The predictive value of 18F-FDG PET/CT in an EGFR-mutated lung adenocarcinoma population
Original Article

The predictive value of 18F-FDG PET/CT in an EGFR-mutated lung adenocarcinoma population

Jian Wang1#, Xiaolian Wen2#, Guirong Yang3, Yong Cui4, Mingyan Hao5, Xiaoyuan Qiao2, Baoli Jin6, Bo Li6, Jing Wu6, Xiaomin Li6, Xiaolu Ren6

1Department of Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China; 2Department of Oncology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China; 3Department of Endoscopy, The Fifth People’s Hospital of Datong, Datong, China; 4Department of Surgery, Otuoke Qianqi People’s Hospital, Ordos, China; 5Department of Administration, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China; 6Department of Radiation Oncology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China

Contributions: (I) Conception and design: X Ren; (II) Administrative support: J Wu, X Li; (III) Provision of study materials or patients: M Hao, J Wang, Y Cui, B Li, G Yang; (IV) Collection and assembly of data: J Wang, B Li, B Jin, M Hao, X Wen; (V) Data analysis and interpretation: X Ren, G Yang, Y Cui, X Qiao; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Xiaolu Ren. Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, 3th Zhigong New Street, Taiyuan 030013, China. Email: 13934131048@139.com.

Background: A non-invasive, simple, and convenient method to evaluate the presence of epidermal growth factor receptor (EGFR) mutations is important for initial treatment decisions in lung adenocarcinoma.

Methods: We retrospectively reviewed 297 untreated primary lung adenocarcinoma patients with exact EGFR status. Based on their EGFR status, the patients were divided into a mutant-type group (138 patients) and wild-type group (159 patients). General patient characteristics and possible factors reflecting the status of EGFR were also evaluated.

Results: Of the 297 lung adenocarcinoma patients analyzed for EGFR status who underwent positron emission tomography (PET)/computed tomography (CT) between January 2013 and December 2017, mutations in the EGFR gene were detected in 138 patients (46.5%). EGFR mutations were more frequently associated with women, never smokers, and low 18F-fluoro-2-deoxy-glucose (18F-FDG) PET/CT maximal standard uptake value of the primary tumor (pSUVmax). Multivariate analysis indicated that women [odds ratio (OR) =2.853; 95% confidence interval (CI): 1.451–5.611; P=0.002], never smokers (OR =2.414; 95% CI: 1.217–4.789; P=0.012), tumor size <3.5 cm (OR, 2.170; 95% CI: 1.205–3.908; P=0.010), and pSUVmax <8.2 (OR =1.904; 95% CI: 1.098–3.302; P=0.022) were effective predictors of EGFR mutation. In addition, the area under the curve (AUC) of pSUVmax and tumor size was 0.623 and 0.600, respectively. Combined with clinical characteristics, including sex and smoking status, the AUC of the 4 predictors was 0.770.

Conclusions: These indicators could be helpful for enhancing predictive accuracy of EGFR mutations in lung adenocarcinoma patients, especially in those for whom EGFR detection is unavailable.

Keywords: Epidermal growth factor receptor (EGFR); lung adenocarcinoma; standard uptake value (SUV); mutation


Submitted May 17, 2022. Accepted for publication Jul 12, 2022.

doi: 10.21037/tcr-22-1726


Introduction

The introduction of novel therapies, especially the use of tyrosine kinase inhibitors (TKIs), has improved patient outcomes and enabled a significant paradigm shift in the therapeutic agent management of non-small cell lung cancer (NSCLC) (1-5). Numerous randomised clinical trials have shown that mutations in epidermal growth factor receptor (EGFR) are frequently present in lung adenocarcinoma. While progression-free survival (PFS) is longer when using TKIs than when using chemotherapy in patients harbouring EGFR mutations (6-8), the effectiveness of TKIs is affected by the presence of EGFR mutations (9,10). Therefore, molecular profiling of EGFR or anaplastic lymphoma kinase (ALK) has been recommended as standard care for patients with advanced-stage adenocarcinoma regardless of race, sex, smoking history, or other clinical factors (1). However, obtaining sufficiently high-quality tumor tissues for EGFR testing is challenging in many situations due to a deficiency of biopsy samples and the physical condition of patients. Therefore, a non-invasive, simple, and convenient method to evaluate the presence of EGFR mutations is necessary when managing treatment strategies, especially in making initial treatment decisions.

Positron emission tomography (PET) using 18F-fluoro-2-deoxyglucose (18F-FDG) is a widely applied, non-invasive, functional imaging modality based on differential glucose metabolism and is important for diagnosis, staging, assessment of therapeutic efficacy, and radiotherapy planning in lung cancer (11). High rates of 18F-FDG uptake illustrate both active glucose metabolism and cancer proliferation of malignant cells (12,13). The EGFR signalling pathway regulates glucose metabolism and inhibits apoptosis in EGFR-mutated lung adenocarcinoma cells, and TKIs act to decrease glucose consumption (14,15). Studies have shown that maximal standard uptake values (SUVmax) are associated with EGFR mutational status in both cellular and animal models (16,17). Clinical studies have demonstrated an association between EGFR mutations and 18F-FDG uptake, but the results are conflicting (18-25). We retrospectively analyzed patients with lung adenocarcinoma to investigate whether 18F-FDG PET could be an accurate indicator for predicting EGFR mutations in lung adenocarcinomas. We present the following article in accordance with the STARD reporting checklist (available at https://tcr.amegroups.com/article/view/10.21037/tcr-22-1726/rc).


Methods

The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by committee of Shanxi Province Cancer Hospital (No. 201835). Individual consent for this retrospective analysis was waived.

Study design and inclusion criteria

We retrospectively examined all untreated primary lung adenocarcinoma patients who had undergone EGFR detection and PET/CT simultaneously between January 2013 and December 2017. In total, 432 cases without diseases which influence the standard uptake value (such as diabetes, acute inflammation, and metabolic disease, etc.) were identified, and 297 individuals were ultimately selected. Of these, 135 cases were excluded for one of the following reasons: (I) 73 patients had a history of another malignancy or mixed pathological types; (II) 54 cases had incomplete data; (III) the period between PET/CT and EGFR detection exceeded 1 month in 8 individuals.

The parameters of 18F-FDG PET/CT and clinical information, including age, sex, smoking history, immunohistochemistry (IHC) analysis, EGFR status, and tumor/node/metastasis (TNM) stage were obtained from the hospital’s medical records. Patients who never smoked or had smoked no more than 100 cigarettes in total were defined as never smokers. The patients were divided into a mutant group (n=138) and a wild-type group (n=159) based on EGFR mutational status. TNM staging was based on the International Association for the Study of Lung Cancer (IASLC) 8th TNM Lung Cancer Staging System and clinical testing results. All procedures were performed by at least 2 experienced clinicians according to international guidelines.

18F-FDG PET/CT scanning and analysis

A Discovery STE PET/CT system (GE Healthcare, Waukesha, WI, USA) was used to perform PET/CT. The PET/CT center of Shanxi Cancer Hospital provided 18F-FDG with a radiochemical purity of >95%. Patients fasted for >6 h before examination. The fasting blood glucose concentration was checked and confirmed to be <11 mmol/L before examination. Image scanning was performed 60 min after intravenous injection of 18F-FDG (0.12–0.15 mCi/kg). PET was performed in the 3-dimensional mode using 3.75 mm per slice. CT was performed using the following parameters: 120 kV, 200 mA, 0.8 s/lap, and 22.5 mm/s bed speed. The images were immediately obtained from the top of the skull to the upper femur (6–8 bed positions and 3 min per bed position). The PET images were reconstructed and attenuation-corrected by CT images. The images were then observed on an Xeleris Workstation (GE Healthcare) for assessment. Two experienced nuclear medicine radiologists reviewed all PET/CT data independently and reached a consensus on the results. The reviewers were blinded to the EGFR mutational status.

EGFR mutation analysis

EGFR mutations were detected according to the amplification refractory mutation system (ARMS). Four paraffin-embedded wax slices with a continuous 7-µm thickness were taken to extract DNA using the QIAamp DNA FFPE Kit (Qiagen NV, Venlo, The Netherlands) according to the manufacturer’s instructions. Polymerase chain reaction was performed to detect EGFR mutations using the EGFR 29 Mutation Detection Kit (Amoy Diagnostics, Xiamen, People’s Republic of China). All steps were completed according to the ADx-EGFR detection instructions. The results of EGFR mutation analyses were interpreted by at least 2 experienced pathologists.

IHC analysis

Tissue samples were acquired through surgical resection or biopsy of the primary tumor, primary or distant lymph node, or local or distant metastases. The samples were fixed in 10% neutral formalin and embedded in paraffin wax. Slices 3-µm thick were taken, one of which was used for hematoxylin and eosin (HE staining), and the rest were used for IHC staining. The process was performed using the fully automated Ventana IHC assay. Positive and negative controls were included in each experiment. The results of pathological diagnoses and IHC tests were interpreted by at least 2 experienced pathologists as being positive for a brown or tan nuclear appearance. Positive results for thyroid transcription factor-1 (TTF-1) were defined as >5% of tumor cells with nuclear staining and positive expression of napsin A. Cytokeratin 7 (CK7) positivity was defined as >50% of tumor cells with nuclear staining. The percentage of nuclei staining positive for Ki67 in tumor cells was denoted with a Ki67 score.

Statistical analysis

All data were analyzed using SPSS for Windows (version 22.0). Clinical characteristics, including age, sex, smoking history, IHC results, and staging, were compared according to the EGFR mutational status using the chi-squared test and the t-test. A significant difference was defined as a two-sided P value <0.05. Receiver operating characteristic (ROC) curves were plotted to acquire the cut-off value of maximal standard uptake value of primary tumor (pSUVmax) for predicting EGFR mutations. Logistic regression analysis was used to identify independent predictors of EGFR mutation.

All variables with P<0.05 in the univariate analysis were further analyzed by multivariate regression analysis. Indicators with P<0.05 in the multivariate analysis were considered to be independent predictors, and the odds ratios (OR) and 95% confidence intervals (CI) of the independent predictors were obtained. In addition, ROC curves and area under the curves (AUCs) were plotted for the combined predictors of EGFR mutational status.


Results

Patients and tumor characteristics

Of the 297 lung adenocarcinoma patients analyzed for EGFR status who underwent PET/CT between January 2013 and December 2017, 138 (46.5%) were identified as having mutant EGFR, and 159 participants were wild type. The basic characteristics of the 2 groups are summarized in Table 1. The median age of the 297 patients was 59.27 years (range, 30–88 years), and there were 131 women (44.1%) and 166 men (55.9%). In total, 168 (76.1%) were never smokers. In addition, 66 (22.2%), 31 (10.4%), 59 (19.9), and 141 (47.5%) patients had stage I, II, III, and IV disease, respectively. The mutant group contained 69 cases (50.0%) with an exon 19 mutation, 62 patients (44.9%) with an exon 21 mutation, and 7 cases (5.1%) with rare mutations (Table 1). The median pSUVmax was 9.52 (range, 0.67–39.20).

Table 1

Associations between the 2 groups

Variables EGFR Total (N=297) P value
Mutant (N=138) Wild-type (N=159)
Age (years), mean ± SD [range] 58.95±9.87 [31–83] 59.54±10.52 [30–88] 59.27±10.21 [30–88] 0.619a
Gender, n (%) <0.001b
   Male 48 (34.8) 118 (74.2) 166 (55.9)
   Female 90 (65.2) 41 (25.8) 131 (44.1)
Smoking status, n (%) <0.001b
   Never smoker 105 (76.1) 63 (39.6) 168 (76.1)
   Ever smoker 33 (23.9) 96 (60.4) 129 (43.4)
pSUVmax, mean ± SD 8.24±4.51 10.64±5.77 9.53±5.35 <0.001a
nSUVmax, mean ± SD 5.64±3.77 7.25±4.53 6.58±4.29 0.009a
mSUVmax, mean ± SD 7.20±3.86 7.32±4.72 7.27±4.37 0.870a
Tumor size, mean ± SD 2.89±1.46 3.54±1.90 3.24±1.74 0.001a
Nodal metastasis, n (%) 0.141b
   Negative 51 (37.0) 46 (28.9) 97 (32.7)
   Positive 113 (71.7) 87 (63.0) 200 (67.3)
Distant metastasis, n (%) 0.293b
   Negative 77 (55.8) 79 (49.7) 156 (52.5)
   Positive 61 (44.2) 80 (50.3) 141 (47.5)
Stage, n (%) 0.138b
   I 38 (27.5) 28 (17.6) 66 (22.2)
   II 16 (11.6) 15 (9.4) 31 (10.4)
   III 23 (16.7) 36 (22.6) 59 (19.9)
   IV 61 (44.2) 80 (50.3) 141 (47.5)
TTF-1, n (%) 0.001b
   Negative 2 (4.0) 24 (24.5) 26 (17.6)
   Positive 48 (96.0) 74 (75.5) 122 (82.4)
Napsin A, n (%) <0.001b
   Negative 2 (4.2) 33 (37.5) 35 (25.7)
   Positive 46 (95.8) 55 (82.5) 101 (74.3)
CK7 (n, %) 0.548b
   Negative 0 (0.0) 3 (4.9) 3 (3.3)
   Positive 30 (100.0) 58 (95.1) 88 (96.7)
Ki67 score, mean 35.95±19.07 49.08±23.96 44.60±23.19 0.003a
EGFR mutations, n (%)
   Exon 18 3 (2.2)
   Exon 19 69 (50.0)
   Exon 20 4 (2.9)
   Exon 21 62 (44.9)

a, t-test calculation; b, Chi-Square calculation. EGFR, epidermal growth factor receptor; SD, standard deviation; pSUVmax, maximal standard uptake value of primary tumor; nSUVmax, maximal standard uptake value of lymph node; mSUVmax, maximal standard uptake value of distant metastasis; TTF-1, thyroid transcription factor 1.

Association between EGFR mutations and clinical characteristics

To analyze the association between EGFR mutations and clinical information, the characteristics of the 2 groups were compared. EGFR mutations were detected more frequently in women (65.2% vs. 34.8%, P>0.001), never smokers (76.1% vs. 23.9%, P<0.001), and those with positive expression of TTF-1 (96.0% vs. 4.0%, P=0.001) and napsin A (95.8% vs. 4.2%, P<0.001). In addition, the Ki67 scores were lower in the EGFR-mutant group (35.95±19.07) than in the wild-type group (49.08±23.96, P=0.003; Table 1). The means of the pSUVmax (8.24±4.51 vs. 10.64±5.77; P<0.001) and lymph node SUVmax (5.64±3.77 vs. 7.25±4.53; P=0.009) of PET/CT were lower, respectively, in the mutant group compared to the wild-type group. The mean tumor size of the primary mass (2.89±1.46 vs. 3.54±1.90; P=0.001) was smaller in the mutant group than that in the wild-type group. There were no statistical differences in the parameters of PET/CT among the EGFR-mutation subtypes.

A ROC curve was plotted to calculate the cut-off point (8.20) of the pSUVmax with 64.8% sensitivity, 55.8% specificity, a 62.8% positive predictive value, a 57.9% negative predictive value, and 60.6% accuracy; the AUC was 0.623 (95% CI: 0.560–0.686; P<0.001). The cut-off point for tumor size was 3.5 cm with 46.5% sensitivity, 71.2% specificity, 71.2% positive predictive value, 56.0% negative predictive value, and 61.3% accuracy; the AUC was 0.600 (95% CI: 0.536–0.665; P=0.003). Thus, even though EGFR mutations were detected more frequently in individuals with a pSUVmax <8.2 (P<0.001) and tumor size <3.5 cm, the 2 indicators had a marginal predictive value for EGFR mutations.

Prediction of EGFR mutations

To confirm the predictive factors of EGFR mutations, univariate and multivariate logistic regression analyses were used. Univariate regression analysis indicated that sex, smoking status, tumor size, pSUVmax, and stage were correlated with EGFR mutations. Multivariate regression indicated that sex, smoking status, tumor size, and pSUVmax remained independent factors for predicting EGFR mutation. Women (OR =2.853; 95% CI: 1.451–5.611; P=0.002), never smokers (OR =2.414; 95% CI: 1.217–4.789; P=0.012), tumor size <3.5 cm (OR =2.170; 95% CI: 1.205–3.908; P=0.010), and pSUVmax <8.2 (OR =1.904; 95% CI: 1.098–3.302; P=0.022) were effective predictors of EGFR mutation (Table 2). In addition, the AUCs of the pSUVmax and tumor size were 0.623 and 0.600, respectively. When using pSUVmax combined with tumor size to calculate the ROC curve, the AUC was 0.656. Combined with clinical characteristics, including sex and smoking status, the AUC of the 4 predictors increased to 0.770 (Figure 1).

Table 2

Univariate and multivariate analysis of various predictive indicators for EGFR status

Characteristics Univariate analysis Multivariate analysis
OR (95% CI) P value OR (95% CI) P value
Age 0.230
   <60 years Reference
   ≥60 years 1.323 (0.838–2.090)
Sex <0.001 0.002
   Male Reference Reference
   Female 5.396 (3.276–8.888) 2.853 (1.451–5.611)
Smoking status <0.001 0.012
   Never 4.848 (2.929–8.026) 2.414 (1.217–4.789)
   Ever Reference Reference
Tumor size <0.001 0.010
   <3.5 cm 3.314 (1.881–5.222) 2.170 (1.205–3.908)
   ≥3.5 cm Reference Reference
pSUVmax <0.001 0.022
   <8.2 2.322 (1.454–3.706) 1.904 (1.098–3.302)
   ≥8.2 Reference Reference
Nodal metastasis 0.142
   Negative Reference
   Positive 0.694 (0.427–1.130)
Distant metastasis 0.293
   Negative Reference
   Positive 0.782 (0.495–1.236)
Stage 0.046
   I Reference
   II 0.786 (0.334–1.852) 0.582
   III 0.471 (0.230–0.963) 0.039
   IV 0.562 (0.311–1.015) 0.056

EGFR, epidermal growth factor receptor; OR, odds ratio; CI, confidence interval; pSUVmax, maximal standard uptake value of primary tumor.

Figure 1 ROC curves of EGFR mutation. pSUVmax gained 0.623 (continuous line) of AUC, and the value increased to 0.770 when combined with all predictive factors (gender, smoking status, tumor size, and pSUVmax, dotted line). P value <0.05. AUC, area under curve; ROC, receiver operating characteristic; EGFR, epidermal growth factor receptor; pSUVmax, maximal standard uptake value of primary tumor.

Discussion

TKIs have played an important role in improving the outcomes of a particular group of patients and enabled a significant paradigm shift in the therapeutic agent management of NSCLC (1-5). EGFR mutations are mostly found in lung adenocarcinoma, which is the predominant subtype of lung cancer (24). Nevertheless, due to the deficiency of biopsy samples and the physical condition of patients, obtaining sufficient high-quality tumor tissues for EGFR testing remains challenging in many situations. Therefore, a noninvasive, simple, and convenient method to detect the presence of EGFR mutations is needed for managing treatment strategies, especially when making initial treatment decisions. In this study, we showed that lung adenocarcinoma patients harboring EGFR mutations had lower pSUVmax and smaller tumor size than wild-type cases according to 18F-FDG PET/CT. As a noninvasive method, pSUVmax and tumor size based on PET/CT together with clinical characteristics such as gender and smoking status could enhance EGFR status discriminability in those patients for whom EGFR detection is not feasible.

Previous studies have revealed a relationship between EGFR mutations and 18F-FDG uptake, but the results have been conflicting (18-29). The majority of studies demonstrated that a lower pSUVmax was an indicator for predicting EGFR mutations in either lung adenocarcinomas or NSCLC, regardless of tumor stage (18-25). The present study also showed a lower pSUVmax was an independent indicator in lung adenocarcinoma. Our results were consistent with the findings of multiple studies (18-25) but contradicted with 2 studies (26,27). Notably, one of the 2 studies that showed a higher pSUVmax was a predictor of EGFR mutations involved 132 advanced adenocarcinoma (ADC) cases regardless of tumor stage (26), while the other study included 77 stage III and IV ADC patients (27). Further, retrospective analyses conducted by Lee et al. (28) and Caicedo et al. (29) found there was no relationship between pSUVmax and EGFR mutations, which also differed from our results. Further studies of ADC patients in stage IV demonstrated that there was no difference in pSUVmax between EGFR-mutant patients and wild-type patients (21,28).

Several possible factors may have contributed to these contradictory results. First, these studies involved either NSCLC or lung adenocarcinomas with different tumor stages. Thus, the histological type and tumor stage of the included cases could have influenced the results. Second, of the 2 studies showing results that were contrary to ours (26,27), one included 132 ADC cases regardless of tumor stage and also required pretreatment serum indicator and CT assessments (26). The other study, which involved 77 cases of stage III and IV ADC patients, reported an EGFR-mutation rate of 64% (27). The pretreatment requirement, small sample size, and high EGFR mutation rate of both studies may reflect the bias in patient selection. Moreover, although pSUVmax was significantly related to EGFR mutation in univariate regression analysis, no relationship was found in multivariate regression analysis in the study conducted by Lee et al. (28). In our study, despite the fact that pSUVmax showed predictive value in both univariate and multivariate regression analysis, the AUC was only 0.623, which was consistent with the results of Lv et al. (AUC =0.557) (21). Hence, the marginal discriminating ability of pSUVmax could have led to discrepancy among results.

The results of tumor size as a predictive indicator for EGFR mutations were also conflicting and there are several possible reasons for this (21,28,30). The studies performed by Lee et al. (28) and Guan et al. (30) showed a smaller tumor size could predict EGFR mutations in both univariate and multivariate regression analysis, which was consistent with our study. The results from Lv et al. (21) revealed no relationship between tumor size and EGFR status in a separate analysis of an ADC-only group. However, tumor size was smaller in EGFR-mutant individuals than that in wild-type patients when ADC and non-ADC cases were analyzed as a whole in Lv’s study. In our study, the rate of EGFR mutations was 46.5%, and a smaller tumor size was weakly associated with EGFR mutations (AUC =0.600). Further, due to the limited availability of testing technology, the detection rate of EGFR mutation was only 9.6% in China about a decade ago (31). One large-sample survey reported the EGFR-mutation rate was 42.5% in a noninterventional, real-world study of IIIB/IV NSCLC patients in northern China (32). However, patients with early-stage disease and a sound economic background were more likely to undergo genetic detection. Therefore, the modest discriminating power of tumor size and genetic-testing patient selection bias may have affected the results.

Although detecting EGFR mutations from tissue is the gold standard for therapeutic agent management using EGFR-TKIs for first-line treatment, sufficient tissue for EGFR-mutation detection is not available in many situations. Previous randomized head-to-head data (33,34) have demonstrated the potential benefit of EGFR-TKIs in patients who did not undergo EGFR-status detection and were selected based on clinical characteristics only. More and more diagnoses of NSCLC rely on cytologic specimens or small biopsies which result in insufficient quality and quantity of tumor cells for EGFR detection. A retrospective study including 11 Asian Pacific countries showed 71.4% (53.8% in China) of samples used for detecting EGFR mutations were cytology samples and/or small biopsies (35). Overall failure rates for EGFR testing using cytology samples and/or small biopsies have been reported to be 5–30% (36,37). Therefore, EGFR-mutation detection from tissue rather than cytology samples and/or small biopsies is highly recommended before first-line treatment with EGFR-TKIs. In addition to the limitations of clinical resources and testing technology mentioned above, the subjective willingness of patients also restrict invasive examination and obtainment of sufficient high-quality tumor tissues for EGFR testing.

While EGFR-mutation testing from tissue is important, noninvasive methods to predict EGFR mutations is also necessary when tissue detection is not available in order to identify patients likely to benefit from EGFR-TKIs. This study aimed to investigate the predictive value of pSUVmax for EGFR mutations in those patients for whom EGFR testing was not feasible. The results showed that a lower pSUVmax and a smaller tumor size could be helpful for distinguishing EGFR mutations from wild type. Although both indicators had marginal predictive value for EGFR mutations, they could be combined with other clinical characteristics to enhance patient stratification in cases where EGFR detection is not feasible. Reasonable application of clinical factors and parameters of PET/CT could be used to manage therapeutic strategies in lung adenocarcinomas without available EGFR detection.

Our study had several limitations. Above all, the retrospective nature of the study may have led to sample availability bias and patient selection bias. The nature of a single-center study can also induce bias in the results. Thus, larger samples and multicenter prospective studies should be performed to validate the results. In addition, maximal standard uptake value of lymph node (nSUVmax) and maximal standard uptake value of distant metastasis (mSUVmax) were not included in this study for patients without lymph node and distant metastasis. Hence, individuals with a similar background (stage of disease, race, gender, smoking status, etc.) should be compared to verify the results. Finally, the study did not detect all mutations, and several drive mutations may exist in a patient concurrently. Each driver mutation may influence the glucose metabolism process through different signaling pathway activations. Therefore, more comprehensive genetic detection is needed to confirm the results.


Conclusions

This study aimed to reveal whether pSUVmax could be a noninvasive indicator to predict EGFR mutations in untreated primary lung adenocarcinomas. The results identified that pSUVmax <8.2 was an independent predictor of EGFR mutations, and the AUC was 0.629. When using all the predictors, including female, never smoker, tumor size <3.5 cm, and pSUVmax <8.2, the AUC increased to 0.770. Other biomarkers could be integrated with the parameters of PET/CT to predict EGFR status and efficiency of TKI treatment in the future in cases where genetic detection is not feasible.


Acknowledgments

Funding: This work was supported by the Youth Foundation of Health Commission of Shanxi Province (No. 2020058) and the Youth Foundation of Science and Technology Commission of Shanxi Province (No. 202103021223447).


Footnote

Reporting Checklist: The authors have completed the STARD reporting checklist. Available at https://tcr.amegroups.com/article/view/10.21037/tcr-22-1726/rc

Data Sharing Statement: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-22-1726/dss

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-22-1726/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by committee of Shanxi Province Cancer Hospital (No. 201835). Individual consent for this retrospective analysis was waived.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Lindeman NI, Cagle PT, Beasley MB, et al. Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J Mol Diagn 2013;15:415-53. [Crossref] [PubMed]
  2. Wu SG, Shih JY. Management of acquired resistance to EGFR TKI-targeted therapy in advanced non-small cell lung cancer. Mol Cancer 2018;17:38. [Crossref] [PubMed]
  3. Jänne PA, Yang JC, Kim DW, et al. AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer. N Engl J Med 2015;372:1689-99. [Crossref] [PubMed]
  4. Buddharaju LNR, Ganti AK. Immunotherapy in lung cancer: the chemotherapy conundrum. Chin Clin Oncol 2020;9:59. [Crossref] [PubMed]
  5. Ettinger DS, Wood DE, Aisner DL, et al. Non-Small Cell Lung Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2022;20:497-530. [Crossref] [PubMed]
  6. Akamatsu H, Katakami N, Okamoto I, et al. Osimertinib in Japanese patients with EGFR T790M mutation-positive advanced non-small-cell lung cancer: AURA3 trial. Cancer Sci 2018;109:1930-8. [Crossref] [PubMed]
  7. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 2009;361:947-57. [Crossref] [PubMed]
  8. Pao W, Miller V, Zakowski M, et al. EGF receptor gene mutations are common in lung cancers from "never smokers" and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc Natl Acad Sci U S A 2004;101:13306-11. [Crossref] [PubMed]
  9. Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med 2010;362:2380-8. [Crossref] [PubMed]
  10. Mitsudomi T, Morita S, Yatabe Y, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol 2010;11:121-8. [Crossref] [PubMed]
  11. Stroobants S, Verschakelen J, Vansteenkiste J. Value of FDG-PET in the management of non-small cell lung cancer. Eur J Radiol 2003;45:49-59. [Crossref] [PubMed]
  12. Higashi K, Ueda Y, Yagishita M, et al. FDG PET measurement of the proliferative potential of non-small cell lung cancer. J Nucl Med 2000;41:85-92. [PubMed]
  13. Vesselle H, Schmidt RA, Pugsley JM, et al. Lung cancer proliferation correlates with F-18fluorodeoxyglucose uptake by positron emission tomography. Clin Cancer Res 2000;6:3837-44. [PubMed]
  14. Makinoshima H, Takita M, Matsumoto S, et al. Epidermal growth factor receptor (EGFR) signaling regulates global metabolic pathways in EGFR-mutated lung adenocarcinoma. J Biol Chem 2014;289:20813-23. [Crossref] [PubMed]
  15. Baselga J. Why the epidermal growth factor receptor? The rationale for cancer therapy. Oncologist 2002;7:2-8. [Crossref] [PubMed]
  16. Sun X, Xiao Z, Chen G, et al. A PET imaging approach for determining EGFR mutation status for improved lung cancer patient management. Sci Transl Med 2018;10:eaan8840. [Crossref] [PubMed]
  17. De Rosa V, Iommelli F, Monti M, et al. Reversal of Warburg Effect and Reactivation of Oxidative Phosphorylation by Differential Inhibition of EGFR Signaling Pathways in Non-Small Cell Lung Cancer. Clin Cancer Res 2015;21:5110-20. [Crossref] [PubMed]
  18. Na II, Byun BH, Kim KM, et al. 18F-FDG uptake and EGFR mutations in patients with non-small cell lung cancer: a single-institution retrospective analysis. Lung Cancer 2010;67:76-80. [Crossref] [PubMed]
  19. Cho A, Hur J, Moon YW, et al. Correlation between EGFR gene mutation, cytologic tumor markers, 18F-FDG uptake in non-small cell lung cancer. BMC Cancer 2016;16:224. [Crossref] [PubMed]
  20. Mak RH, Digumarthy SR, Muzikansky A, et al. Role of 18F-fluorodeoxyglucose positron emission tomography in predicting epidermal growth factor receptor mutations in non-small cell lung cancer. Oncologist 2011;16:319-26. [Crossref] [PubMed]
  21. Lv Z, Fan J, Xu J, et al. Value of 18F-FDG PET/CT for predicting EGFR mutations and positive ALK expression in patients with non-small cell lung cancer: a retrospective analysis of 849 Chinese patients. Eur J Nucl Med Mol Imaging 2018;45:735-50. [Crossref] [PubMed]
  22. Takamochi K, Mogushi K, Kawaji H, et al. Correlation of EGFR or KRAS mutation status with 18F-FDG uptake on PET-CT scan in lung adenocarcinoma. PLoS One 2017;12:e0175622. [Crossref] [PubMed]
  23. Chen L, Zhou Y, Tang X, et al. EGFR mutation decreases FDG uptake in non-small cell lung cancer via the NOX4/ROS/GLUT1 axis. Int J Oncol 2019;54:370-80. [PubMed]
  24. Kim YI, Paeng JC, Park YS, et al. Relation of EGFR Mutation Status to Metabolic Activity in Localized Lung Adenocarcinoma and Its Influence on the Use of FDG PET/CT Parameters in Prognosis. AJR Am J Roentgenol 2018;210:1346-51. [Crossref] [PubMed]
  25. Gu J, Xu S, Huang L, et al. Value of combining serum carcinoembryonic antigen and PET/CT in predicting EGFR mutation in non-small cell lung cancer. J Thorac Dis 2018;10:723-31. [Crossref] [PubMed]
  26. Ko KH, Hsu HH, Huang TW, et al. Value of 18F-FDG uptake on PET/CT and CEA level to predict epidermal growth factor receptor mutations in pulmonary adenocarcinoma. Eur J Nucl Med Mol Imaging 2014;41:1889-97. Erratum in: Eur J Nucl Med Mol Imaging 2014;41:1989-90. [Crossref] [PubMed]
  27. Huang CT, Yen RF, Cheng MF, et al. Correlation of F-18 fluorodeoxyglucose-positron emission tomography maximal standardized uptake value and EGFR mutations in advanced lung adenocarcinoma. Med Oncol 2010;27:9-15. [Crossref] [PubMed]
  28. Lee SM, Bae SK, Jung SJ, et al. FDG uptake in non-small cell lung cancer is not an independent predictor of EGFR or KRAS mutation status: a retrospective analysis of 206 patients. Clin Nucl Med 2015;40:950-8. [Crossref] [PubMed]
  29. Caicedo C, Garcia-Velloso MJ, Lozano MD, et al. Role of [18F]FDG PET in prediction of KRAS and EGFR mutation status in patients with advanced non-small-cell lung cancer. Eur J Nucl Med Mol Imaging 2014;41:2058-65. Erratum in: Eur J Nucl Med Mol Imaging 2014;41:2164. [Crossref] [PubMed]
  30. Guan J, Chen M, Xiao N, et al. EGFR mutations are associated with higher incidence of distant metastases and smaller tumor size in patients with non-small-cell lung cancer based on PET/CT scan. Med Oncol 2016;33:1. [Crossref] [PubMed]
  31. Xue C, Hu Z, Jiang W, et al. National survey of the medical treatment status for non-small cell lung cancer (NSCLC) in China. Lung Cancer 2012;77:371-5. [Crossref] [PubMed]
  32. Cheng Y, Wang Y, Zhao J, et al. Real-world EGFR testing in patients with stage IIIB/IV non-small-cell lung cancer in North China: A multicenter, non-interventional study. Thorac Cancer 2018;9:1461-9. [Crossref] [PubMed]
  33. Han JY, Park K, Kim SW, et al. First-SIGNAL: first-line single-agent iressa versus gemcitabine and cisplatin trial in never-smokers with adenocarcinoma of the lung. J Clin Oncol 2012;30:1122-8. [Crossref] [PubMed]
  34. Fukuoka M, Wu YL, Thongprasert S, et al. Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS). J Clin Oncol 2011;29:2866-74. [Crossref] [PubMed]
  35. Yatabe Y, Kerr KM, Utomo A, et al. EGFR mutation testing practices within the Asia Pacific region: results of a multicenter diagnostic survey. J Thorac Oncol 2015;10:438-45. [Crossref] [PubMed]
  36. Shiau CJ, Babwah JP, da Cunha Santos G, et al. Sample features associated with success rates in population-based EGFR mutation testing. J Thorac Oncol 2014;9:947-56. [Crossref] [PubMed]
  37. Leary AF, Castro DG, Nicholson AG, et al. Establishing an EGFR mutation screening service for non-small cell lung cancer - sample quality criteria and candidate histological predictors. Eur J Cancer 2012;48:61-7. [Crossref] [PubMed]

(English Language Editor: A. Muylwyk)

Cite this article as: Wang J, Wen X, Yang G, Cui Y, Hao M, Qiao X, Jin B, Li B, Wu J, Li X, Ren X. The predictive value of 18F-FDG PET/CT in an EGFR-mutated lung adenocarcinoma population. Transl Cancer Res 2022;11(7):2338-2347. doi: 10.21037/tcr-22-1726

Download Citation