Skip to main content

C-Myc protein expression indicates unfavorable clinical outcome in surgically resected small cell lung cancer

Abstract

Background

By being highly involved in the tumor evolution and disease progression of small cell lung cancer (SCLC), Myc family members (C-Myc, L-Myc, and N-Myc) might represent promising targetable molecules. Our aim was to investigate the expression pattern and prognostic relevance of these oncogenic proteins in an international cohort of surgically resected SCLC tumors.

Methods

Clinicopathological data and surgically resected tissue specimens from 104 SCLC patients were collected from two collaborating European institutes. Tissue sections were stained by immunohistochemistry (IHC) for all three Myc family members and the recently introduced SCLC molecular subtype-markers (ASCL1, NEUROD1, POU2F3, and YAP1).

Results

IHC analysis showed C-Myc, L-Myc, and N-Myc positivity in 48%, 63%, and 9% of the specimens, respectively. N-Myc positivity significantly correlated with the POU2F3-defined molecular subtype (r = 0.6913, p = 0.0056). SCLC patients with C-Myc positive tumors exhibited significantly worse overall survival (OS) (20 vs. 44 months compared to those with C-Myc negative tumors, p = 0.0176). Ultimately, in a multivariate risk model adjusted for clinicopathological and treatment confounders, positive C-Myc expression was confirmed as an independent prognosticator of impaired OS (HR 1.811, CI 95% 1.054–3.113, p = 0.032).

Conclusions

Our study provides insights into the clinical aspects of Myc family members in surgically resected SCLC tumors. Notably, besides showing that positivity of Myc family members varies across the patients, we also reveal that C-Myc protein expression independently correlates with worse survival outcomes. Further studies are warranted to investigate the role of Myc family members as potential prognostic and predictive markers in this hard-to-treat disease.

Introduction

Small cell lung cancer, one of the most aggressive types of lung cancer, is characterized by a rapid doubling time, strong predilection for early metastasis and dismal prognosis [1]. Worldwide, more than 300,000 new small cell lung cancer (SCLC) cases are being detected [2]. Despite all efforts to improve the clinical outcomes, the prognosis of SCLC patients did not change significantly over the past decades and the 5-year survival rates are still way below 10% [3]. Accordingly, new and more effective therapeutic strategies are clearly needed for this devastating disease.

Although SCLC has long been regarded both clinically and biologically as a homogenous entity, recent evidence arising from preclinical and clinical profiling studies indicates that SCLCs can be divided into distinct molecular subtypes. This novel subtype classification is primarily based on the relative expression of four key transcriptional factors: ASCL1 (SCLC-A subtype), NEUROD1 (SCLC-N), POU2F3 (SCLC-P), and YAP1 (SCLC-Y) [4]. Of note, beyond their prognostic significance, these major SCLC subtypes have distinct neuroendocrine profiles and unique therapeutic vulnerabilities [4,5,6,7]. Moreover, it has been recently indicated that the presence of these molecular subtypes is greatly influenced by strong intra-tumoral heterogeneity and that a temporal plasticity between the subtypes might also exists [5]. Importantly, we are only at the beginning to understand whether tumor evolution between subtypes reflects lineage plasticity or differential selection among pre-existing subclones due therapy. Nevertheless, it is anticipated that besides the cytotoxic effects of chemotherapy, Myc oncogene family members also contribute to this dynamic progress of subtype switching [5, 8, 9].

The Myc family consists of three distinct members (MYC, MYCL, and MYCN), each of them coding for three distinct transcription factors (C-Myc, L-Myc, and N-Myc). As for its functions, Myc regulates a wide range of physiological cellular processes such as cell cycle, growth, or metabolism and strongly contributes to the progression of most human tumors including SCLC [10, 11]. Notably, Myc family members have been previously demonstrated to be critical factors in SCLC initiation and progression both in vitro and in vivo [9]. Moreover, they represent potent regulators of several clinically relevant processes, such as the development of chemoresistance, or the temporal evolution of SCLC molecular subtypes [8, 12, 13].

Interestingly, increased Myc expression has been demonstrated to associate with poor prognosis in various malignancies [14,15,16]. While targeting oncogenic Myc has been considered a promising novel therapeutic strategy for malignancies, finding effective ways for its inhibition has remained a challenge for decades [17]. Fortunately, recent efforts have resulted in the development of highly effective direct Myc inhibitors and initial clinical trials have already started to evaluate the feasibility of certain Myc inhibitors in patients with various malignancies [18, 19]. Importantly, there is a clear rationale for targeting Myc in SCLC as well, and the first preclinical results concerning the efficacy of Myc-inhibition are indeed promising [20, 21]. Here, we evaluated the expression profile and clinical relevance of Myc, as well as its co-expression pattern with the subtype-defining transcription factors in a cohort of surgically-treated Caucasian SCLC.

Materials and methods

Patients and study design

In this multi-institutional retrospective study, formalin-fixed paraffin-embedded (FFPE) SCLC blocks and corresponding clinicopathological data were collected from patients undergoing surgery between January 2000 and December 2019 at the Medical University of Vienna (Austria) or in the National Koranyi Institute of Pulmonology (Hungary). Clinical follow-up and survival data were retrospectively extracted from institutional medical records and/or archives of the national statistical offices. Notably, all patients have been clinically managed based on the recommendations of a multidisciplinary tumor-board consisting of board-certified pulmonary-, radiation-, and surgical-oncologists in accordance with the recommendations of the contemporary National Comprehensive Cancer Network (NCCN) guidelines [22]. In particular, routine preoperative staging included the following conventional radiological examinations: whole-body computed tomography scan (CT), CT or magnetic-resonance-imaging (MRI) scan of the brain, whole-body fluorodeoxyglucose (FDG)-positron emission tomography (PET), or whole-body skeletal scintigraphy. Adjuvant therapy was defined as when at least 4 cycles of adjuvant chemotherapy have been conducted either in combination with adjuvant irradiation or without adjuvant irradiation. Chemotherapy regimens comprised platinum-based agents (cisplatin or carboplatin) in combination with the topoisomerase-inhibitor etoposide. Routine oncological follow-up was carried out for all patients, comprising of regular (i.e., every 3 months in the first postoperative year and then every 6–12 months) appointments in an ambulatory setting including blood tests, X-rays, and/or thoracic CT scans. If recurrence or metastasis (as defined below) was suspected, further clinical evaluations (e.g., bronchoscopical/ultra-sound/CT-guided transthoracic biopsy, follow-up PET-CT/MRI scan or skeletal scintigraphy) were performed according to the individual scenario. Information on overall survival (OS) was provided by the institutional statistical offices and calculated as the elapsed time between surgery and death or last follow-up in months. Information on disease-free survival (DFS) was achieved from the medical records and calculated as the time between surgery and clinical evidence of recurrent disease (defined as tumor recurrence in mediastinal/hilar lymph nodes/ipsilateral lung) or distant organ metastases in months.

Surgically resected samples, immunohistochemistry and evaluation

FFPE blocks were cut into 4-µm-thick sections and stained with commercially available antibodies according to the manufacturer’s protocols for C-Myc (Abcam AB32072, dilution 1:100), L-Myc (Thermofisher PA5-41114, dilution 1:200), N-Myc (Cell Signaling D42BY, dilution 1:100), ASCL1 (BD Bioscience #555604, dilution 1:50), NEUROD1 (Abcam AB213725, dilution 1:100), YAP1 (Cell Signaling Technology #4912, dilution 1:200), and POU2F3 (Santa Cruz Biotechnology SC-293402, dilution 1:100). Briefly, after deparaffinization and incubation of the sections with 0.3% H2O2 for 30 min at room temperature, primary antibodies were applied on the tissue slides overnight at 4 °C. On the next day, tissue slides were incubated with secondary antibodies for 1 h at room temperature. Finally, DAB + Substrate Chromogen System (Dako, K3469) and counterstaining with hematoxylin was performed to visualize the staining results. Ultimately, all sections were scanned using the 3DHistech Midi Scanner System in order to provide digital evaluation by the Panoramic CaseViewer Software (Budapest, Hungary, Version 2.4.0.119028). Immunohistochemistry (IHC) results were independently evaluated and scored by two board-certified pathologists with high experience in pulmonary pathology. In case of relevant differences between the two ratings, the slide was re-evaluated. Notably, there are currently no standardized protocols for the evaluation of C-Myc, L-Myc, N-Myc, ASCL1, NEUROD1, YAP1, or POU2F3 expressions for surgically resected SCLC specimens. Therefore, we defined C-Myc, L-Myc, N-Myc, ASCL1, NEUROD1, YAP1, and POU2F3 expressions as positive if more than 1% of the tumor cells demonstrated prominent nucleocytoplasmic staining or as previously described [5].

Statistical analysis

Continuous variables were presented as median, and in distinct cases (e.g., age of the study cohort), as range including minimum and maximum values. Categorical variables were presented as total numbers and frequencies, and the groups were compared by using the Chi-square test. If the expected frequency was < 5 in one group, the Fisher’s exact test was applied. Correlation analysis between categorical variables was achieved by using Pearson’s chi-squared and Cramér’s V approach.

For risk factor estimation, univariate Cox regression model analysis was applied and the outcomes were presented as hazard ratios (HR) including the corresponding 95% confidence intervals (CI). All risk factors with significant results on univariate analysis were included for multivariate analysis to be tested for robustness and independency. For survival curve estimation of OS and DFS, the Kaplan–Meier method was applied. For comparison of significant differences of Kaplan–Meier curves, the log-rank test was used. All tests were calculated in a two-sided manner and p values below 0.05 were considered statistically significant. In this study, all statistical and graphical illustrations were done by using GraphPad Prism 8 (GraphPad Software, Inc., USA) and SPSS Statistics Version 27.0 (IBM Corp., USA).

Results

Demographic description of the study cohort

In total, 104 patients were included in the current study (Table 1). Median age of the patients at time of surgery was 64 (range 41–83) years. Fifty-four patients (52%) were male and 87 (83.7%) were former or current tobacco smokers. Hypertension (50.9%), chronic obstructive pulmonary disease (COPD, 41.3%), and diabetes mellitus (16.3%) were the most common comorbidities within the study cohort. Regarding the type of surgical resection, lobectomy (50.9%) was the most commonly performed procedure, followed by segmentectomy (13.5%), wedge resection (11.5%), and pneumonectomy (10.6%). According to the histopathological findings, 57 (54.8%) patients had limited-stage (= TNM stage I or II) SCLC, 44 (42.3%) patients had negative lymph nodes (= N0), and 42 (40.4%) individuals had T1 lesions. The majority of the study cohort (n = 63, 60.6%) received adjuvant treatment (n = 54 adjuvant chemotherapy without radiotherapy, n = 2 radiotherapy without chemotherapy, and n = 7 combined chemo-radiotherapy). All patients had an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1. Ultimately, the most common molecular SCLC subtypes among the evaluated specimens was SCLC-A (ASCL1-dominant) with 54%, SCLC-QN (quadruple negative) with 27%, SCLC-AN (combined ASCL1/NEUROD1-dominant) and SCLC-P (POU2F3) with 7%, and SCLC-N (NEUROD1) with 6% of the cases (Fig. 1). Notably, no unique SCLC-Y (YAP1-dominant) subtype was identified.

Table 1 Clinicopathological characteristics of the study cohort. In total, 104 individuals undergoing surgical resection for SCLC were included in this study
Fig. 1
figure 1

Frequency and distribution of major molecular SCLC subtypes within the study cohort after immunohistochemistry for ASCL1, NEUROD1, POU2F3, and YAP1. (SCLC-A: ASCL1-dominant subtype; SCLC-QN: quadruple-negative subtype; SCLC-AN: combined ASCL1/NEUROD1-dominant subtype; SCLC-P: POU2F3-dominant subtype; SCLC-N: NEUROD1-dominant subtype)

Impact of clinicopathological characteristics on clinical outcome

Next, we aimed to determine whether distinct clinicopathological features of surgically treated SCLC patients associate with the postoperative outcome (Fig. 2). Male patients had significantly worse OS than female individuals (median OSs were 29 and 44 months, respectively, p = 0.0054). Similarly, patients with COPD had significantly shorter median OS (vs. non-COPD patients, 30 vs. 44 months, p = 0.0266). In contrast, patients receiving adjuvant treatment had a remarkably improved OS compared to patients undergoing surgery alone (40 vs. 16 months, p = 0.0066). Finally, none of the clinicopathological characteristics showed a statistically significant association with disease-free survival (DFS). However, patients with more advanced lymph node status (N1 or N2 vs. N0, 13 vs. 30 months, p = 0.1709) and patients receiving sublobar resection (i.e., wedge resection or anatomic segmentectomy) showed a tendency toward shorter DFS (compared to patients receiving lobectomy or pneumonectomy, 10 vs. 29 months, p = 0.0694).

Fig. 2
figure 2

Clinicopathological features correlating with overall (OS) or disease-free survival (DFS) following surgery in SCLC patients. Men (A), patients with COPD (B), and those not receiving any type of adjuvant therapy (C) tended to have significantly worse OS. Patients with advanced lymph node stage (D) and those receiving sublobar resection (defined as wedge resection or segmentectomy (E), showed a statistically non-significant but clear tendency toward shorter DFS

Expression patterns of Myc and their association with clinicopathological characteristics

Positive C-Myc expression by IHC was found in 48% of all cases (Fig. 3). L-Myc expression was even more pronounced present with positive rate of 63%. In contrast, N-Myc expression was clearly weaker since 9% of all specimens were classified as positive for N-Myc. Although none of the Myc family members were associated with distinct demographic features (Supplementary Table 1), N-Myc positivity significantly correlated with the SCLC-P (POU2F3 dominant) subtype (Pearson correlation: 0.6913, p = 0.0056, Table 2).

Fig. 3
figure 3

Representative images of C-Myc, L-Myc, and N-Myc expression detected by IHC in surgically resected SCLC specimens. The pie charts illustrate the relative frequencies of positivity concerning distinct MYC family members. Representative negative controls are shown at the bottom

Table 2 Correlation between Myc family member positivity and major molecular SCLC subtypes within the study cohort

Prognostic value of Myc and validation of independent predictors for clinical outcome

Finally, we evaluated the prognostic value of Myc members in surgically resected SCLC patients. Regarding OS, positive C-Myc expression was significantly associated with worse overall outcome (20 months vs. 44 months compared to C-Myc negative patients, p = 0.0176, Fig. 4). Likewise, C-Myc positivity correlated with poor DFS (14 months vs. 32 months compared to C-Myc negative cases, p = 0.0842) by just nearly missing the level of statistical significance. In contrast, neither the expression of L-Myc or N-Myc demonstrated an association with OS or DFS.

Fig. 4
figure 4

Kaplan–Meier survival curves for overall survival (AC) and disease-free survival (DF) according to the IHC protein expression status of MYC family members in surgically resected SCLC

Ultimately, in order to test whether C-Myc positivity influences the survival outcomes independently of other clinicopathological variables, we performed univariate and multivariate analysis for all available characteristics for OS (Fig. 5 and Supplementary Table 2) and DFS (Fig. 6 and Supplementary Table 3). In univariate analysis, gender (male vs. female, hazard ratio (HR) = 1.954, confidence interval (CI) 95% 1.209–3.158, p = 0.006), COPD (HR 1.712, CI 95% 1.059–2.77, p = 0.028), and positive C-Myc expression (HR 1.774, CI 95% 1.099–2.865, p = 0.019) were clear risk factors for shorter OS while adjuvant treatment (HR 0.456, CI 95% 0.255–0.815, p = 0.008) represented a significant predictor of longer OS. In multivariate analysis, only positive C-Myc expression (HR 1.811, CI 95% 1.054–3.113, p = 0.032) and adjuvant treatment (HR 0.374, CI 95% 0.193–0.726, p = 0.004) remained reliable prognostic factors for OS.

Fig. 5
figure 5

Forest map of univariate analysis for the evaluation of distinct clinicopathological features and differential Myc family members’ expression as predictors of overall survival (OS) following surgical resection in SCLC. Detailed results are included in Supplementary Table 2. Forest map of multivariate analysis for the evaluation of distinct clinicopathological features and differential Myc family members’ expression as predictors of overall survival (OS) following surgical resection in SCLC. Detailed results are included in Supplementary Table 2

Fig. 6
figure 6

Univariate analysis for the evaluation of distinct clinicopathological factors and differential Myc family members’ expression status as predictors of disease-free survival (DFS) following surgical resection in SCLC. Detailed results are included in Supplementary Table 3

Regarding DFS, univariate analysis did not demonstrate a statistically significant prognostic value for distinct clinicopathological factors or expression of any Myc member. Nevertheless, lobar resection (vs. sublobar resection, HR 0.348, CI 95% 0.106–1.148, p = 0.083) slightly missed the level of significance as a favorable predictor for DFS. In addition, univariate analysis also revealed a statistically non-significant but clearly increased risk for shorter DFS in case of C-Myc positivity (HR 2.770, CI 95% 0.829–9.259, p = 0.098).

Discussion

In this study, we investigated the protein expression and prognostic impact of Myc family members in an international cohort of 104 surgically treated SCLC patients. C-Myc, L-Myc, and N-Myc showed heterogeneous expressions with the highest positivity rate for L-Myc, followed by C-Myc and N-Myc. In addition, positive C-Myc expression was identified as a novel prognostic biomarker robustly indicating impaired clinical outcome among surgically managed SCLC patients.

Therapeutic opportunities in various thoracic malignancies have gone through a remarkable improvement with continuously increasing OS rates over the past decades [23,24,25]. In contrast, only few therapeutic advancements with very modest survival benefits have been added to the treatment armamentarium in SCLC. Indeed, SCLC has remained one of the most disastrous thoracic malignancies with 5-year survival rates constantly plateauing below 10% since decades [26]. Although surgical resection combined with adjuvant platinum-based chemotherapy has been widely proved to offer acceptable long-term results, only a small number of SCLC patients can be treated with this approach [27, 28]. Unfortunately, the vast majority of SCLC patients already has advanced disease and metastasis at the time of initial diagnosis when local resection is not feasible anymore [29]. Inevitably, this results in a low number of surgically managed SCLC patients and thus a limited availability of surgically resected whole tissue specimens for research purposes. Regarding our study cohort, the overall number of a comparatively large group of 104 whole tissue (WTS) specimens of SCLC patients was achieved through a collaboration between two high-volume thoracic surgical departments. Also, we selected a study period of 20 years and decided to include specimens even from advanced stage SCLC patients to increase the total number of available specimens. Interestingly, we found remarkable associations between distinct clinicopathological characteristics and clinical outcomes. Male patients, patients with COPD and those not receiving an adjuvant treatment had clearly worse OS. Also, advanced lymph node stage and sublobar surgical resection showed a clear association with early recurrence. Our findings underline previous reports from comparable cohorts, demonstrating a clear impact of gender, lymph node status, lobar resection, and adjuvant treatment on clinical outcome [27, 30, 31]. Interestingly, the reason why females demonstrate a remarkably longer OS—irrespective of possible confounders such as age, stage, therapy, smoking history—in various entities including SCLC, remains a matter of ongoing debate [32]. One possible explanation has been linked to the protective effects of estrogen, which underlines the longer natural life expectancy of females both in humans and animals [33]. However, ongoing debates necessitate further elucidation for alternative explanations. Adjuvant chemotherapy should be mandatory for all surgically managed SCLC patients, yet we had a high proportion of cases not receiving adjuvant chemotherapy, which is similar to the findings in comparable cohorts [34, 35]. Additionally, nodal upstaging is a common phenomenon in surgically managed SCLC, which might have also resulted in a significant number of advanced-stage patients in our study cohort [36, 37]. With regards to the potentially increasing number of limited-stage lung cancer cases in nationwide lung screening programs and the ongoing debate to reinforce the role of surgery in current SCLC guidelines, these findings should endorse practicing clinicians to carefully perform preoperative staging, take individual clinicopathological factors into account, and consequently, manage all patients selected for surgery in a multidisciplinary manner to offer optimal long-term results [38,39,40,41].

For the evaluation of Myc expression in our surgically resected specimens, we decided to perform IHC as it is one the most well-established methods for protein expression determination and which has been previously proven to be a valuable approach for investigating Myc in cancerous tissue [42, 43]. Clearly, gene amplification rate might also be relevant when investigating the role of Myc, but there is already sufficient data showing a clear correlation between increased amplification of Myc and a poor outcome in various malignancies including SCLC [44,45,46]. Moreover, increased gene amplification rates of Myc have been previously shown to clearly correlate with increased Myc protein level and detectability by IHC [47]. Therefore, we focused on IHC and divided our cohort into completely negative samples versus samples showing at least 1% positivity. With this approach, we found that 63% of patients were positive for L-Myc followed by 48% for C-Myc and 9% for N-Myc. These rates are in line with previously reported tendencies toward higher L-Myc and C-Myc presence compared to a generally weaker N-Myc presence in SCLC [48]. Recent studies on mouse and human models demonstrate that Myc family members drive dynamic evolution of SCLC subtypes. In this context, MYC directly activates NOTCH signaling, thus reprogramming SCLC from ASCL1 + to NEUROD1 + and, finally, to non-neuroendocrine states [8, 9]. Moreover, in vivo and in vivo SCLC models have also shown direct associations between the abundance of distinct Myc family members and molecular SCLC subtypes (e.g., ASCL1/SCLC-A and MYCL/L-Myc or NEUROD1/SCLC-N and MYC/C-Myc) [4]. Although we did not identify clear relations between positivity for Myc and clinicopathological characteristics, there was weak evidence that patients with N-Myc positivity were more likely to have a POU2F3-dominant molecular SCLC subtype. However, considering the low total number of N-Myc positive and POU2F3 SCLC cases in our cohort, this finding should be interpreted with caution. Altogether, because surgery is rarely performed in SCLC and thus only a few studies consisting of small cohorts provided controversial data on the expression and prognostic role of Myc family members in surgically managed SCLC patients, caution must be taken when interpreting our findings [49, 50]. Of note, Qin et al. analyzed the expression levels of C-Myc and L-Myc in n = 46 resected SCLC cases by IHC. Notably, they used deviant definitions for positive staining results (in case of C-Myc: ≥ 40% positive cells, in case of L-Myc: ≥ 10% positive cells) and found an overall low prevalence of C-Myc (4/46) and L-Myc positivity. Moreover, in their study, patients with positive C-Myc and L-Myc expression had a tendency toward a favorable clinical outcome. In contrast, another recent study analyzed C-Myc, L-Myc, and N-Myc expression levels in n = 83 surgically resected high-grade neuroendocrine carcinoma (HGNEC) lung specimens, of whom n = 36 specimens were SCLC cases [50]. In this study, specimens were defined as positive if ≥ 10% of the cells showed a positive staining for C-Myc, L-Myc, or N-Myc. Ultimately, comparable to the findings of our study, C-Myc positivity was found to be an independent prognostic factor indicating unfavorable clinical outcome. Clearly, further studies are warranted to evaluate the prognostic value of C-Myc in surgically resected SCLC patients.

The prognostic and predictive relevance of molecular SCLC subtypes has been just recently evaluated by our group in a large-scaled multicentric study including n = 386 surgically resected specimens and 26 human cell lines [5]. In that study, high ASCL1 expression was an independent negative prognosticator in surgically treated SCLC patients. In contrast, in a univariate model, high POU2F3 expression was associated with improved survival outcomes. In addition, we found remarkable associations between distinct molecular SCLC subtypes and the efficacy of standard-of-care and targeted therapeutics in vitro [5]. Notably, most patients in the current study had SCLC-A tumors, and no distinct YAP1-defined subtype could be distinguished. This is in line with our previous findings. Indeed, the results of other protein-based studies are also questioning the subtype-defining potential of YAP1 and arguing in favor of a unique subtype showing negative staining with the subtype markers ASCL1, NEUROD1, and POU2F3 [5, 51]. However, since the patient pool of the current study (all 104 cases) overlapped with our multicenter research [5], we did not aim to republish our previous findings on the prognostic role of molecular SCLC subtypes in the smaller current dataset.

Regarding their prognostic impact, L-Myc and N-Myc did not demonstrate any correlation with the clinical outcome in our surgical SCLC cohort. However, patients with C-Myc positive tumors had significantly impaired survival outcomes and also a clearly increased risk for shorter OS according to multivariate Cox proportional analysis. These findings might be explicable by the solid preclinical evidence demonstrating that Myc plays a key role in SCLC disease progression [8, 9]. Accordingly, tumors with higher C-Myc presence may have a more aggressive nature and result in accelerated progression, resulting in early recurrence and shorter OS. In line with our findings, there are numerous methodically comparable studies reporting on similar results in further thoracic and extra-thoracic malignancies [14, 52, 53]. In addition, preclinical evidence highlights a potential link between increased C-Myc activity and acquired resistance to standard-of-care chemotherapeutics, which might serve as a potential explanation why SCLC patients with a higher C-Myc abundance face a dismal overall prognosis [12, 54]. Altogether, Myc represents a promising targetable molecule in SCLC. Indeed, preclinical SCLC models clearly demonstrated efficacy of indirect or direct inhibitory strategies for Myc [20, 21, 55]. Of note, the first clinical trials including patients with various solid tumor types have recently been started to evaluate the therapeutic potency of direct Myc inhibition with OMO-103 [19, 56]. Altogether, clinical evaluation of Myc targeting in SCLC is highly warranted. We believe that our results on the prognostic role of C-Myc positivity in surgically treated patients are hypothesis-generating in their current form. However, in order to consider C-Myc positivity as a novel biomarker for the selecting eligible SCLC patients for surgical resection, further validation is needed. Nevertheless, C-Myc expression can still be considered a prognostic marker that can aid in defining the optimal follow-up strategy for patients who underwent surgical resection.

Our study has certain limitations that must be alluded. First, although we collected a comparatively large cohort of surgically treated SCLC cases, the overall number of included patients still remained relatively small. Second, due to the retrospective study design, the extent of information on clinicopathological and follow-up data was limited. Third, as there are no standardized protocols for the evaluation of Myc or the subtype-specific SCLC marker expressions by IHC, reproducibility of our results might be weakened when applying differing approaches of evaluation. Fourth, the current study solely included FFPE samples and thus Myc expressions may significantly differ in fresh-frozen samples [57]. Finally, although they could have provided valuable information on the role of these molecules in surgically managed SCLC patients, genomic data on MYC, MYCL, MYCN, ASCL1, NEUROD1, POU2F3, and YAP1 expressions were not available in our cohort.

Conclusion

Protein expression pattern of C-Myc, L-Myc, N-Myc, and their correlation with molecular SCLC subtypes is heterogeneous in surgically resected SCLC patients. In addition, C-Myc positivity is a robust prognosticator of impaired OS among surgically managed SCLC patients. More studies are warranted to investigate the role and clinical implications of Myc family members and subtype-specific markers in SCLC.

Availability of data and materials

Data were generated by the authors and available on reasonable request.

Abbreviations

CI:

Confidence intervals

DFS:

Disease-free survival

FFPE:

Formalin-fixed paraffin-embedded

HR:

Hazard ratios

IHC:

Immunohistochemistry

NCCN:

National Comprehensive Cancer Network

OS:

Overall survival

SCLC:

Small cell lung cancer

SCLC-A:

ASCL1-dominant subtype

SCLC-AN:

Combined ASCL1/NEUROD1-dominant subtype

SCLC-P:

POU2F3 dominant subtype

SCLC-N:

NEUROD1-dominant subtype

SCLC-QN:

Quadruple negative subtype

SCLC-Y:

YAP1-dominant subtype

WTS:

Whole tissue specimens

References

  1. Schwendenwein A, Megyesfalvi Z, Barany N, Valko Z, Bugyik E, Lang C, et al. Molecular profiles of small cell lung cancer subtypes: therapeutic implications. Molecular therapy oncolytics. 2021;20:470–83.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.

    Article  PubMed  Google Scholar 

  3. Karachaliou N, Pilotto S, Lazzari C, Bria E, de Marinis F, Rosell R. Cellular and molecular biology of small cell lung cancer: an overview. Transl Lung Cancer Res. 2016;5(1):2–15.

    PubMed Central  CAS  Google Scholar 

  4. Rudin CM, Poirier JT, Byers LA, Dive C, Dowlati A, George J, et al. Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat Rev Cancer. 2019;19(5):289–97.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Megyesfalvi Z, Barany N, Lantos A, Valko Z, Pipek O, Lang C, et al. Expression patterns and prognostic relevance of subtype-specific transcription factors in surgically resected small-cell lung cancer: an international multicenter study. J Pathol. 2022;257(5):674–86.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Qu S, Fetsch P, Thomas A, Pommier Y, Schrump DS, Miettinen MM, et al. Molecular subtypes of primary SCLC tumors and their associations with neuroendocrine and therapeutic markers. J Thoracic Oncol. 2022;17(1):141–53.

    Article  CAS  Google Scholar 

  7. Szeitz B, Megyesfalvi Z, Woldmar N, Valkó Z, Schwendenwein A, Bárány N, et al. In-depth proteomic analysis reveals unique subtype-specific signatures in human small-cell lung cancer. Clin Transl Med. 2022;12(9):e1060.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Ireland AS, Micinski AM, Kastner DW, Guo B, Wait SJ, Spainhower KB, et al. MYC drives temporal evolution of small cell lung cancer subtypes by reprogramming neuroendocrine fate. Cancer Cell. 2020;38(1):60-78.e12.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Mollaoglu G, Guthrie MR, Böhm S, Brägelmann J, Can I, Ballieu PM, et al. MYC drives progression of small cell lung cancer to a variant neuroendocrine subtype with vulnerability to aurora kinase inhibition. Cancer Cell. 2017;31(2):270–85.

    Article  PubMed Central  CAS  Google Scholar 

  10. Meyer N, Penn LZ. Reflecting on 25 years with MYC. Nat Rev Cancer. 2008;8(12):976–90.

    Article  PubMed  CAS  Google Scholar 

  11. Brägelmann J, Böhm S, Guthrie MR, Mollaoglu G, Oliver TG, Sos ML. Family matters: how MYC family oncogenes impact small cell lung cancer. Cell Cycle. 2017;16(16):1489–98.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Chen J, Guanizo AC, Jakasekara WSN, Inampudi C, Luong Q, Garama DJ, et al. MYC drives platinum resistant SCLC that is overcome by the dual PI3K-HDAC inhibitor fimepinostat. J Exp Clin Cancer Res. 2023;42(1):100.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Grunblatt E, Wu N, Zhang H, Liu X, Norton JP, Ohol Y, et al. MYCN drives chemoresistance in small cell lung cancer while USP7 inhibition can restore chemosensitivity. Genes Dev. 2020;34(17–18):1210–26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Feng W, Dean DC, Hornicek FJ, Spentzos D, Hoffman RM, Shi H, et al. Myc is a prognostic biomarker and potential therapeutic target in osteosarcoma. Ther Adv Med Oncol. 2020;12:1758835920922055.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Ambrosio MR, Lazzi S, Bello GL, Santi R, Porro LD, de Santi MM, et al. MYC protein expression scoring and its impact on the prognosis of aggressive B-cell lymphoma patients. Haematologica. 2019;104(1):e25–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Qu J, Zhao X, Wang J, Liu X, Yan Y, Liu L, et al. MYC overexpression with its prognostic and clinicopathological significance in breast cancer. Oncotarget. 2017;8(55):93998–4008.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Chen H, Liu H, Qing G. Targeting oncogenic Myc as a strategy for cancer treatment. Signal Transduct Target Ther. 2018;3(1):5.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Llombart V, Mansour MR. Therapeutic targeting of “undruggable” MYC. EBioMedicine. 2022;75:103756.

    Article  PubMed  CAS  Google Scholar 

  19. Whitfield JR, Soucek L. The long journey to bring a Myc inhibitor to the clinic. J Cell Biol. 2021;220(8):e202103090.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Fiorentino FP, Tokgün E, Solé-Sánchez S, Giampaolo S, Tokgün O, Jauset T, et al. Growth suppression by MYC inhibition in small cell lung cancer cells with TP53 and RB1 inactivation. Oncotarget. 2016;7(21):31014–28.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Cargill KR, Stewart CA, Park EM, Ramkumar K, Gay CM, Cardnell RJ, et al. Targeting MYC-enhanced glycolysis for the treatment of small cell lung cancer. Cancer & Metabolism. 2021;9(1):33.

    Article  Google Scholar 

  22. Ganti AKP, Loo BW, Bassetti M, Blakely C, Chiang A, D’Amico TA, et al. Small cell lung cancer, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Cancer Netw. 2021;19(12):1441–64.

    Article  CAS  Google Scholar 

  23. Majeed U, Manochakian R, Zhao Y, Lou Y. Targeted therapy in advanced non-small cell lung cancer: current advances and future trends. J Hematol Oncol. 2021;14(1):108.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Malhotra J, Jabbour SK, Aisner J. Current state of immunotherapy for non-small cell lung cancer. Transl Lung Cancer Res. 2017;6(2):196–211.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Hsu JC, Wei CF, Yang SC, Lin PC, Lee YC, Lu CY. Lung cancer survival and mortality in Taiwan following the initial launch of targeted therapies: an interrupted time series study. BMJ Open. 2020;10(5):e033427.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Wang S, Tang J, Sun T, Zheng X, Li J, Sun H, et al. Survival changes in patients with small cell lung cancer and disparities between different sexes, socioeconomic statuses and ages. Sci Rep. 2017;7(1):1339.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  27. Martucci N, Morabito A, La Rocca A, De Luca G, De Cecio R, Botti G, et al. Surgery in small-cell lung cancer. Cancers (Basel). 2021;13(3):390.

    Article  PubMed  CAS  Google Scholar 

  28. Dingemans AC, Früh M, Ardizzoni A, Besse B, Faivre-Finn C, Hendriks LE, et al. Small-cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up(). Ann Oncol. 2021;32(7):839–53.

    Article  PubMed  Google Scholar 

  29. Bernhardt EB, Jalal SI. Small Cell Lung Cancer. Cancer Treat Res. 2016;170:301–22.

    Article  PubMed  Google Scholar 

  30. Ju MH, Kim HR, Kim JB, Kim YH, Kim DK, Park SI. Surgical outcomes in small cell lung cancer. Korean J Thorac Cardiovasc Surg. 2012;45(1):40–4.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Nakamura H, Kato Y, Kato H. Outcome of surgery for small cell lung cancer—response to induction chemotherapy predicts survival. Thorac Cardiovasc Surg. 2004;52(4):206–10.

    Article  PubMed  CAS  Google Scholar 

  32. Wheatley-Price P, Ma C, Ashcroft LF, Nankivell M, Stephens RJ, White SC, et al. The strength of female sex as a prognostic factor in small-cell lung cancer: a pooled analysis of chemotherapy trials from the Manchester Lung Group and Medical Research Council Clinical Trials Unit. Ann Oncol. 2010;21(2):232–7.

    Article  PubMed  CAS  Google Scholar 

  33. Viña J, Borrás C, Gambini J, Sastre J, Pallardó FV. Why females live longer than males: control of longevity by sex hormones. Sci Aging Knowledge Environ. 2005;2005(23):pe17.

    Article  PubMed  Google Scholar 

  34. Yang CF, Chan DY, Speicher PJ, Gulack BC, Wang X, Hartwig MG, et al. Role of Adjuvant therapy in a population-based cohort of patients with early-stage small-cell lung cancer. J Clin Oncol. 2016;34(10):1057–64.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Zhou N, Bott M, Park BJ, Vallières E, Wilshire CL, Yasufuku K, et al. Predictors of survival following surgical resection of limited-stage small cell lung cancer. J Thorac Cardiovasc Surg. 2021;161(3):760-71.e2.

    Article  PubMed  Google Scholar 

  36. Thomas DC, Arnold BN, Rosen JE, Salazar MC, Blasberg JD, Detterbeck FC, et al. Defining outcomes of patients with clinical stage I small cell lung cancer upstaged at surgery. Lung Cancer. 2017;103:75–81.

    Article  Google Scholar 

  37. Takei H, Kondo H, Miyaoka E, Asamura H, Yoshino I, Date H, et al. Surgery for small cell lung cancer: a retrospective analysis of 243 patients from Japanese Lung Cancer Registry in 2004. J Thorac Oncol. 2014;9(8):1140–5.

    Article  PubMed  Google Scholar 

  38. Hoda MA, Klikovits T, Klepetko W. Controversies in oncology: surgery for small cell lung cancer? It’s time to rethink the case. ESMO Open. 2018;3(3):e000366.

    Article  PubMed Central  Google Scholar 

  39. de Koning HJ, van der Aalst CM, de Jong PA, Scholten ET, Nackaerts K, Heuvelmans MA, et al. Reduced lung-cancer mortality with volume CT screening in a randomized trial. N Engl J Med. 2020;382(6):503–13.

    Article  PubMed  Google Scholar 

  40. Kerpel-Fronius A, Monostori Z, Kovacs G, Ostoros G, Horvath I, Solymosi D, et al. Nationwide lung cancer screening with low-dose computed tomography: implementation and first results of the HUNCHEST screening program. Eur Radiol. 2022;32(7):4457–67.

    Article  PubMed  CAS  Google Scholar 

  41. Georgia H, Armin F, Christina T, Elli K, Elena B, Andreas F, et al. Multidisciplinary care models for patients with lung cancer. Breathe. 2020;16(4):200076.

    Article  Google Scholar 

  42. Oberley MJ, Rajguru SA, Zhang C, Kim K, Shaw GR, Grindle KM, et al. Immunohistochemical evaluation of MYC expression in mantle cell lymphoma. Histopathology. 2013;63(4):499–508.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Tapia G, Lopez R, Muñoz-Mármol AM, Mate JL, Sanz C, Marginet R, et al. Immunohistochemical detection of MYC protein correlates with MYC gene status in aggressive B cell lymphomas. Histopathology. 2011;59(4):672–8.

    Article  PubMed  Google Scholar 

  44. Deming SL, Nass SJ, Dickson RB, Trock BJ. C-myc amplification in breast cancer: a meta-analysis of its occurrence and prognostic relevance. Br J Cancer. 2000;83(12):1688–95.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Shi X, Zhou R, Zheng L, Jiang M. Prognostic significance of C-MYC dysregulation in esophageal squamous cell carcinoma: a meta-analysis. Biomark Med. 2020;14(7):599–609.

    Article  PubMed  CAS  Google Scholar 

  46. Alves Rde C, Meurer RT, Roehe AV. MYC amplification is associated with poor survival in small cell lung cancer: a chromogenic in situ hybridization study. J Cancer Res Clin Oncol. 2014;140(12):2021–5.

    Article  PubMed  Google Scholar 

  47. Blancato J, Singh B, Liu A, Liao DJ, Dickson RB. Correlation of amplification and overexpression of the c-myc oncogene in high-grade breast cancer: FISH, in situ hybridisation and immunohistochemical analyses. Br J Cancer. 2004;90(8):1612–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Takahashi T, Obata Y, Sekido Y, Hida T, Ueda R, Watanabe H, et al. Expression and amplification of myc gene family in small cell lung cancer and its relation to biological characteristics1. Can Res. 1989;49(10):2683–8.

    CAS  Google Scholar 

  49. Qin J, Xie F, Li C, Han N, Lu H. MYCL1 Amplification and expression of L-Myc and c-Myc in surgically resected small-cell lung carcinoma. Pathol Oncol Res. 2021;27:1609775.

    Article  PubMed Central  Google Scholar 

  50. Oshima Y, Haruki T, Matsui S, Makishima K, Sakabe T, Umekita Y, et al. Clinical significance of MYC family protein expression in surgically resected high-grade neuroendocrine carcinoma of the lung. Thoracic cancer. 2023;14(8):758–65.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Gay CM, Stewart CA, Park EM, Diao L, Groves SM, Heeke S, et al. Patterns of transcription factor programs and immune pathway activation define four major subtypes of SCLC with distinct therapeutic vulnerabilities. Cancer Cell. 2021;39(3):346-60.e7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Tsiatis AC, Herceg ME, Keedy VL, Halpern JL, Holt GE, Schwartz HS, et al. Prognostic significance of c-Myc expression in soft tissue leiomyosarcoma. Mod Pathol. 2009;22(11):1432–8.

    Article  PubMed  CAS  Google Scholar 

  53. Zhou C, Che G, Zheng X, Qiu J, Xie Z, Cong Y, et al. Expression and clinical significance of PD-L1 and c-Myc in non-small cell lung cancer. J Cancer Res Clin Oncol. 2019;145(11):2663–74.

    Article  PubMed  CAS  Google Scholar 

  54. Drapkin BJ, George J, Christensen CL, Mino-Kenudson M, Dries R, Sundaresan T, et al. Genomic and functional fidelity of small cell lung cancer patient-derived xenografts. Cancer Discov. 2018;8(5):600–15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Chalishazar MD, Wait SJ, Huang F, Ireland AS, Mukhopadhyay A, Lee Y, et al. MYC-driven small-cell lung cancer is metabolically distinct and vulnerable to arginine depletion. Clin Cancer Res. 2019;25(16):5107–21.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. S.L. P. Phase 1/2 study to evaluate safety, PK and efficacy of the MYC-inhibitor OMO-103 in solid tumours. 2021. https://ClinicalTrials.gov/show/NCT04808362.

  57. LüderRipoli F, Mohr A, Conradine Hammer S, Willenbrock S, Hewicker-Trautwein M, Hennecke S, et al. A comparison of fresh frozen vs. formalin-fixed, paraffin-embedded specimens of canine mammary tumors via branched-DNA assay. Int J Mol Sci. 2016;17(5):724.

    Article  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Open access funding provided by Semmelweis University. BD was supported by the Austrian Science Fund (FWF I3522, FWF I3977, and I4677) and the “BIOSMALL” EU HORIZON-MSCA-2022-SE-01 project. BD and ZM were supported by funding from the Hungarian National Research, Development, and Innovation Office (TKP2021‐EGA‐33, 2020‐1.1.6‐JÖVŐ, FK‐143751, and FK-147045). ZM was supported by the New National Excellence Program of the Ministry for Innovation and Technology of Hungary (UNKP‐20‐3, UNKP‐21‐3 and UNKP-23–5), and by the Bolyai Research Scholarship of the Hungarian Academy of Sciences. ZM is also the recipient of an International Association for the Study of Lung Cancer/International Lung Cancer Foundation Young Investigator Grant (2022). BF is a recipient of the Semmelweis 250 + Excellence PhD Scholarship (EFOP-3.6.3-VEKOP-16–2017- 00009) of the Semmelweis University. VL is a recipient of the Bolyai Research Scholarship of the Hungarian Academy of Sciences and the UNKP-19–4 New National Excellence Program of the Ministry for Innovation and Technology. KS was supported by the Austrian Science Fund (FWF No. T 1062-B33) and the City of Vienna Fund for Innovative Interdisciplinary Cancer Research.

Author information

Authors and Affiliations

Authors

Contributions

CL: Study conception and design, Administrative support, Collection and assembly of data, Data analysis and interpretation, Manuscript writing, Final approval of manuscript; ZM: Administrative support, Collection and assembly of data, Manuscript writing, Final approval of manuscript; AL: Administrative support, Data analysis and interpretation, Manuscript writing, Final approval of manuscript; FO: Administrative support, Provision of study materials, Manuscript writing, Final approval of manuscript; MAH: Administrative support, Provision of study materials, Manuscript writing, Final approval of manuscript; AS: Administrative support, Manuscript writing, Final approval of manuscript; BF: Administrative support, Manuscript writing, Final approval of manuscript; JF: Administrative support, Provision of study materials, Manuscript writing, Final approval of manuscript; AST: Administrative support, Manuscript writing, Final approval of manuscript; ASQ: Administrative support, Collection and assembly of data, Manuscript writing, Final approval of manuscript; FE: Administrative support, Collection and assembly of data, Manuscript writing, Final approval of manuscript; KB: Administrative support, Manuscript writing, Final approval of manuscript; TK: Administrative support, Provision of study materials, Manuscript writing, Final approval of manuscript; GT: Administrative support, Manuscript writing, Final approval of manuscript; FRV: Administrative support, Provision of study materials, Manuscript writing, Final approval of manuscript; CA: Administrative support, Provision of study materials, Manuscript writing, Final approval of manuscript; KH: Administrative support, Provision of study materials, Manuscript writing, Final approval of manuscript; VL: Administrative support, Manuscript writing, Final approval of manuscript; KS: Study conception and design, Administrative support, Manuscript writing, Final approval of manuscript; BD: Study conception and design, Administrative support, Data analysis and interpretation, Manuscript writing, Final approval of manuscript.

Corresponding authors

Correspondence to Zsolt Megyesfalvi or Balazs Dome.

Ethics declarations

Ethics approval and consent to participate

The study was conducted in accordance with the Declaration of Helsinki and the Good Scientific Practice statements of the participating institutions. Moreover, study approval was provided by the institutional Ethics Committees (Medical University of Vienna EK#: 2196/2019; National Koranyi Institute of Pulmonology ETT-TUKEB 23636–2/2018, 23636/10/2018/EÜIG).

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

Additional file 1: Supplementary Table 1.

Correlation of Myc family members with clinicopathological characteristics of the study population (n = 104).

Additional file 2: Supplementary Table 2.

Univariate and multivariate analysis for the evaluation of distinct clinicopathological features and differential Myc family members expression as predictors of overall survival (OS) following surgical resection in SCLC.

Additional file 3: Supplementary Table 3.

Univariate and multivariate analyses for the evaluation of distinct clinicopathological factors and differential Myc family members’ expression status as predictors of disease-free survival (DFS) following surgical resection in SCLC.

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

Lang, C., Megyesfalvi, Z., Lantos, A. et al. C-Myc protein expression indicates unfavorable clinical outcome in surgically resected small cell lung cancer. World J Surg Onc 22, 57 (2024). https://doi.org/10.1186/s12957-024-03315-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12957-024-03315-7

Keywords