[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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (697)

Search Parameters:
Keywords = nomogram

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 5482 KiB  
Article
Topographic Pattern-Based Nomogram to Guide Keraring Implantation in Eyes with Mild to Moderate Keratoconus: Visual and Refractive Outcome
by Ugo de Sanctis, Paolo Caselgrandi, Carlo Gennaro, Cecilia Tosi, Enrico Borrelli, Paola Marolo and Michele Reibaldi
J. Clin. Med. 2025, 14(3), 870; https://doi.org/10.3390/jcm14030870 - 28 Jan 2025
Abstract
Background: To assess the outcome of Keraring (Mediphacos, Brazil) implantation according to a topographic pattern-based nomogram in eyes with mild to moderate keratoconus. Materials and Methods: A topographic pattern-based nomogram was used to guide Keraring selection in 47 consecutive eyes with stage [...] Read more.
Background: To assess the outcome of Keraring (Mediphacos, Brazil) implantation according to a topographic pattern-based nomogram in eyes with mild to moderate keratoconus. Materials and Methods: A topographic pattern-based nomogram was used to guide Keraring selection in 47 consecutive eyes with stage I-II keratoconus (Amsler-Krumeich staging), which underwent femtosecond laser-assisted implantation at a single center. Electronic data of LogMar uncorrected distance visual acuity (UDVA) and corrected distance visual acuity (CDVA) manifest refraction and tomographic analysis (Pentacam HR, Oculus, Germany) measured preoperatively and at the last postoperative examination were retrospectively analyzed. Results: Mean follow-up was 18.8 months. (range 3–35). Mean UDVA improved (p < 0.001) from 0.87 ± 0.27 to 0.35 ± 0.21. UDVA increased on average by 5.13 lines. Mean CDVA improved from 0.21 ± 0.10 to 0.09. ± 0.07, and the proportion of eyes with CDVA ≥ 20/25 increased from 29.8% to 85.1% after surgery. No eyes lost lines of CDVA. The Alpins correction index of astigmatism was 0.77 and the mean refractive cylinder decreased from 4.99 ± 1.89 to −2.31 ± 1.47 D (p < 0.001). Mean and maximal keratometry was reduced on average by −2.10 ± 1.42 D and −3.02 ± 3.68 D, respectively (p < 0.001). The RMS of corneal high-order aberrations dropped from 3.296 ± 1.180 µm to 2.192 ± 0.919 µm, and that of vertical coma from −2.656 ± 1.189 µm to −1.427 ± 1.024 µm (p < 0.001). All topometric indices improved after surgery. Conclusions: Planning Keraring implantation using the topographic pattern-based nomogram is very effective and safe in eyes with mild to moderate keratoconus. Using that nomogram of UDVA and CDVA are clinically significant. Full article
(This article belongs to the Section Ophthalmology)
Show Figures

Figure 1

Figure 1
<p>Cumulative percentage of preoperative CDVA versus postoperative UDVA.</p>
Full article ">Figure 2
<p>Changes in UDVA Snellen Lines.</p>
Full article ">Figure 3
<p>Cumulative percentage of preoperative CDVA versus postoperative CDVA.</p>
Full article ">Figure 4
<p>Changes in CDVA Snellen Lines.</p>
Full article ">Figure 5
<p>Preoperative and postoperative sphere, cylinder, and sphero-equivalent data.</p>
Full article ">Figure 6
<p>Overview of Alpins vectorial analysis.</p>
Full article ">Figure 7
<p>Examples of differences between postoperative vs. preoperative axial curvature maps in eyes with topographic patterns such as a croissant (<b>A</b>), duck (<b>B</b>), snowman (<b>C</b>), and bowtie (<b>D</b>).</p>
Full article ">Figure 7 Cont.
<p>Examples of differences between postoperative vs. preoperative axial curvature maps in eyes with topographic patterns such as a croissant (<b>A</b>), duck (<b>B</b>), snowman (<b>C</b>), and bowtie (<b>D</b>).</p>
Full article ">Figure 7 Cont.
<p>Examples of differences between postoperative vs. preoperative axial curvature maps in eyes with topographic patterns such as a croissant (<b>A</b>), duck (<b>B</b>), snowman (<b>C</b>), and bowtie (<b>D</b>).</p>
Full article ">
18 pages, 6491 KiB  
Article
An Integrated Approach Utilizing Single-Cell and Bulk RNA-Sequencing for the Identification of a Mitophagy-Associated Genes Signature: Implications for Prognostication and Therapeutic Stratification in Prostate Cancer
by Yuke Zhang, Li Ding, Zhijin Zhang, Liliang Shen, Yadong Guo, Wentao Zhang, Yang Yu, Zhuoran Gu, Ji Liu, Aimaitiaji Kadier, Jiang Geng, Shiyu Mao and Xudong Yao
Biomedicines 2025, 13(2), 311; https://doi.org/10.3390/biomedicines13020311 - 27 Jan 2025
Abstract
Introduction: Prostate cancer, notably prostate adenocarcinoma (PARD), has high incidence and mortality rates. Although typically resistant to immunotherapy, recent studies have found immune targets for prostate cancer. Stratifying patients by molecular subtypes may identify those who could benefit from immunotherapy. Methods: [...] Read more.
Introduction: Prostate cancer, notably prostate adenocarcinoma (PARD), has high incidence and mortality rates. Although typically resistant to immunotherapy, recent studies have found immune targets for prostate cancer. Stratifying patients by molecular subtypes may identify those who could benefit from immunotherapy. Methods: We used single-cell and bulk RNA sequencing data from GEO and TCGA databases. We characterized the tumor microenvironment at the single-cell level, analyzing cell interactions and identifying fibroblasts linked to mitophagy. Target genes were narrowed down at the bulk transcriptome level to construct a PARD prognosis prediction nomogram. Unsupervised consensus clustering classified PARD into subtypes, analyzing differences in clinical features, immune infiltration, and immunotherapy. Furthermore, the cellular functions of the genes of interest were verified in vitro. Results: We identified ten cell types and 160 mitophagy-related single-cell differentially expressed genes (MR-scDEGs). Strong interactions were observed between fibroblasts, endothelial cells, CD8+ T cells, and NK cells. Fibroblasts linked to mitophagy were divided into six subtypes. Intersection of DEGs from three bulk datasets with MR-scDEGs identified 26 key genes clustered into two subgroups. COX regression analysis identified seven prognostic key genes, enabling a prognostic nomogram model. High and low-risk groups showed significant differences in clinical features, immune infiltration, immunotherapy, and drug sensitivity. In prostate cancer cell lines, CAV1, PALLD, and ITGB8 are upregulated, while CLDN7 is downregulated. Knockdown of PALLD significantly inhibits the proliferation and colony-forming ability of PC3 and DU145 cells, suggesting the important roles of this gene in prostate cancer progression. Conclusions: This study analyzed mitophagy-related genes in PARD, predicting prognosis and aiding in subtype identification and immunotherapy response analysis. This approach offers new strategies for treating prostate cancer with specific molecular subtypes and helps develop potential biomarkers for personalized medicine strategies. Full article
(This article belongs to the Section Cancer Biology and Oncology)
16 pages, 3472 KiB  
Article
The Development and Validation of a Nomogram for Predicting Sepsis Risk in Diabetic Patients with Urinary Tract Infection
by Hua-Qiao Tan, Xiang-Jie Duan, Wan Qu, Mi Shu, Guang-Yao Zhong, Li-Hong Liang, Dong-Mei Bin and Yu-Ming Chen
Medicina 2025, 61(2), 225; https://doi.org/10.3390/medicina61020225 - 27 Jan 2025
Abstract
Background and Objectives: Urinary tract infection (UTI) is a common comorbidity in diabetic patients, making up one of the causes of sepsis. This study aims to develop a nomogram to predict the risk probability of sepsis in diabetic patients with UTI (DPUTIs). [...] Read more.
Background and Objectives: Urinary tract infection (UTI) is a common comorbidity in diabetic patients, making up one of the causes of sepsis. This study aims to develop a nomogram to predict the risk probability of sepsis in diabetic patients with UTI (DPUTIs). Materials and Methods: This is a retrospective observational study. Clinical data for DPUTIs were extracted from the Medical Information Mart for Intensive Care IV database. Eligible DPUTIs were randomly divided into training and validation cohorts in a 7:3 ratio. Independent prognostic factors for sepsis risk were determined using least absolute shrinkage and selection operator (LASSO) regression and multivariate logistic regression. A corresponding nomogram based on these factors was constructed to predict sepsis occurrence in DPUTIs. The discrimination of the nomogram was assessed by multiple indicators, including the area under the receiver operating characteristic curve (AUC), net reclassification improvement index (NRI), and integrated discrimination improvement (IDI). In addition, a calibration curve and decision curve analysis (DCA) were used to evaluate the performance of the nomogram. Results: A total of 1990 DPUTIs were included. Nine independent prognostic factors were identified as predictive factors for sepsis risk in DPUTIs. The prognostic factors included urine red blood cell classification (urine RBC cat), urine white blood cell classification (urine WBC cat), blood glucose, age, temperature, white blood cells (WBCs), sequential organ failure assessment (SOFA) score, lymphocytes, and hematocrit. The AUC, NRI, and IDI of the nomogram indicated robust discrimination. The calibration curve and Hosmer–Lemeshow test showed good calibration of the nomogram. The DCA curve demonstrated a better clinical utility of the nomogram. Conclusions: The nomogram established in this study helps clinicians predict the probability of sepsis in DPUTIs, providing evidence for optimizing the management of related risk factors. Full article
Show Figures

Figure 1

Figure 1
<p>Flowchart of screening.</p>
Full article ">Figure 2
<p>(<b>A</b>) illustrates the relationship between the variable coefficients and log(lambda) values. Each line corresponds to a different variable. As log(lambda) increases, the coefficients of the variables trend towards zero. (<b>B</b>) shows the relationship between Binomial Deviance and log(lambda). We plotted vertical lines at the optimal values using λ.min (left dashed line) and λ.1se (right dashed line, 1−SE standard). In this study, we selected the λ value according to the 1−SE criterion.</p>
Full article ">Figure 3
<p>A nomogram based on lymphocytes, blood glucose, temperature, age, urine RBC cat, urine WBC cat, hematocrit, WBCs, and SOFA. * represents 0.01 &lt; <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> 0.001–0.01, and *** represents <span class="html-italic">p</span> &lt; 0.001. An example of the application of the nomogram is shown above. The corresponding score of each variable is represented by a red dot. When the total score is 266 points, the probability of a DPUTI developing sepsis is 0.655.</p>
Full article ">Figure 4
<p>Performance of the predictive models for sepsis risk. (<b>A</b>,<b>B</b>) ROC curves of the nomogram, SOFA score, and APACHE Ⅱ score for predicting the likelihood of sepsis in DPUTIs. (<b>A</b>) is the training cohort; (<b>B</b>) is the test cohort. The dashed lines in (<b>A</b>,<b>B</b>) represent the baseline.</p>
Full article ">Figure 5
<p>The calibration curves for the training cohort (<b>A</b>) and the validation cohort (<b>B</b>) indicated that the nomogram predictions were consistent with the actual observed outcomes. The Hosmer–Lemeshow test results suggested no statistical significance (both <span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 6
<p>Decision curve analysis (DCA). DCA of the nomogram, SOFA score, and APACHE II score predicts the sepsis risk in DPUTIs. The pink dotted line represents the “treat−none” strategy, while the blue dashed line represents the “treat−all” strategy. (<b>A</b>) The training cohort; (<b>B</b>) the validation cohort.</p>
Full article ">Figure A1
<p>Missing percent of each variable.</p>
Full article ">
19 pages, 8598 KiB  
Article
Identification of Hepatocellular Carcinoma Subtypes Based on Global Gene Expression Profiling to Predict the Prognosis and Potential Therapeutic Drugs
by Cunzhen Zhang, Jiyao Wang, Lin Jia, Qiang Wen, Na Gao and Hailing Qiao
Biomedicines 2025, 13(1), 236; https://doi.org/10.3390/biomedicines13010236 - 20 Jan 2025
Viewed by 584
Abstract
Background: Hepatocellular carcinoma (HCC) is a highly heterogeneous tumor, and distinguishing its subtypes holds significant value for diagnosis, treatment, and the prognosis. Methods: Unsupervised clustering analysis was conducted to classify HCC subtypes. Subtype signature genes were identified using LASSO, SVM, and logistic regression. [...] Read more.
Background: Hepatocellular carcinoma (HCC) is a highly heterogeneous tumor, and distinguishing its subtypes holds significant value for diagnosis, treatment, and the prognosis. Methods: Unsupervised clustering analysis was conducted to classify HCC subtypes. Subtype signature genes were identified using LASSO, SVM, and logistic regression. Survival-related genes were identified using Cox regression, and their expression and function were validated via qPCR and gene interference. GO, KEGG, GSVA, and GSEA were used to determine enriched signaling pathways. ESTIMATE and CIBERSORT were used to calculate the stromal score, tumor purity, and immune cell infiltration. TIDE was employed to predict the patient response to immunotherapy. Finally, drug sensitivity was analyzed using the oncoPredict algorithm. Results: Two HCC subtypes with different gene expression profiles were identified, where subtype S1 exhibited a significantly shorter survival time. A subtype scoring formula and a nomogram were constructed, both of which showed an excellent predictive performance. COL11A1 and ACTL8 were identified as survival-related genes among the signature genes, and the downregulation of COL11A1 could suppress the invasion and migration of HepG2 cells. Subtype S1 was characterized by the upregulation of pathways related to collagen and the extracellular matrix, as well as downregulation associated with the xenobiotic metabolic process and fatty acid degradation. Subtype S1 showed higher stromal scores, immune scores, and ESTIMATE scores and infiltration of macrophages M0 and plasma cells, as well as lower tumor purity and infiltration of NK cells (resting/activated) and resting mast cells. Subtype S2 was more likely to benefit from immunotherapy. Subtype S1 appeared to be more sensitive to BMS-754807, JQ1, and Axitinib, while subtype S2 was more sensitive to SB505124, Pevonedistat, and Tamoxifen. Conclusions: HCC patients can be classified into two subtypes based on their gene expression profiles, which exhibit distinctions in terms of signaling pathways, the immune microenvironment, and drug sensitivity. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Figure 1

Figure 1
<p>Flow chart of this study.</p>
Full article ">Figure 2
<p>Identification of HCC subtypes. (<b>A</b>) Heatmap of the consensus matrix for two clusters in TCGA-LIHC (k = 2). (<b>B</b>) Heatmap of the consensus matrix for two clusters in GSE14520 (k = 2). (<b>C</b>) Principal component analysis (PCA) of two subtypes. (<b>D</b>) Clustering heatmap of S1 and S2 subtypes. (<b>E</b>) Differentially expressed genes between S1 and S2 subtypes. (<b>F</b>,<b>G</b>) K-M survival analysis of S1 and S2 subtypes based on OS (log-rank test) in the TCGA training cohort (<b>F</b>) and GSE14520 validation cohort (<b>G</b>). <span class="html-italic">p</span>-values were determined using Student’s <span class="html-italic">t</span>-test and the Wilcoxon test.</p>
Full article ">Figure 3
<p>Identification of characteristic genes. (<b>A</b>,<b>B</b>) Screening of characteristic genes in S1 and S2 subtypes using LASSO regression (<b>A</b>) and SVM (<b>B</b>). (<b>C</b>) Cross-validation of LASSO regression and SVM. (<b>D</b>) Univariate and multivariate logistic regression in S1 and S2 subtypes. (<b>E</b>) Protein–protein interaction (PPI) analysis of characteristic genes.</p>
Full article ">Figure 4
<p>Construction and evaluation of the subtype prediction formula. (<b>A</b>) Distribution of predictor in S1 and S2 subtypes. (<b>B</b>) ROC analysis of predictor. (<b>C</b>) Nomogram for the identification of S1 and S2 subtypes. (<b>D</b>) K-M survival analysis between low and high predictor groups based on OS (log-rank test). (<b>E</b>) Univariate and multivariate Cox regressions on the characteristic genes. (<b>F</b>) Expression of COL11A1 and ACTL8 in the TCGA cohort. (<b>G</b>) Expression of COL11A1 and ACTL8 between normal and HCC patients (normal n = 5, HCC n = 6).</p>
Full article ">Figure 5
<p>Downregulated Col11a1 inhibited the invasion and migration of HepG2 cells. (<b>A</b>) Col11a1 mRNA expression was significantly downregulated in HepG2 cells by col11a1 siRNA (<span class="html-italic">t</span>-test). (<b>B</b>) Invasion of HepG2 cells detected using the trans-well assay (<span class="html-italic">t</span>-test). (<b>C</b>) Migration of HepG2 detected using the scratch assay (<span class="html-italic">t</span>-test).</p>
Full article ">Figure 6
<p>Enrichment analysis of signaling pathways in S1 and S2 subtypes. (<b>A</b>) Gene Ontology (GO) analysis of DEGs between two subtypes. (<b>B</b>) Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of DEGs between two subtypes.</p>
Full article ">Figure 7
<p>Enrichment analysis of signaling pathways in S1 and S2 subtypes. (<b>A</b>) Gene set variation analysis (GSVA)-GO between two subtypes. (<b>B</b>) GSVA-KEGG between two subtypes. (<b>C</b>) Gene set enrichment analysis (GSEA) between two subtypes.</p>
Full article ">Figure 8
<p>Immune microenvironment characteristics in S1 and S2 subtypes. (<b>A</b>,<b>B</b>) Stromal score, immune score, ESTIMATE score, and tumor purity (<b>A</b>) and their correlation with the subtype predictor (<b>B</b>). (<b>C</b>) Immune cell infiltration characteristics of two subgroups. (<b>D</b>) TIDE scores of S1 and S2 subtypes. <span class="html-italic">p</span>-values were determined via Student’s <span class="html-italic">t</span>-test and the Wilcoxon test; correlation analysis was performed via Spearman analysis.</p>
Full article ">
15 pages, 1077 KiB  
Article
A Novel Nomogram for Estimating a High-Risk Result in the EndoPredict® Test for Estrogen Receptor-Positive/Human Epidermal Growth Factor Receptor 2 (HER2)-Negative Breast Carcinoma
by Víctor Macarrón, Itsaso Losantos-García, Alberto Peláez-García, Laura Yébenes, Alberto Berjón, Laura Frías, Covadonga Martí, Pilar Zamora, José Ignacio Sánchez-Méndez and David Hardisson
Cancers 2025, 17(2), 273; https://doi.org/10.3390/cancers17020273 - 16 Jan 2025
Viewed by 429
Abstract
Background/Objectives: The EndoPredict® assay has been widely used in recent years to estimate the risk of distant recurrence and the absolute chemotherapy benefit for patients with estrogen (ER)-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer. However, there are no well-defined [...] Read more.
Background/Objectives: The EndoPredict® assay has been widely used in recent years to estimate the risk of distant recurrence and the absolute chemotherapy benefit for patients with estrogen (ER)-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer. However, there are no well-defined criteria for selecting patients who may benefit from the test. The aim of this study was to develop a novel nomogram to estimate the probability of obtaining a high-risk EndoPredict® result in clinical practice. Methods: The study cohort comprised 348 cases of T1-3/N0-1a/M0 ER-positive/HER2-negative breast carcinoma. A multivariate analysis was conducted using a training cohort (n = 270) based on clinicopathological features that demonstrated a statistically significant correlation with the EndoPredict® result in a univariate analysis. The predictive model was subsequently represented as a nomogram to estimate the probability of obtaining a high-risk result in the EndoPredict® assay. The predictive model was then validated using a separate validation cohort (n = 78). Results: The clinicopathological features incorporated into the nomogram included tumor size, tumor grade, sentinel lymph node status, pN stage, and Ki67. The internal validation of the model yielded an area under the curve (AUC) of 0.803 (95% CI = 0.751, 0.855) in the receiver operating characteristic (ROC) curve for the training cohort, with an optimal sensitivity and specificity at a threshold of 0.536. The external validation yielded an AUC of 0.789 (95% CI = 0.689, 0.890) in its ROC curve, with optimal sensitivity and specificity achieved at a threshold of 0.393. Conclusions: This study presents, for the first time, the development of a clinically accessible nomogram designed to estimate the probability of obtaining a high-risk result in the EndoPredict® assay. The use of easily available clinicopathological features allows for the optimization of patient selection for the EndoPredict® assay, ensuring that those who would most benefit from undergoing the test are identified. Full article
(This article belongs to the Special Issue Insights from the Editorial Board Member)
Show Figures

Figure 1

Figure 1
<p>Nomogram to predict for high-risk EndoPredict<sup>®</sup> score. Sentinel lymph node status, tumor grade, pN stage, Ki67 levels, and tumor size were finally selected to develop the model.</p>
Full article ">Figure 2
<p>Receiver operating characteristic (ROC) curve of the nomogram for the (<b>a</b>) training cohort and (<b>b</b>) validation cohort.</p>
Full article ">Figure 2 Cont.
<p>Receiver operating characteristic (ROC) curve of the nomogram for the (<b>a</b>) training cohort and (<b>b</b>) validation cohort.</p>
Full article ">
12 pages, 1447 KiB  
Article
Development of Novel Nomograms to Predict 5- and 7-Year Biochemical-Recurrence-Free Survival in High-Risk Prostate Cancer Patients After Carbon-Ion Radiotherapy and Androgen Deprivation Therapy
by Takanobu Utsumi, Hiroyoshi Suzuki, Masaru Wakatsuki, Kana Kobayashi, Atsushi Okato, Mio Nakajima, Shuri Aoki, Taisuke Sumiya, Tomohiko Ichikawa, Koichiro Akakura, Hiroshi Tsuji, Shigeru Yamada and Hitoshi Ishikawa
Appl. Sci. 2025, 15(2), 804; https://doi.org/10.3390/app15020804 - 15 Jan 2025
Viewed by 424
Abstract
Background: The aim of this study was to develop nomograms predicting 5- and 7-year biochemical-recurrence (BCR)-free survival in high-risk prostate cancer (PCa) patients treated with carbon-ion radiotherapy (CIRT) and androgen deprivation therapy (ADT). Methods: We retrospectively evaluated 785 high-risk PCa patients treated with [...] Read more.
Background: The aim of this study was to develop nomograms predicting 5- and 7-year biochemical-recurrence (BCR)-free survival in high-risk prostate cancer (PCa) patients treated with carbon-ion radiotherapy (CIRT) and androgen deprivation therapy (ADT). Methods: We retrospectively evaluated 785 high-risk PCa patients treated with CIRT and ADT. Based on the least absolute shrinkage and selection operator model, two nomograms predicting 5- and 7-year BCR-free survival were developed and internally validated. The ability of each nomogram to predict BCR-free survival was determined by calculating the area under the survival curve (AUC). Results: The 5- and 7-year BCR-free survival rates were 92.1% and 89.3%, respectively. Age, prostate-specific antigen level, clinical T stage, and Gleason score were incorporated into the nomogram predicting 5-year BCR-free survival. In addition to these variables, the percentage of positive biopsy cores was also added to the nomogram predicting 7-year BCR-free survival. The AUC value of each nomogram showed suboptimal-to-good discrimination. Conclusions: We developed the first nomograms accurately predicting BCR-free survival in high-risk PCa patients treated with CIRT and ADT. These nomograms will enable adequate understanding and explanation of BCR-free survival to patients when clinicians use them. Full article
(This article belongs to the Special Issue Nuclear Medicine and Radiotherapy in Cancer Treatment)
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier curves for BCR-free survival according to each clinical variable. BCR: biochemical recurrence, PSA: prostate-specific antigen, %PC: percentage of biopsy positive cores.</p>
Full article ">Figure 2
<p>Nomograms predicting BCR-free survival after CIRT with ADT. Directions: A line was drawn upwards to the number of points in each category. The points were summed, and then a line of total points was drawn downwards to determine the probability on the bottom line. ADT: androgen deprivation therapy, BCR: biochemical recurrence, CIRT: carbon-ion radiotherapy, PSA: prostate-specific antigen.</p>
Full article ">Figure 3
<p>Kaplan–Meier survival curves stratified by risk group, defined by calculated cut-off values. BCR: biochemical recurrence.</p>
Full article ">
10 pages, 2138 KiB  
Article
A Nomogram Built on Clinical Factors and CT Attenuation Scores for Predicting Treatment Response of Acute Myeloid Leukemia Patients
by Linna Liu, Wenzheng Lu, Li Xiong, Han Qi, Robert Peter Gale and Bin Yin
Biomedicines 2025, 13(1), 198; https://doi.org/10.3390/biomedicines13010198 - 15 Jan 2025
Viewed by 412
Abstract
Background: Acute myeloid leukemia (AML) is an aggressive cancer with variable treatment responses. While clinical factors such as age and genetic mutations contribute to prognosis, recent studies suggest that CT attenuation scores may also predict treatment outcomes. This study aims to develop a [...] Read more.
Background: Acute myeloid leukemia (AML) is an aggressive cancer with variable treatment responses. While clinical factors such as age and genetic mutations contribute to prognosis, recent studies suggest that CT attenuation scores may also predict treatment outcomes. This study aims to develop a nomogram combining clinical and CT-based factors to predict treatment response and guide personalized therapy for AML patients. Methods: This retrospective study included 74 newly diagnosed AML patients who underwent unenhanced abdominal CT scans within one week before receiving their first induction chemotherapy. Clinical biomarkers of tumor burden were also collected. Patients were classified into two groups based on treatment response: complete remission (CR; n = 24) and non-complete remission (NCR; n = 50). Multivariable logistic regression was used to identify independent predictors of treatment response. Predictive performance was evaluated using receiver operating characteristic (ROC) curves, and model consistency was assessed through calibration and decision curve analysis (DCA). Results: Significant differences in hemoglobin (Hb), platelets (Plt), and CT attenuation scores were observed between the CR and NCR groups (all p < 0.05). Multivariable logistic regression identified Hb, Plt, and CT attenuation scores as independent predictors of treatment response. A nomogram incorporating these factors demonstrated excellent predictive performance, with an area under the curve (AUC) of 0.912 (95% CI: 0.842–0.983), accuracy of 0.865 (95% CI: 0.765–0.933), sensitivity of 0.880 (95% CI: 0.790–0.970), and specificity of 0.833 (95% CI: 0.684–0.982). The CR nomogram displayed significant clinical value and excellent goodness of fit. Conclusions: The nomogram, which incorporates Hb, Plt, and CT attenuation scores, provides valuable insights into predicting treatment response in AML patients. This model offers strong discriminatory ability and could enhance personalized treatment planning and prognosis prediction for AML. Full article
(This article belongs to the Special Issue Pathogenesis, Diagnosis and Treatment of Hematologic Malignancies)
Show Figures

Figure 1

Figure 1
<p>Flow chart of inclusion and exclusion of patients.</p>
Full article ">Figure 2
<p>Pretreatment CT images of representative AML patients who achieved complete remission (CR) and those who did not achieve complete remission (NCR). (<b>A</b>): Axial unenhanced CT with an abdominal window, and (<b>B</b>): a bone window showed an increased bone marrow density of both iliac bones (CT: 212.03 HU). (<b>C</b>,<b>D</b>): A representative AML patient who achieved NCR after the therapy. (<b>C</b>): Axial unenhanced CT with an abdominal window, and (<b>D</b>): a bone window showed a lower bone marrow density of both iliac bones (CT: 116.08 HU). AML, acute myeloid leukemia; CR, complete remission; NCR, non-CR.</p>
Full article ">Figure 3
<p>The nomogram that was employed to forecast the probability of the complete remission for patients with AML. The points by which two factors (Hb, Plt, and CT), respectively, make a vertical line to the topmost line are added to obtain total points; then, the total points correspond to the probability of treatment response of the AML patient with treatment response of the bottom line. AML, acute myeloid leukemia; Hb, hemoglobin; Plt, platelets; CT: CT attenuation scores.</p>
Full article ">Figure 4
<p>ROC curves of the models in predicting the complete remission. The nomogram was composed of clinical test indicators (Hb and Plt) and CT attenuation scores. Clinical was composed of Hb and Plt in clinical laboratory indicators. The CT model was composed of the measured CT attenuation scores.</p>
Full article ">Figure 5
<p>The calibration curve of the nomogram. The Hosmer–Lemeshow goodness-of-fit test is a method used to assess the goodness of fit of binary logistic regression models. <span class="html-italic">p</span> = 0.885 represented good fitting efficacy.</p>
Full article ">Figure 6
<p>Decision curve analyses for the simple-to-use model predicting complete remission in the nomogram.</p>
Full article ">
18 pages, 1214 KiB  
Review
PK/PD-Guided Strategies for Appropriate Antibiotic Use in the Era of Antimicrobial Resistance
by Tetsushu Onita, Noriyuki Ishihara and Takahisa Yano
Antibiotics 2025, 14(1), 92; https://doi.org/10.3390/antibiotics14010092 - 14 Jan 2025
Viewed by 580
Abstract
Antimicrobial resistance (AMR) poses a critical global health threat, necessitating the optimal use of existing antibiotics. Pharmacokinetic/pharmacodynamic (PK/PD) principles provide a scientific framework for optimizing antimicrobial therapy, particularly to respond to evolving resistance patterns. This review examines PK/PD strategies for antimicrobial dosing optimization, [...] Read more.
Antimicrobial resistance (AMR) poses a critical global health threat, necessitating the optimal use of existing antibiotics. Pharmacokinetic/pharmacodynamic (PK/PD) principles provide a scientific framework for optimizing antimicrobial therapy, particularly to respond to evolving resistance patterns. This review examines PK/PD strategies for antimicrobial dosing optimization, focusing on three key aspects. First, we discuss the importance of drug concentration management for enhancing efficacy while preventing toxicity, considering various patient populations, including pediatric and elderly patients with their unique physiological characteristics. Second, we analyze different PK modeling approaches: the classic top-down approach exemplified by population PK analysis, the bottom-up approach represented by physiologically based PK modeling, and hybrid models combining both approaches for enhanced predictive performance. Third, we explore clinical applications, including nomogram-based dosing strategies, Bayesian estimation, and emerging artificial intelligence applications, for real-time dose optimization. Critical challenges in implementing PK/PD simulation are addressed, particularly the selection of appropriate PK models, the optimization of PK/PD indices, and considerations concerning antimicrobial concentrations at infection sites. Understanding these principles and challenges is crucial for optimizing antimicrobial therapy and combating AMR through improved dosing strategies. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of key pharmacokinetic and pharmacodynamic (PK/PD) parameters in relation to antimicrobial resistance prevention: (<b>A</b>) bolus infusion, (<b>B</b>) intermittent infusion, and (<b>C</b>) extended infusion.</p>
Full article ">Figure 1 Cont.
<p>Schematic representation of key pharmacokinetic and pharmacodynamic (PK/PD) parameters in relation to antimicrobial resistance prevention: (<b>A</b>) bolus infusion, (<b>B</b>) intermittent infusion, and (<b>C</b>) extended infusion.</p>
Full article ">
23 pages, 24936 KiB  
Article
TLR7: A Key Prognostic Biomarker and Immunotherapeutic Target in Lung Adenocarcinoma
by Feiming Hu, Chenchen Hu, Yuanli He, Yuanjie Sun, Chenying Han, Xiyang Zhang, Lingying Yu, Daimei Shi, Yubo Sun, Junqi Zhang, Dongbo Jiang, Shuya Yang and Kun Yang
Biomedicines 2025, 13(1), 151; https://doi.org/10.3390/biomedicines13010151 - 9 Jan 2025
Viewed by 442
Abstract
Background: The tumor microenvironment (TME) plays a crucial role in the progression of lung adenocarcinoma (LUAD). However, understanding its dynamic immune and stromal modulation remains a complex challenge. Methods: We utilized the ESTIMATE algorithm to evaluate the immune and stromal components of the [...] Read more.
Background: The tumor microenvironment (TME) plays a crucial role in the progression of lung adenocarcinoma (LUAD). However, understanding its dynamic immune and stromal modulation remains a complex challenge. Methods: We utilized the ESTIMATE algorithm to evaluate the immune and stromal components of the LUAD TME from the TCGA database. Correlations between these components and clinical characteristics and patient prognosis were analyzed. Toll-like receptor 7 (TLR7) was identified as a key prognostic biomarker through PPI network and COX regression analysis. Validation of TLR7 expression was conducted using GEO data, qPCR, WB, and IHC. A prognostic model was developed using a nomogram, incorporating TLR7 expression. Enrichment analysis, the Tumor Immune Estimation Resource database, and single-sample gene set enrichment analysis were used to explore TLR7’s potential function. The response of the TLR7 subgroup to immunotherapy and drug sensitivity was observed. Results: We found significant associations between the immune and stromal components of LUAD TME and clinical features and prognosis. Specifically, TLR7 was identified as a prognostic biomarker, where lower expression in tumor tissues was linked to worse outcomes. This finding was further confirmed by comparing TLR7 expression in LUAD cells to normal bronchial epithelial cells, revealing lower expression in the tumor cells. Incorporating TLR7 into a nomogram prognostic model resulted in a good predictor of patient survival. Additionally, TLR7 was associated with immune function and positively correlated with various immune cells. Importantly, patients with high TLR7 expression were more likely to benefit from anti-PD-1 checkpoint blockade therapy. We also identified four treatment candidates for patients with high TLR7 expression. Conclusion: TLR7 is a powerful clinical feature that predicts patient prognosis, immunotherapeutic response, and drug candidates, providing additional insights for the treatment of LUAD. Full article
(This article belongs to the Section Immunology and Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>Flow chart of the framework for this study. Step 1: An in-depth analysis of lung adenocarcinoma samples from the TCGA database using the ESTIMATE algorithm revealed significant correlations between scores on immune and stromal components of the tumor microenvironment and clinical features and patient prognosis, and identified key genes associated with these scores. Step 2: TLR7, identified by PPI and Cox analysis, is a key biomarker for LUAD prognosis, and lower tumor expression is associated with poorer prognosis, as validated by GEO. qPCR, WB, and IHC confirmed that TLR7 expression is lower in LUAD compared to normal cells. A nomogram-based prognostic model incorporating TLR7 can effectively predict patient survival. Step 3: By enrichment analysis, TLR7 was found to be closely associated with immune function. Further immune infiltration analysis showed that TLR7 was positively correlated with the infiltration levels of multiple immune cells. These findings may explain the ability of the tumor microenvironment (TME) to maintain an immunodominant state. Step 4: The response of TLR7 subgroups to immunotherapy was analyzed and patients with high TLR7 were found to be more likely to benefit from anti-PD-1 checkpoint blockade therapy. Four therapeutic candidates for patients with high TLR7 were identified.</p>
Full article ">Figure 1 Cont.
<p>Flow chart of the framework for this study. Step 1: An in-depth analysis of lung adenocarcinoma samples from the TCGA database using the ESTIMATE algorithm revealed significant correlations between scores on immune and stromal components of the tumor microenvironment and clinical features and patient prognosis, and identified key genes associated with these scores. Step 2: TLR7, identified by PPI and Cox analysis, is a key biomarker for LUAD prognosis, and lower tumor expression is associated with poorer prognosis, as validated by GEO. qPCR, WB, and IHC confirmed that TLR7 expression is lower in LUAD compared to normal cells. A nomogram-based prognostic model incorporating TLR7 can effectively predict patient survival. Step 3: By enrichment analysis, TLR7 was found to be closely associated with immune function. Further immune infiltration analysis showed that TLR7 was positively correlated with the infiltration levels of multiple immune cells. These findings may explain the ability of the tumor microenvironment (TME) to maintain an immunodominant state. Step 4: The response of TLR7 subgroups to immunotherapy was analyzed and patients with high TLR7 were found to be more likely to benefit from anti-PD-1 checkpoint blockade therapy. Four therapeutic candidates for patients with high TLR7 were identified.</p>
Full article ">Figure 2
<p>Screening for immune-related differential genes. (<b>A</b>,<b>B</b>) Graphical representation of differentially expressed genes (DEGs) as a volcano plot, contrasting the high and low groups in terms of ImmuneScore and StromalScore. (|log2FoldChange| &gt;1 and adjusted <span class="html-italic">p</span>-value &lt; 0.05). (<b>C</b>,<b>D</b>) Graphical representation of DEGs as a heat plot, contrasting the high and low groups in terms of ImmuneScore and StromalScore. (<b>E</b>,<b>F</b>) Venn diagrams illustrating the overlap of DEGs that are either up- or downregulated in both ImmuneScore and StromalScore. (<b>G</b>,<b>H</b>) TCGA-LUAD cohort scale independence and average connectivity. (<b>I</b>) The cluster dendrogram clusters similar gene expressions into modules, where each color represents a gene. (<b>J</b>) Pearson correlation analysis of the merged modules with ImmuneScore, StromalScore, ESTIMATEScore, and TumorPurity. (<b>K</b>) The Venn diagram illustrates the DEGs common to upregulation and WGCNA in ImmuneScore and StromalScore. (<b>L</b>) Circular plots depicting the GO enrichment analysis for 117 DEGs, highlighting terms significantly enriched at a <span class="html-italic">p</span>-adjusted threshold of less than 0.05, focusing on the top 20 GO-BP terms. (<b>M</b>) Circular plot for KEGG enrichment analysis of 117 DEGs, featuring terms significantly enriched at a <span class="html-italic">p</span>-adjusted threshold of less than 0.05, with an emphasis on the top 20 KEGG terms. (<b>N</b>) UpsetR and Venn plots of previously filtered immune and stromal shared upregulated DEGs and related genes screened by WGCNA and IRGs.</p>
Full article ">Figure 3
<p>PPI and univariate Cox analysis of DEGs, and association of TLR7 expression with clinical characteristics and survival. (<b>A</b>) A network with nodes above 0.7 was constructed. (<b>B</b>) The 20 most significant genes. (<b>C</b>) A univariate Cox regression on 38 DEGs identified 12 genes with <span class="html-italic">p</span> &lt; 0.05. (<b>D</b>) The Venn diagram shows the factors common to the 20 top PPI nodes and the univariate Cox model. (<b>E</b>) TCGA data were analyzed in all normal and tumor samples for TLR7 expression. (<b>F</b>) TCGA data were analyzed in paired normal and tumor samples from the same patient for TLR7 expression. (<b>G</b>) Association of TLR7 expression with stage classification in early and mid-late clinical stages. (<b>H</b>) Association of TLR7 expression with early and mid-late clinical T stage. (<b>I</b>–<b>K</b>) TLR7 expression at mRNA and protein levels in bronchial epithelial cells and lung adenocarcinoma cell lines. (<b>L</b>,<b>M</b>) TLR7 expression in paraneoplastic and tumor tissues. (<b>N</b>) Survival analysis of LUAD patients with different TLR7 expression, marking patients as high or low expression based on comparison with the median expression level. <span class="html-italic">p</span> = 0.002 by log-rank test. ***: <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 4
<p>Construction and calibration of a nomogram. (<b>A</b>) A prognostic nomogram was developed by integrating TLR7 with multiple clinical variables to predict survival in LUAD patients. (<b>B</b>) A calibration plot assessed the accuracy of the prognostic model for estimating survival probabilities. (<b>C</b>) The area under the ROC curve, AUC, was used to predict the overall survival of LUAD patients at 1, 3, and 5 years. (<b>D</b>) Patients were categorized into high-risk and low-risk groups based on the median RiskScore expression level, and the log-rank test was used for survival analysis, <span class="html-italic">p</span> &lt; 0.001. (<b>E</b>) TIDE scores in different risk subgroups, by <span class="html-italic">t</span>-test, <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, -: not significant.</p>
Full article ">Figure 4 Cont.
<p>Construction and calibration of a nomogram. (<b>A</b>) A prognostic nomogram was developed by integrating TLR7 with multiple clinical variables to predict survival in LUAD patients. (<b>B</b>) A calibration plot assessed the accuracy of the prognostic model for estimating survival probabilities. (<b>C</b>) The area under the ROC curve, AUC, was used to predict the overall survival of LUAD patients at 1, 3, and 5 years. (<b>D</b>) Patients were categorized into high-risk and low-risk groups based on the median RiskScore expression level, and the log-rank test was used for survival analysis, <span class="html-italic">p</span> &lt; 0.001. (<b>E</b>) TIDE scores in different risk subgroups, by <span class="html-italic">t</span>-test, <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, -: not significant.</p>
Full article ">Figure 5
<p>GO, KEGG, and GSEA of high- and low-expressing TLR7 groups in LUAD. (<b>A</b>) Volcano plots of DEGs generated by comparing TLR7 groups in LUAD. (|log2FoldChange| &gt; 1 and Adjust <span class="html-italic">p</span>-value &lt; 0.05). (<b>B</b>) Heatmap of TLR7 high- and low-expression differential genes. The row names of the heatmaps are gene names and the column names are the IDs of samples not shown in the graphs. (<b>C</b>,<b>D</b>) Differential genes were analyzed for GO and KEGG enrichment, and terms with <span class="html-italic">p</span>-adjust &lt; 0.05 were considered significantly enriched. (<b>E</b>) The high-expressing TLR7 samples were enriched in the gene set of the HALLMARK collection and only the gene sets with NOM <span class="html-italic">p</span> &lt; 0.05 and FDR q &lt; 0.05 were regarded as significant. (<b>F</b>) Low TLR7 samples showed enrichment in HALLMARK sets.</p>
Full article ">Figure 6
<p>TLR7 expression correlates with infiltrating immune cells in LUAD. (<b>A</b>) TLR7 gene copy number variations impact immune cell infiltration. (<b>B</b>) TLR7 is linked to lung adenocarcinoma tumor purity and immune cell expression. (<b>C</b>) Survival analysis showed that the overall survival of lung adenocarcinoma patients was positively correlated with the expression of B cell, DC, and TLR7. (<b>D</b>) Multi-group stacked histogram showing the proportion of 25 tumor-infiltrating immune cells in LUAD tumor samples. (<b>E</b>) Bar graph showing the distribution of 28 immune cells between LUAD tumor samples with high or low TLR7 expression. (<b>F</b>) Heatmap of the correlation between 28 tumor-infiltrating lymphocytes and TLR7, with the chromaticity of each small colored box representing the corresponding correlation value between the two cells. ***: <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, ns: not significant.</p>
Full article ">Figure 6 Cont.
<p>TLR7 expression correlates with infiltrating immune cells in LUAD. (<b>A</b>) TLR7 gene copy number variations impact immune cell infiltration. (<b>B</b>) TLR7 is linked to lung adenocarcinoma tumor purity and immune cell expression. (<b>C</b>) Survival analysis showed that the overall survival of lung adenocarcinoma patients was positively correlated with the expression of B cell, DC, and TLR7. (<b>D</b>) Multi-group stacked histogram showing the proportion of 25 tumor-infiltrating immune cells in LUAD tumor samples. (<b>E</b>) Bar graph showing the distribution of 28 immune cells between LUAD tumor samples with high or low TLR7 expression. (<b>F</b>) Heatmap of the correlation between 28 tumor-infiltrating lymphocytes and TLR7, with the chromaticity of each small colored box representing the corresponding correlation value between the two cells. ***: <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, ns: not significant.</p>
Full article ">Figure 7
<p>Drug sensitivity and immunotherapy analysis for high and low TLR7 expression. (<b>A</b>,<b>B</b>) Drug sensitivity analysis of LUAD patients in TCGA and GSE75037. (<b>C</b>) Venn analysis of drugs common to (<b>A</b>,<b>B)</b>. (<b>D</b>) IPS analysis of LUAD patients in TCGA. (***: <span class="html-italic">p</span> &lt; 0.001, *: <span class="html-italic">p</span> &lt; 0.05, ns: not significant).</p>
Full article ">
13 pages, 2303 KiB  
Article
Early Identification of the Non-Transplanted Functional High-Risk Multiple Myeloma: Insights from a Predictive Nomogram
by Yanjuan Li, Lifen Kuang, Beihui Huang, Junru Liu, Meilan Chen, Xiaozhe Li, Jingli Gu, Tongyong Yu and Juan Li
Biomedicines 2025, 13(1), 145; https://doi.org/10.3390/biomedicines13010145 - 9 Jan 2025
Viewed by 343
Abstract
Background: Patients with multiple myeloma (MM) who have a suboptimal response to induction therapy or early relapse are classified as functional high-risk (FHR) patients and have been shown to have a dismal prognosis. The aim of this study was to establish a [...] Read more.
Background: Patients with multiple myeloma (MM) who have a suboptimal response to induction therapy or early relapse are classified as functional high-risk (FHR) patients and have been shown to have a dismal prognosis. The aim of this study was to establish a predictive nomogram for patients with non-transplanted FHR MM. Materials and Methods: The group comprised 215 patients in our center between 1 January 2006 and 1 March 2024. To identify independent risk factors, univariate and multivariate logistic regression analyses were performed, and a nomogram was constructed to predict non-transplant FHR MM. To evaluate the nomogram’s predictive accuracy, we utilized bias-corrected AUC, calibration curves, decision curve analysis (DCA), and clinical impact curves (CIC). Results: Multivariate logistic regression demonstrated that younger age at onset, a higher proportion of LDH (more than 220 U/L), pattern A + C of M protein decline patterns, a lower proportion of patients with induction treatment efficacy than VGPR, and those undergoing maintenance therapies were independent risk factors for patients with non-transplanted FHR MM. The AUC scores for the training and internal validation groups were 0.940 (95% CI 0.893–0.986) and 0.978 (95% CI 0.930–1.000). DCA and CIC curves were utilized to further verify the clinical efficacy of the nomogram. Conclusions: We developed a nomogram that enables early prediction of non-transplant FHR MM patients. Younger age at onset, LDH ≥ 220 U/L, an A + C pattern of M-protein decline, and induction therapy efficacy not reaching VGPR are more likely to be FHR MM patients. Patients who do not undergo maintenance therapy are prone to early progression or relapse. Full article
(This article belongs to the Special Issue Pathogenesis, Diagnosis and Treatment of Hematologic Malignancies)
Show Figures

Figure 1

Figure 1
<p>The flowchart of the study.</p>
Full article ">Figure 2
<p>Survival of the three groups. (<b>a</b>). PFS of the three groups. (<b>b</b>). OS of the three groups. OS of the SR group was not reached. The dashed lines in the figures represent the median survival time for each group.</p>
Full article ">Figure 3
<p>Nomogram for predicting the probability of non-transplant FHR MM. The nomogram can be used by finding each patient’s unique point on each variable axis. To calculate the points that each variable receives, upward-pointing lines and dots are drawn. In order to determine the probability of non-transplant FHR MM, a line is drawn downward from the sum of the points on the Total Points axis.</p>
Full article ">Figure 4
<p>AUC and ROC curves of this nomogram. Training group (<b>a</b>) and internal validation group (<b>b</b>).</p>
Full article ">Figure 5
<p>Calibration curves of this nomogram in (<b>a</b>) the training group and (<b>b</b>) the internal validation group.</p>
Full article ">Figure 6
<p>DCA curves of this nomogram in (<b>a</b>) the training group and (<b>b</b>) the internal validation group.</p>
Full article ">Figure 7
<p>CIC of this nomogram in (<b>a</b>) the training group and (<b>b</b>) the internal validation group.</p>
Full article ">
11 pages, 931 KiB  
Article
Elderly Prostate Cancer Patients Treated with Robotic Surgery Are More Likely to Harbor Adverse Pathology Features and Experience Disease Progression: Analysis of the Prognostic Impact of Adverse Pathology Risk Score Patterns Using Briganti’s 2012 Nomogram and EAU Risk Groups
by Antonio Benito Porcaro, Emanuele Serafin, Francesca Montanaro, Sonia Costantino, Lorenzo De Bon, Alberto Baielli, Francesco Artoni, Luca Roggero, Claudio Brancelli, Michele Boldini, Alberto Bianchi, Alessandro Veccia, Riccardo Rizzetto, Matteo Brunelli, Maria Angela Cerruto, Riccardo Giuseppe Bertolo and Alessandro Antonelli
J. Clin. Med. 2025, 14(1), 193; https://doi.org/10.3390/jcm14010193 - 31 Dec 2024
Viewed by 521
Abstract
Background/Objectives: Prostate cancer (PCa) is prevalent among men over 70. Treatment may involve interventions like radical prostatectomy. The objective of this study was to investigate the combination of adverse pathology patterns on PCa progression through the Briganti 2012 nomogram and EAU risk classes [...] Read more.
Background/Objectives: Prostate cancer (PCa) is prevalent among men over 70. Treatment may involve interventions like radical prostatectomy. The objective of this study was to investigate the combination of adverse pathology patterns on PCa progression through the Briganti 2012 nomogram and EAU risk classes in elderly patients treated with robotic surgery. Methods: A cohort of 1047 patients treated from January 2013 to December 2021 was categorized as being older if aged 70 or above. The adverse pathology risk scores were ranked from zero to three. These scores were then analyzed for correlations with the Briganti 2012 nomogram via EAU risk groups and for PCa progression. Results: Overall, older age was detected in 287 patients who had higher rates of adverse pathology features combined into a pattern risk score of 3. Within each age group, the adverse pathology risk score patterns were positively predicted by the Briganti 2012 nomogram across EAU prognostic groups. After a median (95% CI) follow-up period of 95 months, PCa progression occurred in 237 patients, of whom 68 were elderly and more likely to progress as adverse pathology patterns increased, particularly for a risk score of 3 (p < 0.0001), which was almost three times higher than that in younger patients (p < 0.0001). Conclusions: Managing PCa in elderly patients is challenging due to their increasing life expectancy. The Briganti 2012 nomogram effectively predicts disease progression in this population. Elderly prostate cancer patients have higher severe pathology rates predicted independently by the Briganti 2012 nomogram, with nearly triple the risk of progression compared to that in younger cases, necessitating tailored treatment approaches. Full article
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier survival risk curves of prostate cancer (PCa) progression in 760 patients aged less than 70 years, treated with robotic surgery, and stratified through adverse pathology risk score patterns in the surgical specimen. Accordingly, median survival time of PCa progression decreased from adverse pathology pattern risk score zero (106 months; 95% CI: 94.2–117.7 months), one (82 months, 95% CI: 60.2–103.6 months), two (53 months; 95% CI: 42.4–63.5 months), and three (44 months; 95% CI: 21.0–66.9 months), with the difference being significant (Mantel–Cox log rank test: <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 2
<p>Kaplan–Meier survival risk curves of prostate cancer (PCa) progression in 287 patients aged at least 70 years, treated with robotic surgery, and stratified through adverse pathology risk score patterns in the surgical specimen. Accordingly, median survival time of PCa progression decreased from adverse pathology pattern risk score zero (103 months; 95% CI: 91.5–114.4 months), one (67 months, 95% CI: 65.4–68.5 months), two (61 months; 95% CI: 48.5–73.4 months), and three (34 months; 95% CI: 29.2–38.7 months), with the difference being significant (Mantel–Cox log rank test: <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
11 pages, 497 KiB  
Article
Role of Age, Comorbidity, and Frailty in the Prediction of Postoperative Complications After Surgery for Vulvar Cancer: A Retrospective Cohort Study with the Development of a Nomogram
by Giovanni Delli Carpini, Francesco Sopracordevole, Camilla Cicoli, Marco Bernardi, Lucia Giuliani, Mariasole Fichera, Nicolò Clemente, Anna Del Fabro, Jacopo Di Giuseppe, Luca Giannella, Enrico Busato and Andrea Ciavattini
Curr. Oncol. 2025, 32(1), 21; https://doi.org/10.3390/curroncol32010021 - 31 Dec 2024
Viewed by 596
Abstract
Surgery is the cornerstone of vulvar cancer treatment, but it is associated with a significant risk of complications that may impact prognosis, particularly in older patients with multiple comorbidities. The objective of this study was to evaluate the role of age, comorbidities, and [...] Read more.
Surgery is the cornerstone of vulvar cancer treatment, but it is associated with a significant risk of complications that may impact prognosis, particularly in older patients with multiple comorbidities. The objective of this study was to evaluate the role of age, comorbidities, and frailty in predicting postoperative complications after vulvar cancer surgery and to develop a predictive nomogram. A retrospective cohort study was conducted, including patients who underwent surgery for vulvar cancer at two Italian institutions from January 2018 to December 2023. A logistic regression model for the rate of Clavien-Dindo 2+ 30-days complications was run, considering the age-adjusted Charlson Comorbidity Index (AACCI), body mass index (BMI), and frailty as exposures. Lesion characteristics and surgical procedures were considered as confounders. Among the 225 included patients, 50 (22.2%) had a grade 2+ complication. The predictive score of the nomogram ranged from 44 to 140. The AACCI (0–64 points) and BMI (0–100 points) were independently associated with a risk of complications. A nomogram including the AACCI and BMI predicts the risk of complications for patients undergoing surgery for vulvar cancer. The preoperative determination of the risk of complications enables surgical planning and allows a tailored peri- and postoperative management plan. Full article
(This article belongs to the Section Gynecologic Oncology)
Show Figures

Figure 1

Figure 1
<p>Nomogram for prediction of Clavien–Dindo 2+ postoperative complications.</p>
Full article ">
21 pages, 17082 KiB  
Article
Single-Cell and Bulk Transcriptomics Reveal the Immunosenescence Signature for Prognosis and Immunotherapy in Lung Cancer
by Yakun Zhang, Jiajun Zhou, Yitong Jin, Chenyu Liu, Hanxiao Zhou, Yue Sun, Han Jiang, Jing Gan, Caiyu Zhang, Qianyi Lu, Yetong Chang, Yunpeng Zhang, Xia Li and Shangwei Ning
Cancers 2025, 17(1), 85; https://doi.org/10.3390/cancers17010085 - 30 Dec 2024
Viewed by 876
Abstract
Background: Immunosenescence is the aging of the immune system, which is closely related to the development and prognosis of lung cancer. Targeting immunosenescence is considered a promising therapeutic approach. Methods: We defined an immunosenescence gene set (ISGS) and examined it across 33 TCGA [...] Read more.
Background: Immunosenescence is the aging of the immune system, which is closely related to the development and prognosis of lung cancer. Targeting immunosenescence is considered a promising therapeutic approach. Methods: We defined an immunosenescence gene set (ISGS) and examined it across 33 TCGA tumor types and 29 GTEx normal tissues. We explored the 46,993 single cells of two lung cancer datasets. The immunosenescence risk model (ISRM) was constructed in TCGA LUAD by network analysis, immune infiltration analysis, and lasso regression and validated by survival analysis, cox regression, and nomogram in four lung cancer cohorts. The predictive ability of ISRM for drug response and immunotherapy was detected by the oncopredict algorithm and XGBoost model. Results: We found that senescent lung tissues were significantly enriched in ISGS and revealed the heterogeneity of immunosenescence in pan-cancer. Single-cell and bulk transcriptomics characterized the distinct immune microenvironment between old and young lung cancer. The ISGS network revealed the crucial function modules and transcription factors. Multiplatform analysis revealed specific associations between immunosenescence and the tumor progression of lung cancer. The ISRM consisted of five risk genes (CD40LG, IL7, CX3CR1, TLR3, and TLR2), which improved the prognostic stratification of lung cancer across multiple datasets. The ISRM showed robustness in immunotherapy and anti-tumor therapy. We found that lung cancer patients with a high-risk score showed worse survival and lower expression of immune checkpoints, which were resistant to immunotherapy. Conclusions: Our study performed a comprehensive framework for assessing immunosenescence levels and provided insights into the role of immunosenescence in cancer prognosis and biomarker discovery. Full article
(This article belongs to the Section Cancer Informatics and Big Data)
Show Figures

Figure 1

Figure 1
<p>The complete workflow of this study. Our study describes immunosenescence in human transcriptomes, including three sections: function analysis of the immunosenescence gene set in bulk and single-cell transcriptomes, candidate features for immunosenescence prognostic models in lung cancer, and development and validation of the immunosenescence risk model in lung cancer.</p>
Full article ">Figure 2
<p>ISGS functional properties associated with aging in TCGA and GTEX. (<b>A</b>) The landscape for differential expression and GSEA enrichment scores between age groups in TCGA (left) and GTEX (right). NES represents normalized enrichment score, red means NES &gt; 0, blue means NES &lt; 0; <span class="html-italic">p</span> value represents the significance of gene set enrichment; Up represented the number of upregulated genes in old groups; Common represents the number of genes shared by ISGS genes and upregulated genes in the old group. (<b>B</b>) Based on GSEA enrichment curves, the ISGS is significantly enriched during the aging process in lung tumors and lung tissues (<span class="html-italic">p</span> &lt; 0.05, NES &gt; 1). Red bar means old group; blue bar means young group. (<b>C</b>) Radar charts show the log2 (fold change) of ISGS genes in TCGA cancer types (top) and in GTEX normal tissues (bottom). (<b>D</b>) Bar plot represents the difference in ssgsea score of the ISGS between TCGA tumors and normal tissues.</p>
Full article ">Figure 3
<p>Four ISGS-related cell clusters were identified in single-cell data of lung cancer. (<b>A</b>) The tSNE plot of single-cell clustering analysis of GSE144945. (<b>B</b>) The number of overlapping genes of senescence-related gene sets. (<b>C</b>) To assess whether gene sets were enriched in cell subsets, we scored individual cells using four gene set enrichment methods and then calculated the differentially expressed gene sets for each cell subset. Finally, we used the robust rank aggregation (RRA) algorithm to screen out the gene sets that were significantly enriched in most gene set enrichment analysis methods. (<b>D</b>) The top 10 expression (left) and enriched (right) pathways of the markers of immunosenescence clusters. Red Hallmark represents the upregulated pathway, blue Hallmark represents the downregulated pathway. (<b>E</b>) The dot plot shows the average expression of cell molecules in the immunosenescence clusters. (<b>F</b>) The pseudotime trajectory of immunosenescence clusters annotated by states (top), clusters (middle), and pseudotime (bottom). (<b>G</b>) The expression of the top 1 marker of immunosenesence clusters during the pseudotime. (<b>H</b>) The different regulation of switch genes between two branches (state1–2 and state1–3).</p>
Full article ">Figure 4
<p>Immunosenescence-associated characteristics in tumor immune microenvironments. (<b>A</b>) Uniform manifold approximation and projection (UMAP) plot showing the main cell types in single-cell datasets of lung cancer patients (top). Proportions of cell clusters, with the numbers in parentheses indicating the number of cells (bottom). (<b>B</b>) The enrichment score (ES) for the ISGS within cell clusters, represented in the UMAP plot. (<b>C</b>) The distribution of ISGS enrichment scores of cell clusters. (<b>D</b>) The number of shared marker genes among the top 5 ISGS enriched cells. (<b>E</b>) KEGG pathways that were significantly enriched by the markers of the top 5 ISGS enriched cells. (<b>F</b>) Box plot of ES between the old and young groups. (<b>G</b>) The proportions of cell sub-populations between age groups. (<b>H</b>) The proportions of cell sub-populations among samples. (<b>I</b>) The inferred interaction number (top) and strength (bottom) between old and young groups. (<b>J</b>) Inferred cell–cell interactions among cell clusters in groups. (<b>K</b>) The crosstalk of the tumor-infiltrating lymphocyte cells (cytotoxic CD8 + T cells, CD4+ T cells, and B cells). The numbers represent the relative interaction strength as the sum of interaction weights. Edge weights are proportional to interaction strength; a thicker line refers to a stronger signal. (<b>L</b>) Dot plot for LRIs between B cells and other cells comparing old and young groups.</p>
Full article ">Figure 5
<p>ISGS activity was significantly correlated with the immune infiltration in lung adenocarcinoma. (<b>A</b>) Fraction of immune cell infiltration between old and young samples in the TCGA LUAD cohort. Green represents the old group; yellow represents the young group. (<b>B</b>) The overall activity of the ISGS is positively correlated with immune cells (<span class="html-italic">p</span> &lt; 0.05, R &gt; 6.0). The scatter represents the correlation coefficient. (<b>C</b>) The heat map shows that the expression of ISGS genes was significantly upregulated in old samples. Green represents the old group; yellow represents the young group. (<b>D</b>) Functional annotation of upregulated ISGS genes. GO terms show the biological process (BP). Red bar means log2 (fold change). (<b>E</b>) Correlation between upregulated ISGS genes and immune cell infiltration. (<b>F</b>) Scatter plots between ISGS genes and immune factors. (<b>G</b>) GSVA scores for the ISGS differed significantly between old and young groups (<span class="html-italic">p</span> &lt; 0.05, Wilcoxon rank sum test). (<b>H</b>) Scatter plots of GSVA scores in age groups. (<b>I</b>) Pearson correlation between the immune infiltration score and the GSVA score of the ISGS. GSVA, gene set variation analysis.</p>
Full article ">Figure 6
<p>Protein-protein interaction (PPI) network and TF-target network associated with the ISGS. (<b>A</b>) The PPI network consists of ISGS genes via string analysis. (<b>B</b>) Overall expression levels of ISGS genes between young and old groups (<span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05). (<b>C</b>) The lollipop chart shows the degrees of nodes in the PPI network. (<b>D</b>) Hub genes significantly enriched in GO terms (BP, biological process; CC, cellular component; MF, molecular function). (<b>E</b>) Boxplots of the expressions of TFs between old and young groups. (<b>F</b>) The TF-target network consists of ISGS genes and TFs.</p>
Full article ">Figure 7
<p>Construction and validation of the immunosenescence risk model (ISRM) in lung cancer cohorts. (<b>A</b>) Key ISGS genes were selected as candidate features for the model. (<b>B</b>) KEGG pathways are enriched by feature genes, showing the top ten pathways. (<b>C</b>) LASSO regression analysis identified 5 risk genes for the ISRM. (<b>D</b>) IHC staining of risk genes (CD40LG, CX3CR1, IL7, TLR3) in LUAD. (<b>E</b>) Kaplan–Meier plots of overall survival grouped by the median of the risk scores. Blue represents the high-risk group; red represents the low-risk group. (<b>F</b>) Kaplan–Meier plots of overall survival grouped by the median of the GSVA scores. Yellow represents the high-score group; light blue represents the low-score group. (<b>G</b>) ROC curves for one-year survival rate in TCGA LUAD patients. Red means the IRSM prediction model, blue means the GSVA prediction model. (<b>H</b>) Univariate Cox regression analysis for the ISRM and clinical factors. (<b>I</b>) Multivariate Cox regression analysis for the ISRM and clinical factors. ***: <span class="html-italic">p</span> &lt; 0.001. (<b>J</b>) A constructed nomogram for prognostic prediction of a patient with LUAD. The importance of each variable was ranked according to the standard deviation along nomogram scales. ***: <span class="html-italic">p</span> &lt; 0.001. (<b>K</b>) Kaplan–Meier curves for overall survival grouped by the risk scores in GSE68465 and GSE72094 (<span class="html-italic">p</span> &lt; 0.5, log-rank test). (<b>L</b>) ROC curves for one-year survival rate in GSE68465, GSE72094, GSE26939, and GSE68571. TCGA, The Cancer Genome Atlas; ROC, receiver operating characteristic; LUAD, lung adenocarcinoma; AUC, the area under the ROC curve.</p>
Full article ">Figure 8
<p>Application of the ISRM in anti-tumor therapy and immunotherapy of lung cancer. (<b>A</b>) Density plots and boxplots of high-risk group-specific anti-tumor drugs predicted by the ISRM model. Blue represents the high-risk group; orange represents the low-risk group. (<b>B</b>) Anti-tumor drugs (left) with significant IC50 differences between risk groups, targets (middle), and pathways (right). (<b>C</b>) Boxplots of immune checkpoint molecules grouped by risk scores. Blue represents the high-risk group, orange represents the low-risk group. (<b>D</b>) Heatmap showing the expression of the risk genes and PDCD1 in the GSE93157 cohort of 35 patients. Blue represents the high-risk group; orange represents the low-risk group. (<b>E</b>) Violin plots of risk genes and PDCD1 grouped by the ISRM prediction model. Blue represents the high-risk group; orange represents the low-risk group. (<b>F</b>) Violin plots of risk genes and PDCD1 grouped by the response to anti-PD-1 immunotherapy. Blue means responder (NPD), and yellow means non-responder (PD). (<b>G</b>) XGBoost evaluated the predictive ability of ISRM on immunotherapy. IC50, the half maximal inhibitory concentration.</p>
Full article ">
13 pages, 2705 KiB  
Article
Prognostic Role of Lymphocyte-to-C-Reactive Protein Ratio in Patients with Pulmonary Arterial Hypertension
by Meng-Qi Chen, Chuan-Xue Wan, Jun Tong, An Wang, Bin-Qian Ruan and Jie-Yan Shen
J. Clin. Med. 2024, 13(24), 7855; https://doi.org/10.3390/jcm13247855 - 23 Dec 2024
Viewed by 532
Abstract
Background: Inflammation plays a critical role in the prognosis of patients with pulmonary arterial hypertension (PAH). The lymphocyte-to-C-reactive protein ratio (LCR), as a novel inflammatory marker, has not been studied in patients with PAH. The objective of this study was to investigate the [...] Read more.
Background: Inflammation plays a critical role in the prognosis of patients with pulmonary arterial hypertension (PAH). The lymphocyte-to-C-reactive protein ratio (LCR), as a novel inflammatory marker, has not been studied in patients with PAH. The objective of this study was to investigate the prognostic value of the LCR in patients with PAH. Methods: A retrospective cohort study was conducted on 116 patients with PAH diagnosed in Renji Hospital, School of Medicine, Shanghai Jiao Tong University, from January 2014 to December 2018. The primary outcome was a composite endpoint that included lung transplantation, rehospitalization for PAH, and all-cause death. The LCR is the ratio of the blood lymphocyte count to the C-reactive protein concentration. Results: A total of 116 patients with PAH were included in this study, with an average age of 41.53 years; 92.2% were female, and the event rate was 57.8%. Restricted cubic spline analysis confirmed a linear association between the LCR and the risk of clinical worsening events. Multivariate Cox proportional hazards analysis showed that the LCR was significantly negatively associated with clinical worsening events, with hazard ratios and 95% confidence intervals of 0.772 (0.614–0.970). The Kaplan–Meier curve showed that event-free survival decreased significantly when the LCR was less than 1.477. LASSO regression selected four potential predictors, including the LCR, to construct a nomogram. The nomogram had a high predictive strength, with an area under the ROC curve of 0.805 (0.713–0.896). The calibration curves and decision curve analysis indicated that the nomogram had good predictive performance and the ability to guide clinical management. Conclusions: The LCR is a valuable prognostic marker for predicting long-term clinical events in patients with PAH, and the nomogram incorporating the LCR could effectively stratify risk and guide clinical decision making. Full article
(This article belongs to the Special Issue Clinical Insights into Pulmonary Hypertension)
Show Figures

Figure 1

Figure 1
<p>Restricted cubic spline analysis of LCR and risk of clinical worsening events in patients with PAH. Changes in hazard ratios for 1-year (<b>A</b>), 3-year (<b>B</b>), and 5-year (<b>C</b>) clinical worsening events across different baseline levels of the LCR. Hazard ratios and 95% confident intervals were estimated using Cox proportional hazards models. LCR, lymphocyte-to-C-reactive protein ratio; PAH, pulmonary arterial hypertension.</p>
Full article ">Figure 2
<p>Kaplan–Meier survival analysis of LCR and the risk of clinical worsening events in patients with PAH. The LCR was expressed as a categorical variable according to optimal <span class="html-italic">p</span>-value method: LCR &lt; 1.477 and LCR ≥ 1.477. Differences between the two groups were compared using the log-rank test. LCR, lymphocyte-to-C-reactive protein ratio; PAH, pulmonary arterial hypertension.</p>
Full article ">Figure 3
<p>Selection of potential predictors of LASSO regression in patients with PAH. (<b>A</b>) The coefficient shrinkage process for 28 variables, with colored lines representing the changes in coefficients of different features at various levels of shrinkage. (<b>B</b>) A 10-fold cross-validation to determine the optimal penalty parameter lambda. A vertical line is drawn at the point of 1 standard error (1-SE) of the minimum criterion. LASSO, least absolute shrinkage and selection operator; PAH, pulmonary arterial hypertension.</p>
Full article ">Figure 4
<p>Nomogram for clinical worsening events prediction in patients with PAH. The nomogram was based on Cox proportional hazards models, integrating four variables: lymphocyte-to-C-reactive protein ratio (LCR), PAH subtype (subgroups), 6 min walk distance (6MWD), and WHO cardiac functional class (WHO-FC). PAH, pulmonary arterial hypertension.</p>
Full article ">Figure 5
<p>Receiver operating characteristic curves of four models for 1-year (<b>A</b>), 3-year (<b>B</b>) and 5-year (<b>C</b>) clinical worsening events. The dotted line represents the reference line. Clinical model included variables of PAH subtype, 6 min walk distance, and WHO cardiac functional class. European Society of Cardiology (ESC) model included variables of B-type natriuretic peptide, 6 min walk distance, and WHO cardiac functional class.</p>
Full article ">Figure 6
<p>Validation of the four models through calibration curve and decision curve analysis. (<b>A</b>–<b>C</b>) Calibration curves of the four models for 1-year (<b>A</b>), 3-year (<b>B</b>), and 5-year (<b>C</b>) clinical worsening events, with the x-axes representing actual event probabilities and the y-axes representing predicted event probabilities. The dotted line represents the ideal prediction. (<b>D</b>–<b>F</b>) Decision curve analysis of the four models for 1-year (<b>D</b>), 3-year (<b>E</b>), and 5-year (<b>F</b>) clinical worsening events. Clinical model includes variables of PAH subtype, 6 min walk distance, and WHO cardiac functional class. European Society of Cardiology (ESC) model includes variables of B-type natriuretic peptide, 6 min walk distance, and WHO cardiac functional class.</p>
Full article ">
15 pages, 1295 KiB  
Article
Predictive Factors of the Degrees of Malnutrition According to GLIM Criteria in Head and Neck Cancer Patients: Valor Group
by Francisco Javier Vílchez-López, María González-Pacheco, Rocío Fernández-Jiménez, María Teresa Zarco-Martín, Montserrat Gonzalo-Marín, Jesús Cobo-Molinos, Alba Carmona-Llanos, Araceli Muñoz-Garach, Pedro Pablo García-Luna, Aura D. Herrera-Martínez, Felisa Pilar Zarco-Rodríguez, María del Carmen Galindo-Gallardo, Luis Miguel-Luengo, María Luisa Fernández-Soto and José Manuel García-Almeida
Cancers 2024, 16(24), 4255; https://doi.org/10.3390/cancers16244255 - 21 Dec 2024
Viewed by 683
Abstract
Background: Malnutrition is highly prevalent in patients with head and neck cancer, with relevant consequences in the treatment results. Methods: Multicenter observational study including 514 patients diagnosed with HNC. The morphofunctional assessment was carried out during the first 2 weeks of radiotherapy treatment. [...] Read more.
Background: Malnutrition is highly prevalent in patients with head and neck cancer, with relevant consequences in the treatment results. Methods: Multicenter observational study including 514 patients diagnosed with HNC. The morphofunctional assessment was carried out during the first 2 weeks of radiotherapy treatment. A correlation analysis between nutritional variables and groups of malnutrition, a multivariate logistic regression analysis, and a random forest analysis to select the most relevant variables to predict malnutrition were performed. Results: In total, 51.6% were undernourished (26.3% moderately and 25.3% severely). There was a negative correlation between morphofunctional variables and a positive correlation between hsCRP and well vs. moderate and well vs. severe malnutrition groups. The increase in different bioelectrical and ultrasound parameters was associated with a lower risk of moderate and severe malnutrition when groups with different degrees of malnutrition were compared. To predict the importance of morphofunctional variables on the risk of undernutrition, a nomogram, a random forest, and decision tree models were conducted. For the well vs. moderate, for the well vs. severe, and for the moderate vs. severe malnutrition groups, FFMI (cut-off < 20 kg/m2), BCMI (cut-off < 7.6 kg/m2), and RF-Y-axis (cut-off < 0.94 cm), respectively, were the most crucial variables, showing a greater probability of mortality in the two last comparisons. Conclusions: Malnutrition is very prevalent in HNC patients. Morphofunctional assessment with simple tools such as electrical impedance and muscle ultrasound allows an early nutritional diagnosis with an impact on survival. Therefore, these techniques should be incorporated into the daily clinical attention of patients with HNC. Full article
(This article belongs to the Section Cancer Causes, Screening and Diagnosis)
Show Figures

Figure 1

Figure 1
<p>Significant correlations between body composition parameters assessed by BIVA and ultrasound nutritional evaluation, biochemical nutritional parameters, and sarcopenia (* <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). BIVA: Bioelectrical impedance vector analysis.</p>
Full article ">Figure 2
<p>Clinical algorithm for predicting malnutrition in HNC patients using body composition parameters determined by BIVA ultrasound nutritional evaluation. (<b>A</b>) Random forest evaluating the most important variable between well-nourished and moderate malnutrition groups. (<b>B</b>) Decision tree model for well-nourished vs. moderate malnutrition group. (<b>C</b>) Survival analysis comparing the group of well-nourished vs. moderate malnutrition group. (<b>D</b>) Random forest evaluating the most important variable between well-nourished and severe malnutrition groups. (<b>E</b>) Decision tree model for well-nourished vs. severe malnutrition groups. (<b>F</b>) Survival analysis comparing the well-nourished group with the severe malnutrition group. (<b>G</b>) Random forest evaluating the most important variable between moderate and severe malnutrition groups. (<b>H</b>) Decision tree model for moderate vs. severe malnutrition groups. (<b>I</b>). Survival analysis comparing the moderate and severe malnutrition groups.</p>
Full article ">
Back to TopTop