparatus, and endoplasmic reticulum of lung cancer cells were observed by the immunohistochemical method. (<b>a</b>) Bright field images of Rab21, Golgi apparatus, and endoplasmic reticulum, scale 100 μm. (<b>b</b>) Rab21, (<b>c</b>) Golgi, (<b>d</b>) positive rates of endoplasmic reticulum. The data were mean ± SD (n = 6), * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Scheme 1
<p>Scheme of FFPG-modified liposomes for targeted delivery of PTX in vivo.</p>
Full article ">
15 pages, 1190 KiB  
Article
MCF-DTI: Multi-Scale Convolutional Local–Global Feature Fusion for Drug–Target Interaction Prediction
by Jihong Wang, Ruijia He, Xiaodan Wang, Hongjian Li and Yulei Lu
Molecules 2025, 30(2), 274; https://doi.org/10.3390/molecules30020274 - 12 Jan 2025
Viewed by 212
Abstract
Predicting drug–target interactions (DTIs) is a crucial step in the development of new drugs and drug repurposing. In this paper, we propose a novel drug–target prediction model called MCF-DTI. The model utilizes the SMILES representation of drugs and the sequence features of targets, [...] Read more.
Predicting drug–target interactions (DTIs) is a crucial step in the development of new drugs and drug repurposing. In this paper, we propose a novel drug–target prediction model called MCF-DTI. The model utilizes the SMILES representation of drugs and the sequence features of targets, employing a multi-scale convolutional neural network (MSCNN) with parallel shared-weight modules to extract features from the drug side. For the target side, it combines MSCNN with Transformer modules to capture both local and global features effectively. The extracted features are then weighted and fused, enabling comprehensive feature representation to enhance the predictive power of the model. Experimental results on the Davis dataset demonstrate that MCF-DTI achieves an AUC of 0.9746 and an AUPR of 0.9542, outperforming other state-of-the-art models. Our case study demonstrates that our model effectively validated several known drug–target relationships in lung cancer and predicted the therapeutic potential of certain preclinical compounds in treating lung cancer. These findings contribute valuable insights for subsequent drug repurposing efforts and novel drug development. Full article
27 pages, 796 KiB  
Review
Immunotherapy in Oncogene-Addicted NSCLC: Evidence and Therapeutic Approaches
by Lorenzo Foffano, Elisa Bertoli, Martina Bortolot, Sara Torresan, Elisa De Carlo, Brigida Stanzione, Alessandro Del Conte, Fabio Puglisi, Michele Spina and Alessandra Bearz
Int. J. Mol. Sci. 2025, 26(2), 583; https://doi.org/10.3390/ijms26020583 - 11 Jan 2025
Viewed by 475
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. The discovery of specific driver mutations has revolutionized the treatment landscape of oncogene-addicted NSCLC through targeted therapies, significantly improving patient outcomes. However, immune checkpoint inhibitors (ICIs) have demonstrated limited effectiveness [...] Read more.
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. The discovery of specific driver mutations has revolutionized the treatment landscape of oncogene-addicted NSCLC through targeted therapies, significantly improving patient outcomes. However, immune checkpoint inhibitors (ICIs) have demonstrated limited effectiveness in this context. Emerging evidence, though, reveals significant heterogeneity among different driver mutation subgroups, suggesting that certain patient subsets may benefit from ICIs, particularly when combined with other therapeutic modalities. In this review, we comprehensively examine the current evidence on the efficacy of immunotherapy in oncogene-addicted NSCLC. By analyzing recent clinical trials and preclinical studies, along with an overview of mechanisms that may reduce immunotherapy efficacy, we explored potential strategies to address these challenges, to provide insights that could optimize immunotherapy approaches and integrate them effectively into the treatment algorithm for oncogene-addicted NSCLC. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

Figure 1
<p>Emerging therapeutic strategies to enhance immunotherapy efficacy in oncogene-addicted NSCLC. This figure illustrates the interaction of various combination approaches with the tumor microenvironment (TME) in lung cancer. The approaches highlighted include tyrosine kinase inhibitors (TKIs), antibody–drug conjugates (ADCs), Vascular Endothelial Growth Factor Inhibitors (VEGFis), immune checkpoint inhibitors (ICIs), chimeric antigen receptor T cell therapy (CAR-T), cancer vaccines, and oncolytic viruses. Each therapeutic strategy is depicted in relation to its mechanism of action within the TME.</p>
Full article ">
19 pages, 2508 KiB  
Article
Machine Learning-Based Radiomics Analysis for Identifying KRAS Mutations in Non-Small-Cell Lung Cancer from CT Images: Challenges, Insights and Implications
by Mirjam Schöneck, Nicolas Rehbach, Lars Lotter-Becker, Thorsten Persigehl, Simon Lennartz and Liliana Lourenco Caldeira
Life 2025, 15(1), 83; https://doi.org/10.3390/life15010083 (registering DOI) - 11 Jan 2025
Viewed by 205
Abstract
Kirsten Rat Sarcoma viral oncogene homolog (KRAS) is a frequently occurring mutation in non-small-cell lung cancer (NSCLC) and influences cancer treatment and disease progression. In this study, a machine learning (ML) pipeline was applied to radiomic features extracted from public and internal CT [...] Read more.
Kirsten Rat Sarcoma viral oncogene homolog (KRAS) is a frequently occurring mutation in non-small-cell lung cancer (NSCLC) and influences cancer treatment and disease progression. In this study, a machine learning (ML) pipeline was applied to radiomic features extracted from public and internal CT images to identify KRAS mutations in NSCLC patients. Both datasets were analyzed using parametric (ttest) and non-parametric statistical tests (Mann–Whitney U test) and dimensionality reduction techniques. Afterwards, the proposed ML pipeline was applied to both datasets using a five-fold cross-validation on the training set (70/30 train/test split) before being validated on the other dataset. The results show that the radiomic features are significantly different (Mann–Whitney U test; p < 0.05) between the two datasets, despite the use of identical feature extraction methods. Model transferability is therefore difficult to achieve, which became evident during external testing (F1 score = 0.41). Oversampling, undersampling, clustering and harmonization techniques were applied to balance and harmonize the datasets, but did not improve the classification of KRAS mutation presence. In general, due to only a single moderate result (highest test F1 score = 0.67), the accuracy of KRAS prediction is not sufficient for clinical application. In future work, the complexity of KRAS mutation might be addressed by taking submutations into consideration. Larger multicentric datasets with balanced tumor stages, including multi-scanner datasets, seem to be necessary for building robust predictive models. Full article
13 pages, 727 KiB  
Article
The Potential Role of sPD-L1 as a Predictive Biomarker in EGFR-Positive Non-Small-Cell Lung Cancer
by Vesna Ćeriman Krstić, Dragana Jovanović, Natalija Samardžić, Milija Gajić, Jelena Kotur Stevuljević, Aleksandra Klisic, Ivan Soldatović, Damir Radončić, Marina Roksandić Milenković, Biljana Šeha, Nikola Čolić, Katarina Lukić and Milan Savić
Curr. Issues Mol. Biol. 2025, 47(1), 45; https://doi.org/10.3390/cimb47010045 - 11 Jan 2025
Viewed by 217
Abstract
Background/Objectives: A significant breakthrough in non-small-cell lung cancer (NSCLC) treatment has occurred with the introduction of targeted therapies and immunotherapy. However, not all patients treated with these therapies would respond to treatment, and patients who respond to treatment would acquire resistance at some [...] Read more.
Background/Objectives: A significant breakthrough in non-small-cell lung cancer (NSCLC) treatment has occurred with the introduction of targeted therapies and immunotherapy. However, not all patients treated with these therapies would respond to treatment, and patients who respond to treatment would acquire resistance at some time point. This is why we need new biomarkers that can predict response to therapy. The aim of this study was to investigate whether soluble programmed cell death-ligand 1 (sPD-L1) could be a predictive biomarker in patients with epidermal growth factor receptor (EGFR)-positive NSCLC. Materials and Methods: Blood samples from 35 patients with EGFR-mutated (EGFRmut) adenocarcinoma who achieved disease control with EGFR tyrosine kinase inhibitor (EGFR TKI) therapy were collected for sPD-L1 analysis. We analyzed sPD-L1 concentrations in 30 healthy middle-aged subjects, as a control population, to determine the reference range. Adenocarcinoma patients were divided into two groups, i.e., a group with low sPD-L1 (≤182.5 ng/L) and a group with high sPD-L1 (>182.5 ng/L). Results: We found that progression-free survival (PFS) was 18 months, 95% CI (11.1–24.9), for patients with low sPD-L1 and 25 months, 95% CI (8.3–41.7), for patients with high sPD-L1. There was no statistically significant difference in PFS between the groups (p = 0.100). Overall survival (OS) was 34.4 months, 95% CI (26.6–42.2), for patients with low sPD-L1 and 84.1 months, 95% CI (50.6–117.6), for patients with high sPD-L1; there was also no statistically significant difference between the groups (p = 0.114). Conclusion: In our study, we found that patients with high sPD-L1 had numerically better PFS and OS, but this has no statistical significance. Further studies with a larger number of patients are needed to evaluate the role of sPD-L1 as a predictive biomarker in patients with EGFRmut NSCLC. Full article
(This article belongs to the Special Issue Targeting Tumor Microenvironment for Cancer Therapy, 3rd Edition)
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier curves of PFS in patients with low and high sPD-L1. Patients with high sPD-L1 levels had better PFS compared to patients with low sPD-L1, but there was no statistical significance (<span class="html-italic">p</span> = 0.100).</p>
Full article ">Figure 2
<p>Kaplan–Meier curves of OS in patients with low and high sPD-L1. Patients with high sPD-L1 levels had better OS compared to patients with low sPD-L1, but there was no statistical significance (<span class="html-italic">p</span> = 0.114).</p>
Full article ">
12 pages, 1520 KiB  
Article
Incidence and Prevalence of Bone Metastases in Different Solid Tumors Determined by Natural Language Processing of CT Reports
by Niamh Long, David Woodlock, Robert D’Agostino, Gary Nguyen, Natalie Gangai, Varadan Sevilimedu and Richard Kinh Gian Do
Cancers 2025, 17(2), 218; https://doi.org/10.3390/cancers17020218 - 11 Jan 2025
Viewed by 263
Abstract
Background/Objectives: Improved survival due to advances in medical therapy has resulted in increasing numbers of cancer patients living with bone metastases; however, our understanding of the prognostic implications of bone metastases requires larger population-based studies outlining their incidence and prevalence in different primary [...] Read more.
Background/Objectives: Improved survival due to advances in medical therapy has resulted in increasing numbers of cancer patients living with bone metastases; however, our understanding of the prognostic implications of bone metastases requires larger population-based studies outlining their incidence and prevalence in different primary cancer types, including those with lower incidence. This study aimed to evaluate the incidence and prevalence of bone metastases in solid organ tumors by analyzing reports of staging CT studies with natural language processing (NLP). Methods: In this retrospective study, 639,470 reports representing 129,326 unique patients were analyzed; 6279 randomly selected reports were manually annotated and labeled for the presence or absence of bone metastases. From these data, a BERT-based NLP model was developed and applied to the patient database. The cumulative incidence at 5 years and prevalence of bone metastases in each cancer type were calculated. Results: The accuracy of the NLP model on a validation set was 97.1%, with a positive predictive value (precision) of 88.0% and a sensitivity (recall) of 86.3%. The 5-year incidence rate of bone metastases was highest in prostate, breast, head and neck, and lung cancer (52%, 41%, 36%, 33%). Incidence was lowest in central nervous system cancer and testicular cancer (8%, 5%). Prevalence was highest in prostate, breast, and lung cancer (32%, 25% and 23%), and lowest in central nervous system cancer and testicular cancer (4%, 4%). Conclusions: NLP was utilized to demonstrate patterns of bone metastases in a broad range of cancer types and is a valuable tool in population-based assessment of bone metastases. Full article
Show Figures

Figure 1

Figure 1
<p>Flowchart detailing the sequential steps in the development of the natural language processing model. There were 639,470 consecutive CT reports first identified between 2009 and 2021. Manual curation was performed on about 1% of reports (6279 report), which were split into training (80%) and testing sets (20%) to build a BERT-based natural language processing model. The final model was evaluated on a validation set of 448 reports and applied to the remaining unlabeled reports. Rule-based labelling was used on a subset of records where the default language (e.g., “unremarkable”) was used in our structured reports.</p>
Full article ">Figure 2
<p>Bone metastases from primary cancers with the highest incidence rates. (<b>A</b>) Sclerotic pelvic metastases in a 76-year-old male patient with prostate cancer. (<b>B</b>) Lytic metastasis in the left third rib in a 60-year-old female patient with breast cancer. (<b>C</b>) Lytic metastasis left pubic bone in a 48-year-old female patient with thyroid cancer. (<b>D</b>) Sclerotic metastasis in an L1 vertebral body in a 59-year-old male patient with adenoid cystic carcinoma of the tongue. (<b>E</b>) Lytic metastases in the left sacral ala and left ilium in a 65-year-old male patient with poorly differentiated lung adenocarcinoma. (<b>F</b>) Multiple sclerotic pelvic bone metastases in a 57-year-old male with a lung carcinoid tumor. (<b>G</b>) Sclerotic right iliac metastasis in a 63-year-old female patient with melanoma. (<b>H</b>) Lytic left sacral metastasis in a 55-year-old female patient with melanoma. (<b>I</b>) Lytic left second rib metastasis in a 60-year-old female patient with hepatocellular carcinoma. (<b>J</b>) Multiple lumbar spine sclerotic metastases in a 92-year-old male patient with hepatocellular carcinoma. (<b>K</b>) Lytic left iliac metastasis in a 67-year-old male patient with esophageal cancer. (<b>L</b>) Follow-up demonstrating interval sclerosis of the left iliac metastasis representing treatment effect.</p>
Full article ">Figure 3
<p>Bone metastases from primary cancers with the highest incidence rates. (<b>A</b>) Sclerotic pelvic metastases in a 76-year-old male patient with prostate cancer. (<b>B</b>) Left 5th rib lytic metastasis in an 84-year-old male with colorectal cancer. (<b>C</b>) Lytic metastasis T5 vertebra in a 64-year-old female patient with ovarian cancer. (<b>D</b>) Sclerotic metastasis right acetabulum in a 62-year-old male patient with urothelial cancer. (<b>E</b>) Lytic metastasis left ilium in a 54-year-old female patient with renal cell cancer. (<b>F</b>) Lytic metastasis left ilium in a 44-year-old male patient with renal cell carcinoma. (<b>G</b>) Multiple sclerotic lumbar spine metastases in a 54-year-old male patient with urothelial carcinoma of the renal pelvis. (<b>H</b>) Sagittal CT lumbar spine of multifocal sclerotic osseous metastases in a 35-year-old female patient with HER2-negative gastric cancer.</p>
Full article ">Figure 4
<p>Kaplan–Meier curves of eight primary tumors: prostate, pancreas, ovary, lung, hepatobiliary, colorectal, stomach, and central nervous system (CNS). 50% of patients with prostate cancer developed bone metastases within five years (dashed lines).</p>
Full article ">
19 pages, 6027 KiB  
Article
The X-Linked Tumor Suppressor TSPX Regulates Genes Involved in the EGFR Signaling Pathway and Cell Viability to Suppress Lung Adenocarcinoma
by Tatsuo Kido, Hui Kong and Yun-Fai Chris Lau
Genes 2025, 16(1), 75; https://doi.org/10.3390/genes16010075 (registering DOI) - 11 Jan 2025
Viewed by 251
Abstract
Background: TSPX is an X-linked tumor suppressor that was initially identified in non-small cell lung cancer (NSCLC) cell lines. However, its expression patterns and downstream mechanisms in NSCLC remain unclear. This study aims to investigate the functions of TSPX in NSCLC by identifying [...] Read more.
Background: TSPX is an X-linked tumor suppressor that was initially identified in non-small cell lung cancer (NSCLC) cell lines. However, its expression patterns and downstream mechanisms in NSCLC remain unclear. This study aims to investigate the functions of TSPX in NSCLC by identifying its potential downstream targets and their correlation with clinical outcomes. Methods: RNA-seq transcriptome and pathway enrichment analyses were conducted on the TSPX-overexpressing NSCLC cell lines, A549 and SK-MES-1, originating from lung adenocarcinoma and squamous cell carcinoma subtypes, respectively. In addition, comparative analyses were performed using the data from clinical NSCLC specimens (515 lung adenocarcinomas and 502 lung squamous cell carcinomas) in the Cancer Genome Atlas (TCGA) database. Results: TCGA data analysis revealed significant downregulation of TSPX in NSCLC tumors compared to adjacent non-cancerous tissues (Wilcoxon matched pairs signed rank test p < 0.0001). Notably, the TSPX expression levels were inversely correlated with the cancer stage, and higher TSPX levels were associated with better clinical outcomes and improved survival in lung adenocarcinoma, a subtype of NSCLC (median survival extended by 510 days; log-rank test, p = 0.0025). RNA-seq analysis of the TSPX-overexpressing NSCLC cell lines revealed that TSPX regulates various genes involved in the cancer-related signaling pathways and cell viability, consistent with the suppression of cell proliferation in cell culture assays. Notably, various potential downstream targets of TSPX that correlated with patient survival (log-rank test, p = 0.016 to 4.3 × 10−10) were identified, including EGFR pathway-related genes AREG, EREG, FOSL1, and MYC, which were downregulated. Conclusions: Our results suggest that TSPX plays a critical role in suppressing NSCLC progression by downregulating pro-oncogenic genes, particularly those in the EGFR signaling pathway, and upregulating the tumor suppressors, especially in lung adenocarcinoma. These findings suggest that TSPX is a potential biomarker and therapeutic target for NSCLC management. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>TSPX expression levels in relation to the pathologic stage and survival ratios of lung adenocarcinoma and squamous cell carcinoma patients. (<b>A</b>) TSPX expression levels in 58 lung adenocarcinoma tumor (T)/non-tumor (NT) paired samples from TCGA. Expression values (RSEM normalized count values) were plotted, with paired samples linked by a solid line; blue, decrease; red, increase. The <span class="html-italic">p</span>-value of the Wilcoxon matched pairs signed rank test is shown. (<b>B</b>) TSPX expression levels in 59 NT and 515 lung adenocarcinoma cases. The latter were divided into the TSPX-high group (highest 25%, <span class="html-italic">n</span> = 129), TSPX-low (lowest 25% cases, <span class="html-italic">n</span> = 129), and TSPX-mid (<span class="html-italic">n</span> = 257) group. (<b>C</b>) Distributions of pathologic stages (I-IV) across the TSPX-low, TSPX-mid, and TSPX-high groups. Chi-squared test <span class="html-italic">p</span>-value is indicated. (<b>D</b>) Survival curves for the TSPX-high (red), TSPX-mid (brown), and TSPX-low (blue) groups. Log-rank test <span class="html-italic">p</span>-value is indicated. (<b>E</b>) TSPX expression levels in 51 lung squamous cell carcinoma tumor/non-tumor paired samples from TCGA, similar to A. (<b>F</b>) TSPX expression levels in 51 NT and 502 lung squamous cell carcinoma cases, categorized into TSPX-high (highest 25% cases, <span class="html-italic">n</span> = 126), TSPX-low (lowest 25% cases, <span class="html-italic">n</span> = 126), and TSPX-mid (<span class="html-italic">n</span> = 246) groups. (<b>G</b>) Distributions of pathologic stages between the TSPX-low, TSPX-mid, and TSPX-high groups for lung squamous cell carcinoma, similar to C. Red indicates Stage-IV. Chi-squared test <span class="html-italic">p</span>-value is indicated. (<b>H</b>) Survival curves for the TSPX-high (red), TSPX-mid (brown), and TSPX-low (blue) groups in lung squamous cell carcinoma. Log-rank test <span class="html-italic">p</span>-value is indicated.</p>
Full article ">Figure 2
<p>Overexpression of TSPX inhibits cell proliferation and causes cell death in A549 cells. (<b>A</b>) Diagram of the tet-ON transgene activation system. Addition of doxycycline (Dox) in the culture medium recruited the transactivator rtTA onto the tetracycline responsive (TRE) promoter and activated the target transgene. (<b>B</b>) The expressions of EGFP and TSPX in the respective transduced A549 cells with (+) and without (−) Dox induction were confirmed by Western blot, using β-actin as a reference. (<b>C</b>) Immunofluorescence of EGFP (green, left), TSPX (red, middle), and DAPI staining (blue) in the respective transduced A549 cells at 24 h after Dox induction. Far right panels show the merged images of TSPX, EGFP, and DAPI staining. (<b>D</b>) Scratch tests for A549-tetON-EGFP cells (left) and A549-tetON-TSPX cells (right) under a Dox-induction condition. Far right panel shows a magnified image of the boxed area. Yellow line indicates the wound edges at 0 h. Pink arrows indicate detached A549-tetON-TSPX cells. (<b>E</b>) Annexin-V binding assay (red) at 48 h after Dox induction showed that the detached A549-tetON-TSPX cells were positively stained by Annexin-V conjugated with Alexa Fluor 594 (red), corresponding to dead or apoptotic cells. Yellow arrows indicate cells stained with Annexin-V/Alexa Fluor 594. (<b>F</b>) Cell proliferation of A549-tetON-TSPX cells was inhibited under the Dox-induction condition (+Dox, right) but not under uninduced condition (−Dox, left), comparing with A549-tetON-EGFP cells, indicating that the TSPX overexpression inhibited cell proliferation. Asterisks indicate the significant difference at Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span>-value &lt; 0.05. Scale bar = 50 µm in (<b>C</b>), 100 µm in (<b>D</b>,<b>E</b>).</p>
Full article ">Figure 3
<p>Overexpression of TSPX inhibits cell proliferation in SK-MES-1 cells. (<b>A</b>) The expressions of EGFP and TSPX in the respective transduced SK-MES-1 cells with (+) and without (−) Dox induction were confirmed by Western blot. β-actin was analyzed as a reference. (<b>B</b>) Immunofluorescence of EGFP (green), TSPX (red), and DAPI staining (blue) in the respective transduced SK-MES-1 cells at 24 h after Dox-induction. Right panels show the merged images of TSPX and DAPI staining. (<b>C</b>) Cell proliferation of MES1-tetON-TSPX cells was inhibited under the Dox-induction condition (+Dox, right) but not under uninduced condition (−Dox, left), comparing with MES1-tetON-EGFP cells, indicating that the TSPX overexpression inhibited cell proliferation in SK-MES-1 cells. Asterisks indicate the significant difference at Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span>-value &lt; 0.05. (<b>D</b>) Scratch tests for MES1-tetON-TSPX cells (top panels) and MES1-tetON-EGFP cells (bottom panels) under a Dox-induction condition. Phase contrast images show the representative gaps at 0 h and 48 h after scratch and Dox-induction. Far right panels show digitally magnified images of the boxed areas, respectively. The scratched area was completely healed by MES1-tetON-EGFP, but not by MES1-tetON-TSPX cells at 48 h. Scale bar = 100 µm in (<b>B</b>), 200 µm in (<b>D</b>).</p>
Full article ">Figure 4
<p>Identification of the TSPX downstream genes and their biological processes in A549 and SK-MES-1 cells. (<b>A</b>) Volcano plots representing DEGs between TSPX-overexpressing and control cells in A549 (left) and SK-MES-1 (right, abbreviated as MES1) cell lines. The list of DEGs is shown in <a href="#app1-genes-16-00075" class="html-app">Supplementary Table S1</a>. (<b>B</b>) Results of the DAVID pathway enrichment analysis for DEGs in TSPX-overexpressing A549 and SK-MES-1 cells. The pathways that were commonly enriched in both A549 and SK-MES-1 cells are shown on the right. A magnified image of all enriched pathways, including the cell line-specific pathways, is shown in <a href="#app1-genes-16-00075" class="html-app">Supplementary Figure S1</a>. (<b>C</b>) Gene expression changes in DEGs involved in the selected pathways in (<b>B</b>). A549, DEGs in TSPX-overexpressing A549 cells; MES1, DEGs in TSPX-overexpressing SK-MES-1 cells. Only DEGs shared between A549 and SK-MES-1 cells are shown. Arrows indicate the genes analyzed in (<b>D</b>). (<b>D</b>) Validation of the gene expression changes induced by TSPX overexpression in A549 cells and SK-MES-1 cells by quantitative RT-PCR. Values represent the relative expression levels of AREG, DKK1, EREG, FOSL1, and MYC in A549-tetON-TSPX cells or MES1-tetON-TSPX cells (TSPX) at 24 h after Dox induction (mean ± standard error, <span class="html-italic">n</span> = 3). A549-tetON-EGFP cells and MES1-tetON-EGFP cells were used as references (EGFP), respectively. Values were normalized against GAPDH as an internal control. **, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span>-value ≤ 0.001; ***, <span class="html-italic">p</span>-value ≤ 0.0001.</p>
Full article ">Figure 5
<p>Gene expression patterns of TSPX-downstream genes in clinical lung adenocarcinoma from TCGA datasets. (<b>A</b>) Expression levels (RSEM normalized read counts) of <span class="html-italic">AREG</span>, <span class="html-italic">BIRC3</span>, <span class="html-italic">CXCL5</span>, <span class="html-italic">DKK1</span>, <span class="html-italic">EREG</span>, <span class="html-italic">FOSL1</span>, <span class="html-italic">MYC</span>, <span class="html-italic">PLAU</span>, and <span class="html-italic">CACNA2D2</span>, in non-tumor lung tissues (NT), TSPX-low lung adenocarcinoma group (TSPX-low), and TSPX-high lung adenocarcinoma group (TSPX-high) are shown. Statistical significance was determined by one-way ANOVA with Tukey’s multiple comparison test; * <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01; *** <span class="html-italic">p</span> ≤ 0.001; **** <span class="html-italic">p</span> ≤ 0.0001; ns, not significant. Error bars represent mean ± SEM. (<b>B</b>) Correlation between the expression levels of the indicated genes and patient survival. Survival curves for high expressors (orange) or low expressors (green) are shown. Log-rank test <span class="html-italic">p</span>-values were obtained from TCGA datasets via the Human Protein Atlas (HPA).</p>
Full article ">Figure 6
<p>Schematic diagram illustrating the functions of the identified TSPX-downstream genes in the EGFP-signaling and other pathways involved in cell survival in lung adenocarcinoma. Details of respective functions are described in the body text.</p>
Full article ">
22 pages, 2196 KiB  
Article
Anti-Inflammatory and Anti-Migratory Effects of Morin on Non-Small-Cell Lung Cancer Metastasis via Inhibition of NLRP3/MAPK Signaling Pathway
by Punnida Arjsri, Kamonwan Srisawad, Sonthaya Umsumarng, Pilaiporn Thippraphan, Songyot Anuchapreeda and Pornngarm Dejkriengkraikul
Biomolecules 2025, 15(1), 103; https://doi.org/10.3390/biom15010103 - 10 Jan 2025
Viewed by 323
Abstract
Non-small-cell lung cancer (NSCLC) remains the leading cause of cancer-related deaths globally, with a persistently low five-year survival rate of only 14–17%. High rates of metastasis contribute significantly to the poor prognosis of NSCLC, in which inflammation plays an important role by enhancing [...] Read more.
Non-small-cell lung cancer (NSCLC) remains the leading cause of cancer-related deaths globally, with a persistently low five-year survival rate of only 14–17%. High rates of metastasis contribute significantly to the poor prognosis of NSCLC, in which inflammation plays an important role by enhancing tumor growth, angiogenesis, and metastasis. Targeting inflammatory pathways within cancer cells may thus represent a promising strategy for inhibiting NSCLC metastasis. This study evaluated the anti-inflammatory and anti-metastatic properties of morin, a bioactive compound derived from a Thai medicinal herb, focusing on its effects on NLRP3 inflammasome-mediated pathways in an in vitro NSCLC model. The A549 and H1299 cell lines were stimulated with lipopolysaccharide (LPS) and adenosine triphosphate (ATP) to activate the NLRP3 pathway. The inhibition effects exhibited by morin in reducing pro-inflammatory secretion in LPS- and ATP-stimulated NSCLC cells were assessed by ELISA, while wound healing and trans-well invasion assays evaluated its impact on cell migration and invasion. RT-qPCR measurement quantified the expression of inflammatory genes, and zymography and Western blotting were used to examine changes in invasive protein levels, epithelial-to-mesenchymal transition (EMT) markers, and underlying molecular mechanisms. Our findings demonstrated the significant ability of morin to decrease the production of IL-1β, IL-18, and IL-6 in a dose-dependent manner (p < 0.05), as well as suppress NSCLC cell migration and invasion. Morin downregulated invasive proteins (MMP-2, MMP-9, u-PAR, u-PA, MT1-MMP) and EMT markers (fibronectin, N-cadherin, vimentin) (p < 0.01) while also reducing the mRNA levels of NLRP3, IL-1β, IL-18, and IL-6. Mechanistic investigations revealed that morin suppressed NLRP3 inflammasome activity and inactivated MAPK pathways. Specifically, it decreased the expression of NLRP3 and ASC proteins and reduced caspase-1 activity, while reducing the phosphorylation of ERK, JNK, and p38 proteins. Collectively, these findings suggest that morin’s inactivation of the NLRP3 inflammasome pathway could offer a novel therapeutic strategy for counteracting pro-tumorigenic inflammation and metastatic progression in NSCLC. Full article
(This article belongs to the Special Issue Inflammation—The Surprising Bridge between Diseases)
25 pages, 406 KiB  
Review
Matters of the Heart: Cardiotoxicity Related to Target Therapy in Oncogene-Addicted Non-Small Cell Lung Cancer
by Sara Torresan, Martina Bortolot, Elisa De Carlo, Elisa Bertoli, Brigida Stanzione, Alessandro Del Conte, Michele Spina and Alessandra Bearz
Int. J. Mol. Sci. 2025, 26(2), 554; https://doi.org/10.3390/ijms26020554 - 10 Jan 2025
Viewed by 336
Abstract
The treatment of Non Small Cell Lung Cancer (NSCLC) has been revolutionised by the introduction of targeted therapies. With the improvement of response and frequently of overall survival, however, a whole new set of adverse events emerged. In fact, due to the peculiar [...] Read more.
The treatment of Non Small Cell Lung Cancer (NSCLC) has been revolutionised by the introduction of targeted therapies. With the improvement of response and frequently of overall survival, however, a whole new set of adverse events emerged. In fact, due to the peculiar mechanism of action of each one of the tyrosine kinase inhibitors and other targeted therapies, every drug has its own specific safety profile. In addition, this safety profile could not fully emerge from clinical trials data, as patients in clinical practice usually have more comorbidities and frailties. Cardiotoxicity is a well-known and established adverse event of anti-cancer therapies. However, only recently it has become a central topic for targeted therapies in NSCLC, due to the unknown real range and frequency. Management of this toxicity begins with prevention, and must balance the need of continuing an effective anticancer treatment versus low risk of even fatal events and the preservation of long-term quality of life. The aim of this review is to summarise the current knowledge focusing on currently used targeted therapies in NSCLC. Full article
(This article belongs to the Section Molecular Oncology)
14 pages, 1040 KiB  
Study Protocol
Peripheral Extracellular Vesicles for Diagnosis and Prognosis of Resectable Lung Cancer: The LUCEx Study Protocol
by Jorge Rodríguez-Sanz, Nadia Muñoz-González, José Pablo Cubero, Pablo Ordoñez, Victoria Gil, Raquel Langarita, Myriam Ruiz, Marta Forner, Marta Marín-Oto, Elisabet Vera, Pedro Baptista, Francesca Polverino, Juan Antonio Domingo, Javier García-Tirado, José María Marin and David Sanz-Rubio
J. Clin. Med. 2025, 14(2), 411; https://doi.org/10.3390/jcm14020411 - 10 Jan 2025
Viewed by 302
Abstract
Background/Objectives: Lung cancer is the primary cause of cancer-related deaths. Most patients are typically diagnosed at advanced stages. Low-dose computed tomography (LDCT) has been proven to reduce lung cancer mortality, but screening programs using LDCT are associated with a high number of false [...] Read more.
Background/Objectives: Lung cancer is the primary cause of cancer-related deaths. Most patients are typically diagnosed at advanced stages. Low-dose computed tomography (LDCT) has been proven to reduce lung cancer mortality, but screening programs using LDCT are associated with a high number of false positives and unnecessary thoracotomies. It is therefore imperative that a certain diagnosis is refined, especially in cases of solitary pulmonary nodules that are difficult to technically access for an accurate preoperative diagnosis. Extracellular vesicles (EVs) involved in intercellular communication may be an innovative biomarker for diagnosis and therapeutic strategies in lung cancer, regarding their ability to carry tumor-specific cargo. The aim of the LUCEx study is to determine if extracellular vesicle cargoes from both lung tissue and blood could provide complementary information to screen lung cancer patients and enable personalized follow-up after the surgery. Methods: The LUCEx study is a prospective study aiming to recruit 600 patients with lung cancer and 50 control subjects (false positives) undergoing surgery after diagnostic imaging for suspected pulmonary nodules using computed tomography (CT) scans. These patients will undergo curative surgery at the Department of Thoracic Surgery of the Miguel Servet Hospital in Zaragoza, Spain, and will be followed-up for at least 5 years. At baseline, samples from both tumor distal lung tissue and preoperative peripheral blood will be collected and processed to compare the quantity and content of EVs, particularly their micro-RNA (miRNA) cargo. At the third and fifth years of follow-up, CT scans, functional respiratory tests, and blood extractions will be performed. Discussion: Extracellular vesicles and their miRNA have emerged as promising tools for the diagnosis and prognosis of several diseases, including cancer. The LUCEx study, based on an observational clinical cohort, aims to understand the role of these vesicles and their translational potential as complementary tools for imaging diagnosis and prognosis. Full article
(This article belongs to the Section Pulmonology)
Show Figures

Figure 1

Figure 1
<p>Exosome isolation process in solid tissue (created in <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">Figure 2
<p>Exosome isolation process in blood plasma (created in <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">Figure 3
<p>Planned flowchart for the sample size described.</p>
Full article ">
14 pages, 1588 KiB  
Article
Natural Killer Cell-Secreted IFN-γ and TNF-α Mediated Differentiation in Lung Stem-like Tumors, Leading to the Susceptibility of the Tumors to Chemotherapeutic Drugs
by Kawaljit Kaur, Angie Perez Celis and Anahid Jewett
Cells 2025, 14(2), 90; https://doi.org/10.3390/cells14020090 - 10 Jan 2025
Viewed by 409
Abstract
We demonstrate that natural killer (NK) cells induce a higher cytotoxicity against lung cancer stem-like cells (hA549) compared to differentiated lung cancer cell lines (H292). The supernatants from split-anergized NK cells (IL-2 and anti-CD16 mAb-treated NK cells) induced differentiation in hA549. Differentiated lung [...] Read more.
We demonstrate that natural killer (NK) cells induce a higher cytotoxicity against lung cancer stem-like cells (hA549) compared to differentiated lung cancer cell lines (H292). The supernatants from split-anergized NK cells (IL-2 and anti-CD16 mAb-treated NK cells) induced differentiation in hA549. Differentiated lung cancer cell line (H292) and NK cells differentiated hA549 expressed reduced NK cell-mediated cytotoxicity but expressed higher sensitivity to chemotherapeutic drugs. This finding validated our previous reports demonstrating that the levels of tumor killing by NK cells and by chemotherapeutic drugs correlate directly and indirectly, respectively, with the stage and levels of tumor differentiation. We also demonstrate the role of IFN-γ and TNF-α in inducing tumor differentiation. NK cells’ supernatants or IFN-γ and TNF-α-induced tumor differentiation was blocked when we used antibodies against IFN-γ and TNF-α. Therefore, IFN-γ and TNF-α released from NK cells play a significant role in differentiating tumors, resulting in increased susceptibility of tumors to chemotherapeutic drugs. We also observed the different effects of MHC-class I antibodies in CSCs vs. differentiated tumors. Treatment with anti-MHC-class I decreased NK cell-mediated cytotoxicity in hA549 tumors, whereas it increased NK cell-mediated cytotoxicity when differentiated tumors were treated with antibodies against MHC-class I. Full article
(This article belongs to the Special Issue Spotlight on Natural Killer Cells in Immuno-Oncology)
Show Figures

Figure 1

Figure 1
<p>Different levels of killing of stem-like vs. differentiated tumors by NK cells and chemo-drugs. Untreated, IL-2 treated, and IL-2 + anti-CD16 mAb-treated NK cells (1 × 10<sup>6</sup> cells/mL) of healthy individuals were incubated overnight before they were used as effector cells against <sup>51</sup>Cr-labeled hA549 or H292 lung cancer cell lines at different NK-to-tumor (Effector: Target) ratios using a 4-h <sup>51</sup>Cr release assay. The lytic unit (LU) 30/10<sup>6</sup> cells were calculated based on an inverse number of NK cells being needed to kill 30% of the target cells × 100 (<b>A</b>). hA549 and H292 lung cancer cell lines were treated with paclitaxel (40 µg/mL) (<b>B</b>) and cisplatin (CDDP) (40 µg/mL) (<b>C</b>) overnight before the tumors were stained with propidium iodide (PI); percent cell viability was determined using a flow cytometer (<b>B</b>,<b>C</b>). *** (<span class="html-italic">p</span> value &lt; 0.001); ** (<span class="html-italic">p</span> value 0.001–0.01); * (<span class="html-italic">p</span> value 0.01–0.05).</p>
Full article ">Figure 2
<p>IL-2 and anti-CD16 mAb-treated NK cells’ supernatants induced differentiation in hA549 tumors. As described in the Materials and Methods, hA549 tumors were differentiated using untreated and IL-2 + anti-CD16 mAb-treated NK cells’ supernatants. hA549 tumors were analyzed for IgG2, MHC-class I, B7H1 (PD-L1), and CD54 surface expression using flow cytometer (<b>A</b>). Untreated, IL-2 treated and IL-2 + anti-CD16 mAb-treated NK cells (1 × 10<sup>6</sup> cells/mL) from healthy individuals were incubated overnight before they were used as effector cells against <sup>51</sup>Cr-labeled CSCs and differentiated hA549 at different NK-to-tumor (Effector: Target) ratios using a 4-h <sup>51</sup>Cr release assay. The lytic unit (LU) 30/10<sup>6</sup> cells were calculated based on an inverse number of NK cells needed to kill 30% of the target cells × 100 (<b>B</b>). hA549 and NK cell-differentiated hA549 were treated with paclitaxel (40 µg/mL) (<b>C</b>) or with cisplatin (CDDP) (40 µg/mL) (<b>D</b>) overnight before the tumors were stained with propidium iodide (PI), and percent cell viability was determined using flow cytometric analysis (<b>C</b>,<b>D</b>). *** (<span class="html-italic">p</span> value &lt; 0.001); ** (<span class="html-italic">p</span> value 0.001–0.01); * (<span class="html-italic">p</span> value 0.01–0.05).</p>
Full article ">Figure 3
<p>Treatment with IFN-γ and TNF-α induced differentiation in hA549 tumors. hA549 cells treated with rhTNF-α (20 ng/mL), rhIFN-γ (200 U/mL), and rhTNF-α (20 ng/mL) + rhIFN-γ (200 U/mL) were incubated overnight before they were used as targets for untreated and IL-2-treated NK cells in a 4-h <sup>51</sup>Cr release assay. The lytic unit (LU) 30/10<sup>6</sup> cells were calculated based on an inverse number of NK cells needed to kill 30% of the target cells × 100 (<b>A</b>). Untreated and rhTNF-α (20 ng/mL), rhIFN-γ (200 U/mL), and rhTNF-α (20 ng/mL) + rhIFN-γ (200 U/mL)-treated hA549 cells were treated with paclitaxel (40 µg/mL) (<b>B</b>) and cisplatin (CDDP) (40 µg/mL) (<b>C</b>) overnight before they were stained with propidium iodide (PI), amd percent cell viability was determined using flow cytometric analysis (<b>B</b>,<b>C</b>). *** (<span class="html-italic">p</span> value &lt; 0.001); ** (<span class="html-italic">p</span> value 0.001–0.01); * (<span class="html-italic">p</span> value 0.01–0.05).</p>
Full article ">Figure 4
<p>Treatments with antibodies against IFN-γ and TNF-α blocked NK cells’ supernatant-induced, as well as the IFN-γ- and TNF-α-induced, differentiation of hA549 cells. As described in the Materials and Methods, hA549 tumors were treated with supernatants from IL-2 + anti-CD16 mAb-treated NK cells alone or in combination with rhTNF-α (20 ng/mL), rhIFN-γ (200 U/mL), and αTNFα mAbs (1:100) + αIFNγ mAbs (1:100) for six days. On day 6, tumors were analyzed for surface marker levels of IgG2, CD54, MHC-class I, B7H1 (PD-L1), and CD54 using flow cytometric analysis (<b>A</b>). hA549 cells were treated as described in (<b>A</b>) and were stained with propidium iodide (PI) and percent cell viability was determined using flow cytometer (<b>B</b>). hA549 cells were treated as described in (<b>A</b>). Untreated, IL-2 (1000 U/mL)-treated, and IL-2 + anti-CD16 mAb (3 μg/mL)-treated NK cells were incubated overnight before they were used as effectors against hA549 cells in a 4-h <sup>51</sup>Cr release assay. The lytic unit (LU) 30/10<sup>6</sup> cells were calculated based on an inverse number of NK cells needed to kill 30% of the target cells × 100 (<b>C</b>). hA549 tumors were treated with rhTNF-α (20 ng/mL), rhIFN-γ (200 U/mL), rhTNF-α (20 ng/mL) + rhIFN-γ (200 U/mL), in the presence and absence of αIFN-γ mAbs (1:100), and αTNFα mAbs (1:100) for six days, and were used as targets for untreated, IL-2 treated, and IL-2 + anti-CD16 mAb-treated NK cells in a 4-h <sup>51</sup>Cr release assay. The lytic unit (LU) 30/10<sup>6</sup> cells were calculated based on an inverse number of NK cells needed to kill 30% of the target cells × 100 (<b>D</b>).</p>
Full article ">Figure 5
<p>Treatment of hA549 with anti-MHC-class I (αPA2.6mAb, 1:100) decreased the sensitivity of hA549 CSCs and increased the sensitivity of differentiated hA549 cells, respectively, to NK cell-induced tumor killing. As described in the Materials and Methods, IL-2 and anti-CD16 mAb-treated NK cells’ supernatant was used to differentiate hA549 cells. Untreated and IL-2 (1000 U/mL)-treated NK cells were incubated overnight before they were used as effectors against hA549 cells in a 4-h <sup>51</sup>Cr release assay. The lytic unit (LU) 30/10<sup>6</sup> cells were calculated based on an inverse number of NK cells needed to kill 30% of the target cells × 100.</p>
Full article ">
Back to TopTop