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
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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (669)

Search Parameters:
Keywords = nomogram

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 1892 KiB  
Article
Performance of EuroSCORE II in Octogenarians Undergoing Coronary Artery Surgery (from the KROK Registry)
by Grzegorz Hirnle, Sleiman Sebastian Aboul-Hassan, Piotr Knapik, Zdzisław Tobota, Bohdan Maruszewski, Jan Rogowski, Wojciech Pawliszak, Paweł Bugajski, Marek Deja, Tomasz Hrapkowicz and on behalf of the KROK Investigators
J. Clin. Med. 2024, 13(22), 6863; https://doi.org/10.3390/jcm13226863 - 14 Nov 2024
Viewed by 252
Abstract
Background: Octogenarians constitute the fastest-growing segment within contemporary cardiac surgery, yet precise risk assessment in this age group remains challenging. Aims: This study aimed to evaluate EuroSCORE II reliability in octogenarians undergoing isolated coronary surgery and to create an adjustment formula if necessary. [...] Read more.
Background: Octogenarians constitute the fastest-growing segment within contemporary cardiac surgery, yet precise risk assessment in this age group remains challenging. Aims: This study aimed to evaluate EuroSCORE II reliability in octogenarians undergoing isolated coronary surgery and to create an adjustment formula if necessary. Patients and Methods: All octogenarians who had isolated coronary surgery in Poland from January 2012 to December 2023, recorded in the Polish National Registry of Cardiac Surgical Procedures (KROK registry), were retrospectively assessed. EuroSCORE II’s reliability was measured using the ROC curve area and observed-to-predicted mortality ratio, differentiating on-pump and off-pump cases. A nomogram was developed to enhance predictive accuracy. Results: Among 5771 octogenarians, 2729 (47.3%) underwent on-pump and 3042 (52.7%) underwent off-pump surgery. EuroSCORE II demonstrated reliability in off-pump patients (AUC:0.72, O/E ratio:0.98) but underestimated mortality for on-pump cases (AUC:0.73, O/E ratio:1.62). The lowest predicted mortality group (0.50–1.82%) showed the greatest discrepancies. Calibration was improved by adding a coefficient and creating a nomogram. Conclusions: EuroSCORE II was dependable in predicting outcomes for Polish octogenarians undergoing isolated coronary surgery. Observed mortality following on-pump surgeries was higher than expected, which was corrected by adding a coefficient to the initial EuroSCORE II calculation. Full article
(This article belongs to the Section Cardiovascular Medicine)
Show Figures

Figure 1

Figure 1
<p>Distribution of EuroSCORE II in the analyzed population.</p>
Full article ">Figure 2
<p>The number of coronary artery procedures (ONCAB and OPCAB) performed in octogenarian patients in the consecutive years during the observation period.</p>
Full article ">Figure 3
<p>The results for each quartile of EuroSCORE II for patients undergoing ONCAB (<b>A</b>) and OPCAB (<b>B</b>).</p>
Full article ">Figure 4
<p>A nomogram of the calculations of predicted mortality.</p>
Full article ">
17 pages, 15430 KiB  
Article
CLIC4 Is a New Biomarker for Glioma Prognosis
by Zhichun Liu, Junhui Liu, Zhibiao Chen, Xiaonan Zhu, Rui Ding, Shulan Huang and Haitao Xu
Biomedicines 2024, 12(11), 2579; https://doi.org/10.3390/biomedicines12112579 - 11 Nov 2024
Viewed by 345
Abstract
Background: Chloride Intracellular Channel 4 (CLIC4) plays a versatile role in cellular functions beyond its role in primary chloride ion transport. Notably, many studies found an association between CLIC4 expression and cancers. However, the correlation between CLIC4 and glioma remains to [...] Read more.
Background: Chloride Intracellular Channel 4 (CLIC4) plays a versatile role in cellular functions beyond its role in primary chloride ion transport. Notably, many studies found an association between CLIC4 expression and cancers. However, the correlation between CLIC4 and glioma remains to be uncovered. Methods: A total of 3162 samples from nine public datasets were analyzed to reveal the relationship between CLIC4 expression and glioma malignancy or prognosis. Immunohistochemistry (IHC) staining was performed to examine the results in an in-house cohort. A nomogram model was constructed to predict the prognosis. Functional enrichment analysis was employed to find CLIC4-associated differentially expressed genes in glioma. Immune infiltration analysis, correlation analysis, and IHC staining were employed, aiming to examine the correlation between CLIC4 expression, immune cell infiltration, and ECM (extracellular matrix)-related genes. Results: The expression level of CLIC4 was correlated with the malignancy of glioma and the prognosis of patients. More aggressive gliomas and mesenchymal GBM are associated with a high expression of CLIC4. Gliomas with IDH mutation or 1p19q codeletion express a low level of CLIC4, and a high expression of CLIC4 correlates with poor prognosis. The nomogram model shows a good predictive performance. The DEGs (differentially expressed genes) in gliomas with high and low CLIC4 expression are enriched in extracellular matrix and immune functions. On the one hand, gliomas with high CLIC4 expression have a greater presence of macrophages, neutrophils, and eosinophils; on the other hand, a high CLIC4 expression in gliomas is positively associated with ECM-related genes. Conclusions: Compared to glioma cells with low CLIC4 expression, gliomas with high CLIC4 expression exhibit greater malignancy and poorer prognosis. Our findings indicate that a high level of CLIC4 correlates with high expression of ECM-related genes and the infiltration of macrophages, neutrophils, and eosinophils within glioma tissues. Full article
(This article belongs to the Special Issue Glioblastoma: Pathogenetic, Diagnostic and Therapeutic Perspectives)
Show Figures

Figure 1

Figure 1
<p>Transcription level of <span class="html-italic">CLIC4</span> in gliomas of public datasets. (<b>A</b>) <span class="html-italic">CLIC4</span> mRNA data of glioma tissues and normal brain tissues from six public datasets. (<b>B</b>) <span class="html-italic">CLIC4</span> mRNA data of glioma tissues from four public datasets. Each dataset was divided into three groups according to WHO grading. (<b>C</b>) <span class="html-italic">CLIC4</span> mRNA data of glioma tissues from two public datasets. Each one was divided into four groups according to IDH statue and 1p19q deletion. ns &gt; 0.05, * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. NBT means normal brain tissue.</p>
Full article ">Figure 2
<p>Transcription level of <span class="html-italic">CLIC4</span> in different subtypes of GBM. (<b>A</b>) <span class="html-italic">CLIC4</span> mRNA in different subtypes of GBM from five datasets. (<b>B</b>) The correlation between CLIC4 expression and mesenchymal-related genes. ns &gt; 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>Transcription level of CLIC4 in gliomas of clinic samples. (<b>A</b>) Representative IHC images of CLIC4 in normal brain tissue, LGG, and GBM. Scale bars were 500 µm, 100 µm, 50 µm, and 20 µm. (<b>B</b>) The average optical density of IHC staining. (<b>C</b>) Representative IHC images in IDH mut LGG, IDH WT LGG, and IDH WT GBM. Scale bars were 50 µm and 500 µm. ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Relationship between <span class="html-italic">CLIC4</span> mRNA expression and prognosis in patients with glioma. Differences in survival of different <span class="html-italic">CLIC4</span> mRNA expression levels were analyzed by Kaplan–Meier analysis. HR, hazard ratio.</p>
Full article ">Figure 5
<p>The nomogram model that includes clinicopathological factors (age, WHO grade, and IDH status) and <span class="html-italic">CLIC4</span> expression levels to predict the 1-, 3-, and 5-year survival rates of glioma patients.</p>
Full article ">Figure 6
<p>An analysis of the nomogram model’s calibration curve for predicting 1-, 3-, and 5-year survival rates.</p>
Full article ">Figure 7
<p>Based on the <span class="html-italic">CLIC4</span> expression levels in gliomas, functional enrichment analysis of differentially expressed genes (DEGs) was conducted. (<b>A</b>) GO enrichment analysis of the <span class="html-italic">CLIC4</span>-associated DEGs. (<b>B</b>) In glioma tissues, a Gene Set Enrichment Analysis was conducted using the <span class="html-italic">CLIC4</span>-associated DEGs between groups with high- and low-<span class="html-italic">CLIC4</span> expression.</p>
Full article ">Figure 8
<p>Inflammatory cells infiltrating into gliomas are correlated with <span class="html-italic">CLIC4</span> expression in cancer patients with <span class="html-italic">CLIC4</span> expression. (<b>A</b>) Spearman’s correlation analysis results in 24 immune cell types. (<b>B</b>) The infiltration levels of macrophage, neutrophils, eosinophils, pDCs, NK CD56bright cells, and treg cells. (<b>C</b>) The correlation chord diagram results between <span class="html-italic">CLIC4</span> expression and the immune cell markers. ns &gt; 0.05 * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 9
<p>IHC staining of CD68 (<b>A</b>), VIM (<b>B</b>), SNAI1 (<b>C</b>), and MMP (<b>D</b>) in a low or high expression of CLIC4 glioma patients. Scale bars were 500 µm, 100 µm, and 20 µm.</p>
Full article ">Figure 10
<p>The correlation analysis between ECM-related genes and <span class="html-italic">CLIC4</span> expression in glioma patients.</p>
Full article ">
20 pages, 3583 KiB  
Article
Lunar Satellite Constellations in Frozen Low Orbits
by Mikhail Ovchinnikov, Maksim Shirobokov and Sergey Trofimov
Aerospace 2024, 11(11), 918; https://doi.org/10.3390/aerospace11110918 - 8 Nov 2024
Viewed by 390
Abstract
This research studies the potential of frozen low lunar orbits to be used in the design of constellations for global and regional communication or navigation. We introduce a robust two-stage approach to the frozen low lunar orbit design based on the successive application [...] Read more.
This research studies the potential of frozen low lunar orbits to be used in the design of constellations for global and regional communication or navigation. We introduce a robust two-stage approach to the frozen low lunar orbit design based on the successive application of non-gradient techniques, the Bayesian optimization and the Nelder–Mead method. The developed methodology has a number of advantages over existing numerical design techniques and allows revealing orbits with the periodic behavior of the eccentricity vector over long propagation intervals in the full dynamical model. By leveraging a convenient nomogram with constellation visibility parameters and lower bound coverage curves, we have identified most suitable low-altitude orbital configurations of Walker type and then adjust them to be frozen. The frozenness condition can be achieved without changing the orientation of orbital planes. Visibility and coverage metrics (multiplicity of continuous coverage for specified sites, polar regions, or the whole lunar surface; position dilution of precision) of candidate constellations are analyzed. Several promising designs of frozen constellations in near-circular low lunar orbits are singled out. The frozen orbit stability and the station-keeping cost are discussed. Full article
(This article belongs to the Section Astronautics & Space Science)
Show Figures

Figure 1

Figure 1
<p>Degree and order of the lunar gravitational potential expansion required to meet the truncated perturbing acceleration threshold <math display="inline"><semantics> <mrow> <mi>ε</mi> <mo>=</mo> <msup> <mn>10</mn> <mrow> <mo>−</mo> <mn>7</mn> </mrow> </msup> </mrow> </semantics></math>.</p>
Full article ">Figure 2
<p>Relative magnitude of perturbations as a function of the orbital altitude.</p>
Full article ">Figure 3
<p>Eccentricity vector evolution for the 73–cycle RGT periodic orbit (Adapted from [<a href="#B21-aerospace-11-00918" class="html-bibr">21</a>], reprinted by permission of the American Institute of Aeronautics and Astronautics, Inc.). (<b>a</b>) Retrieved from [<a href="#B21-aerospace-11-00918" class="html-bibr">21</a>], (<b>b</b>) reproduced.</p>
Full article ">Figure 4
<p>Eccentricity vector evolution for the 328–cycle RGT periodic orbit (Adapted from [<a href="#B21-aerospace-11-00918" class="html-bibr">21</a>], reprinted by permission of the American Institute of Aeronautics and Astronautics, Inc.). (<b>a</b>) Retrieved from [<a href="#B21-aerospace-11-00918" class="html-bibr">21</a>], (<b>b</b>) reproduced.</p>
Full article ">Figure 5
<p>Ten-year evolution of lunar orbit eccentricity and argument of periapsis (522 km altitude, <math display="inline"><semantics> <msup> <mn>0</mn> <mo>∘</mo> </msup> </semantics></math> RAAN), considering Solar System planetary and lunar (up to degree/order 50) gravitational perturbations. The critical eccentricity (0.12) significantly exceeds the maximum observed value.</p>
Full article ">Figure 6
<p>Ten-year evolution of lunar orbit eccentricity and argument of periapsis (261 km altitude, <math display="inline"><semantics> <msup> <mn>0</mn> <mo>∘</mo> </msup> </semantics></math> RAAN), considering Solar System planetary and lunar (up to degree/order 50) gravitational perturbations. The critical eccentricity (0.07) significantly exceeds the maximum observed value.</p>
Full article ">Figure 7
<p>Relation between the minimum elevation angle, the orbital altitude, and the footprint size.</p>
Full article ">Figure 8
<p>CoDe nomogram for basic coverage parameters as a function of the footprint size: the altitude of orbits, in <span class="html-italic">R</span> (blue); the free-space path loss (red) normalized to its value at <math display="inline"><semantics> <mrow> <mi>h</mi> <mo>=</mo> <mi>R</mi> </mrow> </semantics></math>; the theoretical minimum constellation size, in 100 s of satellites, for 1-fold (maroon) and 4-fold (violet) coverage. The 5 deg minimum elevation angle is assumed.</p>
Full article ">Figure 9
<p>Quasi-uniform surface grid of 1200 points with 8 sites included by default; among them are the North and South lunar poles and the Boguslawsky (green) and Manzinus (purple) craters.</p>
Full article ">Figure 10
<p>Five candidate LLO constellations marked on the CoDe nomogram.</p>
Full article ">Figure 11
<p>One-year evolution of orbital elements for orbits in the <math display="inline"><semantics> <mrow> <msup> <mn>84</mn> <mo>∘</mo> </msup> <mspace width="-2.0pt"/> <mo>:</mo> <mn>54</mn> <mo>/</mo> <mn>3</mn> <mo>/</mo> <mn>1</mn> </mrow> </semantics></math> constellation (<math display="inline"><semantics> <mrow> <msub> <mi>h</mi> <mi>ref</mi> </msub> <mo>=</mo> <mn>261</mn> </mrow> </semantics></math> km). Different colors correspond to orbits with different orbital planes.</p>
Full article ">Figure 12
<p>One-year evolution of orbital elements for orbits in the <math display="inline"><semantics> <mrow> <msup> <mn>84</mn> <mo>∘</mo> </msup> <mspace width="-2.0pt"/> <mo>:</mo> <mn>70</mn> <mo>/</mo> <mn>5</mn> <mo>/</mo> <mn>1</mn> </mrow> </semantics></math> constellation (<math display="inline"><semantics> <mrow> <msub> <mi>h</mi> <mi>ref</mi> </msub> <mo>=</mo> <mn>261</mn> </mrow> </semantics></math> km). Different colors correspond to orbits with different orbital planes.</p>
Full article ">Figure 13
<p>One-year evolution of orbital elements for orbits in the <math display="inline"><semantics> <mrow> <msup> <mn>84</mn> <mo>∘</mo> </msup> <mspace width="-2.0pt"/> <mo>:</mo> <mn>36</mn> <mo>/</mo> <mn>3</mn> <mo>/</mo> <mn>1</mn> </mrow> </semantics></math> constellation (<math display="inline"><semantics> <mrow> <msub> <mi>h</mi> <mi>ref</mi> </msub> <mo>=</mo> <mn>522</mn> </mrow> </semantics></math> km). Different colors correspond to orbits with different orbital planes.</p>
Full article ">Figure 14
<p>Three-month evolution of the eccentricity vector for a 522 km orbit. Every month is marked by a different color. Two-impulse station-keeping maneuvers are executed on a monthly basis, with the associated eccentricity vector jump being indicated by a red arrow.</p>
Full article ">
15 pages, 2425 KiB  
Article
The Demographic and Clinical Characteristics, Prognostic Factors, and Survival Outcomes of Head and Neck Carcinosarcoma: A SEER Database Analysis
by Wanting Hou, Ouying Yan and Hong Zhu
Biomedicines 2024, 12(11), 2556; https://doi.org/10.3390/biomedicines12112556 - 8 Nov 2024
Viewed by 337
Abstract
Background: Head and neck carcinosarcoma (HNCS) is a rare and highly aggressive malignancy with limited research, resulting in an incomplete understanding of disease progression and a lack of reliable prognostic tools. This study aimed to retrospectively analyze the clinical characteristics and outcomes of [...] Read more.
Background: Head and neck carcinosarcoma (HNCS) is a rare and highly aggressive malignancy with limited research, resulting in an incomplete understanding of disease progression and a lack of reliable prognostic tools. This study aimed to retrospectively analyze the clinical characteristics and outcomes of HNCS patients using data from the Surveillance, Epidemiology, and End Results (SEER) database and to develop a nomogram to predict overall survival (OS) and cancer-specific survival (CSS). Methods: Patients diagnosed with HNCS from 1975 to 2020 were identified in the SEER database. Univariate and multivariate Cox regression analyses were conducted to identify independent prognostic indicators, with the optimal model selected using the minimal Akaike Information Criterion (AIC). The identified prognostic factors were incorporated into nomograms to predict OS and CSS. Model performance was assessed using the concordance index (C-index), area under the curve (AUC), calibration curves, and decision curve analysis (DCA). Survival curves were generated using Kaplan–Meier analysis and compared via the log-rank test. Results: A total of 152 HNCS patients were included, with 108 assigned to the training cohort and 44 to the validation cohort in a 7:3 ratio. Prognostic factors including age, primary tumor site, marital status, radiotherapy, chemotherapy, tumor size, pathological grade, and tumor stage were incorporated into the nomogram models. The models demonstrated strong predictive performance, with C-index values for OS and CSS of 0.757 and 0.779 in the training group, and 0.777 and 0.776 in the validation group, respectively. AUC values for predicting 3-, 5-, and 10-year OS were 0.662, 0.713, and 0.761, and for CSS the values were 0.726, 0.703, and 0.693. Kaplan–Meier analysis indicated significantly improved survival for patients with lower risk scores. The 3-, 5-, and 10-year OS rates for the entire cohort were 54.1%, 45.6%, and 35.1%, respectively, and the CSS rates were 62.9%, 57.5%, and 52.2%, respectively. Conclusions: This study provides validated nomograms for predicting OS and CSS in HNCS patients, offering a reliable tool to support clinical decision-making for this challenging malignancy. These nomograms enhance the ability to predict patient prognosis and personalize treatment strategies. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

Figure 1
<p>Survival outcomes for HNCS by cohort (<b>a</b>,<b>b</b>), tumor site (<b>c</b>), and disease stage (<b>d</b>).</p>
Full article ">Figure 2
<p>Nomograms constructed to predict 3-, 5-, and 10-year OS (<b>a</b>) and CSS (<b>b</b>) rates in patients with HNCS.</p>
Full article ">Figure 3
<p>Receiver operating characteristic (ROC) curves for evaluating the predictive ability of independent prognostic factors for 3-, 5-, and 10-year OS and CSS in HNCS patients in the training cohort: (<b>a</b>) 3-year OS; (<b>b</b>) 5-year OS; (<b>c</b>) 10-year OS; (<b>d</b>) 3-year CSS; (<b>e</b>) 5-year CSS; (<b>f</b>) 10-year CSS.</p>
Full article ">Figure 4
<p>Calibration curves for the nomogram in the training group: (<b>a</b>) 3-year OS, (<b>b</b>) 5-year OS, (<b>c</b>) 10-year OS, (<b>d</b>) 3-year CSS, (<b>e</b>) 5-year CSS, and (<b>f</b>) 10-year CSS.</p>
Full article ">Figure 5
<p>Decision curve analysis for the nomograms in the training group: (<b>a</b>) 3-year OS, (<b>b</b>) 5-year OS, (<b>c</b>) 10-year OS, (<b>d</b>) 3-year CSS, (<b>e</b>) 5-year CSS, and (<b>f</b>) 10-year CSS. The <span class="html-italic">x</span>-axis represents the threshold probability, and the <span class="html-italic">y</span>-axis represents the net benefit.</p>
Full article ">Figure 6
<p>Kaplan–Meier (K-M) curves for the nomogram in the training group: (<b>a</b>) OS and (<b>b</b>) CSS.</p>
Full article ">
25 pages, 18080 KiB  
Article
Comprehensive Analysis and Verification of the Prognostic Significance of Cuproptosis-Related Genes in Colon Adenocarcinoma
by Yixiao Gu, Chengze Li, Yinan Yan, Jingmei Ming, Yuanhua Li, Xiang Chao and Tieshan Wang
Int. J. Mol. Sci. 2024, 25(21), 11830; https://doi.org/10.3390/ijms252111830 - 4 Nov 2024
Viewed by 535
Abstract
Colon adenocarcinoma (COAD) is a frequently occurring and lethal cancer. Cuproptosis is an emerging type of cell death, and the underlying pathways involved in this process in COAD remain poorly understood. Transcriptomic and clinical data for COAD patients were collected from The Cancer [...] Read more.
Colon adenocarcinoma (COAD) is a frequently occurring and lethal cancer. Cuproptosis is an emerging type of cell death, and the underlying pathways involved in this process in COAD remain poorly understood. Transcriptomic and clinical data for COAD patients were collected from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. We investigated alterations in DNA and chromatin of cuproptosis-related genes (CRGs) in COAD. In order to identify predictive differentially expressed genes (DEGs) and various molecular subtypes, we used consensus cluster analysis. Through univariate, multivariate, and Lasso Cox regression analyses, four CRGs were identified. A risk prognostic model for cuproptosis characteristics was constructed based on four CRGs. This study also examined the association between the risk score and the tumor microenvironment (TME), the immune landscape, and drug sensitivity. We distinguished two unique molecular subtypes using consensus clustering analysis. We discovered that the clinical characteristics, prognosis, and TME cell infiltration characteristics of patients with multilayer CRG subtypes were all connected. The internal and external evaluations of the predicted accuracy of the prognostic model built using data derived from a cuproptosis risk score were completed at the same time. A nomogram and a clinical pathological analysis make it more useful in the field of medicine. A significant rise in immunosuppressive cells was observed in the high cuproptosis risk score group, with a correlation identified between the cuproptosis risk score and immune cell infiltration. Despite generally poor prognoses, the patients with a high cuproptosis risk but low tumor mutation burden (TMB), cancer stem cell (CSC) index, or microsatellite instability (MSI) may still benefit from immunotherapy. Furthermore, the cuproptosis risk score positively correlated with immune checkpoint gene expression. Analyzing the potential sensitivity to medications could aid in the development of clinical chemotherapy regimens and decision-making. CRGs are the subject of our in-depth study, which exposed an array of regulatory mechanisms impacting TME. In addition, we performed additional data mining into clinical features, prognosis effectiveness, and possible treatment medications. COAD’s molecular pathways will be better understood, leading to more precise treatment options. Full article
(This article belongs to the Special Issue Molecular Advances in Cancer and Cell Metabolism)
Show Figures

Figure 1

Figure 1
<p>Genetic and transcriptional alteration of CRGs in COAD. (<b>A</b>) Mutation rates of 12 CRGs in 447 patients from the TCGA-COAD dataset; (<b>B</b>) frequency of CNVs involving gains and losses in CRGs; (<b>C</b>) chromosomal locations of CNV-altered CRGs; (<b>D</b>) differential expression of 8 CRGs in normal versus COAD tissues; significance levels are denoted as follows: ***, <span class="html-italic">p</span> &lt; 0.001; **, <span class="html-italic">p</span> &lt; 0.01; (<b>E</b>) interaction network diagram illustrating relationships among CRGs in COAD. Edges between genes indicate interactions among CRGs, with blue denoting negative correlations and pink indicating positive correlations. Line thickness indicates correlation strength. Circle size indicates the prognostic significance of CRGs, with <span class="html-italic">p</span>-values classified as <span class="html-italic">p</span> &lt; 0.0001, <span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">p</span> &lt; 0.01, <span class="html-italic">p</span> &lt; 0.05, and <span class="html-italic">p</span> &lt; 1. Purple circles denote risk factors, while green circles indicate protective factors.</p>
Full article ">Figure 2
<p>Characteristics of the CRGs in terms of subtype, clinicopathological factors, and biological aspects. (<b>A</b>) Consensus cluster analysis generated the consensus matrix diagram for the two related regions; (<b>B</b>) PCA emphasized the differences between the two subtypes; (<b>C</b>) OS rates were compared between the two subtypes; (<b>D</b>) evaluation of immune cell infiltration differences between subtypes; significance levels are denoted as follows: ***, <span class="html-italic">p</span> &lt; 0.001; *, <span class="html-italic">p</span> &lt; 0.05; and ns, not significant; (<b>E</b>) heatmap illustrating clinicopathological disparities and CRG expression levels across subtypes; (<b>F</b>) GSVA of biological pathways in subtypes, with red for activation and blue for inhibition; (<b>G</b>,<b>H</b>) GO and KEGG analyses of DEGs among CRG subtypes.</p>
Full article ">Figure 3
<p>Gene subtypes were identified through differential expression analysis. (<b>A</b>) A consensus clustering matrix (k = 3) categorized COAD patients into three unique genomic subtypes; (<b>B</b>) the overall survival (OS) rates differed among these gene clusters; (<b>C</b>) CRG expression levels varied significantly across the clusters; (<b>D</b>) differences in immune cell infiltration levels across the three gene clusters; significance levels are denoted as follows: ***, <span class="html-italic">p</span> &lt; 0.001; *, <span class="html-italic">p</span> &lt; 0.05; (<b>E</b>) association between the three gene clusters and clinicopathological characteristics.</p>
Full article ">Figure 4
<p>Development and validation of the cuproptosis risk score. (<b>A</b>) Sankey plots depict the relationships between cuproptosis clusters, gene clusters, cuproptosis risk scores, and survival status; (<b>B</b>) a comparison of cuproptosis risk scores across the two cuproptosis clusters; (<b>C</b>) analysis of cuproptosis risk score differences across three gene clusters; (<b>D</b>) examination of risk distribution, survival status, and related gene expression; (<b>E</b>) evaluation of overall survival (OS) rates between high-risk and low-risk groups; (<b>F</b>) ROC curve analysis for 1-, 3-, and 5-year survival predictions based on cuproptosis risk score; (<b>G</b>) comparison of survival rates between high-risk and low-risk patient groups.</p>
Full article ">Figure 5
<p>Correlation between cuproptosis risk score and clinicopathological subtype in COAD patients. (<b>A</b>,<b>B</b>) Age (≤65, &gt;65); (<b>C</b>,<b>D</b>) gender (female, male); (<b>E</b>,<b>F</b>) T classification (T1–2, T3–4); (<b>G</b>,<b>H</b>) N classification (N0, N1–2); (<b>I</b>,<b>J</b>) M classification (M0, M1); (<b>K</b>,<b>L</b>) stage (I–II, III–IV).</p>
Full article ">Figure 6
<p>Clinical utility and independent prognostic assessment. (<b>A</b>) Heatmap showing the correlation between risk groups and clinicopathological characteristics; significance levels are denoted as follows: ***, <span class="html-italic">p</span> &lt; 0.001; (<b>B</b>–<b>D</b>) proportions of risk groups across clinicopathological characteristics, including stage and T and N classifications; (<b>E</b>) univariate Cox regression analysis based on cuproptosis risk score and clinicopathological characteristics; (<b>F</b>) multivariate Cox regression analysis using the cuproptosis risk score and clinicopathological characteristics; (<b>G</b>–<b>I</b>) ROC curve assessing the risk model’s predictive capability at 1, 3, and 5 years.</p>
Full article ">Figure 7
<p>Development and validation of the nomogram. (<b>A</b>) Nomogram for predicting 1-, 3-, and 5-year overall survival (OS) in COAD patients; significance levels are denoted as follows: ***, <span class="html-italic">p</span> &lt; 0.001; *, <span class="html-italic">p</span> &lt; 0.05; (<b>B</b>) calibration curve of the nomogram; (<b>C</b>–<b>E</b>) ROC curves for 1-, 3-, and 5-year OS prediction; (<b>F</b>–<b>H</b>) DCA curves for 1-, 3-, and 5-year OS prediction.</p>
Full article ">Figure 8
<p>Examination of the immune landscape based on risk attributes. (<b>A</b>) The ESTIMATE algorithm to evaluate the association between cuproptosis risk groups and TME scores; (<b>B</b>) Analyzed immune cell abundance and immune function score disparities between high-risk and low-risk groups using ssGSEA; (<b>C</b>) Heatmap is provided to depict variations in immune cell content between these groups across different algorithms; (<b>D</b>) Correlation between immune cell abundance and the four genes in the constructed model is examined. Significance levels are denoted as follows: ***, <span class="html-italic">p</span> &lt; 0.001; **, <span class="html-italic">p</span> &lt; 0.01; *, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 9
<p>Relationship between cuproptosis risk score and MSI, TMB, and CSC index in COAD patients. (<b>A</b>,<b>B</b>) Correlation between cuproptosis risk scores and MSI; (<b>C</b>) relationship between cuproptosis risk scores and CSC index; (<b>D</b>) TMB across various risk groups; (<b>E</b>) Spearman’s correlation analysis of cuproptosis risk scores and TMB; (<b>F</b>,<b>G</b>) oncoplots of somatic mutation characteristics established with high and low cuproptosis risk scores; (<b>H</b>) prognostic analysis of TMB; (<b>I</b>) prognostic analysis between cuproptosis risk score and TMB.</p>
Full article ">Figure 10
<p>Examination of the relationship between prognostic features derived from cuproptosis risk scores and immune checkpoint genes. (<b>A</b>) Relationship between cuproptosis risk scores and immune checkpoints; (<b>B</b>–<b>G</b>) Correlation between the cuproptosis risk scores and the 6 key immune check-point genes (CD274, CTLA-4, HAVCR2, IDO1, PDCD1, and PDCD1LG2); (<b>H</b>) Differential ex-pression of immune checkpoint genes between high- and low-risk groups. Significance levels are denoted as follows: ***, <span class="html-italic">p</span> &lt; 0.001; **, <span class="html-italic">p</span> &lt; 0.01; *, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 11
<p>Analysis of immunotherapy and drug sensitivity in IPS. (<b>A</b>) No use of PD-1 or CTLA-4 inhibitors; (<b>B</b>) use of a CTLA-4 inhibitor alone; (<b>C</b>) use of a PD-1 inhibitor alone; (<b>D</b>) combined use of CTLA-4 and PD-1 inhibitors; (<b>E</b>–<b>H</b>) association between cuproptosis risk scores and drug sensitivity.</p>
Full article ">Figure 12
<p>Biological validation results of key genes CDKN2A, CKMT2 and MSLN. (<b>A</b>) Differences in three convenient expressions of CDKN2A, CKMT2 and MSLN between human normal tissues and tumor tissues obtained based on IHC from HPA database; (<b>B</b>) H-score of IHCs; (<b>C</b>) results of RT-qPCR to detect the differential expression of transcript levels of CDKN2A, CKMT2, and MSLN in normal and tumor cell lines. Significance levels are denoted as follows: ***, <span class="html-italic">p</span> &lt; 0.001; **, <span class="html-italic">p</span> &lt; 0.01; *, <span class="html-italic">p</span> &lt; 0.05; and ns, not significant.</p>
Full article ">
17 pages, 1278 KiB  
Article
Severity Patterns in COVID-19 Hospitalised Patients in Spain: I-MOVE-COVID-19 Study
by Miriam Latorre-Millán, María Mar Rodríguez del Águila, Laura Clusa, Clara Mazagatos, Amparo Larrauri, María Amelia Fernández, Antonio Rezusta and Ana María Milagro
Viruses 2024, 16(11), 1705; https://doi.org/10.3390/v16111705 - 30 Oct 2024
Viewed by 452
Abstract
In the frame of the I-MOVE-COVID-19 project, a cohort of 2050 patients admitted in two Spanish reference hospitals between March 2020 and December 2021 was selected and a range of clinical factor data were collected at admission to assess their impact on the [...] Read more.
In the frame of the I-MOVE-COVID-19 project, a cohort of 2050 patients admitted in two Spanish reference hospitals between March 2020 and December 2021 was selected and a range of clinical factor data were collected at admission to assess their impact on the risk COVID-19 severity outcomes through a multivariate adjusted analysis and nomograms. The need for ventilation and intensive care unit (ICU) admission were found to be directly associated with a higher death risk (OR 6.9 and 3.2, respectively). The clinical predictors of death were the need for ventilation and ICU, advanced age, neuromuscular disorders, thrombocytopenia, hypoalbuminemia, dementia, cancer, elevated creatin phosphokinase (CPK), and neutrophilia (OR between 1.8 and 3.5), whilst the presence of vomiting, sore throat, and cough diminished the risk of death (OR 0.5, 0.2, and 0.1, respectively). Admission to ICU was predicted by the need for ventilation, abdominal pain, and elevated lactate dehydrogenase (LDH) (OR 371.0, 3.6, and 2.2, respectively) as risk factors; otherwise, it was prevented by advanced age (OR 0.5). In turn, the need for ventilation was predicted by low oxygen saturation, elevated LDH and CPK, diabetes, neutrophilia, obesity, and elevated GGT (OR between 1.7 and 5.2), whilst it was prevented by hypertension (OR 0.5). These findings could enhance patient management and strategic interventions to combat COVID-19. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Spanish COVID-19 hospitalised cases by month (I-MOVE-COVID19 study).</p>
Full article ">Figure 2
<p>Predictive factors associated with ventilation, ICU admission, and death in Spanish hospitalised patients. I-MOVE-COVID19 study. Note ↓ and ↑ indicates that the predictive factor is over or under the normal range respectively.</p>
Full article ">Figure 3
<p>Nomograms for predicting COVID-19 severity outcomes in Spanish hospitalised patients. I-MOVE-COVID19 study. (<b>A</b>) Death, (<b>B</b>) ICU admission, and (<b>C</b>) need for ventilation. Note ↓ and ↑ indicates that the predictive factor is over or under the normal range respectively.</p>
Full article ">
14 pages, 1088 KiB  
Article
Predicting Additional Metastases in Axillary Lymph Node Dissection After Neoadjuvant Chemotherapy: Ratio of Positive/Total Sentinel Nodes
by Isaac Cebrecos, Ines Torras, Helena Castillo, Claudia Pumarola, Sergi Ganau, Carla Sitges, Sergi Vidal-Sicart, Francesco Schettini, Esther Sanfeliu, Ignacio Loinaz, Marta Garcia, Gabriela Oses, Meritxell Molla, Maria Vidal and Eduard Mension
Cancers 2024, 16(21), 3638; https://doi.org/10.3390/cancers16213638 - 29 Oct 2024
Viewed by 402
Abstract
Background/Objectives: The aim of the study was to determine the clinical value of the sentinel lymph node ratio (SLN-R) in predicting additional positive lymph nodes during axillary lymph node dissection (ALND) in breast cancer patients following neoadjuvant chemotherapy (NAC). Methods: A cross-sectional study [...] Read more.
Background/Objectives: The aim of the study was to determine the clinical value of the sentinel lymph node ratio (SLN-R) in predicting additional positive lymph nodes during axillary lymph node dissection (ALND) in breast cancer patients following neoadjuvant chemotherapy (NAC). Methods: A cross-sectional study was performed at a single institution evaluating data from 1521 BC patients. Inclusion criteria comprised cT1/cT4, cN0/cN1 status with positive post-NAC axillary staging by SLN/TAD, respectively, and subsequent ALND. Results: The study included 118 patients, divided into two groups based on the presence or absence of additional node metastasis at ALND: 39 in the residual disease group (RD) and 79 in the non-residual disease group (nRD). Univariate logistic regression analysis of SLN-R was conducted to assess its predictive value, yielding an odds ratio (OR) of 7.79 (CI 1.92–29.5, p = 0.003). An SLN-R cut-off point of <0.35 was identified using ROC curve analysis, with a false-negative rate of 10.2%, as a predictor for no additional metastasis at ALND following post-NAC SLN/TAD positivity. Conclusions: The study concludes that SLN-R is a valuable predictor for determining the omission of ALND in cases where SLN/TAD is positive after NAC. This metric, in combination with other clinical variables, could help develop a nomogram to spare patients from ALND. Full article
(This article belongs to the Special Issue Rare Breast Tumors)
Show Figures

Figure 1

Figure 1
<p>Flowchart of patients according to initial cN status undergoing NAC. Bold boxes refer to excluded patients. List of abbreviations: BC—breast cancer. HCB—Hospital Clinic of Barcelona. NAC—neoadjuvant chemotherapy. <span class="html-italic">c</span>—clinical. SLN—sentinel lymph node biopsy. TAD—targeted axillary dissection. ALND—axillary lymph node dissection. US—ultrasound. MRI—magnetic resonance imaging. FNAC—fine-needle aspiration cytology. CNB—core needle biopsy.</p>
Full article ">Figure 2
<p>Patterns of SLN, SLN-R and non-SLN involvement at ALND. (<b>A</b>) Number of non-SLN affected at ALND, according to cN status at diagnosis and size of SLN metastasis in the overall population. (<b>B</b>) Pattern of non-SLN involvement after ALND, according to SLN-R cut-off ≤ 0.35. (<b>C</b>) Number of non-SLN affected at ALND, according to SLN-R cut-off ≤ 0.35 and size of SLN metastasis. (<b>D</b>) Number of non-SLN affected at ALND, according to SLN-R cut-off ≤ 0.35 within each IHC subtype. List of abbreviations: nRD—non-residual disease; RD—residual disease; IHC—immunohistochemistry; SLN—sentinel lymph node; ALND—axillary lymph node dissection; ITC—isolated tumor cells; Micro—micrometastases; Macro—macrometastases; HR—hormone receptor; + positive; − negative; TN—triple negative breast cancer. <span class="html-italic">p</span> values refer to Chi-squared tests.</p>
Full article ">
9 pages, 590 KiB  
Article
Preoperative Briganti Nomogram Score and Risk of Prostate Cancer Progression After Robotic Surgery Beyond EAU Risk Categories
by Antonio Benito Porcaro, Rossella Orlando, Andrea Panunzio, Alessandro Tafuri, Alberto Baielli, Francesco Artoni, Claudio Brancelli, Luca Roggero, Sonia Costantino, Andrea Franceschini, Michele Boldini, Lorenzo Pierangelo Treccani, Francesca Montanaro, Sebastian Gallina, Alberto Bianchi, Emanuele Serafin, Giovanni Mazzucato, Francesco Ditonno, Mariana Finocchiaro, Alessandro Veccia, Riccardo Rizzetto, Matteo Brunelli, Vincenzo De Marco, Salvatore Siracusano, Maria Angela Cerruto, Riccardo Bertolo and Alessandro Antonelliadd Show full author list remove Hide full author list
Medicina 2024, 60(11), 1763; https://doi.org/10.3390/medicina60111763 - 27 Oct 2024
Viewed by 635
Abstract
Background and Objectives: We sought to investigate whether the 2012 Briganti nomogram may represent a potential prognostic factor of prostate cancer (PCa) progression after surgical treatment beyond European Association of Urology (EAU) risk categories. Materials and Methods: From January 2013 to [...] Read more.
Background and Objectives: We sought to investigate whether the 2012 Briganti nomogram may represent a potential prognostic factor of prostate cancer (PCa) progression after surgical treatment beyond European Association of Urology (EAU) risk categories. Materials and Methods: From January 2013 to December 2021, data on PCa patients treated with robot-assisted radical prostatectomy at a single tertiary referral center were extracted. The 2012 version of the Briganti nomogram assessing the risk of pelvic lymph node invasion was used. Here, the nomogram score was evaluated both as a continuous and a categorical variable. The association between variables and disease progression after surgery was evaluated through Cox regression models. Results: Overall, 1047 patients were identified. According to the EAU classification system, 297 (28.4%) patients were low-risk, 527 (50.3%) intermediate-risk, and 223 (21.3%) high-risk. The median (interquartile range) 2012 Briganti nomogram score within the investigated population was 3% (2–8%). Median (95% Confidence Interval [CI]) follow-up was 95 (91.9–112.4) months. Disease progression occurred in 237 (22.6%) patients, who were more likely to have an increasing 2012 Briganti nomogram score (Hazard Ratio [HR]: 1.03; 95%CI: 1.01–1.81; p = 0.015), independently of unfavorable issues at clinical presentation. Moreover, the nomogram score stratified according to tertiles (<3% vs. 3–8% vs. ≥8%) hold significance beyond EAU risk categories: accordingly, the risk of disease progression increased as the score increased from the first (reference) to the second (HR: 1.50; 95%CI: 1.67–3.72; p < 0.001) up to the third (HR: 3.26; 95%CI: 2.26–4.72; p < 0.001) tertile. Conclusions: Beyond EAU risk categories, the 2012 Briganti nomogram represented an independent predictor of PCa progression after surgery. Likewise, as the nomogram score increased so patients were more likely to experience disease progression. Accordingly, it may allow further stratification of patients within each risk category to modulate appropriate treatment paradigms. Full article
(This article belongs to the Section Urology & Nephrology)
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier plots depicting PCa progression-free survival in 1047 patients belonging to all European Association of Urology (EAU) prognostic risk categories stratified according to the 2012 Briganti nomogram score distribution based on tertiles: less than 3% vs. from 3% to less than 8% vs. at least 8% or greater.</p>
Full article ">
15 pages, 2006 KiB  
Article
Prognostic Value of Myosteatosis and Albumin–Bilirubin Grade for Survival in Hepatocellular Carcinoma Post Chemoembolization
by Kittipitch Bannangkoon, Keerati Hongsakul, Teeravut Tubtawee and Natee Ina
Cancers 2024, 16(20), 3503; https://doi.org/10.3390/cancers16203503 - 17 Oct 2024
Viewed by 754
Abstract
Objective: This study aimed to investigate the prognostic value of preoperative myosteatosis and the albumin–bilirubin (ALBI) grade in patients with hepatocellular carcinoma (HCC) undergoing transarterial chemoembolization (TACE) and develop a robust prognostic score based on these factors. Methods: Patients with HCC who underwent [...] Read more.
Objective: This study aimed to investigate the prognostic value of preoperative myosteatosis and the albumin–bilirubin (ALBI) grade in patients with hepatocellular carcinoma (HCC) undergoing transarterial chemoembolization (TACE) and develop a robust prognostic score based on these factors. Methods: Patients with HCC who underwent TACE between January 2009 and December 2020 were included. Multivariate Cox regression analysis identified prognostic factors. CT-based body composition parameters were acquired from baseline abdominal CT images at the level of the third lumbar vertebra. A prognostic score (Myo-ALBI) was developed based on the presence of preoperative myosteatosis and the ALBI grade, and its prognostic value was evaluated. Results: Of 446 patients, 63% were male, and the mean age was 62.4 years. Preoperative myosteatosis was present in 41.5% of patients. The BCLC stages were mostly B (67.9%). Multivariate analysis shows that preoperative myosteatosis, ALBI grade 2, and ALBI grade 3 were independent prognostic factors. The Myo-ALBI grade was incorporated into a prognostic model, including alpha-fetoprotein and up-to-seven criteria, to generate a nomogram. The C-index of the nomogram based on the Myo-ALBI grade (0.743) was significantly higher than the non-Myo-ALBI nomogram (0.677), the up-to-seven criteria (0.653), the ALBI grade (0.616), and the Child–Pugh class (0.573) (all p < 0.05). The t-ROC curve for the nomogram was consistently superior to the other models throughout the observation period in all patients and the BCLC-B subgroup. Conclusions: The combination of preoperative CT-derived myosteatosis and ALBI grade enhances prognostication for patients with unresectable HCC undergoing TACE. The Myo-ALBI nomogram constructed in this study could support individualized prognosis prediction, assisting in treatment decision-making for HCC patients. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

Figure 1
<p>Association between the skeletal muscle density and clinical parameters. Scatter plots demonstrate that skeletal muscle density is positively associated with sarcopenia (<b>A</b>) while negatively correlated with body mass index (<b>B</b>).</p>
Full article ">Figure 2
<p>Kaplan–Meier analysis for overall survival of patients with hepatocellular carcinoma according to preoperative myosteatosis and ALBI grade. Kaplan–Meier analysis for OS according to (<b>A</b>) preoperative myosteatosis, (<b>B</b>) ALBI grade, (<b>C</b>) combination of preoperative ALBI grade and myosteatosis, and (<b>D</b>) Myo-ALBI grade. ALBI grade, albumin–bilirubin grade.</p>
Full article ">Figure 3
<p>Nomogram for predicting 1-, 2-, and 3-year overall survival in hepatocellular carcinoma patients undergoing TACE. (<b>A</b>): Nomogram to predict 1-, 2-, and 3-year overall survival in HCC patients who underwent TACE. Calibration plot of the nomogram for 2-year survival (<b>B</b>) and 3-year survival (<b>C</b>). ALBI grade, albumin–bilirubin grade.</p>
Full article ">Figure 4
<p>Time-dependent receiver operating characteristic curves for the Myo-ALBI nomogram, non-Myo-ALBI nomogram, up-to-seven criteria, ALBI grade, and Child–Pugh class for the prediction of overall survival in all patients (<b>A</b>) and BCLC-B patients (<b>B</b>). AUC, area under the curve; ALBI grade, albumin–bilirubin grade.</p>
Full article ">
11 pages, 1321 KiB  
Article
A Risk Correlative Model for Sleep Disorders in Chinese Older Adults Based on Blood Micronutrient Levels: A Matched Case-Control Study
by Cheng Cheng, Xukun Chen, Liyang Zhang, Zehao Wang, Huilian Duan, Qi Wu, Ruiting Yan, Di Wang, Zhongxia Li, Ruikun He, Zhenshu Li, Yongjie Chen, Fei Ma, Yue Du, Wen Li and Guowei Huang
Nutrients 2024, 16(19), 3306; https://doi.org/10.3390/nu16193306 - 29 Sep 2024
Viewed by 796
Abstract
Background: The physical abilities of older adults decline with age, making them more susceptible to micronutrient deficiency, which may affect their sleep quality. Objectives: This study aimed to construct a risk correlative model for sleep disorders in Chinese older adults based on blood [...] Read more.
Background: The physical abilities of older adults decline with age, making them more susceptible to micronutrient deficiency, which may affect their sleep quality. Objectives: This study aimed to construct a risk correlative model for sleep disorders in Chinese older adults based on blood micronutrient levels. Methods: In this matched case-control study, we recruited 124 participants with sleep disorders and 124 matched controls from the Tianjin Elderly Nutrition and Cognition cohort in China. Micronutrient levels in whole blood were measured using the dried blood spot technique. We compared the differences in micronutrient levels between the two groups and also constructed a receiver operating characteristic (ROC) model and nomogram for sleep disorders. Results: In comparison to the control group, the sleep disorders group showed lower levels of blood vitamin A, vitamin E (VE), folate, magnesium, copper, iron, and selenium (Se) in the univariate analysis (p < 0.05). The ROC curve analysis indicated that the combination of VE + folate + Se may have an excellent diagnostic effect on sleep disorders, with an area under the curve of 0.964. This VE + folate + Se was integrated into a nomogram model to demonstrate their relationship with sleep disorders. The consistency index of the model was 0.88, suggesting that the model assessed sleep disorders well. Conclusions: The sleep disorders risk correlative model constructed by the levels of VE, folate, and Se in whole blood might show good performance in assessing the risk of sleep disorders in older adults. Full article
(This article belongs to the Section Geriatric Nutrition)
Show Figures

Figure 1

Figure 1
<p>Flow chart of the study population.</p>
Full article ">Figure 2
<p>The importance of micronutrients for sleep disorders. *, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>ROC for assessing sleep disorders. ROC, receiver operating characteristic; AUC, area under the curve.</p>
Full article ">Figure 4
<p>Nomogram to assess the probability of sleep disorder events in participants. C index = 0.880.</p>
Full article ">
13 pages, 1317 KiB  
Article
Prognostic Index for Liver Radiation (PILiR)
by Laura Callan, Haddis Razeghi, Natalie Grindrod, Stewart Gaede, Eugene Wong, David Tan, Jason Vickress, John Patrick and Michael Lock
Curr. Oncol. 2024, 31(10), 5862-5872; https://doi.org/10.3390/curroncol31100436 - 29 Sep 2024
Viewed by 646
Abstract
A Prognostic Index for Liver Radiation (PILiR) for improved patient selection for stereotactic liver-directed radiotherapy (SBRT) was developed. Using a large single-center database, 195 patients treated with SBRT for local control, including 66 with hepatocellular carcinoma (HCC) and 129 with metastatic liver disease, [...] Read more.
A Prognostic Index for Liver Radiation (PILiR) for improved patient selection for stereotactic liver-directed radiotherapy (SBRT) was developed. Using a large single-center database, 195 patients treated with SBRT for local control, including 66 with hepatocellular carcinoma (HCC) and 129 with metastatic liver disease, were analyzed. Only patients ineligible for alternative treatments were included. Overall survival was 11.9 months and 9.4 months in the HCC group and metastatic groups, respectively. In the combined dataset, Child–Pugh Score (CPS) (p = 0.002), serum albumin (p = 0.039), and presence of extrahepatic disease (p = 0.012) were significant predictors of early death on multivariable analysis and were included in the PILiR (total score 0 to 5). Median survival was 23.8, 9.1, 4.5, and 2.6 months for patients with 0, 1–2, 3, and 4–5 points, respectively. In the HCC dataset, CPS (p < 0.001) and gross tumor volume (p = 0.013) were predictive of early death. In the metastatic dataset, serum albumin (p < 0.001) and primary disease site (p = 0.003) were predictive of early death. The AUC for the combined, HCC, and metastatic datasets are 0.78, 0.84, and 0.80, respectively. Poor liver function (defined by CPS and serum albumin) and extrahepatic disease were most predictive of early death, providing clinically important expected survival information for patients and caregivers. Full article
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier Survival curves for HCC and metastatic datasets. Median overall survival is 11.9 months for the HCC dataset and 9.4 months for the metastatic dataset.</p>
Full article ">Figure 2
<p>PILiR score to estimate survival based on pre-treatment characteristics. Median survival (months) in the combined group based on PILiR: 0 = 23.8; 1–2 = 9.1; 3 = 4.5; 4–5 = 2.6. Median survival (months) in the HCC group based on PILiR: 0 = 20.9; 1–3 = 7.8; 4 = 1.8. Median survival (months) in the metastatic group based on PILiR: 0 = 13.9; 1–2 = 13.0; 3–4 = 2.9.</p>
Full article ">Figure 3
<p>Kaplan–Meier Survival curves based on PILiR score for the combined dataset. Patients can be classified as Good Prognosis (0 points) with a median survival of 23.8 months, Fair Prognosis (1 or 2 points) with a median survival of 9.1 months, Borderline Prognosis (3 points) with a median survival of 4.5 months, or Poor Prognosis (4 or 5 points) with a median survival of 2.6 months.</p>
Full article ">Figure 4
<p>ROC for combined dataset, HCC dataset and metastatic dataset. The area under the curve (AUC) was 0.74, 0.79, and 0.75, respectively.</p>
Full article ">
12 pages, 9170 KiB  
Article
A Novel Prognostic Model of Hepatocellular Carcinoma per Two NAD+ Metabolic Synthesis-Associated Genes
by Luo Dai, Shiliu Lu, Linfeng Mao, Mingbei Zhong, Gangping Feng, Songqing He and Guandou Yuan
Int. J. Mol. Sci. 2024, 25(19), 10362; https://doi.org/10.3390/ijms251910362 - 26 Sep 2024
Viewed by 627
Abstract
Hepatocellular carcinoma (HCC) is a formidable challenge to global human health, while recent years have witnessed the important role of NAD+ in tumorigenesis and progression. However, the expression pattern and prognostic value of NAD+ in HCC still remain elusive. Gene expression files and [...] Read more.
Hepatocellular carcinoma (HCC) is a formidable challenge to global human health, while recent years have witnessed the important role of NAD+ in tumorigenesis and progression. However, the expression pattern and prognostic value of NAD+ in HCC still remain elusive. Gene expression files and corresponding clinical pathological files associated with HCC were obtained from the Cancer Genome Atlas (TCGA) database, and genes associated with NAD+ were retrieved from the GSEA and differentially analyzed in tumor and normal tissues. A consensus clustering analysis was conducted by breaking down TCGA patients into four distinct groups, while Kaplan–Meier curves were generated to investigate the disparity in clinical pathology and endurance between clusters. A prognostic model based on NAD+-associated genes was established and assessed by combining LASSO-Cox regression, uni- and multi-variate Cox regression, and ROC curve analyses. Investigations were conducted to determine the expression of distinct mRNAs and proteins in both HCC and non-tumor tissues. A novel two-gene signature including poly (ADP-Ribose) polymerase 2 (PARP2) and sirtuin 6 (SIRT6) was obtained through LASSO-Cox regression and was identified to have favorable prognostic performance in HCC patients from TCGA. Analyses of both single and multiple variables showed that the prognostic model was a distinct prognostic factor in the endurance of liver cancer patients in both the training and trial groups. The nomogram also exhibited clinical significance in the prognosis of HCC patients. Immunohistochemistry, qRT-PCR, and Western blotting revealed that HCC samples exhibited higher PARP2 and SIRT6 expression levels than those of normal controls. This study identified a robust prognostic model comprising two NAD+-associated genes using bioinformatic methods, which is accurate in predicting the survival outcome of HCC patients. This model might benefit the early diagnosis of HCC and further facilitate the management of individualized medical service and clinical decision-making. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

Figure 1
<p>The structures of NAD+ and NADH.</p>
Full article ">Figure 2
<p>Expression of 32 NAD+-related genes in hepatocellular carcinoma and their correlation, as well as biological regulation of interacted proteins. (<b>A</b>) Heat map of expression difference in NAD+-related genes. (<b>B</b>) Heat map of correlation between DEGs. (<b>C</b>) PPI network map of DEGs. (<b>D</b>) DEGs involved in biological regulation of related proteins.</p>
Full article ">Figure 3
<p>The consensus clustering analysis of the NAD+ regulators. (<b>A</b>) The most appropriate selection with clustering stability when k = 4 was used. (<b>B</b>) The overall survival rates of different clusters were different. (<b>C</b>) Heat maps of the relationship between different clusters and clinicopathology.</p>
Full article ">Figure 4
<p>The risk model of the NAD+ regulatory factor was constructed based on TCGA hepatocellular carcinoma (HCC) data. (<b>A</b>) With <span class="html-italic">p</span> &lt; 0.2 as the standard, 4 genes related to OS were screened by uni-variate regression analysis. (<b>B</b>) Target genes were screened as prognostic models by LASSO. (<b>C</b>) Cross-validation is intended to adjust parameter selection in LASSO regression. (<b>D</b>) A heat map of the relationship between the risk score and 2 genes. (<b>E</b>) HCC patient distribution based on the risk score. (<b>F</b>) HCC patient survival status (a dotted line is used to identify patients with high or low risk scores). (<b>G</b>) A Kaplan–Meier plot was used to compare OS between the high- and low-risk groups. (<b>H</b>) A PCA diagram based on the risk score. (<b>I</b>) The ssGSEA method was applied to the HCC samples to evaluate the distribution of 23 immune cell types. * <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 5
<p>GESA analysis based on GO and KEGG. (<b>A</b>) The top 5 most significantly enriched KEGG pathways in the high-risk group. (<b>B</b>) The top 5 most significantly enriched GO terms in the high-risk group. (<b>C</b>) The top 5 most significantly enriched KEGG pathways in the low-risk group. (<b>D</b>) The top 5 most significantly enriched GO terms in the low-risk group.</p>
Full article ">Figure 6
<p>Uni-variate and multi-variate regression analyses were used to assess the prognostic value of risk scores in TCGA. (<b>A</b>,<b>B</b>) Uni-variate and multi-variate regression analyses in TCGA. (<b>C</b>–<b>I</b>) The correlation between the risk score and clinical features of patients from TCGA.</p>
Full article ">Figure 7
<p>Nomogram for evaluating prognosis. (<b>A</b>) Nomogram was applied to assess prognosis of HCC patients in TCGA. (<b>B</b>) Calibration curve for predicting 1-, 3-, and 5-year prognosis of HCC patients.</p>
Full article ">Figure 8
<p>Verification of clinical tissue samples. (<b>A</b>,<b>B</b>) Expression of PARP2 and SIRT6 in HCC tissues and non-tumor tissues analyzed by GEPIA2.0. (<b>C</b>,<b>D</b>) RT-qPCR analysis of PARP2 and SIRT6 in HCC and non-tumor tissues. (<b>E</b>) Protein level of PARP2 and SIRT6 in HCC and non-tumor tissues. (<b>F</b>) Characteristic pictures of PARP2 and SIRT6 in HCC and adjacent tumor. (<b>G</b>) Statistical diagram of F. (<b>H</b>) Survival analysis between patients in high-risk and low-risk group. (<b>I</b>) ROC curve was used to evaluate predictive effectiveness. All data are shown as mean ± standard deviation. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
25 pages, 19339 KiB  
Article
Identifying Key Genes as Progression Indicators of Prostate Cancer with Castration Resistance Based on Dynamic Network Biomarker Algorithm and Weighted Gene Correlation Network Analysis
by Siyuan Liu, Yi Hu, Fei Liu, Yizheng Jiang, Hongrui Wang, Xusheng Wu and Dehua Hu
Biomedicines 2024, 12(9), 2157; https://doi.org/10.3390/biomedicines12092157 - 23 Sep 2024
Viewed by 1001
Abstract
Background: Androgen deprivation therapy (ADT) is the mainstay of treatment for prostate cancer, yet dynamic molecular changes from hormone-sensitive to castration-resistant states in patients treated with ADT remain unclear. Methods: In this study, we combined the dynamic network biomarker (DNB) method and the [...] Read more.
Background: Androgen deprivation therapy (ADT) is the mainstay of treatment for prostate cancer, yet dynamic molecular changes from hormone-sensitive to castration-resistant states in patients treated with ADT remain unclear. Methods: In this study, we combined the dynamic network biomarker (DNB) method and the weighted gene co-expression network analysis (WGCNA) to identify key genes associated with the progression to a castration-resistant state in prostate cancer via the integration of single-cell and bulk RNA sequencing data. Based on the gene expression profiles of CRPC in the GEO dataset, the DNB method was used to clarify the condition of epithelial cells and find out the most significant transition signal DNB modules and genes included. Then, we calculated gene modules associated with the clinical phenotype stage based on the WGCNA. IHC was conducted to validate the expression of the key genes in CRPC and primary PCa patients Results:Nomograms, calibration plots, and ROC curves were applied to evaluate the good prognostic accuracy of the risk prediction model. Results: By combining single-cell RNA sequence data and bulk RNA sequence data, we identified a set of DNBs, whose roles involved in androgen-associated activities indicated the signals of a prostate cancer cell transition from an androgen-dependent state to a castration-resistant state. In addition, a risk prediction model including the risk score of four key genes (SCD, NARS2, ALDH1A1, and NFXL1) and other clinical–pathological characteristics was constructed and verified to be able to reasonably predict the prognosis of patients receiving ADT. Conclusions: In summary, four key genes from DNBs were identified as potential diagnostic markers for patients treated with ADT and a risk score-based nomogram will facilitate precise prognosis prediction and individualized therapeutic interventions of CRPC. Full article
(This article belongs to the Topic Recent Advances in Anticancer Strategies)
Show Figures

Figure 1

Figure 1
<p>Flowchart of this study.</p>
Full article ">Figure 2
<p>Ten cell clusters with different annotations based on CRPC scRNA-seq data, revealing cellular heterogeneity in CRPC. (<b>A</b>,<b>B</b>) Dimensionality reduction based on t-SNE algorithm and the distribution of 6 CRPC samples from GSE137829 dataset and 21 clusters were acquired; (<b>C</b>) expression level of marker genes in each cell cluster; (<b>D</b>) cell cluster annotation based on the composition of marker genes; and (<b>E</b>) proportion of different cell types in each sample.</p>
Full article ">Figure 3
<p>Cell annotation and construction of differentiation trajectories in epithelial cells based on single cell sequence data. (<b>A</b>) Annotating epithelial cells according to marker genes; (<b>B</b>) proportion of epithelial cell subgroups in different samples; (<b>C</b>) marker gene expression level of each epithelial cell subgroup; (<b>D</b>) pseudotime differentiation trajectories of epithelial cells; and (<b>E</b>) differentiation trajectory states of different subgroups.</p>
Full article ">Figure 4
<p>Key transformed cell subgroups and gene modules of epithelial cells. (<b>A</b>) Assumed key transforming signal in different cellular differentiation states in epithelial cells. (<b>B</b>,<b>C</b>) Random score of key transforming signals after shuffling gene labels; (<b>D</b>,<b>E</b>) random score of key transforming signals after shuffling sample labels; (<b>F</b>) expression of key transforming module genes in different epithelial cell states; (<b>G</b>) GO BP enrichment state of key transforming module genes in different epithelial cell states; and (<b>H</b>,<b>I</b>) cancer hallmark enrichment and significance of key transforming module genes in different subgroups of epithelial cells.</p>
Full article ">Figure 5
<p>Heterogeneity in cell communication of S1 and S2 epithelial cells in CRPC. (<b>A</b>) Cell communication network of S1 and S2 epithelial cells with other cell types (the S2 group is at the top and the S1 group is at the bottom. The size of the spot indicates the number of cells). (<b>B</b>) Comparison of the communication strength in different signaling pathways in S1 and S2 epithelial cells. The color “red” on the vertical axis indicates that cell communication was more active in S2 and the color “blue” indicates cell communication was more active in S1. The color “black” indicates that there was no significance between the two groups. (<b>C</b>) Cell communication number and strength among different cell types. The color indicates the difference. The color “red” indicates cell communication was more active in S2 and the color “blue” indicates that cell communication was more active in S1. The bar chart on the right indicates that the outgoing signal and the bar chart on the top indicates the incoming signal. (<b>D</b>) Heatmap of signaling pathway strength in epithelial cells in the S1 and S2 groups.</p>
Full article ">Figure 6
<p>Ligand–receptor differences between S1 and S2 epithelial cells in CRPC. (<b>A</b>,<b>B</b>) Differences in functional ligand–receptor interactions in epithelial cells in S1 and S2 groups to other cell subgroups. The color “red” on the horizontal axis indicates communication of epithelial cells in the S2 group and the color “cyan” indicates communication of epithelial cells in the S1 group. The color of the spot indicates the cellular communication probability and the size of the spot indicates the significance of the <span class="html-italic">p</span> value. (<b>C</b>,<b>D</b>) Differences in functional ligand–receptors of other cell subgroups to epithelial cells in the S1 and S2 groups. The color “red” on the horizontal axis indicates communication of epithelial cells in the S2 group and color “cyan” indicates communication of epithelial cells in the S1 group. The color of the spot indicates the cellular communication probability and the size of the spot indicates the significance of the <span class="html-italic">p</span> value.</p>
Full article ">Figure 7
<p>Identification of co-expression modules of androgen-related key genes based on CRPC bulk seq. data. (<b>A</b>,<b>B</b>) Cluster dendrogram of co-expression network modules in GSE70770 and GSE80609 (1-TOM). (<b>C</b>) GO analysis of “midnightblue” co-expressed gene modules in GSE70770. (<b>D</b>) GO analysis of “blue” co-expressed gene modules in GSE80609.</p>
Full article ">Figure 8
<p>Fuzzy clustering and evaluation of key biomarkers in the key transformation subgroups of epithelial cells. (<b>A</b>) Using Mfuzz to clarify the dynamic change in neighboring genes in the key transforming signaling modules in different states of cytodifferentiation in epithelial cells. (<b>B</b>) Hallmark enrichment analysis of the cluster 4 gene set. (<b>C</b>) Intersection Venn plot of genes in the WGCNA, DNB, and soft clustering analysis.</p>
Full article ">Figure 9
<p>Key transformed cell subgroups and gene modules of epithelial cells. (<b>A</b>) Risk score of the 4 key biomarkers. (<b>B</b>) Kaplan–Meier curve of the TCGA training cohort. (<b>C</b>) Kaplan–Meier curve of the GSE111177 validation cohort. (<b>D</b>) A nomogram combining the risk score, age, Gleason grade, and tumor stage was developed to predict the 1-, 2-, and 3-year PFIs of patients who underwent ADT in the TCGA cohort. (<b>E</b>) A 1-year calibration analysis of the TCGA cohort nomogram. (<b>F</b>) ROC curves of multiple time points (1 year, 2 years, 3 years) of the PFI in the TCGA cohort.</p>
Full article ">Figure 10
<p>Representative photo images and histograms of ALDH1A1, SCD, NARS2, or NFXL1 protein expression levels in CRPC or HSPC sample tissues. (<b>A</b>) Representative IHC images showing high ALDH1A1 and SCD expression in CRPC tissue and high NARS2 expression in HSPC tissue. (<b>B</b>) Histograms of ALDH1A1, SCD, NARS2, and NFXL1 expression levels. Scale bars, 625 μm and 100 μm. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01; ns, not significant.</p>
Full article ">
14 pages, 5888 KiB  
Article
Comprehensive Analysis of Characteristics of Cuproptosis-Related LncRNAs Associated with Prognosis of Lung Adenocarcinoma and Tumor Immune Microenvironment
by Feihong Chen, Xin Wen, Jiani Wu, Min Feng and Shicheng Feng
Pharmaceuticals 2024, 17(9), 1244; https://doi.org/10.3390/ph17091244 - 21 Sep 2024
Viewed by 661
Abstract
As a novel discovered mechanism of cell death, cuproptosis is copper-dependent and induces protein toxicity related to advanced tumors, disease prognosis, and human innate and adaptive immune response. However, it has not yet been fully established how the prognosis of lung adenocarcinoma (LUAD) [...] Read more.
As a novel discovered mechanism of cell death, cuproptosis is copper-dependent and induces protein toxicity related to advanced tumors, disease prognosis, and human innate and adaptive immune response. However, it has not yet been fully established how the prognosis of lung adenocarcinoma (LUAD) is related to the immune microenvironment of cuproptosis-related lncRNAs using several bioinformatic techniques. In the study, 19 genes related to cuproptosis were collected. Subsequently, 783 lncRNAs related to the co-expression of cuproptosis were obtained. Moreover, the Cox model revealed and constructed four lncRNA (AC012020.1, AC114763.1, AL161431.1, AC010260.1) prognostic markers related to cuproptosis. Based on the median risk score (RS) values, patients were categorized into two groups: high risk and low risk. The Kaplan–Meier (KM) survival curve depicted a statistically significant overall survival (OS) rate among two groups. Principal component analysis (PCA) and receiver operator characteristic curve (ROC) proved that the model had promising ability in prognosis. The analysis of univariate and multivariate Cox regression revealed that RS served as an independent prognostic factor. Moreover, multivariate Cox regression was employed for the establishment of a nomogram of prognostic indicators. The tumor mutational burden (TMB) depicted a considerable difference between the two risk groups. The immunotherapy response of LUAD patients with high risk was improved compared to low risk patients. The study also revealed that drug sensitivity associated with LUAD was significantly linked to RS. The findings could be helpful to establish a good diagnosis, prognosis, and management regime for patients with LUAD. Full article
(This article belongs to the Special Issue Data-Driven Biomarker and Drug Discovery for Complex Disease)
Show Figures

Figure 1

Figure 1
<p>Sankey diagram and heat map. (<b>A</b>) Sankey diagram of co-expression between 19 cuproptosis-related genes and 240 cuproptosis-related lncRNAs. (<b>B</b>) Correlation of 19 cuproptosis-related genes and 4 prognostic cuproptosis-related lncRNAs. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>Construction of the prognostic cuproptosis-related lncRNA risk model in LUAD. (<b>A</b>) Univariate Cox regression analysis for identifying the prognostic cuproptosis-related lncRNAs. (<b>B,C</b>) Lasso–Cox regression analysis was performed to construct prognostic prediction models. (<b>D</b>) Kaplan–Meier curves for survival analysis in the high- and low-risk groups in the entire cohort. (<b>E</b>) Kaplan–Meier curves for survival analysis in the validation set. (<b>F</b>) Kaplan–Meier curves for survival analysis in the training set. (<b>G</b>) Kaplan–Meier curves of progression-free survival (PFS).</p>
Full article ">Figure 3
<p>Validation of the risk model in the cohort and principal component analysis. Risk score distribution, survival status, and heatmap of the prognostic markers and overall survival in the cohort. (<b>A</b>) The training set. (<b>B</b>) The validation set. (<b>C</b>) The entire cohort. PCA between the high- and low-risk groups based on (<b>D</b>) all genes, (<b>E</b>) cuproptosis-related genes, (<b>F</b>) cuproptosis-related lncRNAs, and (<b>G</b>) cuproptosis-related lncRNA prognostic markers.</p>
Full article ">Figure 4
<p>Independent prognostic analysis of LUAD overall survival (OS). (<b>A</b>) Univariate Cox analysis. Age, stage, and risk score were statistically significant. (<b>B</b>) Multivariate Cox analysis. Age, stage, and risk score were statistically significant. (<b>C</b>) ROC demonstrated the predictive accuracy of the risk model. (<b>D</b>) Time ROC curve predicted 1, 3, and 5 years of OS for LUAD patients. (<b>E</b>) C-index showed the predictive accuracy of the risk model and stage was superior to other clinical parameters.</p>
Full article ">Figure 5
<p>Construction and evaluation of a nomogram based on lncRNAs. (<b>A</b>) A nomogram used to predict prognosis was constructed based on lncRNAs. ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001. (<b>B</b>) Calibration curves are used to predict 1-, 3-, and 5-year overall survival. (<b>C</b>) Kaplan–Meier curves of patients with stage I–II. (<b>D</b>) Kaplan–Meier curves of patients with stage III–IV.</p>
Full article ">Figure 6
<p>Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. (<b>A</b>) Circle diagram of GO enrichment analysis. (<b>B</b>) Bar plot of the top 10 GO enrichment terms. (<b>C</b>) Bubble chart of the top 10 GO enrichment terms. (<b>D</b>) Circle diagram of KEGG enrichment analysis. (<b>E</b>) Bar plot of the top 30 KEGG enrichment terms. (<b>F</b>) Bubble chart of the top 30 KEGG enrichment terms.</p>
Full article ">Figure 7
<p>Immunological landscape in LUAD patients and relationship between tumor mutation burden (TMB) and risk score. (<b>A</b>) Heatmap of the tumor-infiltrating lymphocytes based on single-sample gene set enrichment analysis algorithms among the high- and low-risk groups in LUAD. (<b>B</b>) Multi-GSEA showed the enrichment pathway in high-risk groups. (<b>C</b>) Waterfall plot of top 15 mutant genes in the high-risk group in LUAD. (<b>D</b>) Waterfall plot of top 15 mutant genes in the low-risk group in LUAD. (<b>E</b>) Analysis of TMB differences between the high- and low-risk groups in LUAD. (<b>F</b>) Comparison of TIDE prediction score between the high- and low-risk groups. (<b>G</b>) Survival analysis curves of the high- and low-TMB groups. (<b>H</b>) TMB risk combined with survival curve in LUAD.</p>
Full article ">Figure 8
<p>Drug effectiveness of different risk groups.</p>
Full article ">
11 pages, 1099 KiB  
Article
Analysis of Calculated Liver Scores for Long-Term Outcome in 423 Cutaneous Melanoma Patients
by Nessr Abu Rached, Mariana Marques da Silva Reis, Eggert Stockfleth, Riina Käpynen and Thilo Gambichler
Cancers 2024, 16(18), 3217; https://doi.org/10.3390/cancers16183217 - 21 Sep 2024
Viewed by 532
Abstract
Background: Neoadjuvant and adjuvant therapies are currently getting increasingly important in cutaneous melanoma (CM) management. However, there is still a lack of prognostic tools to identify which patients have a poor prognosis. There is increasing evidence that the liver score may be a [...] Read more.
Background: Neoadjuvant and adjuvant therapies are currently getting increasingly important in cutaneous melanoma (CM) management. However, there is still a lack of prognostic tools to identify which patients have a poor prognosis. There is increasing evidence that the liver score may be a potential prognostic parameter in different tumour types. The aim was to investigate whether established liver scores can establish the prognosis of CM. Methods: According to established methods, the APRI, the MELD score, the MELD-Na score and the De Ritis ratio were calculated from the laboratory values at the time of the initial diagnosis. Survival was compared with the Kaplan–Meier curve and tested with log-rank tests. Risk factors associated with cutaneous melanoma-specific survival (CMSS) and progression-free survival (PFS) were assessed by using the Cox proportional hazards regression model. To determine the diagnostic accuracy, we performed a time-dependent ROC analysis. Results: A total of 423 patients were included, including 141 patients in AJCC stage (2017) I (33.3%), 82 in stage II (19.4%), 128 in stage III (30.3%) and 72 in stage IV (17%). Median time until melanoma-specific death was 99 months (IQR: 37–126). In addition, 37.6% of patients relapsed with a median time to relapse of 88 months (IQR: 17.5–126). In all stages, tumour thickness and ulceration were independent markers for predicting CMSS and PFS (p < 0.05). The multivariable analysis with all stages showed no significant association with CM outcome for liver scores (p > 0.05). The subgroup analysis revealed that the APRI (≥0.2241) was associated with CMSS and PFS in melanoma stages I and II, independently of tumour thickness, age and ulceration (HR 2.57, 95% CI 1.14–5.75; HR 2.94, 95% CI 1.42–6.09, respectively). Conclusions: The 20-year prognosis of AJCC stage I and II CM was dependent on tumour thickness and the APRI. High tumour thickness and an APRI ≥ 0.2241 at the initial diagnosis were associated with a worse prognosis. Future studies should investigate the independent prognostic value of the APRI in low-stage CM. Furthermore, the APRI score could be a potential biomarker for nomograms. Full article
(This article belongs to the Special Issue Advances in Skin Cancer: Diagnosis, Treatment and Prognosis)
Show Figures

Figure 1

Figure 1
<p>The 20-year survival probabilities are shown by Kaplan–Meier curves for patients with cutaneous melanoma in AJCC I and II. The curves show that an APRI ≥ 0.2241 was significantly associated with decreased cutaneous melanoma-specific survival (log-rank test: <span class="html-italic">p</span> = 0.001).</p>
Full article ">Figure 2
<p>The 20-year progression-free survival rates are shown by Kaplan–Meier curves for patients with cutaneous melanoma in AJCC I and II. The curves show that an APRI ≥ 0.2241 was significantly associated with decreased progression-free survival rates (log-rank test: <span class="html-italic">p</span> = 0.001).</p>
Full article ">Figure 3
<p>Time-dependent ROC analyses: (<b>a</b>) APRI ≥ 0.2241 in relation to cutaneous melanoma-specific survival (CMSS), (<b>b</b>) APRI ≥ 0.2241 in relation to progression-free survival (PFS), (<b>c</b>) tumour thickness ≥ 1.66 in relation to CMSS and (<b>d</b>) tumour thickness ≥ 1.66 in relation to PFS.</p>
Full article ">
Back to TopTop