You are viewing the site in preview mode

Skip to main content

Habitat radiomics predicts occult lymph node metastasis and uncovers immune microenvironment of head and neck cancer

Abstract

Background

Occult lymph node metastasis (LNM) is a key prognostic factor for patients with head and neck squamous cell carcinoma (HNSCC). This study was to establish radiomics models derived from intratumoral, peritumoral, and habitat regions for identifying occult LNM in HNSCC.

Methods

Patients with pathologically confirmed HNSCC from three medical Centers (from March 2014 to April 2024) and The Cancer Genome Atlas (TCGA) were enrolled. Center 1 was split into training (n = 330) and internal test sets (n = 154), while Center 2 and Center 3 served as the external test set (n = 183). Genomic set (n = 50) from TCGA and single-cell RNA sequencing set (n = 6) from Center 1 were used for biological analysis. We used the intratumoral, peritumoral, and habitat volumes of interest (VOIs) to extract radiomics features, respectively. Based on Logistic Regression (LR), Support Vector Machine (SVM), and Random Forest (RF) classifiers, nine radiomics models were built to confirm the optimal predictive performance. The best-performing model, along with clinical-radiologic data, was combined to develop a hybrid model. The log-rank test was used to evaluate the model’s prognostic performance. Additionally, bulk and single-cell RNA sequencing were applied for investigating the biological mechanisms underlying the optimal model.

Results

The RF-habitat radiomics model showed the best performance, achieving AUCs of 0.835–0.919 across all datasets. Survival analysis further confirmed the prognostic value of the RF-habitat radiomics model. The RF-habitat radiomics model and the hybrid model notably surpassed the clinical model in predictive performance. Moreover, the RF-habitat radiomics model was associated with the abundance level of exhaustion-associated CD8 + T cells, uncovering the immune microenvironment characteristics contributing to occult LNM in HNSCC.

Conclusions

The RF-habitat radiomics model demonstrated excellent performance for predicting occult LNM in HNSCC across three cohorts, providing a non-invasive solution for occult LNM. Furthermore, radiogenomic analysis further revealed the biological associations of the model, primarily related to T cell dysfunction.

Introduction

Head and neck squamous cell carcinoma (HNSCC) is the most common malignant neoplasm in the head and neck region, characterized by high invasiveness and metastatic potential. Despite remarkable advancements in diagnostic techniques and treatment strategies in recent years, the 5-year survival rate of patients has not significantly improved [1, 2]. This phenomenon is primarily attributed to the high heterogeneity of HNSCC, along with the high risk of local recurrence and distant metastasis. Cervical lymph node metastasis (LNM) further complicates the condition and serves as a critical determinant of poor prognosis in HNSCC [3].

Imaging examinations (ultrasound, CT, MRI, etc.), are routinely applied for assessing LN status by evaluating LN morphology and blood supply. However, a subset of patients classified as clinically LN negative (cN0) present no obvious signs on clinical and imaging evaluation. Postoperative pathological analysis often reveals that 30–40% of these cN0 patients harbor LNM [4]. The appropriate management of cN0 patients remains a topic of ongoing debate. Redaelli et al. [5] proposed routine prophylactic neck dissection for laryngeal cancer patients who were identified as T2 or higher to mitigate the risk of occult LNM. However, this approach may lead to overtreatment, with more than 70% of patients undergoing unnecessary surgery [6]. Though PET-CT is highly sensitive in assessing occult LNM, misinterpretations resulting from inflammation or infection are not uncommon [7]. Various molecular biomarkers show potential for predicting occult LNM in HNSCC, including epithelial-mesenchymal transition markers, tumor microenvironment/immune regulators, and genetic mutation signatures [8, 9]. However, their clinical translation still encounters major hurdles, particularly in standardization of detection protocols and robustness of clinical validation. Consequently, striking a balance between prophylactic neck dissection and avoiding overtreatment for cN0 patients remains a significant clinical challenge.

By transforming medical imaging data into a multitude of quantitative and complex features, radiomics provides deep insights into tumor heterogeneity and has recently gained significant attention in the medical field. Unlike conventional radiomics analysis for the whole tumor region, habitat analysis divides the tumor into subregions based on areas with similar biological features [10]. This approach offers a more accurate quantification of tumor heterogeneity. Verma et al. [11] employed habitat radiomics to MRI data of glioblastoma and identified two subregions—enhancing tumor and FLAIR hyperintensity—that had a major impact on patient prognosis, yielding a concordance index of 0.80 for predicting disease free survival. Furthermore, the two subregions were also linked to tumor aggressiveness, including tumor infiltration and hyperplastic blood vessels. Another study by Huang et al. [12] demonstrated that habitat radiomics model showed better performance than conventional radiomics model in predicting treatment response after radiofrequency ablation of colorectal cancer lung metastasis. The model identified three distinct tumor subregions—necrotic, fluid, and hypervascular—that corresponded with histopathological findings. Growing evidence highlights the close relationship between tumor subregions and biological behavior [13,14,15]. To date, no studies have attempted to address the clinical challenge of occult LNM through radiomics analysis of distinct tumor subregions.

Collectively, our study was intended to construct intratumoral, peritumoral, and habitat radiomics models to predict occult LNM in HNSCC. Additionally, we will further investigate the tumor microenvironment related to the radiomics model through bulk and single-cell RNA sequencing analysis.

Materials and methods

Patients

This study included 723 pathologically confirmed cases of HNSCC from three Centers (from March 2014 to April 2024) and the Cancer Genome Atlas (TCGA). A total of 484 HNSCC patients from Center 1 (The First Affiliated Hospital of Chongqing Medical University) were randomly divided into a training set (n = 330) and an internal test set (n = 154) at a 7:3 ratio. Patients from Center 2 (The People’s Hospital of Hechuan) and Center 3 (Zigong Fourth People’s Hospital) served as the external test set (n = 183). Genomic set (n = 50), including available CT images from The Cancer Imaging Archive (TCIA) and RNA sequencing data from TCGA [16], were obtained to explore radiogenomic correlations. Additionally, six tumor samples from patients at Center 1 were collected for single-cell RNA sequencing. The flowchart and inclusion/exclusion criteria are detailed in Fig. 1. The study was approved by the Institutional Review Board of the First Affiliated Hospital of Chongqing Medical University (approval ID: 2021–433). Written informed consent was waived for the retrospective data sets, whereas it was acquired for single-cell sequencing set. Data collection and review adhered strictly to medical ethical standards, and this study was registered at clinicaltrials.gov (https://www.clinicaltrials.gov/; NCT06757530; January 3, 2025).

Fig. 1
figure 1

Recruitment process of HNSCC patients

Clinical characteristics and radiologic assessment

We comprehensively retrieved and collected patients’ clinical information, including age, sex, smoking, alcohol consumption, histological grade, and clinical T staging. Radiologic assessments were conducted by two radiologists and one otolaryngologist (P.J., J.H., F.C.M., each with 22, 6, and 8 years of experience in head and neck tumors, respectively). The assessors had no access to any clinical information and independently evaluated multi-phase CT imaging, focusing on tumor volume, maximum tumor diameter, tumor enhancement pattern, and intratumoral necrosis. The final tumor volume and maximum tumor diameter were determined by taking the mean of the measurements from three assessors, and any disagreements regarding tumor enhancement patterns or intratumoral necrosis were resolved by majority vote.

Imaging acquisition and tumor segmentation

Image data was sourced from CT scanners at three Centers, and Supplementary Table S1 provides the specific scanning settings. After a plain scan, iodinated contrast agents were injected via the antecubital or hand vein, followed by the acquisition of venous-phase images at 65–70 s post-injection. To minimize differences in CT devices or scanning protocols, image preprocessing, including intensity normalization and resampling, was performed.

To ensure the stability of the extracted radiomic features, accurate identification and delineation of the tumor lesion are essential. With the assistance of ITK-SNAP (version 3.8.0), a widely recognized software for medical image segmentation, reader 1 (J.H.) meticulously depicted the region of interest (ROI) of the tumor on each CT slice. Subsequently, we overlaid the ROIs from all slices to form a three-dimensional (3D) volume of interest (VOI), providing a precise spatial foundation for subsequent analysis. After four weeks, reader 1 (J.H.) and reader 2 (C.X.W., with 6 years of experience in head and neck tumors) independently delineated tumor lesion in a random sample of 30 cases to assess inter- and intraobserver variability. Any uncertainties in tumor segmentation were resolved through consensus review with a senior doctor (P.J.).

Sub-region generation

Division of peri- and intratumoral areas

After obtaining the tumor’s VOI, we isotropically expanded the delineated ROIs of each VOI outward by 1 mm to define the peritumoral region. The choice of expansion length for the peritumoral region was determined based on preliminary experiments using different distances, with 1 mm peritumoral distance ultimately demonstrating the best result (Supplementary Figure S1), which is consistent with other literature [17,18,19]. The expanded peritumoral regions were stacked to form the peritumoral VOI. Any abnormal regions, such as cavities or extracapsular areas, were manually adjusted. The original VOIs were considered as the intratumoral region, which exclusively encompassed the core tumor area without including surrounding tissues. Both the intratumoral and peritumoral VOIs were used for subsequent radiomics analyses. The study flowchart is presented in Fig. 2.

Fig. 2
figure 2

The framework of this retrospective study. Step 1: VOI segmentation. The intratumoral, peritumoral, and habitat VOIs were derived from tumor delineation on the venous-phase CT images. Step 2: Feature extraction/section. Step 3: Model building and evaluation. LR, SVM, and RF classifiers were employed to construct predictive models. Step 4: Biological analysis. Bulk and single-cell RNA sequencing analysis were performed to explore the biological relationship

Habitat region clustering

To further quantify the heterogeneity of different tumor regions, we calculated 18 radiomics features for each voxel within the tumor VOIs, which include quantitative descriptions of image grayscale distribution, spatial relationships, and texture patterns. Detailed explanations of the 18 features are provided in Supplementary Table S2. The K-means algorithm, a commonly used unsupervised clustering algorithm, was exploited to automatically sort the processed voxels into individual subregions. This method performs unsupervised classification of the radiomics feature information for each voxel, dividing the VOIs into distinct subregions based on feature similarity. In addition, we tested cluster numbers from 2 to 10 and utilized the Davis-Bouldin (DB) score [20] to identify the most suitable partitioning of tumor subregions.

Radiomics feature extraction and model development

Based on the intratumoral, peritumoral, and habitat VOIs, radiomics features were separately extracted through the Python package PyRadiomics. To ensure consistency and reliability, features with intra- and interclass correlation coefficients (ICCs) > 0.75 were retained and subjected to Z-score normalization. The Mann-Whitney U test was conducted to determine statistically discrepant features, while Spearman correlation analysis was performed to remove highly correlated features (r > 0.9). The remaining features were chosen using the Least Absolute Shrinkage and Selection Operator (LASSO) method. Finally, founded on the optimal features of intratumoral, peritumoral, and habitat VOIs, models were constructed by using Logistic Regression (LR), Support Vector Machine (SVM), and Random Forest (RF) classifiers to differentiate between cN0 patients with and without LNM. The stability and generalizability of the model were assessed using 5-fold cross-validation.

In the training set, uni- and multivariable LR analyses were conducted between pathological LN positive (pN+) and pathological LN negative (pN-) to identify independent predictive factors among clinical characteristics, radiological indicators, and radiomics scores (the output of the best model). The Variables with P < 0.05 were incorporated into the construction of a hybrid model. Additionally, regarding the results from uni- and multivariate analyses, a clinical model that integrated significant clinical characteristics and radiological indicators were built.

Prognostic evaluation and biological functions exploration

The cutoff value (0.270) was calculated by the maximum Youden index in the area under the receiver operating characteristic (ROC) curve (AUC) analysis of the training set. Patients were categorized into high- and low-risk groups according to the cutoff value. The log-rank test was used to compare 3-year disease-free survival (DFS) and overall survival (OS) between the two groups. Further details on the follow-up strategy can be found in Appendix S1. Additionally, differentially expressed genes (DEGs) between the low- and high-risk groups in the genomic set were analyzed by means of the R package DESeq2. Subsequently, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, Gene Set Enrichment Analysis (GSEA), and Gene Set Variation Analysis (GSVA) were performed using the R package clusterProfiler. Furthermore, single-cell RNA sequencing was utilized to explore cell composition and functional state of the tumor heterogeneity between the two groups. To validate these cellular and molecular findings, tumor samples from both metastasis and non-metastasis groups were employed for multiplex immunofluorescence (mIF) staining to assess the distribution of key cell populations and the expression of important molecules at the tissue level. Supplementary Appendix S1 provides details on the radiotranscriptomic analysis.

Statistical analysis

Statistical analyses were conducted using SPSS (version 24.0) and Python (version 3.8) software. Continuous variables were expressed as mean ± standard deviation and compared between pN + group and pN- group using t-tests or Mann-Whitney U tests. Categorical variables were presented as frequencies and percentages, and compared between the two groups using chi-square tests or Fisher’s exact tests. P value < 0.05 was considered statistically significant. Additionally, the AUC was calculated to assess the diagnostic performance, and DeLong method was employed to compare models’ performance. Calibration curves and decision curves analysis were used to evaluate models’ performance and clinical applicability.

Results

Patient characteristics

The incidence of occult LNM was 26.06% (86 of 330) in the training set, 31.17% (48 of 154) in the internal test set, 33.33% (61 of 183) in the external test set, and 30.00% (15 of 50) in the genomic set, and the LN status was balanced across the four cohorts (P = 0.337). The clinical characteristics of the study population is summarized in Table 1. Grouping by LN status in the training set revealed that pN + patients were more likely to exhibit T3 or T4 stages, poor differentiation, and imaging features such as larger tumor volume, longer maximum tumor diameter, and a greater tendency for heterogeneous enhancement and tumor necrosis (all P < 0.05). On the other hand, there were no significant differences between the two groups in terms of age, sex, smoking, and alcohol consumption (Table 2).

Table 1 Baseline clinical characteristics of the patients in the datasets

Habitat region and feature selection

After applying the K-means method with different numbers of clusters, the optimal DB score was observed with 3 clusters (Supplementary Figure S2). Therefore, the habitat VOIs with 3 subregions were used for subsequent habitat analysis. There were 1034 extracted radiomics features corresponding to the intratumoral, peritumoral, and habitat VOIs, respectively. Of these, 880 features were those with ICCs > 0.75 and were retained to maintain feature reproducibility. Notably, ICCs are unsuitable for evaluating unsupervised habitat radiomics features. Subsequently, the features were further filtered using the Mann-Whitney U test, Spearman correlation, and LASSO method. Ultimately, 28 features from the intratumoral VOIs (Supplementary Figure S3), 25 features from the peritumoral VOIs (Supplementary Figure S4), and 31 features from the habitat VOIs (Supplementary Figure S5) were retained for modeling, respectively.

Performance evaluation of all predictive models

Using LR, SVM, and RF classifiers, nine radiomics models were constructed based on the optimal features extracted from intratumoral, peritumoral, and habitat VOIs. Among them, the RF-based radiomics model demonstrated superior performance than the LR- and SVM-based models in two test sets, with the RF-habitat radiomics model exhibiting excellent results (Supplementary Table S3 and Fig. 3A–C). It achieved AUCs of 0.919 (95% confidence interval [CI]: 0.886–0.951) in the training set, 0.857 (95% CI: 0.780–0.933) in the internal test set, and 0.835 (95% CI: 0.765–0.904) in the external test set. Using Shapley additive explanations (SHAP) method for visualization, the RF-habitat radiomics model’s contribution of individual features was demonstrated (Fig. 4). Among the top 10 features, four originated from region 3, three from region 2, and three from region 1, with features of region 3 showing higher weight in the model development. Moreover, a significant difference in RF-habitat radiomics scores was detected between the pN + and pN- groups in the training set (Table 2). The ROC curves for all predictive models are shown in Fig. 3, and the calibration curves and decision curves analysis are provided in Supplementary Figures S6 and S7.

Fig. 3
figure 3

Performance evaluation of different models for predicting occult LNM in HNSCC patients. ROC curves for intratumoral, peritumoral, and habitat radiomics models in the training (A), internal test (B), and external test sets (C), respectively. Additionally, the predictive performances of the RF-habitat radiomics model, clinical model, and hybrid model in the in the training (D), internal test (E), and external test sets (F), respectively

Fig. 4
figure 4

Model interpretability of the RF-habitat radiomics model through SHAP plot. (A) The SHAP bar chart showed the importance of top 10 features in the model. (B) The SHAP bees-warm plot showed the positive or negative effects of each feature through red and blue colours

Independent predictors of occult LNM, as determined by uni- and multivariate LR analyses, were maximum tumor diameter (odds ratio [OR] = 0.523), histological grade (OR = 0.345), and RF-habitat radiomics score (OR = 6.297) (Table 2). Using these three variables, we constructed a hybrid model to estimate LN status, with AUCs of 0.910 (95% CI: 0.874–0.944), 0.876 (95% CI: 0.806–0.944), 0.826 (95% CI: 0.753–0.898) in the training, internal test and external test sets, respectively (Table 3). Additionally, we found that the hybrid model and the RF-habitat radiomics model significantly outperformed the clinical model, which incorporated maximum tumor diameter and histological grade (both P < 0.05) (Supplementary Figure S8).

Table 2 Uni- and multivariate LR analyses of variables for predicting occult LNM in patients in the training set
Table 3 The performance of predictive models in predicting occult LNM

Clinical outcomes and biological functions related to the RF-habitat radiomics model

We collected patients’ 3-year DFS and OS data to evaluate the prognostic value of the RF-habitat radiomics model. In the training cohort (208 eligible patients), the 3-year DFS and OS rates were 58.1% and 65.9% in the low-risk group, while the 3-year DFS and OS rates in the high-risk group were 39.2% and 49.4%, respectively. In the internal test set (107 eligible patients), the 3-year DFS and OS rates were 65.3% and 72.2% in the low-risk group, compared to 37.1% and 51.4% in the high-risk group. Similarly, in the external test set (123 eligible patients), the 3-year DFS and OS rates were 58.5% and 67.1% in the low-risk group, compared to 31.7% and 48.8% in the high-risk group (Supplementary Table S4). Survival analysis further confirmed that patients stratified by the RF-habitat radiomics model had significantly different prognoses across all sets (Fig. 5).

Fig. 5
figure 5

Kaplan-Meier analyses of 3-year DFS and OS stratified by the RF-habitat radiomics model. (A, B) Training set. (C, D) Internal test set. (E, F) External test set

Four hundred sixty-three DEGs were identified in the genomic set between low- or high-risk groups (Fig. 6A, B). KEGG results showed that DEGs were closely associated with immune-regulatory pathways, including T cell receptor signaling pathway, primary immunodeficiency, and IL-17 signaling pathway. Pathways related to metabolism and signal transduction were also upregulated in high-risk groups (Fig. 6C). Similarly, GSEA indicated that immune regulation pathways and lipid metabolism-related pathways were significantly enriched in the high-risk group (Fig. 6D).

Fig. 6
figure 6

Bulk RNA sequencing analysis for biological basis associated with the RF-habitat radiomics model. (A) Heatmap depicted the expression profiles of DEGs between low and high-risk groups stratified by the RF-habitat radiomics model. (B) Volcano plot showed the distribution of DEGs between two groups. (C) KEGG enrichment barplot displayed the key pathways enriched by DEGs. (D) GSEA based on the Gene Ontology (GO) database revealed immune and metabolism-related pathways enriched in the high-risk group

We conducted single-cell RNA sequencing analysis on 6 HNSCC patients grouped by the RF-habitat radiomics model. After strict quality control, we obtained 68,613 cells in total, which were identified as epithelial cells, endothelial cells, fibroblasts, myeloid cells, NK/T cells, B cells, and plasma cells (Fig. 7A–C). The top five marker genes of each cluster are displayed in Supplementary Figure S9A. Further, malignant epithelial cells were classified as cancer cells using chromosomal copy number variation profile analysis (inferCNV) with B cells as reference (Supplementary Figure S10). Cell populations were unevenly distributed both across samples and between groups, particularly with a distinct difference in the infiltration rate of NK/T cells. It indicated that the high-risk group appeared a more complex immune microenvironment, partially aligned with the findings of bulk RNA sequencing analysis (Fig. 7D–F). Subsequently, we regrouped NK/T cells as CD8_GZMK, CD8_CXCL13, CD8_IGHV, CD4_CCR7, Treg_FOXP3, NK_FCGR3A, DN_KRT6A, DP_MKI67. The top five marker genes across T/NK cell subclusters are displayed in Supplementary Figure S9B. Notably, it showed that the infiltration levels of CD8_CXCL13 and CD8_IGHV cells were heightened in the high-risk group. Though not statistically significant, the two subtypes appeared to be key components contributing to the differences and complexity of the immune microenvironment between the two groups. Further analysis indicated that the high density of CD8_IGHV cells was primarily driven by the imbalanced proportion of the HNSCC6 sample. In contrast, exhaustion-associated CD8_CXCL13 cells exhibited a relatively stable higher abundance, which is consistent with previous research findings [21,22,23]. Additionally, the CD8_CXCL13 cells proportion derived from the CIBERSORTx deconvolution algorithm of 50 bulk RNA data also showed results consistent with single-cell analysis (Supplementary Figure S11). Collectively, it indicated that exhaustion-associated CD8_CXCL13 cells may be pivotal in the immune microenvironment contributing to occult LNM in HNSCC. To further elucidate the molecular characteristics and functional states, we performed DGEs analysis for each subgroup of NK/T cells, as shown in Fig. 7G. Moreover, GSVA revealed that CD8_CXCL13 cells were mainly associated with upregulation of the INF-alpha or gamma response signaling pathway (Fig. 7H). MIF analysis demonstrated that the metastasis group exhibited a relatively higher abundance of exhaustion-associated CD8 + T cells (CD8_CXCL13) (Fig. 8).

Fig. 7
figure 7figure 7

Single-cell landscape of tumor microenvironment related to the RF-habitat radiomics model. (A) Uniform manifold approximation and projection (UMAP) plots color coded for cell types (left), individual samples (middle), and groups (right) (low-risk group: HNSCC1-HNSCC3; high-risk group: HNSCC4 − HNSCC6). (B) Proportions of eight identified cell types presented in bar plots across different samples (left) and groups (right). (C) The infiltration levels of eight cell types in low- and high-risk groups. (D) UMAP plots displayed NK/T cells regrouped into eight subgroups. (E) Proportions of each subgroup presented in bar plots across different samples (left) and groups (right). (F) The infiltration levels of each subgroup in low- and high-risk groups. (G) DGEs analysis showed up- and down-regulated genes of each subgroup. (H) GSVA of Hallmark gene sets exhibited the pathway activity profiles across eight subgroups. Up- and downregulated signaling pathways were shown in red and blue, respectively

Fig. 8
figure 8

MIF results for CD8_CXCL13 cells in pN + and pN- groups. (A) One cN0 patient with HNSCC confirmed as pN+ (score = 0.683) was infiltrated by abundant CD8_CXCL13 cells in tumor microenvironment. (B) Another cN0 patient with HNSCC confirmed as pN- (score = 0.214) showed minimal CD8 + T cell infiltration. Green fluorescent label CD8; Red fluorescent label CXCL13; DAPI blue fluorescent label cell nuclei

Discussion

In this study, the RF-habitat radiomics model outperformed both intratumor and peritumor radiomics models in predicting occult LNM in HNSCC, showing outstanding predictive capability. By quantifying distinct tumor subregions, the model offered a more nuanced understanding of tumor heterogeneity. Furthermore, the RF-habitat radiomics score effectively stratified HNSCC patients in terms of 3-year DFS and OS. Both the RF-habitat radiomics model and the hybrid model surpassed the clinical model in predictive performance. Remarkably, radiogenomic analysis suggested that the RF-habitat radiomics model was linked to exhaustion-associated CD8 + T cell, revealing the immune microenvironment characteristics potentially contributing to occult LNM.

Recently, the rapid development of radiomics has brought a novel and promising perspective for LNM prediction in head and neck tumors. Zhang and his team [24] proposed that a clinical-radiomics nomogram based on iodine maps achieved high predictive accuracy in identifying LNM for HNSCC patients. In addition, a study by Wang and his team [15] collected data from 553 cN0 patients with laryngeal squamous cell carcinoma (LSCC) and compared the performance of deep learning, radiomics, and hybrid models in identifying LN status. It showed that the decision-based hybrid model realized superior predictive accuracy across all study cohorts. Nevertheless, most existing literature primarily concentrates on the features of whole lesion, which may not fully capture the microenvironmental characteristics of tumor’s subregions associated with LNM.

Habitat radiomics decomposes tumors into multiple “habitat” regions with distinct biological characteristics and has gradually become an innovative approach for in-depth analysis of tumor heterogeneity. Wei et al. [25] used multi-sequence MRI data to manually segment the tumor and edema regions for predicting MGMT promoter methylation in grade II-IV astrocytoma. Their results showed that a hybrid model combining two subregions surpassed the one-region models, achieving the best results. However, the manual subregion segmentation process is tedious and heavily relied on radiologists’ subjective experience [26]. Surprisingly, this study applied the K-means method, a classic non-supervised clustering technique, to segregate tumor regions according to voxel feature similarity, which reduced manual intervention and improved the reproducibility of the results [27, 28]. Moreover, unlike previous studies that used imaging grayscale values for clustering, the study calculated 18 radiomics features for tumor voxels, providing a more comprehensive and detailed description of the tumor’s heterogeneity [12, 29]. Our findings revealed that the RF-habitat radiomics model showed excellent precision in the prediction of occult LNM, offering valuable support for preoperative stratification of HNSCC patients. The habitat subregions identified in this study include three areas: region 3, tumor enhancement zone; region 2, tumor necrotic zone; and region 1, transitional zone between region 2 and 3. Among them, region 3 played the most crucial role in model construction and classification. This subregion, corresponding to the area with the most pronounced tumor enhancement on macroscopic appearance, likely reflects increased blood perfusion due to neovascularization, which has been proven to be strongly connected to early metastasis and high invasiveness [30,31,32,33]. Therefore, habitat radiomics achieved a more detailed and precise description of tumor heterogeneity for detecting occult LNM in HNSCC by clustering tumor into subregions.

Both intratumoral and peritumoral radiomics models demonstrated promising performance in predicting occult LNM, with the peritumoral model using 1 mm expansion showing particular efficacy. Some studies suggest 3 mm as the optimal peritumoral distance for cancer research [14, 34]. Considering that HNSCC does not have as large a tumor and peritumoral region as hepatocellular carcinoma or glioma, an excessively large peritumoral region may contain excessive normal tissue. Our preliminary comparative analysis revealed superior predictive performance at 1 mm (vs. 2–3 mm), suggesting this distance optimally captures microenvironmental features relevant to occult LNM development in HNSCC while minimizing extraneous tissue interference [35, 36]. The RF classifier outperformed both LR and SVM classifiers in predicting occult LNM. As an ensemble model, RF classifier improves model stability and robustness by constructing multiple decision trees and combining their outcomes through voting or averaging, especially for high-dimensional and complex datasets like radiomics [33, 37]. In contrast, SVM classifier is more dependent on parameter selection and kernel functions, and an increase in model complexity can lead to overfitting [38]. While LR classifier is simple and interpretable, it struggles with modeling nonlinear relationships [39]. Moreover, our findings displayed that maximum tumor diameter and histological grade were significant predictors for occult LNM, in line with findings from other studies [40, 41]. However, when further integrated into the hybrid model, they provided no additional improvement in model performance.

In the tumor microenvironment, CD8 + T cells serve as key effector cells in antitumor immune responses, eliminating tumor cells through mechanisms such as the perforin/granzyme pathway, Fas/FasL pathway. However, as the tumor microenvironment undergoes dynamic changes, CD8 + T cells may transition into an exhausted phenotype under persistent stimulation from inflammatory or immunosuppressive factors, leading to impaired immune surveillance and fostering early tumor metastasis and progression. Studies have shown that exhaustion-associated CD8 + T cells typically exhibit a range of functional impairments, including reduced cytotoxicity, diminished proliferative capacity, decreased ability to secrete cytokines, and sustained high expression of immune checkpoint molecules [42, 43]. In this study, we conducted bulk and single-cell RNA sequencing analyses to further investigate the underlying biological associations of the RF-habitat radiomics model. The results revealed that patients in the high-risk group exhibited an immunosuppressive microenvironment, primarily characterized by exhaustion-associated CD8_CXCL13 cells, which may represent the potential biological basis for the predictive capability of the RF-habitat radiomics model. Notably, the accumulation of exhaustion-associated CD8 + T cells in high-risk tumors results from two mechanisms: tumor microenvironment-induced T cell exhaustion or recruitment of immunosuppressive cells. Strikingly, radiomic features—including intensity, texture, and wavelet-derived parameters—can effectively captured these immune cell activity patterns [44, 45], providing crucial evidence for understanding the molecular mechanisms related to occult LNM. Furthermore, these findings open new research avenues for developing novel immunotherapeutic strategies for patients with HNSCC.

This study has some limitations. Firstly, one key concern was that its retrospective nature may inherently result in biases. Therefore, a multicenter prospective study is crucial for verifying the applicability of our models. Secondly, validation of the correlation between habitat subregions and pathological results was needed. However, due to the study’s retrospective design, such correlations are technically challenging to achieve. Finally, the sample size of genomic set used in this study was limited. A larger sample size in genomic analysis may be more convincing.

Conclusion

The RF-habitat radiomics model built by venous-phase CT images demonstrated excellent performance in predicting occult LNM in HNSCC patients. Notably, biological analysis showed the RF-habitat radiomics model was connected to the infiltration of exhaustion-related CD8 + T cells, a finding that may uncover tumor microenvironment characteristics contributing to occult LNM. Further experimental validation in cellular or animal studies is needed to test the biological relevance of our genomic findings.

Data availability

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

AUC:

Area under the curve

cN0:

Clinically LN negative

CI:

Confidence interval

CNV:

Copy number variation

DB:

Davis-Bouldin

DEGs:

Differentially expressed genes

DFS:

Disease-free survival

GO:

Gene ontology

GSEA:

Gene set enrichment analysis

GSVA:

Gene set variation analysis

HNSCC:

Head and neck squamous cell carcinoma

ICCs:

Intra- and interclass correlation coefficients

KEGG:

Kyoto encyclopedia of genes and genomes

LASSO:

Least absolute shrinkage and selection operator

LR:

Logistic regression

LSCC:

Laryngeal squamous cell carcinoma

LNM:

Lymph node metastasis

mIF:

Multiplex immunofluorescence

OR:

Odds ratio

OS:

Overall survival

pN-:

Pathological LN negative

pN +:

Pathological LN positive

RF:

Random forest

ROC:

Receiver operating characteristic

ROI:

Region of interest

SHAP:

SHapley Additive exPlanations

SVM:

Support vector machine

TCIA:

Cancer imaging archive

TCGA:

Cancer genome atlas

UMAP:

Uniform manifold approximation and projection

VOI:

Volumes of interest

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424. https://doiorg.publicaciones.saludcastillayleon.es/10.3322/caac.21492.

    Article  PubMed  Google Scholar 

  2. Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE, Grandis JR. Head and neck squamous cell carcinoma. Nat Reviews Disease Primers. 2020;6(1):92. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41572-020-00224-3.

    Article  PubMed  Google Scholar 

  3. Sun P, Zhang Y, Li K, Wang C, Zeng F, Zhu J, et al. Image-guided surgery of head and neck carcinoma in rabbit models by intra-operatively defining tumour-infiltrated margins and metastatic lymph nodes. EBioMedicine. 2019;50:93–102. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ebiom.2019.10.055.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Bluemel C, Rubello D, Colletti PM, de Bree R, Herrmann K. Sentinel lymph node biopsy in oral and oropharyngeal squamous cell carcinoma: current status and unresolved challenges. Eur J Nucl Med Mol Imaging. 2015;42(9):1469–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00259-015-3049-y.

    Article  PubMed  Google Scholar 

  5. Redaelli de Zinis LO, Nicolai P, Tomenzoli D, Ghizzardi D, Trimarchi M, Cappiello J, et al. The distribution of lymph node metastases in supraglottic squamous cell carcinoma: therapeutic implications. Head Neck. 2002;24(10):913–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/hed.10152.

    Article  PubMed  Google Scholar 

  6. Ambrosch P, Kron M, Pradier O, Steiner W. Efficacy of selective neck dissection: a review of 503 cases of elective and therapeutic treatment of the neck in squamous cell carcinoma of the upper aerodigestive tract. Otolaryngol Head Neck Surg. 2001;124(2):180–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1067/mhn.2001.111598.

    Article  CAS  PubMed  Google Scholar 

  7. Bhargava P, Rahman S, Wendt J. Atlas of confounding factors in head and neck PET/CT imaging. Clin Nucl Med. 2011;36(5):e20–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/RLU.0b013e318212c872.

    Article  PubMed  Google Scholar 

  8. Ingruber J, Savic D, Steinbichler TB, Sprung S, Fleischer F, Glueckert R, et al. KLF4, slug and EMT in head and neck squamous cell carcinoma. Cells. 2021;10(3). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells10030539.

  9. Hedberg ML, Goh G, Chiosea SI, Bauman JE, Freilino ML, Zeng Y, et al. Genetic landscape of metastatic and recurrent head and neck squamous cell carcinoma. J Clin Invest. 2016;126(4):1606. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI86862.

    Article  PubMed  PubMed Central  Google Scholar 

  10. O’Connor JP, Rose CJ, Waterton JC, Carano RA, Parker GJ, Jackson A. Imaging intratumor heterogeneity: role in therapy response, resistance, and clinical outcome. Clin Cancer Res. 2015;21(2):249–57. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.Ccr-14-0990.

    Article  PubMed  Google Scholar 

  11. Verma R, Correa R, Hill VB, Statsevych V, Bera K, Beig N, et al. Tumor Habitat-derived radiomic features at pretreatment MRI that are prognostic for Progression-free survival in glioblastoma are associated with key morphologic attributes at histopathologic examination: A feasibility study. Radiol Artif Intell. 2020;2(6):e190168. https://doiorg.publicaciones.saludcastillayleon.es/10.1148/ryai.2020190168.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Huang H, Chen H, Zheng D, Chen C, Wang Y, Xu L, et al. Habitat-based radiomics analysis for evaluating immediate response in colorectal cancer lung metastases treated by radiofrequency ablation. Cancer Imaging. 2024;24(1):44. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40644-024-00692-w.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Kim M, Park JE, Kim HS, Kim N, Park SY, Kim YH, et al. Spatiotemporal habitats from multiparametric physiologic MRI distinguish tumor progression from treatment-related change in post-treatment glioblastoma. Eur Radiol. 2021;31(8):6374–83. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00330-021-07718-y.

    Article  PubMed  Google Scholar 

  14. Lin P, Xie W, Li Y, Zhang C, Wu H, Wan H, et al. Intratumoral and peritumoral radiomics of MRIs predicts pathologic complete response to neoadjuvant chemoimmunotherapy in patients with head and neck squamous cell carcinoma. J Immunother Cancer. 2024;12(11). https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2024-009616.

  15. Wang W, Liang H, Zhang Z, Xu C, Wei D, Li W, et al. Comparing three-dimensional and two-dimensional deep-learning, radiomics, and fusion models for predicting occult lymph node metastasis in laryngeal squamous cell carcinoma based on CT imaging: a multicentre, retrospective, diagnostic study. EClinicalMedicine. 2024;67:102385. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.eclinm.2023.102385.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Zuley ML, Jarosz R, Kirk S, Lee Y, Colen R, Garcia K et al. The Cancer genome atlas Head-Neck squamous cell carcinoma collection (TCGA-HNSC) (Version 6). 2016. The Cancer Imaging Archive. https://doiorg.publicaciones.saludcastillayleon.es/10.7937/K9/TCIA.2016.LXKQ47MS

  17. Wang Y, Dai A, Wen Y, Sun M, Gao J, Yin Z, et al. Prediction of High-risk capsule characteristics for recurrence of pleomorphic adenoma in the Parotid gland based on habitat imaging and peritumoral radiomics: A Two-center study. Acad Radiol. 2025;S1076. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.acra.2025.01.030. -6332(25)00085– 6.

  18. Huang Z, Tu X, Yu T, Zhan Z, Lin Q, Huang X. Peritumoural MRI radiomics signature of brain metastases can predict epidermal growth factor receptor mutation status in lung adenocarcinoma. Clin Radiol. 2024;79(2):e305–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.crad.2023.10.022.

    Article  CAS  PubMed  Google Scholar 

  19. Qi TH, Hian OH, Kumaran AM, Tan TJ, Cong TRY, Su-Xin GL, et al. Multi-center evaluation of artificial intelligent imaging and clinical models for predicting neoadjuvant chemotherapy response in breast cancer. Breast Cancer Res Treat. 2022;193(1):121–38. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10549-022-06521-7.

    Article  CAS  PubMed  Google Scholar 

  20. Davies DL, Bouldin DWA, Cluster Separation, Measure. IEEE Transactions on Pattern Analysis and Machine Intelligence. 1979;PAMI-1(2):224–7; https://doiorg.publicaciones.saludcastillayleon.es/10.1109/TPAMI.1979.4766909

  21. Du Y, Lin Y, Gan L, Wang S, Chen S, Li C, et al. Potential crosstalk between SPP1 + TAMs and CD8 + exhausted T cells promotes an immunosuppressive environment in gastric metastatic cancer. J Transl Med. 2024;22(1):158. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-023-04688-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Quah HS, Cao EY, Suteja L, Li CH, Leong HS, Chong FT, et al. Single cell analysis in head and neck cancer reveals potential immune evasion mechanisms during early metastasis. Nat Commun. 2023;14(1):1680. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-023-37379-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhang J, Liu Y, Xia L, Zhen J, Gao J, Atsushi T. Constructing heterogeneous single-cell landscape and identifying microenvironment molecular characteristics of primary and lymphatic metastatic head and neck squamous cell carcinoma. Comput Biol Med. 2023;165:107459. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.compbiomed.2023.107459.

    Article  CAS  PubMed  Google Scholar 

  24. Zhang W, Liu J, Jin W, Li R, Xie X, Zhao W, et al. Radiomics from dual-energy CT-derived iodine maps predict lymph node metastasis in head and neck squamous cell carcinoma. Radiol Med. 2024;129(2):252–67. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11547-023-01750-2.

    Article  PubMed  Google Scholar 

  25. Wei J, Yang G, Hao X, Gu D, Tan Y, Wang X, et al. A multi-sequence and habitat-based MRI radiomics signature for preoperative prediction of MGMT promoter methylation in Astrocytomas with prognostic implication. Eur Radiol. 2019;29(2):877–88. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00330-018-5575-z.

    Article  PubMed  Google Scholar 

  26. Lambin P, Rios-Velazquez E, Leijenaar R, Carvalho S, van Stiphout RG, Granton P, et al. Radiomics: extracting more information from medical images using advanced feature analysis. Eur J Cancer. 2012;48(4):441–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejca.2011.11.036.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Hansen S, Kuttner S, Kampffmeyer M, Markussen T-V, Sundset R, Øen SK, et al. Unsupervised supervoxel-based lung tumor segmentation across patient scans in hybrid PET/MRI. Expert Syst Appl. 2021;167:114244. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.eswa.2020.114244.

    Article  Google Scholar 

  28. Sinaga KP, Yang MS. Unsupervised K-Means clustering algorithm. IEEE Access. 2020;8:80716–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1109/ACCESS.2020.2988796.

    Article  Google Scholar 

  29. Zhu Y, Zheng D, Xu S, Chen J, Wen L, Zhang Z, et al. Intratumoral habitat radiomics based on magnetic resonance imaging for preoperative prediction treatment response to neoadjuvant chemotherapy in nasopharyngeal carcinoma. Jpn J Radiol. 2024;42(12):1413–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11604-024-01639-8.

    Article  PubMed  Google Scholar 

  30. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407(6801):249–57. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/35025220.

    Article  CAS  PubMed  Google Scholar 

  31. Jansen JF, Koutcher JA, Shukla-Dave A. Non-invasive imaging of angiogenesis in head and neck squamous cell carcinoma. Angiogenesis. 2010;13(2):149–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10456-010-9167-z.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Young MR, Wright MA, Lozano Y, Prechel MM, Benefield J, Leonetti JP, et al. Increased recurrence and metastasis in patients whose primary head and neck squamous cell carcinomas secreted granulocyte-macrophage colony-stimulating factor and contained CD34 + natural suppressor cells. Int J Cancer. 1997;74(1):69–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/(sici)1097-0215(19970220)74:1%3C69::aid-ijc12%3E3.0.co;2-d.

    Article  CAS  PubMed  Google Scholar 

  33. Paul A, Mukherjee DP, Das P, Gangopadhyay A, Chintha AR, Kundu S. Improved random forest for classification. IEEE Trans Image Process. 2018;27(8):4012–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1109/tip.2018.2834830.

    Article  PubMed  Google Scholar 

  34. Kim S, Shin J, Kim DY, Choi GH, Kim MJ, Choi JY. Radiomics on Gadoxetic Acid-Enhanced magnetic resonance imaging for prediction of postoperative early and late recurrence of single hepatocellular carcinoma. Clin Cancer Res. 2019;25(13):3847–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/1078-0432.CCR-18-2861.

    Article  CAS  PubMed  Google Scholar 

  35. Xu S, Han C, Zhou J, Yang D, Dong H, Zhang Y, et al. Distinct maturity and Spatial distribution of tertiary lymphoid structures in head and neck squamous cell carcinoma: implications for tumor immunity and clinical outcomes. Cancer Immunol Immunother. 2025;74(3):107. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00262-025-03952-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Liang H, Zhang Z, Guan Z, Zheng S, Lou J, Liu W, et al. Follicle-like tertiary lymphoid structures: A potential biomarker for prognosis and immunotherapy response in patients with laryngeal squamous cell carcinoma. Front Immunol. 2023;14:1096220. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1096220.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yao J, Jia X, Zhou W, Zhu Y, Chen X, Zhan W, et al. Predicting axillary response to neoadjuvant chemotherapy using peritumoral and intratumoral ultrasound radiomics in breast cancer subtypes. iScience. 2024;27(9):110716. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.isci.2024.110716.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chao CF, Horng MH. The construction of support vector machine classifier using the firefly algorithm. Comput Intell Neurosci. 2015;2015:212719. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2015/212719.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Uddin S, Khan A, Hossain ME, Moni MA. Comparing different supervised machine learning algorithms for disease prediction. BMC Med Inf Decis Mak. 2019;19(1):281. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12911-019-1004-8.

    Article  Google Scholar 

  40. Kane SV, Gupta M, Kakade AC. Depth of invasion is the most significant histological predictor of subclinical cervical lymph node metastasis in early squamous carcinomas of the oral cavity. Eur J Surg Oncol. 2006;32(7):795–803. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejso.2006.05.004.

    Article  CAS  PubMed  Google Scholar 

  41. Zhang Y, Yu J, Zhu H. Risk factors for cervical lymph node metastasis of head and neck squamous cell carcinoma: A retrospective population-based study. J Stomatol Oral Maxillofac Surg. 2022;123(5):e484–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jormas.2022.05.001.

    Article  PubMed  Google Scholar 

  42. Lin Y, Song Y, Zhang Y, Li X, Kan L, Han S. New insights on anti-tumor immunity of CD8+ T cells: cancer stem cells, tumor immune microenvironment and immunotherapy. J Transl Med. 2025;23(1):341. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06291-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12(6):492–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ni.2035.

    Article  CAS  PubMed  Google Scholar 

  44. Huang YH, Shi ZY, Zhu T, Zhou TH, Li Y, Li W, et al. Longitudinal MRI-Driven Multi-Modality approach for predicting pathological complete response and B cell infiltration in breast Cancer. Adv Sci (Weinh). 2025;e2413702. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/advs.202413702.

  45. Feng Z, Li H, Liu Q, Duan J, Zhou W, Yu X, et al. CT radiomics to predict Macrotrabecular-Massive subtype and immune status in hepatocellular carcinoma. Radiology. 2023;307(1):e221291. https://doiorg.publicaciones.saludcastillayleon.es/10.1148/radiol.221291.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Figure 2 was partly generated using Servier Medical Art, provided by Servier (http://smart.servier.com).

Funding

This work was supported by the projects of the National Natural Science Foundation of China (Grant No. 82373040 & 82103145).

Author information

Authors and Affiliations

Authors

Contributions

C.X.W, H.G.H, P.J, P.M and L.Y.S designed the project; J.H, L.L and L.Y.X collected the data; J.H, F.C.M and C.X.W analyzed the data; C.X.W drafted the manuscript; H.G.H, C.L and P.J revised the manuscript. All authors read and approved the manuscript.

Corresponding authors

Correspondence to Juan Peng or Guohua Hu.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the Institutional Review Boards of The First Affiliated Hospital of Chongqing Medical University (Date: September 15, 2021/No: 2021–433). The requirement for written informed consent was waived for the retrospective data sets, while written informed consent was obtained for single-cell sequencing set.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, X., Jiang, H., Pan, M. et al. Habitat radiomics predicts occult lymph node metastasis and uncovers immune microenvironment of head and neck cancer. J Transl Med 23, 498 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06474-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06474-7

Keywords