Skip to main content

Predictive value of radiomic features extracted from primary lung adenocarcinoma in forecasting thoracic lymph node metastasis: a systematic review and meta-analysis

Abstract

Background

The application of radiomics in thoracic lymph node metastasis (LNM) of lung adenocarcinoma is increasing, but diagnostic performance of radiomics from primary tumor to predict LNM has not been systematically reviewed. Therefore, this study sought to provide a general overview regarding the methodological quality and diagnostic performance of using radiomic approaches to predict the likelihood of LNM in lung adenocarcinoma.

Methods

Studies were gathered from literature databases such as PubMed, Embase, the Web of Science Core Collection, and the Cochrane library. The Radiomic Quality Score (RQS) and the Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) were both used to assess the quality of each study. The pooled sensitivity, specificity, and area under the curve (AUC) of the best radiomics models in the training and validation cohorts were calculated. Subgroup and meta-regression analyses were also conducted.

Results

Seventeen studies with 159 to 1202 patients each were enrolled between the years of 2018 to 2022, of which ten studies had sufficient data for the quantitative evaluation. The percentage of RQS was between 11.1% and 44.4% and most of the studies were considered to have a low risk of bias and few applicability concerns in QUADAS-2. Pyradiomics and logistic regression analysis were the most commonly used software and methods for radiomics feature extraction and selection, respectively. In addition, the best prediction models in seventeen studies were mainly based on radiomics features combined with non-radiomics features (semantic features and/or clinical features). The pooled sensitivity, specificity, and AUC of the training cohorts were 0.84 (95% confidence interval (CI) [0.73–0.91]), 0.88 (95% CI [0.81–0.93]), and 0.93(95% CI [0.90–0.95]), respectively. For the validation cohorts, the pooled sensitivity, specificity, and AUC were 0.89 (95% CI [0.82–0.94]), 0.86 (95% CI [0.74–0.93]) and 0.94 (95% CI [0.91–0.96]), respectively.

Conclusions

Radiomic features based on the primary tumor have the potential to predict preoperative LNM of lung adenocarcinoma. However, radiomics workflow needs to be standardized to better promote the applicability of radiomics.

Trial registration

CRD42022375712.

Peer Review reports

Introduction

Lung cancer is currently the second most common cancer in incidence and the leading cause of cancer-related mortality in the world [1]. Adenocarcinoma is the most common histological subtype [2] and lymph node metastasis (LNM) is the main mode of cancer metastasis. Accurate preoperative prediction of LNM is of great significance in the treatment and prognosis prediction of adenocarcinoma [3]. Currently, diagnostic methods are classified as either invasive or non-invasive. Invasive procedures such as mediastinoscopic biopsy, ultrasound-guided bronchial needle aspiration or lymph node sampling, which will carry risks of postoperative complications to the patient [4, 5]. Non-invasive measures on the other hand are commonly the next best test of choice. Radiological studies like computed tomography (CT), magnetic resonance imaging (MRI) and positron emission tomography/computed tomography (PET/CT), have all demonstrated potential diagnostic efficacy in identifying LNM [6, 7]. Yet, false negative and false positive judgments may be occurred on CT and PET/CT due to some clinical and radiological factors, such as micrometastasis or inflammatory hyperplasia [8, 9]. While MRI is non-radiation and can offers apparent diffusion coefficient characteristics, motion artifacts would limit its assessment in tumor heterogeneity [7, 10].

To improve the efficacy of diagnosis, many studies have relied on radiomics to predict LNM of non-small cell lung cancer [11,12,13]. Radiomics is a non-invasive technique which can be applied to traditional imaging modalities to extract and quantify radiomic features [14]. Recently, radiomics has already been applied for the identification of malignancy [15] and histological subtypes [16], prediction of gene expression [17], and assessment of treatment response in lung cancer [18]. Radiomic features can be extracted from different regions of interest (ROIs) such as the intratumoral and/or peritumoral areas [19,20,21,22]. For example, Das SK et al. improved the performance of predicting cT1N0M0 lung adenocarcinoma by combining features of the intratumor region, the peritumoral region and lymph node [23].

With radiomic approaches becoming more common in medical research, it was hypothesized that radiomic features of primary tumor would be instrumental in predicting the possibility of LNM in lung adenocarcinoma. Therefore, the purpose of this review was to provide a general overview of the methodological quality and evaluate diagnostic performance in radiomics for the prediction of LNM in lung adenocarcinoma.

Methods

This systematic review and meta-analysis was reported in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses for Diagnostic Test Accuracy (PRISMA-DTA) guidelines (Additional file 1: Table S1) and was registered on PROSPERO database for systematic reviews (CRD42022375712) [24].

Database search strategy

A comprehensive search of PubMed, Embase, the Web of Science Core Collection and the Cochrane library was conducted until November 16, 2022. Search terms such as “lung adenocarcinoma”, “machine learning”, “radiomics”, and “lymph node metastasis” were included. The detailed search strategy was described in Table S2 (Additional file 1). No language or publication date restrictions were placed on the initial database search.

Study selection

Studies were selected if they met all inclusion criteria: (1) patients with lung adenocarcinoma confirmed by pathology; (2) articles based on CT/MRI/PET-CT radiomics to evaluate the likelihood of preoperative LNM; (3) the ROI for segmentation contained the primary tumor; (4) articles were published in English. Studies were excluded if they met any of the following exclusion criteria: (1) case studies, editorials, letters, review articles and conference abstracts; (2) studies not in the field of interest.

Data extraction

Two independent investigators firstly extracted the following information from each selected study: (1) study details: first author, publication year, country of origin, study design; (2) patient details: the source of data acquisition, criteria for lymph node staging, diameter and density of primary tumor, diagnostic method of LNM, number of patients and negative/positive LNM in the training/internal validation/external validation cohort, clinical stage; (3) imaging details: imaging modality; (4) radiomic details: segmentation method and software, ROI, radiomic feature extraction software and method, number of radiomic features extracted, type of radiomic features extracted, type of models constructed, the best performance model, number of radiomic/non-radiomic features included in the best performance model; (5)diagnostic performance: sensitivity, specificity and area under the curve (AUC)/concordance index (C-index) of the prediction models.

If more than one predictive model was included in a study, the radiomics model with the highest AUC/C-index in the training and validation cohort was included in the quantitative evaluation, respectively [25, 26]. If an internal validation cohort and an external validation cohort were included in a study, we included data from both cohorts.

Risk of bias assessment

The Radiomic Quality Score (RQS) [27] was used to evaluate the procedural validity of each study (Additional file 1: Table S3). The RQS provided rigorous evaluation criteria and reporting guidelines for radiomic studies [27]. The total score ranged from -8 to 36, and sixteen items are assigned corresponding scores [27]. The Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) [28] was used to determine the risk of bias and the applicability of each included study (Additional file 1: Table S4). The QUADAS-2 tools was first divided into two broad categories: the risk of bias and the applicability concerns [28]. The former included features such as patient selection, index test, reference standard, flow and timing [28]. The latter examined similar parameters with patient selection, index test and reference standard [28]. Based on basic answers of "yes", "no", or "unclear" for each item, the level was rated as "low", "high", or "unclear" [28]. The RQS and QUADAS-2 were used to evaluate the quality of the literature independently by two authors. Discrepancies were rediscussed and evaluated to reach a consensus.

Statistical analysis

Firstly, we extracted sample size, sensitivity, and specificity of the best radiomics models in the training and validation cohorts from the studies. Then the number of true positives, false positives, false negatives, and true negatives were calculated by Review Manager 5.4.

Quantitative evaluation was performed using the midas command in Stata 17.0 software. Pooled sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and AUC were calculated, and summary receiver operating characteristic curve (SROC) was created. Heterogeneity was assessed using Cochrane Q-test (two-sides p < 0.05 was considered statistically significant) and I2 statistic (I2 values of 25%, 50% and 75% represent low, moderate and high heterogeneity, respectively) from forest plots [29]. Spearman rank coefficients was performed to determine whether there was heterogeneity caused by threshold effect. The sources of heterogeneity were further analyzed by subgroup and univariate meta-regression analyses.

Results

Literature search and extraction

A total of 7087 studies were obtained by the search strategy of which 1959 remained after removing duplicates. After, 5034 articles did not meet the inclusion criteria based on title and abstract and 94 studies were examined in full text. Among them, 42 studies were not related to radiomics, 34 studies covered patients beyond lung adenocarcinoma, and the imaging modality of 1 study was not of interest (ultrasound). Finally, this systematic review involved 17 studies containing a total of 7,117 patients [23, 30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45]. Seven studies [30, 31, 35, 37,38,39, 44] were excluded due to lack of sufficient data, and 10 studies [23, 32,33,34, 36, 40,41,42,43, 45] were included in the meta-analysis. Figure 1 illustrates the PRISMA flow chart for the included studies in this review.

Fig. 1
figure 1

Flowchart of the study screening and selection process

Patient and study characteristics

Table 1 presents the basic characteristics for all 17 retrospective studies which were published between 2018 and 2022 [23, 30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45]. Most of the studies (14/17, 82.4%) were derived from one center [30,31,32,33,34,35,36,37,38,39, 41, 42, 44, 45]. And almost all of the studies (16/17, 94.1%) were from China [23, 30, 32,33,34,35,36,37,38,39,40,41,42,43,44,45], except for one from the United States [31]. The included studies (11/17, 64.7%) [23, 31,32,33,34, 36, 37, 39,40,41, 44] usually used the 8th edition of tumor-node-metastasis staging system as the standard for lymph node staging [46].

Table 1 Characteristics of included studies

All studies relied on surgical resection for the diagnosis of LNM. One study also included lymph node sampling [43], and one study included CT follow-up validation [44]. The number of patients included ranged from 159 to 1202. Eleven studies (11/17, 64.7%) [23, 32, 34, 35, 37, 38, 41,42,43,44,45] had internal validation cohorts and eight studies [23, 30, 38,39,40, 43,44,45] had external validation cohorts. Eight studies selected patients with clinical stage N0 at enrollment [23, 30, 31, 33, 35, 36, 39, 40].

Radiomics workflow

CT was the primary imaging modality in 13 studies [23, 30,31,32,33,34,35,36,37,38, 40, 42, 45]. In addition, 18F-PET/CT was used in five studies [36, 39, 41, 43, 44]. The ROIs were manually segmented in 11 studies [23, 30, 33, 34, 36,37,38, 40, 42, 44, 45], semi-automatically in five studies [31, 35, 39, 41, 43] and fully automatically in one study [32] (Table 2). There were eight types of ROI segmentation software, among which the most frequently used was ITK-SNAP [23, 37, 41, 44, 45]. All studies included primary tumors in their ROI segmentation [23, 30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45].

Table 2 Radiomics workflow for the included studies

A total of seven different software was applied for the extraction of radiomic features in each study, among which Pyradiomics was the most used [32, 34, 35, 37, 38, 40, 45] (Table 2). The common methods of radiomic feature selection were logistic regression analysis [23, 30,31,32, 36, 39, 40, 42, 44, 45] and least absolute shrinkage and selection operator method [23, 32, 34, 35, 39,40,41,42,43,44]. The number of radiomics features included ranged from 1 to 32 in each of the best models, except for one study in which the best model included only semantic features without radiomic features [36]. The types of prediction models constructed ranged from 1 to 7, and most of the best models (15/17, 88.2%) were models that combined radiomic and non-radiomic features (semantic features and/or clinical features) (Additional file 1: Table S5) [23, 30,31,32, 34, 35, 37,38,39,40,41,42,43,44,45].

Quality assessment

The overall RQS and percent RQS for each study are presented in Table 3 and Fig. 2, along with the scores for the individual components. The median RQS total scores was 14 (range 4 – 16) and 38.9% (range 11.1% – 44.4%). Most studies (8/17, 47.1%) had RQS scores between 30% and 40% (Fig. 2a). No study scored in the four items of “Cost-effectiveness analysis”, “Prospective study” “Biological correlates” and “Imaging at multiple time points” (Fig. 2b).

Table 3 Radiomic quality scores for all included studies
Fig. 2
figure 2

Qualitative quality assessment evaluated through the Radiomics Quality Score (RQS) tool. a Proportion of studies with different RQS percentage score. b Percentage of the 16 components of the included studies with different scores in the RQS

The distribution of the QUADAS-2 scores for each included study was shown in Table S6 (Additional file 1) and Fig. 3. The risk of bias in patient selection was low in 13 studies and unclear in 4 studies. The risk of bias for the index test was low in 10 studies and unclear in 7 studies. The risk of bias for the reference standard test was low in 17 studies. The risk of bias for flow and timing was low in 14 studies, unclear in 2 studies, and high in 1 study. Most studies were assessed as having a low risk of bias and minimal concerns regarding applicability.

Fig. 3
figure 3

The percentage of the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) scoring criteria

Data analysis

Diagnostic performance

The diagnostic efficacy of each study will be presented in Table S7-S9 (Additional file 1:). Ten studies were included in this meta-analysis, in which the pooled sensitivity, specificity, PLR, NLR, DOR and AUC in the training cohorts were 0.84 (95% CI [0.73–0.91]), 0.88 (95% CI [0.81–0.93]), 7.0 (95% CI [4.5–11.0]), 0.18 (95% CI [0.11–0.31]), 39 (95% CI [19–78]), 0.93 (95% CI [0.90–0.95]), respectively. Meanwhile, three studies did not evaluate the diagnostic performance of the validation cohorts due to the lack of validation cohorts [33, 34, 36]. The pooled sensitivity, specificity, PLR, NLR and DOR of 11 internal and external validation cohorts from 7 studies were 0.89 (95%CI [0.82–0.94]), 0.86 (95% CI [0.74–0.93]), 6.3 (95% CI [3.4–11.8]), 0.12 (95% CI [0.08–0.20]), 52 (95% CI [27–97]), 0.94 (95% CI [0.91–0.96]), respectively. Figure 4 and Fig. 5 show the forest plots and SROC plots for the training and validation cohorts, respectively. High heterogeneity was observed in the sensitivity and specificity of the training cohorts (p ≤ 0.01, I2 = 89.98; p ≤ 0.01, I2 = 92.84). Since only seven studies involved the validation cohorts, we mainly explored the sources of heterogeneity of the ten studies for the training cohorts. Spearman correlation coefficient was -0.45 (p = 0.17), indicating that heterogeneity due to threshold effects may be low.

Fig. 4
figure 4

Coupled Forest plots of pooled sensitivity and specificity. a The training cohorts. b The validation cohorts. (internal: an internal validation cohort; external: an external validation cohort)

Fig. 5
figure 5

Summary receiver operating characteristic curves (SROC) of the diagnostic performance. a The training cohorts. b The validation cohorts

Investigation of heterogeneity

Subgroup analysis was performed on the training cohorts of 10 studies, mainly including the following categories: (1) imaging modality: CT, PET/CT; (2) clinical stage: clinical N0, others; (3) sample size: ≤ 300, > 300; (4) primary tumor diameter: ≤ 30 mm, others; (5) segmentation method: manual, semi-automated/automated; (6) ROI: only primary tumor, including peritumoral/lymph node region; (7) radiomic software: Pyradiomics, others. From Table 4, radiomic features based on primary tumor showed high diagnostic performance in predicting LNM of lung adenocarcinoma in all subgroups. Univariable meta-regression analysis further performed, which showed that primary tumor diameter (p < 0.01) was a possible source of heterogeneity in sensitivity. Imaging modalities (p < 0.001), sample size (p < 0.05), and radiomics software (p < 0.05) were possible sources of heterogeneity in terms of specificity (Fig. 6).

Table 4 Diagnostic performance of subgroup analysis
Fig. 6
figure 6

Univariable Meta-regression analysis plot to investigate sources of heterogeneity. (Small Sample Size: sample sizes ≤ 300; Diameter: primary tumor diameter ≤ 30 mm)

Discussion

This study revealed that radiomic features extracted from the primary tumor have the potential to predict preoperative LNM in lung adenocarcinoma. The QUADAS-2 and RQS tools were applied to assess the risk of bias and the quality of the radiomic method. Meta-analysis was used to quantitatively evaluate the diagnostic performance of the best radiomics models. Obviously, the radiomics models achieved satisfactory diagnostic performance in both the training and validation cohorts. However, the low methodological quality of the systematic review and the high heterogeneity of the quantitative meta-analysis suggest that radiomics models still need to be further improved to better assist the clinical practice.

The clinical diagnosis of positive LNM is usually based on imaging findings (e.g., short axis diameter of lymph nodes > 10 mm on CT, maximum standardized uptake value ≥ 2.5 on PET/CT). However, the subjective factors of manual identification and the limits of the naked eye are highly likely to induce unwanted bias, such as occult LNM [8, 9, 47, 48]. Radiomics can directly extract features from the ROIs of macroscopic images (such as primary tumor, peritumoral area, etc.) for quantitative analysis in a high-throughput manner [49]. In this review, radiomics studies based on the primary tumor were included. Based on the characteristics of the primary tumor, the severity of tumor hypoxia and angiogenic effects of the primary lesion can be identified to evaluate tumor heterogeneity [50]. Cancerous cells within the primary tumor can proliferate by generating new lymphatic vessels in a variety of ways [51] or they can metastasize to the mediastinum through abundant subpleural drainage [37, 52].

The RQS was able to assess the quality of the radiomic methods; however, the best score achieved in the included studies was 16 (44.4%) [23, 40, 41, 43]. The reason for this result was that 17 studies had a low score in each item of the RQS, which meant that there was a lack of standardized workflow for radiomics research (Table 3). In terms of imaging, all studies documented good image protocol quality and multiple segmentations. However, few studies explored the differences between various scanners and provided open data sources, which will lead to low reproducibility of radiomics research. The choice of ROI segmentation method also had a certain effect. The accuracy of manual segmentation is high, but it is limited by time consumption and inter-reader variation. In one study, radiomic features were not included in the best prediction model, likely because only three independent features were selected for analysis due to the small sample size [36]. Skewness was incorporated as a radiomics feature in the best prediction models of 5 studies [30, 34, 35, 38, 43], and one study found that the skewness of lymph node positive lesions was significantly lower than that of negative lesions [30]. Meanwhile, the biological validation of models can facilitate the clinical translation of radiomics. Although two studies combined genes or proteins [44, 45], neither of them was statistically significant. Finally, multi-center validation is an important key to reduce overfitting and optimize the model. Therefore, future radiomics studies would be better follow standardized workflows, such as obtaining large and high-quality multi-center datasets, ensuring consistent image acquisition parameters, developing accurate and reproducible segmentation methods, and correlating with genomics or proteomics.

According to the QUADAS-2 results, most studies were of a low risk and had good applicability, which may be due to the inclusion of appropriate patient groups and the selection of gold standards for reference. However, some studies were unclear about the selection of participants and whether the use of gold standards was made uninformed decisions. Thus, future studies are needed to illustrate the exclusion criteria and procedures for patient selection clearly, as well as whether there is an appropriate time interval between the reference standard and imaging examination.

The high heterogeneity of radiomics models in quantitative evaluation cannot be ignored, although they showed good diagnostic performance. We observed whether the primary tumor was ≤ 30 mm as a possible source of heterogeneity in sensitivity. Tumor diameter was also identified as an important predictor among non-radiomic features in this review (Additional file 1: Table S5) [34, 35, 37, 40, 43]. Similarly, patients with a relatively large primary tumor diameter tend to have a relatively high probability of LNM and poor prognosis [46]. Meanwhile, in terms of specificity, imaging modality, sample size and radiomics software were possible sources of heterogeneity. This review mainly included CT-based radiomics models, and its diagnostic performance compared with other imaging modalities (PET or PET/CT) remains to be studied. One of the included studies compared the performance of radiomic prediction models derived from different imaging modalities (CT, PET, or PET/CT) and showed that PET/CT yielded best results than the other [41]. Larger sample size will allow for a more comprehensive assessment of a radiomics study, and public database could expand the sample size for the study [53]. Different radiomics feature extraction software was used in this review, which led to the heterogeneity in specificity. One study showed that discrepancies were present in seven different radiomics feature extraction software [54]. Therefore, for the differences caused by image acquisition, it is necessary to perform image normalization (such as resampling, etc.) or follow the standardization protocol of image acquisition and reconstruction in further studies [55], which will be of great help to the stability of radiomics feature extraction. In addition, the algorithms and codes of radiomics feature software would be better conform to the image biomarker standardization initiative to improve its reproducibility and verify in multiple cohorts [54].

There were also some limitations in this systematic review. Firstly, almost all the included studies were from China. Therefore, some geographic bias may be present due to the greater prevalence of adenocarcinoma in Asian populations. Secondly, all studies were retrospective, and only three studies used multicenter data. This may lead to selection bias. Third, studies on MRI were not included in this review due to a lack of matching studies. Fourthly, low RQS and high QUADAS-2 results may have some impact on the literature quality assessment. Finally, only 10 of the included articles were used for meta-analysis, and they showed high heterogeneity. Although we found possible sources of heterogeneity, more studies are needed to further explore it in the future.

Conclusions

In conclusion, this review summarized that radiomic features based on the primary tumor have the potential to predict preoperative LNM of lung adenocarcinoma. However, future research needs standardized radiomics workflow such as multi-center and prospective studies to promote the applicability of radiomics.

Availability of data and materials

All data generated or analysed during this study are included in this published article [and its supplementary information files].

Abbreviations

AUC:

Area under the curve

C-index:

Concordance index

CI:

Confidence interval

DOR:

Diagnostic odds ratio

LNM:

Lymph node metastasis

NLR:

Negative likelihood ratio

PLR:

Positive likelihood ratio

PRISMA-DTA:

Preferred Reporting Items for Systematic Reviews and Meta-Analyses for Diagnostic Test Accuracy

QUADAS-2:

Quality Assessment of Diagnostic Accuracy Studies-2

ROI:

Region of interest

RQS:

Radiomics quality score

SROC:

Summary receiver operating characteristic

References

  1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33.

    Article  PubMed  Google Scholar 

  2. Nicholson AG, Tsao MS, Beasley MB, Borczuk AC, Brambilla E, Cooper WA, et al. The 2021 WHO classification of lung tumors: impact of advances since 2015. J Thorac Oncol. 2022;17(3):362–87.

    Article  PubMed  Google Scholar 

  3. Shimada Y, Tsuboi M, Saji H, Miyajima K, Usuda J, Uchida O, et al. The prognostic impact of main bronchial lymph node involvement in non-small cell lung carcinoma: suggestions for a modification of the staging system. Ann Thorac Surg. 2009;88(5):1583–8.

    Article  PubMed  Google Scholar 

  4. Sehgal IS, Dhooria S, Aggarwal AN, Behera D, Agarwal R. Endosonography versus mediastinoscopy in mediastinal staging of lung cancer: systematic review and meta-analysis. Ann Thorac Surg. 2016;102(5):1747–55.

    Article  PubMed  Google Scholar 

  5. Mitjà PS, Dos Santos Carvalho FG, Olivé IG, Santos JS, López JJ, Ares AN, et al. Incidence and Risk Factors for Infectious Complications of EBUS-TBNA: Prospective Multicenter Study. Arch Bronconeumol. 2023;59(2):84–9.

    Article  Google Scholar 

  6. Silvestri GA, Gonzalez AV, Jantz MA, Margolis ML, Gould MK, Tanoue LT, et al. Methods for staging non-small cell lung cancer: Diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2013;143(5 Suppl):e211S–e250S.

    Article  PubMed  Google Scholar 

  7. Peerlings J, Troost EG, Nelemans PJ, Cobben DC, de Boer JC, Hoffmann AL, et al. The diagnostic value of MR imaging in determining the Lymph node status of patients with non-small cell lung cancer: a meta-analysis. Radiology. 2016;281(1):86–98.

    Article  PubMed  Google Scholar 

  8. Takamochi K, Nagai K, Yoshida J, Suzuki K, Ohde Y, Nishimura M, et al. The role of computed tomographic scanning in diagnosing mediastinal node involvement in non-small cell lung cancer. J Thorac Cardiovasc Surg. 2000;119(6):1135–40.

    Article  CAS  PubMed  Google Scholar 

  9. Takamochi K, Yoshida J, Murakami K, Niho S, Ishii G, Nishimura M, et al. Pitfalls in lymph node staging with positron emission tomography in non-small cell lung cancer patients. Lung Cancer. 2005;47(2):235–42.

    Article  PubMed  Google Scholar 

  10. Xu L, Tian J, Liu Y, Li C. Accuracy of diffusion-weighted (DW) MRI with background signal suppression (MR-DWIBS) in diagnosis of mediastinal lymph node metastasis of nonsmall-cell lung cancer (NSCLC). J Magn Reson Imaging. 2014;40(1):200–5.

    Article  PubMed  Google Scholar 

  11. He L, Huang Y, Yan L, Zheng J, Liang C, Liu Z. Radiomics-based predictive risk score: A scoring system for preoperatively predicting risk of lymph node metastasis in patients with resectable non-small cell lung cancer. Chin J Cancer Res. 2019;31(4):641–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Akinci D’Antonoli T, Farchione A, Lenkowicz J, Chiappetta M, Cicchetti G, Martino A, et al. CT radiomics signature of tumor and peritumoral lung Parenchyma to predict nonsmall cell lung cancer postsurgical recurrence risk. Acad Radiol. 2020;27(4):497–507.

    Article  PubMed  Google Scholar 

  13. Chen QL, Li MM, Xue T, Peng H, Shi J, Li YY, et al. Radiomics nomogram integrating intratumoural and peritumoural features to predict lymph node metastasis and prognosis in clinical stage IA non-small cell lung cancer: a two-centre study. Clin Radiol. 2023;78(5):e359–67.

    Article  PubMed  Google Scholar 

  14. Gillies RJ, Kinahan PE, Hricak H. Radiomics: Images Are More than Pictures. They Are Data Radiology. 2016;278(2):563–77.

    PubMed  Google Scholar 

  15. Hawkins S, Wang H, Liu Y, Garcia A, Stringfield O, Krewer H, et al. Predicting Malignant Nodules from Screening CTScans. J Thorac Oncol. 2016;11(12):2120–8.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Zhao W, Xu Y, Yang Z, Sun Y, Li C, Jin L, et al. Development and validation of a radiomics nomogram for identifyinginvasiveness of pulmonary adenocarcinomas appearing as subcentimeter ground-glass opacity nodules. Eur J Radiol. 2019;112:161–8.

    Article  PubMed  Google Scholar 

  17. Rossi G, Barabino E, Fedeli A, Ficarra G, Coco S, Russo A, et al. Radiomic Detection of EGFR Mutations in NSCLC. Cancer Res. 2021;81(3):724–31.

  18. Trebeschi S, Drago SG, Birkbak NJ, Kurilova I, Cǎlin AM, Delli Pizzi A, et al. Predicting response to cancer immunotherapyusing noninvasive radiomic biomarkers. Ann Oncol. 2019;30(6):998–1004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yi L, Peng Z, Chen Z, Tao Y, Lin Z, He A, et al. Identification of pulmonary adenocarcinoma and benign lesions in isolated solid lung nodules based on a nomogram of intranodal and perinodal CT radiomic features. Front Oncol. 2022;12:924055.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Wu L, Gao C, Ye J, Tao J, Wang N, Pang P, et al. The value of various peritumoral radiomic features in differentiating the invasiveness of adenocarcinoma manifesting as ground-glass nodules. Eur Radiol. 2021;31(12):9030–7.

    Article  PubMed  Google Scholar 

  21. Choe J, Lee SM, Kim W, Do KH, Kim S, Choi S, et al. CT radiomics-based prediction of anaplastic lymphoma kinase and epidermal growth factor receptor mutations in lung adenocarcinoma. Eur J Radiol. 2021;139:109710.

    Article  PubMed  Google Scholar 

  22. Khorrami M, Khunger M, Zagouras A, Patil P, Thawani R, Bera K, et al. Combination of Peri- and intratumoral radiomic features on baseline CT scans predicts response to Chemotherapy in lung Adenocarcinoma. Radiol Artif Intell. 2019;1(2):e180012.

    Article  PubMed  Google Scholar 

  23. Das SK, Fang KW, Xu L, Li B, Zhang X, Yang HF. Integrative nomogram of intratumoral, peritumoral, and lymph node radiomic features for prediction of lymph node metastasis in cT1N0M0 lung adenocarcinomas. Sci Rep. 2021;11(1):10829.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. McInnes MDF, Moher D, Thombs BD, McGrath TA, Bossuyt PM, Clifford T, et al. Preferred reporting Items for a systematic review and meta-analysis of diagnostic test accuracy studies: The PRISMA-DTA statement. JAMA. 2018;319(4):388–96.

    Article  PubMed  Google Scholar 

  25. Ursprung S, Beer L, Bruining A, Woitek R, Stewart GD, Gallagher FA, et al. Radiomics of computed tomography and magnetic resonance imaging in renal cell carcinoma-a systematic review and meta-analysis. Eur Radiol. 2020;30(6):3558–66.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Mühlbauer J, Egen L, Kowalewski KF, Grilli M, Walach MT, Westhoff N, et al. Radiomics in Renal cell Carcinoma-A systematic review and meta-analysis. Cancers (Basel). 2021;13(6):1348.

    Article  PubMed  Google Scholar 

  27. Lambin P, Leijenaar RTH, Deist TM, Peerlings J, de Jong EEC, van Timmeren J, et al. Radiomics: the bridge between medical imaging and personalized medicine. Nat Rev Clin Oncol. 2017;14(12):749–62.

    Article  PubMed  Google Scholar 

  28. Whiting PF, Rutjes AW, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, et al. QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med. 2011;155(8):529–36.

    Article  PubMed  Google Scholar 

  29. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ. 2003;327(7414):557–60.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Gu YW, She YL, Xie D, Dai CY, Ren YJ, Fan ZW, et al. A texture analysis-based prediction model for Lymph node metastasis in stage IA lung Adenocarcinoma. Ann Thorac Surg. 2018;106(1):214–20.

    Article  PubMed  Google Scholar 

  31. Liu Y, Kim J, Balagurunathan Y, Hawkins S, Stringfield O, Schabath MB, et al. Prediction of pathological nodal involvement by CT-based Radiomic features of the primary tumor in patients with clinically node-negative peripheral lung adenocarcinomas. Med Phys. 2018;45(6):2518–26.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Yang X, Pan X, Liu H, Gao D, He J, Liang W, et al. A new approach to predict lymph node metastasis in solid lung adenocarcinoma: A radiomics nomogram. J Thorac Dis. 2018;10:S807–19.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Zhong Y, Yuan M, Zhang T, Zhang YD, Li H, Yu TF. Radiomics approach to prediction of occult mediastinal lymph node metastasis of lung adenocarcinoma. Am J Roentgenol. 2018;211(1):109–13.

    Article  Google Scholar 

  34. Wang X, Zhao XY, Li Q, Xia W, Peng ZH, Zhang R, et al. Can peritumoral radiomics increase the efficiency of the prediction for lymph node metastasis in clinical stage T1 lung adenocarcinoma on CT? Eur Radiol. 2019;29(11):6049–58.

    Article  PubMed  Google Scholar 

  35. Yang M, She Y, Deng J, Wang T, Ren Y, Su H, et al. CT-based radiomics signature for the stratification of N2 disease risk in clinical stage i lung adenocarcinoma. Transl Lung Cancer Res. 2019;8(6):876–85.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Zhu H, Xu Y, Liang N, Sun H, Huang Z, Xie S, et al. Assessment of clinical stage IA lung adenocarcinoma with PN1/N2 metastasis using CT quantitative texture analysis. Cancer Manag Res. 2020;12:6421–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Li Q, He XQ, Fan X, Zhu CN, Lv JW, Luo TY. Development and validation of a combined model for preoperative prediction of lymph node metastasis in peripheral lung adenocarcinoma. Front Oncol. 2021;11:675877.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Ran J, Cao R, Cai J, Yu T, Zhao D, Wang Z. Development and validation of a nomogram for preoperative prediction of Lymph Node Metastasis in lung adenocarcinoma based on radiomics signature and deep learning signature. Front Oncol. 2021;11:585942.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Wang L, Li T, Hong J, Zhang M, Ouyang M, Zheng X, et al. 18F-FDG PET-based radiomics model for predicting occult lymph node metastasis in clinical N0 solid lung adenocarcinoma. Quant Imaging Med Surg. 2021;11(1):215–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhang R, Zhang R, Luan T, Liu B, Zhang Y, Xu Y, et al. A radiomics nomogram for preoperative prediction of clinical occult lymph node metastasis in ct1-2n0m0 solid lung adenocarcinoma. Cancer Manag Res. 2021;13:8157–67.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Chang C, Ruan M, Lei B, Yu H, Zhao W, Ge Y, et al. Development of a PET/CT molecular radiomics-clinical model to predict thoracic lymph node metastasis of invasive lung adenocarcinoma ≤ 3 cm in diameter. EJNMMI Res. 2022;12(1):23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chen W, Xu M, Sun Y, Ji C, Chen L, Liu S, et al. Integrative predictive models of computed tomography texture parametersand hematological parameters for lymph node metastasis in lung adenocarcinomas. J Comput Assist Tomogr. 2022;46(2):315–24.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Dai M, Wang N, Zhao X, Zhang J, Zhang Z, Zhang J, et al. Value of presurgical 18F-FDG PET/CT radiomics for predicting mediastinal lymph node metastasis in patients with lung adenocarcinoma. Cancer Biother Radiopharm. 2022.

  44. Lv J, Chen X, Liu X, Du D, Lv W, Lu L, et al. Imbalanced data correction based PET/CT radiomics model for predicting lymph node metastasis in clinical stage T1 lung adenocarcinoma. Front Oncol. 2022;12:788968

  45. Ma X, Xia L, Chen J, Wan W, Zhou W. Development and validation of a deep learning signature for predicting lymph node metastasis in lung adenocarcinoma: comparison with radiomics signature and clinical-semantic model. Eur Radiol. 2023;33(3):1949–62.

    Article  PubMed  Google Scholar 

  46. Goldstraw P, Chansky K, Crowley J, Rami-Porta R, Asamura H, Eberhardt WE, et al. The IASLC lung cancer staging project: proposals for revision of the TNM Stage groupings in the forthcoming (Eighth) edition of the TNM classification for lung cancer. J Thorac Oncol. 2016;11(1):39–51.

    Article  PubMed  Google Scholar 

  47. Shimada Y, Kudo Y, Furumoto H, Imai K, Maehara S, Tanaka T, et al. Computed tomography histogram approach to predict Lymph Node Metastasis in patients with clinical stage IA lung cancer. Ann Thorac Surg. 2019;108(4):1021–8.

    Article  PubMed  Google Scholar 

  48. Miyasaka Y, Suzuki K, Takamochi K, Matsunaga T, Oh S. The maximum standardized uptake value of fluorodeoxyglucose positron emission tomography of the primary tumour is a good predictor of pathological nodal involvement in clinical N0 non-small-cell lung cancer. Eur J Cardiothorac Surg. 2013;44(1):83–7.

    Article  PubMed  Google Scholar 

  49. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Ganeshan B, Goh V, Mandeville HC, Ng QS, Hoskin PJ, Miles KA. Non-small cell lung cancer: histopathologic correlates for texture parameters at CT. Radiology. 2013;266(1):326–36.

    Article  PubMed  Google Scholar 

  51. Jalkanen S, Salmi M. Lymphatic endothelial cells of the lymph node. Nat Rev Immunol. 2020;20(9):566–78.

    Article  CAS  PubMed  Google Scholar 

  52. Fidler IJ. The pathogenesis of cancer metastasis: the “seed and soil” hypothesis revisited. Nat Rev Cancer. 2003;3(6):453–8.

    Article  CAS  PubMed  Google Scholar 

  53. Masquelin AH, Alshaabi T, Cheney N, Estépar RSJ, Bates JHT, Kinsey CM. Perinodular Parenchymal Features Improve Indeterminate Lung Nodule Classification. Acad Radiol. 2023;30(6):1073–80.

    Article  PubMed  Google Scholar 

  54. Bettinelli A, Marturano F, Avanzo M, Loi E, Menghi E, Mezzenga E, et al. A Novel Benchmarking Approach to Assess the Agreement among Radiomic Tools. Radiology. 2022;303(3):533–41.

    Article  PubMed  Google Scholar 

  55. Huang EP, O’Connor JPB, McShane LM, Giger ML, Lambin P, Kinahan PE, et al. Criteria for the translation of radiomics into clinically useful tests. Nat Rev Clin Oncol. 2023;20(2):69–82.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all the contributing authors for their great effort on this article.

Funding

This study has received funding by Zhejiang Provincial Natural Science Foundation of China (LTGY23H180001); National Natural Science Foundation of China (82102128); "Pioneer" and "Leading Goose" R&D Program of Zhejiang (2022C03046); Medical Health Science and Technology Project of Zhejiang Province (2022KY230); Research Project of Zhejiang Chinese Medical University (2022FSYYZY08; 2022JKJNTZ19; 2021JKGJYY007). The funding sources had no influence on the analysis and interpretation of data or on the contents of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

T.W.: Conceptualization; Data curation; Investigation; Methodology; Validation; Writing-original draft; Writing-review and; editing. C.G.: Data curation; Investigation; Methodology; Funding acquisition; Resources. X.L.: Formal analysis; Methodology; Visualization. J.W.: Formal analysis; Visualization. M.X.: Conceptualization; Funding acquisition; Project administration; Resources; Software; Supervision; Writing-review and; editing. L.W.: Conceptualization; Funding acquisition; Project administration; Resources; Software; Supervision; Writing-review and; editing. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Maosheng Xu or Linyu Wu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Supplementary Information

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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, T., Gao, C., Lou, X. et al. Predictive value of radiomic features extracted from primary lung adenocarcinoma in forecasting thoracic lymph node metastasis: a systematic review and meta-analysis. BMC Pulm Med 24, 246 (2024). https://doi.org/10.1186/s12890-024-03020-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12890-024-03020-x

Keywords