Skip to main content

Transfer learning–based PET/CT three-dimensional convolutional neural network fusion of image and clinical information for prediction of EGFR mutation in lung adenocarcinoma

Abstract

Background

To introduce a three-dimensional convolutional neural network (3D CNN) leveraging transfer learning for fusing PET/CT images and clinical data to predict EGFR mutation status in lung adenocarcinoma (LADC).

Methods

Retrospective data from 516 LADC patients, encompassing preoperative PET/CT images, clinical information, and EGFR mutation status, were divided into training (n = 404) and test sets (n = 112). Several deep learning models were developed utilizing transfer learning, involving CT-only and PET-only models. A dual-stream model fusing PET and CT and a three-stream transfer learning model (TS_TL) integrating clinical data were also developed. Image preprocessing includes semi-automatic segmentation, resampling, and image cropping. Considering the impact of class imbalance, the performance of the model was evaluated using ROC curves and AUC values.

Results

TS_TL model demonstrated promising performance in predicting the EGFR mutation status, with an AUC of 0.883 (95%CI = 0.849–0.917) in the training set and 0.730 (95%CI = 0.629–0.830) in the independent test set. Particularly in advanced LADC, the model achieved an AUC of 0.871 (95%CI = 0.823–0.919) in the training set and 0.760 (95%CI = 0.638–0.881) in the test set. The model identified distinct activation areas in solid or subsolid lesions associated with wild and mutant types. Additionally, the patterns captured by the model were significantly altered by effective tyrosine kinase inhibitors treatment, leading to notable changes in predicted mutation probabilities.

Conclusion

PET/CT deep learning model can act as a tool for predicting EGFR mutation in LADC. Additionally, it offers clinicians insights for treatment decisions through evaluations both before and after treatment.

Peer Review reports

Background

Non-small cell lung cancer (NSCLC) accounts for nearly 85% of primary lung cancers, and adenocarcinoma (ADC) is the most common subtype [1]. In Asia, epidermal growth factor receptor (EGFR) mutations are found in up to 50% of ADC patients [2]. In recent decades, tyrosine kinase inhibitors (TKI) have been shown to prolong progression-free survival and improve the quality of life in patients with EGFR mutations, especially those with advanced ADC [3]. Currently, molecular pathology is the gold standard for determining EGFR mutation status, but there are limitations. The limitations include sampling bias due to tumor heterogeneity, the requirement for invasive biopsies and related complications, slow detection speed, potentially high costs, and the possibility of unreliable results due to insufficient quantity or quality of tissue [4]. Additionally, during the course of disease treatment and progression, the status of EGFR mutations and the immune landscape may change [5]. Therefore, there is an urgent need for a noninvasive, accurate, simple, and reproducible method to predict EGFR mutations.

18 (18F)-fluorodeoxyglucose (FDG) PET/CT is a widely accepted noninvasive method for evaluating NSCLC [6,7,8,9]. Two recent meta-analyses confirmed the moderate predictive capability of SUVmax for EGFR mutations, with AUC = 0.68–0.69 [10, 11]. Recent studies have focused on 18F-FDG PET/CT radiomics [12, 13]. Heterogeneity is more likely present in tumors with EGFR mutations [12, 14], which radiomics may capture. However, PET/CT-based radiomics features showed remarkable predictive power for EGFR mutations (AUC = 0.50–0.87) [15], yet its clinical application requires further investigation for confirmation and optimization.

Convolutional neural networks (CNNs) have performed well in lesion detection, segmentation, and classification [16,17,18]. Few studies have used PET/CT deep learning models to predict EGFR mutations, mainly due to the data paucity. The only two PET/CT studies with deep learning focused on two-dimensional CNN were trained from scratch [19, 20]; although this method reduces the processing burden, it inevitably affects its performance. Transfer learning (TL) from the pretrained model using ImageNet data has been the standard for deep learning in medical imaging [21]. This method, however, has two limitations: first, the input to the model must be two-dimensional (2D), and thus the rich anatomical three-dimensional (3D) medical images are lost; second, due to the great difference between medical images and natural images, the performance of TL from natural images to medical images is not obvious. To overcome these limitations, we employed Models Genesis, a pretrained model specifically designed for 3D medical imaging data [22]. Unlike other transfer learning strategies, such as pretraining through proxy tasks like lung nodule segmentation or supervised metric learning networks [23], Models Genesis utilizes a self-supervised learning strategy. It focuses on learning from 3D image information to better utilize the spatial information inherent in 3D, demonstrating superiority across multiple 3D medical imaging tasks.

Deep learning offers unique advantages in enhancing the accuracy of medical image diagnosis, especially when integrated with clinical data [24, 25]. These studies underscore the importance of clinical information in constructing efficient deep learning predictive models, highlighting the value of clinical data in understanding and predicting the complex biological influences on EGFR mutation status in lung adenocarcinoma. In this study, we developed a multimodal deep learning model based on 18F-FDG PET/CT, which fully harnesses the inherent 3D characteristics of medical imaging to align more closely with actual clinical scenarios, potentially enhancing the accuracy and reliability of EGFR mutation prediction. This integrative approach provides a comprehensive fusion of radiological and clinical data [26], and also enables efficient classification of EGFR mutation status.

Methods

Adherence to checklists

This study adhered to the CLEAR checklist [27] for the reporting of our radiomics research. The completed CLEAR checklist has been listed in Table S1. To ensure the transparency and reproducibility of our study, we have made all the raw data and analysis code publicly available. For detailed links, please refer to Availability of data and materials.

Participants

This retrospective single-center cohort study used privately sourced data, obtaining information from consecutive patients at the Third Affiliated Hospital of Soochow University. Patients with histologically confirmed lung cancer underwent pretreatment 18F-FDG PET/CT scans in our department between January 2018 and April 2022. The Institutional Review Board approved this study (No. [2022] KD 087) and waived the need for informed consent from the patients. All patient data used in this study were fully de-identified to ensure privacy and confidentiality, in accordance with international data protection guidelines and standards. The sample size was determined based on the consecutive patients available during the study period. Inclusion criteria: (1) the patient was confirmed with lung cancer by surgery or pathological biopsy; (2) the patient underwent 18F-FDG PET/CT examination before surgery, and the interval between surgery and examination was less than 30 days; (3) the patient had definite EGFR test results; and (4) the patient had no history of other malignant tumors. Exclusion criteria: (1) other pathological subtypes except for ADC, (2) lesions with poor image quality or difficulty in measurement, (3) absence of routine chest CT imaging, and (4) severe liver disease or diabetes. Using an internal testing technique, we designated 404 patients from January 2018 to April 2021 as the training set, while the independent test set was formed from 112 patients spanning May 2021 to April 2022. The process of patient enrollment is outlined in Fig. 1. All data used in this study have not been published or used in any other previous publications.

Fig. 1
figure 1

Flowchart of patient enrollment. LADC, lung adenocarcinoma; NSCC-NOS, non‑small cell carcinoma‑not otherwise specified

In this study, we also collected clinical characteristics of the patients, which included: age, gender, smoking history, type of nodules, location of nodules, tumor size, clinical stage (I-IV), carcinoembryonic antigen (CEA), and maximum standardized uptake value (SUVmax).

EGFR mutation detection

The detection of EGFR mutations was conducted in tissue samples obtained by surgical resection or puncture. Mutations in exons 18–21 of the EGFR gene were detected using real-time fluorescent PCR. Real-time fluorescent PCR was conducted per the instructions of the Shanghai Yuanqi EGFR gene mutation detection kit, and the details are described in the Supplementary Material EGFR mutation detection method. EGFR mutation was defined if a mutation was detected in any of those exons; otherwise, wild-type EGFR was defined.

FDG PET/CT image acquisition

The image acquisition protocol was described according to an acquisition protocol based on the Imaging Biomarker Standardization Initiative (IBSI) reporting guidelines [28]. The image acquisition parameters are listed in Table S2. The patient underwent non-contrast chest CT imaging and 18F-FDG PET/CT (Biograph mCT 64, Siemens, Erlangen, Germany) within one month before surgical treatment. Before the administration of 18F-FDG, the blood glucose levels of the patients were checked to ensure they were within the acceptable range (< 150 mg/dL). According to the European Association of Nuclear Medicine (EANM) guidelines 1.0 (version 2.0, published in February 2015) [29], 18F-FDG PET/CT images were acquired 60 ± 5 min after 18F-FDG injection. All PET/CT images were reconstructed on a processing workstation (TrueD software, Siemens Healthcare).

Image segmentation and pre-processing

A nuclear medicine physician with over 10 years of experience selected regions of interest on PET and CT images, and all images were segmented using 3D-Slicer (version 4.11.20200930, www.slicer.org). For CT images (3 mm), we utilized a semi-automatic method with NVIDIA AI-assisted annotation (3D-Slicer built-in) and a boundary-based CT segmentation model to process the lung nodule images. For PET images, 3D masks were generated using a semi-automatic segmentation method developed by Beichel et al. [30]. Please refer to Supplementary Material-PET/CT image pre-processing for deep learning for details.

Development of the deep learning models

The overall approach to developing the deep learning model is summarized in Fig. 2. To initialize the encoder for the target classification task, we employed Models Genesis, an openly available deep learning model on GitHub (https://github.com/MrGiovanni/ModelsGenesis.git). We subsequently fine-tuned it in accordance with the specific requirements of our target task. The structure diagrams of four 3D CNNs, namely CT TL (CT_TL), PET TL (PET_TL), dual-stream TL (DS_TL, fusing PET and CT), and three-stream TL (TS_TL, adding clinical data to PET and CT) networks, are depicted in Fig. S1. Additionally, we trained two models from scratch (CT_origin and PET_origin). Details of the model training and tuning are described in the Supplementary Material-Training of deep learning models.

Fig. 2
figure 2

Overall pipeline for deep learning model development

Visualization of deep learning models

A visualization method, Grad-CAM, was adopted to explain the prediction process of the deep learning models [31]. High-reaction regions (predicted tumor-associated areas) were retained with a cutoff value of 0.5. When the deep learning model predicts the EGFR mutation status, it informs the clinician which areas attracted the model's attention (divided into wild and mutant type-associated activation areas). We selected only 3D input cross-sectional intermediate layer images for visualization.

Statistical analysis

Statistical analysis of clinical data and routine PET/CT metabolic parameters was performed using R software (version 3.4.3; http://www.R-project.org/). The model's performance was assessed using receiver operating characteristic (ROC) curves and quantified by calculating area under the receiver operating characteristic curve (AUC) and 95% confidence intervals (CI). In addition, we further evaluated the model by calculating accuracy, sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) to obtain comprehensive quantitative performance metrics. Due to the presence of class imbalance in the data, we chose AUC as the primary performance metric for the model. A pairwise comparison of the AUC values of the models was performed using the method proposed by Delong et al. [32]. All statistical tests were two-sided, and statistical significance was interpreted as p < 0.05. In this study, no missing data was encountered.

Results

Clinical characteristics of patients

Table 1 describes the clinical characteristics of 516 patients, of which 202 (39.1%) were with wild-type EGFR and 314 (60.9%) exhibited EGFR mutations. During our analysis, we observed significant differences in clinical characteristics such as gender, smoking history, type of nodules, tumor long axis, and tumor short axis between patients with EGFR mutations and those with wild-type EGFR (with p < 0.05 in both the training and test sets). Based on these observations and informed by clinical prior knowledge [33, 34], we decided to incorporate these characteristics as clinical information in constructing TS_TL based on PET/CT images. During this process, we normalized the quantitative indicators to ensure they fall within the range of 0 to 1, aligning with the scale of the normalized PET/CT image data. For categorical variables, we retained their original binary format, allowing for the consideration of clinical variable diversity in the construction of the TS_TL model.

Table 1 Clinical characteristics of patients with different EGFR mutation statuses in the training and test sets

Diagnostic validation of several deep learning models

The predictive performance of several deep learning models in the test set is listed in Table 2 (see Table S3 for training set). In the training set, TS_TL showed the best predictive performance (AUC = 0.883), further confirmed in the independent test set (AUC = 0.730). CT_TL and PET_TL outperformed CT_origin and PET_origin in both training and test sets, with significant improvement in test set for CT_TL (AUC = 0.701 vs. 0.544, p = 0.027) and in training set for PET_TL (AUC = 0.770 vs. 0.619, p < 0.001; Fig. 3 A, B). Also, CT_TL showed high sensitivity and low specificity in both sets, while PET_TL showed low sensitivity and high specificity (Table S3 and Table 2).

Table 2 Predictive performance of several deep learning models in the test set
Fig. 3
figure 3

ROC curves of different transfer learning models in the training and test sets. AUC, area under the receiver operating characteristic curve; CT_origin, CT model from scratch; CT_TL, CT transfer learning; PET_origin, PET model from scratch; PET_TL, PET transfer learning; DS_TL, dual-stream transfer learning; TS_TL, three-stream transfer learning

The tumor long axis, tumor short axis, tumor stage, and SUVmax showed statistical differences between the two sets (all p < 0.05), which might be attributed to the different compositions of patients at different periods in our center (Table S4). To eliminate this discrepancy, we performed a hierarchical analysis to validate the diagnostic performance of the four TL models for different tumor stages (Table S5). For stage I-II tumors, TS_TL exhibited significant overfitting (AUC of training set vs. test set: 0.903 vs. 0.667), while DS_TL had the optimal performance in the test set (AUC of training set vs. test set: 0.862 vs. 0.708); for stage III-IV tumors, TS_TL showed the best performance in both training and test sets (AUC of training set vs. test set: 0.871 vs. 0.760).

Comparisons of deep learning models and radiomics models

We established four radiomics models of CT, PET, PET/CT, and PET/CT combined with clinical features (CT_RS, PET_RS, DS_RS, TS_RS) and compared them with the proposed TL models (Table 3 and Table S6). The specific methods for the development of the radiomics model are described in the Supplementary Material-Development of four radiomics models, Table S7 and Table S8. ROC curves of the four radiomics models and SUVmax are shown in Figure S2. First, PET_RS outperformed SUVmax in both training and test sets but only showed significance in the training set (training set: AUC = 0.651 vs. 0.582, p = 0.014). It was subsequently validated that PET_TL performed significantly better in the training set but slightly inferior in the test set than the PET_RS model (training set: AUC = 0.770 vs. 0.651, p < 0.001; test set: AUC = 0.645 vs. 0.661, p = 0.763). CT_TL performed better than CT_RS in both training and test sets, with a significant improvement in the training set only (training set: AUC = 0.739 vs. 0.655, p < 0.001).

Table 3 Comparison of the prediction performance of deep learning models and radiomics models in the test set

Compared to single-modal radiomics models CT_RS and PET_RS, DS_RS performed slightly better in the training set and worse in the test set, but the differences were not significant (training set: AUC = 0.662 vs. 0.655 vs. 0.651, both p > 0.05; test set: AUC = 0.620 vs. 0.639 vs. 0.661, both p > 0.05). DS_TL performed better than DS_RS in both training and test sets (training set: AUC = 0.823 vs. 0.662, p < 0.001; test set: AUC = 0.722 vs. 0.620, p = 0.033).

By combining DS_RS with clinical features, TS_RS outperformed CT_RS, PET_RS, and DS_RS in both training and test sets, while statistical significance was only noted in the training set (training set: AUC = 0.771 vs. 0.655 vs. 0.651 vs. 0.662, all p < 0.001). Similarly, TS_TL outperformed TS_RS in both training and test sets, with statistical significance in training set only (training set: AUC = 0.883 vs. 0.771, p < 0.001). When DS_TL was combined with clinical features, TS_TL outperformed DS_TL in both training and test sets, with significance shown only in the training set (training set: AUC = 0.883 vs. 0.823, p < 0.001).

TS_TL predicted tumor-associated areas for solid or subsolid lesions of different mutation subtypes

Figure 4 displays six representative solid lesions (three wild-type and three mutant) along with TS_TL's wild and mutant type-associated activation areas. In solid lesions, TS_TL consistently focuses on the local and peripheral areas of the lesion in CT images (Fig. 4a, c, e, g, i, k) and most metabolic areas in PET images (Fig. 4b, d, f, h, j, l), regardless of wild-type or mutant status.

Fig. 4
figure 4

TS_TL predicted tumor-associated areas for solid lesions with either EGFR wild-type or mutation. For each submap, the input CT or PET image, the attention map, and the model-predicted tumor-associated areas are from left to right. For LADC tumors, the deep learning model generated an attention map indicating the importance of each part of the tumor; high-reaction regions (predicted tumor-associated areas) were retained with a cutoff value of 0.5. P and P+ represented the predicted probability of EGFR wild-type and mutant, respectively

Figure 5 displays six representative subsolid lesions (three wild-type and three mutant) along with TS_TL's wild and mutant type-associated activation areas. For subsolid wild-type lesions, the wild type-associated activation areas in CT and PET images are similar to those in solid lesions (Fig. 5a-f). For subsolid mutant lesions, the mutant type-associated activation areas in CT images still robustly capture the local and peripheral areas of the lesion (Fig. 5g, i, k), while the mutant type-associated activation areas in PET images do not effectively capture the metabolic areas of the lesion (Fig. 5, j, l).

Fig. 5
figure 5

TS_TL predicted tumor-associated areas for subsolid lesions with either EGFR wild-type or mutation. For each submap, the input CT or PET image, the attention map, and the model-predicted tumor-associated areas are from left to right. For LADC tumors, the deep learning model generated an attention map indicating the importance of each part of the tumor; high-reaction regions (predicted tumor-associated areas) were retained with a cutoff value of 0.5. P and P+ represented the predicted probability of EGFR wild-type and mutant, respectively

Changes in mutant type-associated activation areas before and after TKI treatment

Figure 6 shows the changes in the mutant type-associated activation areas and the predicted mutation probability (P+) before and after TKI treatment in three EGFR-mutant patients with effective TKI treatment. Both case 1 and case 2 are solid lesions with non-ideal model predictions before TKI treatment (P+ values in Fig. 6a, b and c, d are both less than 0.5, indicating that the model has not yet captured a clear image mutation pattern). The P+ values of both cases significantly increase after TKI treatment (from 0.462 to 0.679 and from 0.299 to 0.485). Looking at the mutant type-associated activation areas after treatment (Fig. 6g, h and i, j), they are also similar to the previously mentioned subsolid mutant lesions (refer to Fig. 5g-l), indicating that the treatment has changed the image pattern captured by the model. For subsolid lesion case 3, there are no obvious mutant type-associated activation areas in either CT or PET pathways before and after treatment (Fig. 6e, f and k, l). However, the P+ value increases significantly after treatment (from 0.373 to 0.620). The changes in classical methods and P+ values before and after TKI treatment in 3 cases can be seen in Table S9.

Fig. 6
figure 6

The mutant type-associated activation areas of the TS_TL in three EGFR-mutant lesions before and after TKI treatment. Case 1 (stage IV): A female non-smoker in the 60–65 age range presented with a solid mass in the left lower lung (41.1 × 29.7 mm) with an EGFR mutation in exon 20 and received oral poziotinib treatment; through PET/CT re-examination after 21 months, the original lesion shrunk (30.2 × 24.6 mm) and its metabolism was lower than before (SUVmax from 14.8 to 10.2). Poziotinib is a novel targeted drug for rare insertion mutations in exon 20 of EGFR and HER2. Case 2 (stage IV): A male smoker in the 65–70 age range presented with a solid mass in the left upper lung (40.9 × 26.7 mm) harboring an exon 21 EGFR mutation. The patient received oral Osimertinib treatment, and after 6 months, a follow-up PET/CT examination revealed a significant reduction in the size of the original lesion, which became subsolid (20.7 × 15.9 mm), and a decrease in metabolism (SUVmax from 11.8 to 3.9). Case 3 (stage IV): A male non-smoker in the 80–85 age range presented with a subsolid mass in the right upper lung (35.5 × 30.5 mm) carrying an exon 19 EGFR mutation. The patient received oral Icotinib hydrochloride treatment, and after 46 months, a follow-up PET/CT examination revealed a slight reduction in the size of the original lesion (28.5 × 27.9 mm) and a decrease in metabolism (SUVmax from 3.1 to 1.9)

Discussion

The development of multimodal biomarkers will be a trend in the field of precision oncology in the future [26]. Anatomical information represented by CT and functional information represented by PET are naturally complementary, and the integration of FDG PET/CT images maintains the sensitivity of CT and the specificity of PET [35]. In our study, CT_TL and PET_TL showed high sensitivity and high specificity, resulting in improved performance of DS_TL, particularly in predicting EGFR in early LADC. This might be attributed to the fact that the important regions available for accurately predicting EGFR mutations could be better and more easily localized using metabolic and anatomical information reflected by PET and CT images, respectively [19].

Many studies have revealed clinical factors associated with EGFR mutation in LADC, such as gender, smoking history, and the presence of ground glass opacity (GGO) [33]. Before CNN training, we standardized the CT and PET images through preprocessing, including resampling to match the CNN's input size. This process may partially lose the original lesion size information, so to compensate, we specifically included tumor long and short axis measurements as additional clinical features into the DS_TL model. This is aimed at preserving crucial information about tumor size, ensuring the model fully considers the actual dimensions of the lesion. By adding gender, smoking history, nodule type, and metadata [34], the performance of the TS_TL model has been improved, especially in predicting advanced LADC.

Previous research has shown that radiomic features extracted from PET/CT and CT images can predict gene expression patterns and EGFR mutation status [14, 36]. In their review, Ge et al. [37] highlighted that machine learning-based radiomics (MLR, specifically shallow learning), have demonstrated high accuracy in predicting EGFR mutations. The advantage of CT_TL and PET_TL over the radiomics models in this study was insignificant, possibly due to the small sample size and different patient compositions in the training set. We further developed the DS_RS model through feature fusion, the performance of which in the test set was lower than expected, while DS_TL better integrated the information of two modes, CT and PET, yielding better generalization. Furthermore, we also found that the input of clinical information significantly improved the diagnostic performance of DS_RS, while the performance of DS_TL showed a mild improvement, which could be explained by performance saturation.

For predicting mutation in solid lesions, both CT and PET images play a stable role in the model, and similar findings were obtained by Yin et al. [20]. Existing studies have reported that the features of the edge of the lesions, such as spiculation sign [38] and lobulation sign [39], are related to EGFR mutation. TS_TL's attention for solid lesions is focused on the tumor periphery with the aforementioned characteristics. For wild-type predictions in subsolid lesions, both CT and PET images still play a role in TS_TL; however, in mutant predictions, CT images take the lead in the model, while PET images fail to capture the metabolic areas of the lesion effectively. This is related to the generally low metabolism of most mutant-type subsolid lesions [40], which cannot effectively activate specific convolutional kernels.

Studies have shown that PET/CT can be used to monitor TKI treatment reactions and evaluate the prognosis of NSCLC patients [41]. This study found that TKI treatment may change the image patterns captured by the model, and the change in P+ value before and after treatment may be superior to traditional indicators, providing an indication of therapeutic efficacy. This is because the P+ value integrates the lesion image and long-short diameter information (which can be considered a weighted combination of these useful factors), closely related to efficacy assessment.

However, this study also comes with its limitations. (1) Being a single-center study, the wider applicability of this model requires external validation, despite our methodical assignment of patients to the training set and independent test set based on time. (2) Some scholars posit that the model's predicted tumor-associated areas could guide clinicians to optimal biopsy locations within the tumor, mitigating the sampling bias due to tumor heterogeneity [18]. Nonetheless, this proposition warrants further exploration through prospective studies, particularly concerning the biological interpretation of the said tumor-associated areas.

Conclusions

Our end-to-end deep learning model integrates CT, PET, and clinical data to effectively predict EGFR mutation in LADC. This model could also find predicted tumor-associated areas strongly linked to EGFR mutation status and help clinicians make patient treatment decisions through pre- and posttreatment qualitative and quantitative assessment.

Availability of data and materials

No datasets were generated or analysed during the current study.

Abbreviations

EGFR:

Epidermal growth factor receptor

LADC:

Lung adenocarcinoma

TKI:

Tyrosine kinase inhibitors

CNN:

Convolutional neural networks

NSCLC:

Non-small cell lung cancer

ADC:

Adenocarcinoma

FDG:

Fluorodeoxyglucose

TL:

Transfer learning

2D:

Two-dimensional

3D:

Three-dimensional

CEA:

Carcinoembryonic antigen

SUV:

Standard uptake value

DS_TL:

Dual-stream transfer learning

TS_TL:

Three-stream transfer learning

HU:

Hounsfield units

ROC:

Receiver operating characteristic

AUC:

Area under the receiver operating characteristic curve

PPV:

Positive predictive value

NPV:

Negative predictive value

References

  1. Travis WD. Pathology of lung cancer. Clin Chest Med. 2011;32(4):669–92.

    Article  PubMed  Google Scholar 

  2. McLoughlin EM, Gentzler RD. Epidermal Growth Factor Receptor Mutations. Thorac Cardiovasc Surg. 2020;30(2):127–36.

    Google Scholar 

  3. Douillard JY, Ostoros G, Cobo M, Ciuleanu T, McCormack R, Webster A, et al. First-line gefitinib in Caucasian EGFR mutation-positive NSCLC patients: a phase-IV, open-label, single-arm study. Br J Cancer. 2014;110(1):55–62.

    Article  CAS  PubMed  Google Scholar 

  4. Taniguchi K, Okami J, Kodama K, Higashiyama M, Kato K. Intratumor heterogeneity of epidermal growth factor receptor mutations in lung cancer and its correlation to the response to gefitinib. Cancer Sci. 2008;99(5):929–35.

    Article  CAS  PubMed  Google Scholar 

  5. Bai H, Wang Z, Chen K, Zhao J, Lee JJ, Wang S, et al. Influence of chemotherapy on EGFR mutation status among patients with non-small-cell lung cancer. J Clin Oncol. 2012;30(25):3077–83.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Ettinger DS, Wood DE, Aisner DL, Akerley W, Bauman JR, Bharat A. Non-small cell lung cancer, version 32022, NCCN clinical practice guidelines in oncology. J Nat Comprehen Cancer Net JNCCN. 2022;20(5):497–530.

    Article  Google Scholar 

  7. Vansteenkiste J, Crino L, Dooms C, Douillard JY, FaivreFinn C, Lim E, et al. 2nd ESMO Consensus Conference on Lung Cancer early-stage non-small-cell lung cancer consensus on diagnosis treatment and follow-up. Ann Oncol. 2014;25(8):1462–74.

    Article  CAS  PubMed  Google Scholar 

  8. Eberhardt WE, De Ruysscher D, Weder W, Le Péchoux C, De Leyn P, Hoffmann H, et al. 2nd ESMO Consensus Conference in Lung Cancer: locally advanced stage III non-small-cell lung cancer. Ann Oncol. 2015;26(8):1573–88.

    Article  CAS  PubMed  Google Scholar 

  9. MacMahon H, Naidich DP, Goo JM, Lee KS, Leung ANC, Mayo JR, et al. Guidelines for Management of Incidental Pulmonary Nodules Detected on CT Images: From the Fleischner Society 2017. Radiology. 2017;284(1):228–43.

    Article  PubMed  Google Scholar 

  10. Du B, Wang S, Cui Y, Liu G, Li X, Li Y. Can (18)F-FDG PET/CT predict EGFR status in patients with non-small cell lung cancer? A systematic review and meta-analysis. BMJ Open. 2021;11(6):e044313.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Guo Y, Zhu H, Yao Z, Liu F, Yang D. The diagnostic and predictive efficacy of (18)F-FDG PET/CT metabolic parameters for EGFR mutation status in non-small-cell lung cancer: A meta-analysis. Eur J Radiol. 2021;141:109792.

    Article  PubMed  Google Scholar 

  12. Li X, Yin G, Zhang Y, Dai D, Liu J, Chen P, et al. Predictive Power of a Radiomic Signature Based on (18)F-FDG PET/CT Images for EGFR Mutational Status in NSCLC. Front Oncol. 2019;9:1062.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Nair JKR, Saeed UA, McDougall CC, Sabri A, Kovacina B, Raidu B, et al. Radiogenomic models using machine learning techniques to predict EGFR mutations in non-small cell lung cancer. Can Assoc Radiol J. 2021;72(1):109–19.

    Article  PubMed  Google Scholar 

  14. Zhang J, Zhao X. Value of pre-therapy (18)F-FDG PET/CT radiomics in predicting EGFR mutation status in patients with non-small cell lung cancer. Eur J Nucl Med Mol Imaging. 2020;47(5):1137–46.

    Article  CAS  PubMed  Google Scholar 

  15. Manafi-Farid R, Askari E, Shiri I, Pirich C, Asadi M, Khateri M, et al. [(18)F]FDG-PET/CT radiomics and artificial intelligence in lung cancer: Technical aspects and potential clinical applications. Semin Nucl Med. 2022;52(6):759–7801.

    Article  PubMed  Google Scholar 

  16. Jemaa S, Fredrickson J, Carano RAD, Nielsen T, de Crespigny A, Bengtsson T. Tumor Segmentation and Feature Extraction from Whole-Body FDG-PET/CT Using Cascaded 2D and 3D Convolutional Neural Networks. J Digit Imaging. 2020;33(4):888–94.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Singadkar G, Mahajan A, Thakur M, Talbar S. Deep Deconvolutional Residual Network Based Automatic Lung Nodule Segmentation. J Digit Imaging. 2020;33(3):678–84.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Wang S, Shi J, Ye Z, Dong D, Yu D, Zhou M, et al. Predicting EGFR mutation status in lung adenocarcinoma on computed tomography image using deep learning. Eur Respi J. 2019;53(3):1800986.

    Article  Google Scholar 

  19. Mu W, Jiang L, Zhang J, Shi Y, Gray JE, Tunali I, et al. Non-invasive decision support for NSCLC treatment using PET/CT radiomics. Nat Commun. 2020;11(1):5228.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yin G, Wang Z, Song Y, Li X, Chen Y, Zhu L, et al. Prediction of EGFR Mutation Status Based on (18)F-FDG PET/CT Imaging Using Deep Learning-Based Model in Lung Adenocarcinoma. Front Oncol. 2021;11:709137.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kim HE, Cosa-Linan A, Santhanam N, Jannesari M, Maros ME, Ganslandt T. Transfer learning for medical image classification: a literature review. BMC Med Imaging. 2022;22(1):69.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Zhou Z, Sodha V, Pang J, Gotway MB, Liang J. Models genesis. Med Image Anal. 2021;67:101840.

    Article  PubMed  Google Scholar 

  23. Thammasorn P, Chaovalitwongse WA, Hippe DS, Wootton LS, Ford EC, Spraker MB, et al. Nearest Neighbor-Based Strategy to Optimize Multi-View Triplet Network for Classification of Small-Sample Medical Imaging Data. IEEE Trans Neural Netw Learn Syst. 2023;34(2):586–600.

    Article  PubMed  Google Scholar 

  24. Chieregato M, Frangiamore F, Morassi M, Baresi C, Nici S, Bassetti C, et al. A hybrid machine learning/deep learning COVID-19 severity predictive model from CT images and clinical data. Sci Rep. 2022;12(1):4329.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhang X, Dong X, Saripan MIB, Du D, Wu Y, Wang Z, et al. Deep learning PET/CT-based radiomics integrates clinical data: A feasibility study to distinguish between tuberculosis nodules and lung cancer. Thorac Cancer. 2023;14(19):1802–11.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Boehm KM, Khosravi P. Harnessing multimodal data integration to advance precision oncology. Nat Rev Cancer. 2022;22(2):114–26.

    Article  CAS  PubMed  Google Scholar 

  27. Kocak B, Baessler B, Bakas S, Cuocolo R, Fedorov A, Maier-Hein L, et al. CheckList for EvaluAtion of Radiomics research (CLEAR): a step-by-step reporting guideline for authors and reviewers endorsed by ESR and EuSoMII. Insights Imaging. 2023;14(1):75.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Zwanenburg A, Vallières M, Abdalah MA, Aerts HJ, Andrearczyk V, Apte A, et al. The image biomarker standardization initiative: standardized quantitative radiomics for high-throughput image-based phenotyping. Radiology. 2020;295(2):328–38.

    Article  PubMed  Google Scholar 

  29. Boellaard R, Delgado-Bolton R, Oyen WJ, Giammarile F, Tatsch K, Eschner W, et al. FDG PET/CT EANM procedure guidelines for tumour imaging version 20. Eur J Nucl Med Molecular Imaging. 2015;42(2):328–54.

    Article  CAS  Google Scholar 

  30. Beichel RR, Van Tol M, Ulrich EJ, Bauer C, Chang T, Plichta KA, et al. Semiautomated segmentation of head and neck cancers in 18F-FDG PET scans: A just-enough-interaction approach. Med Phys. 2016;43(6):2948–64.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Selvaraju RR, Cogswell M, Das A, Vedantam R, Parikh D, Grad-Cam B, editors. Visual explanations from deep networks via gradient-based localization. Proceedings of the 2017 IEEE International Conference on Computer Vision (ICCV); 2021:618–26.

  32. DeLong ER, DeLong DM, Clarke-Pearson DL. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics. 1988;44(3):837–45.

    Article  CAS  PubMed  Google Scholar 

  33. Zhang H, Cai W, Wang Y, Liao M. CT and clinical characteristics that predict risk of EGFR mutation in non-small cell lung cancer: a systematic review and meta-analysis. Int J Clin Oncol. 2019;24(6):649–59.

    Article  CAS  PubMed  Google Scholar 

  34. Park YJ, Choi D, Choi JY, Hyun SH. Performance Evaluation of a Deep Learning System for Differential Diagnosis of Lung Cancer With Conventional CT and FDG PET/CT Using Transfer Learning and Metadata. Clin Nucl Med. 2021;46(8):635–40.

    Article  PubMed  Google Scholar 

  35. Hofman MS, Hicks RJ. How we read oncologic FDG PET/CT. Cancer Imaging. 2016;16(1):35.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Yip SS, Kim J, Coroller TP, Parmar C, Velazquez ER, Huynh E, et al. Associations Between Somatic Mutations and Metabolic Imaging Phenotypes in Non-Small Cell Lung Cancer. J Nucl Med. 2017;58(4):569–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ge X, Gao J, Niu R, Shi Y, Shao X, Wang Y, et al. New research progress on 18F-FDG PET/CT radiomics for EGFR mutation prediction in lung adenocarcinoma: a review. Front Oncol. 2023;13:1242392.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liu Y, Kim J, Qu F, Liu S, Wang H, Balagurunathan Y, et al. CT Features Associated with Epidermal Growth Factor Receptor Mutation Status in Patients with Lung Adenocarcinoma. Radiology. 2016;280(1):271–80.

    Article  PubMed  Google Scholar 

  39. Chen Y, Yang Y, Ma L, Zhu H, Feng T, Jiang S, et al. Prediction of EGFR mutations by conventional CT-features in advanced pulmonary adenocarcinoma. Eur J Radiol. 2019;112:44–51.

    Article  PubMed  Google Scholar 

  40. Guan J, Xiao NJ, Chen M, Zhou WL, Zhang YW, Wang S, et al. 18F-FDG uptake for prediction EGFR mutation status in non-small cell lung cancer. Medicine. 2016;95(30):e4421.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Jiang M, Zhang X, Chen Y, Chen P, Guo X, Ma L, et al. A Review of the Correlation Between Epidermal Growth Factor Receptor Mutation Status and (18)F-FDG Metabolic Activity in Non-Small Cell Lung Cancer. Front Oncol. 2022;12:780186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by Major Project of Changzhou Health Commission (Grant No. ZD202109), Key Laboratory of Changzhou High-tech Research Project (Grant No. CM20193010), Young Talent Development Plan of Changzhou Health Commission (Grant No. CZQM2020012), and 2022 Changzhou "14th Five-Year Plan" Health and Health High-level Talent Training Project-Top-notch talents (2022260).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Xiaonan Shao and Yuetao Wang.  Data curation: Xinyu Ge, Jianxiong Gao, Rong Niu, Yunmei Shi, Xiaoliang Shao, Zhenxing Jiang, and Renyuan Li.  Formal analysis: Xiaonan Shao and Jianxiong Gao.  Funding acquisition: Xiaonan Shao and Yuetao Wang.  Investigation: Xinyu Ge, Jianxiong Gao, Rong Niu, Yunmei Shi, Xiaoliang Shao, Zhenxing Jiang, and Renyuan Li.  Methodology: Xiaonan Shao and Yuetao Wang.  Project administration: Xiaonan Shao and Yuetao Wang.  Software: Xiaonan Shao.  Supervision: Yuetao Wang.  Visualization: Xiaonan Shao.  Writing-original draft: Xiaonan Shao and Rong Niu.  Writing-review & editing: Rong Niu, Xiaoliang Shao, and Yuetao Wang.  All authors have read the final manuscript and approved the version to be published.

Corresponding authors

Correspondence to Xiaonan Shao or Yuetao Wang.

Ethics declarations

Ethics approval and consent to participate

The study protocol followed the Helsinki Declaration and was approved by the Ethics Committee of the Third Affiliated Hospital of Soochow University (No. [2022] KD 087) and waived the need for informed consent from the patients.

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

Shao, X., Ge, X., Gao, J. et al. Transfer learning–based PET/CT three-dimensional convolutional neural network fusion of image and clinical information for prediction of EGFR mutation in lung adenocarcinoma. BMC Med Imaging 24, 54 (2024). https://doi.org/10.1186/s12880-024-01232-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12880-024-01232-5

Keywords