Skip to main content
  • Research article
  • Open access
  • Published:

Low expression of c-Myc protein predicts poor outcomes in patients with hepatocellular carcinoma after resection

Abstract

Background

Embryonic Liver Fodrin (ELF) is an adaptor protein of transforming growth factor (TGF-β) signaling cascade. Disruption of ELF results in mislocalization of Smad3 and Smad4, leading to compromised TGF-β signaling. c-Myc is an important oncogenic transcription factor, and the disruption of TGF-β signaling promotes c-Myc-induced hepatocellular carcinoma (HCC) carcinogenesis. However, the prognostic significance of c-Myc in HCC is less understood

Methods

The expression of c-Myc protein and mRNA were measured by immunohistochemistry (IHC) and qRT- PCR, respectively. IHC was performed to detect TGF-β1 and ELF expression in HCC tissues. Their relationship with clinicopathological factors and overall survival (OS) and disease free survival (DFS) were examined.

Results

The expression of c-Myc protein and mRNA in HCC tissues were significantly higher in HCC area than those in normal liver tissues. However, the expression were low compared with those adjacent to HCC area. c-Myc protein was independently predictive of DFS and OS, and it was negatively correlated with tumor size (P = 0.031), tumor number (P = 0.038), and recurrence (P = 0.001). Low c-Myc expression was associated with short-term recurrence and poor prognosis. The predictive value of c-Myc combined with TGF-β1 or/and ELF was higher than that of any other single marker. Low c-Myc, high TGF-β1 or/and low ELF expression was associated with the worst DFS and OS.

Conclusions

Low expression of c-Myc protein predicts poor outcomes in patients with HCC with hepatectomy. The combination of the expression of c-Myc, TGF-β1, and ELF can be used to accurately predict outcomes of patients with HCC.

Peer Review reports

Background

Hepatocellular carcinoma (HCC) is a very common and serious malignancy with high morbidity and high mortality. It is the sixth most common cancer and the third common cause of cancer-related death worldwide [1,2,3]. Though most cases of HCC happen in Eastern Asia and sub-Saharan Africa, the incidence in some developed countries such as Japan, UK, France, and USA is increasing [4]. The increase of incidence and the lack of effective treatment for HCC have made the disease a major health problem [5]. Therefore, it is important to understand the mechanisms of HCC to find the effective biomarkers and to develop therapies.

c-Myc, an important oncogenic transcription factor, locates on chromosome 8q24.1, and involves in cell proliferation, apoptosis, differentiation and metabolism because it participates in the regulation of 15% of all genes in the human genome, including microRNAs [6,7,8,9,10,11,12]. Furthermore, as a powerful cancer-promoting oncogene, c-Myc has been implicated in the pathogenesis of kinds of malignant tumors, including human solid tumor, leukemia and lymphoma, along with animal tumors [13,14,15,16,17]. Similarly, c-Myc is also identified as a reasonable gene driver and a central regulator of malignant transformation in primary stages of HCC carcinogenesis using gene expression profiling of cirrhotic and dysplastic nodules, and early HCC [18]. In addition, c-Myc gene is commonly found to be overexpressed in advanced HCC tissues and derived HCC cell lines too [19, 20]. Moreover, regression of invasive HCC in animal models could be induced by the inactivation of c-Myc gene, followed by up-regulation of liver cell and liver stem cell markers, and down-regulation of tumor markers [21].

Paradoxically, c-Myc is also found to induce apoptosis and cell senescence, and represent tumor-suppressive through activation of tumor suppressor p53 directly or indirectly [12, 17]. The recent studies indicate that tumorigenesis in mouse models of c-Myc oncogenesis was accelerated by inactivation of the ARF-MDM2-p53 pathway. c-Myc can also change the balance of pro- and anti-apoptotic factors to prime the cells for apoptosis and death [6, 22, 23]. Pelengaris et al. [24] used a switchable form of the c-Myc protein to make a reversible transgenic model of pancreatic β cell oncogenesis, and found that loss of c-Myc protein up-regulated Bcl-xL expression and suppressed apoptosis, finally resulted in a full spectrum of tumor development, even distant metastasis. Murakami et al. [25] and Lee et al. [26] found that c-Myc-induced HCC was marked by a relatively higher degree of cell differentiation, less aggressiveness, a lower rate of genomic instability, and a longer time for carcinogenesis. Other studies have also reported that the level of c-Myc expression determined its oncogenic potential, indicated that low c-Myc expression induced cell proliferation and oncogenesis, whereas high c-Myc expression activated pro-apoptotic programs and accelerated apoptosis [27,28,29].

In this study, we investigated the prognostic value of c-Myc expression in in patients with HCC undergoing curative resection. In addition, it is well-known that overexpression of TGF-β1 correlates with carcinogenesis and progression of HCC. We have observed that the high level of TGF-β1 and low level of ELF (a Smad3/4 adaptor protein) predicted the poor outcome in patients with HCC [30,31,32]. Santoni-Rugiu et al. [33] indicated that disruption of TGF-β1 signaling promoted c-Myc- induced HCC carcinogenesis by TGF-α as well. Therefore, this study further verified the more accurate predictive parameters of HCC using the combination of c-Myc with TGF-β1, and ELF.

Methods

Study population

This study was approved by the Ethics Review Committee of the First Affiliated Hospital of Sun Yat-sen University. Eighty-four patients with HCC receiving hepatectomy at our center from June 2007 to October 2009 were included in this study. The inclusion criteria were patients: 1) with tumor-node-metastasis (TNM) stage I, II, IIIA or IIIB (International Union Against Cancer, the seventh edition); 2) with Child-Pugh class A and class B hepatic function; 3) aged 18–80; and 4) provided written informed consent. The exclusion criteria included patients: 1) with TNM stage IIIC, IVA, or IVB; 2) with Child-Pugh class C hepatic function; 3) with second malignancy or history of second malignancy within 5 years; 4) with perioperative dysfunction of vital organs; 5) with percutaneous ablation, 6) transcatheter arterial chemoembolization (TACE); and 7) received chemotherapy, or radiotherapy within 1 month post-operation. All patients were postoperatively followed-up as described in our previous study [30]. Normal liver tissues, HCC adjacent tissues, and HCC tissues were collected and prepared as described in previous study [30].

Immunohistochemical analysis

Immunohistochemical analysis was performed as previously described [30]. Rabbit polyclonal anti-c-Myc antibody (ab32072, Abcam, USA) was used in the immunohistochemical analysis (1:100 dilution).

Evaluation of Immunohistochemical staining

The immunohistochemical staining in the tissue was scored independently by 2 pathologists blinded to the patients and clinical data, by applying a semiquantitative immunoreactivity score (IRS) reported elsewhere [30]. Category A documented the intensity of immunostaining as 0–3 (0, negative; 1, weak; 2, moderate; 3, strong). Category B documented the percentage of immunoreactive cells as 0 (less than 5%), 1 (6%–25%), 2 (26%–50%), 3 (51%–75%), and 4 (76%–100%). Multiplication of category A and B resulted in an IRS ranging from 0 to 12 for each tumor or non-tumor. Sections with a total score of 0 or 1 or 2 were defined as negative (−), score of 3 or 4 were defined as weakly positive (+), score of 6 or 8 were defined as moderately positive (++), score of 9 or 12 were defined as strongly positive (+++). For categorical analyses, the immunoreactivity was graded as low level (total score < =4) or high level (total score > 4).

Real -time quantitative polymerase chain reaction (qRT-PCR)

Fresh HCC tissues (n = 29), HCC adjacent tissues (n = 29), and normal liver tissues (n = 4) were collected immediately after resection for detecting c-Myc mRNA expression. RNA was extracted by TRIzol reagent (Thermo Fisher Scientific, MA, USA) according to manufacturer’s protocol. The cDNA was synthesized by cDNA synthesis kit (Takara Biotechnology, Japan) in accordance with manufacturer’s instructions. The cDNA was used as templates for qRT-PCR by the SYBR Green PCR kit (Toyobo, Japan) in the Bio-rad IQ5 PCR system (Bio-Rad, CA). GAPDH was used as an internal control. The relative gene expressions were quantified and analyzed. The primers were obtained from Genecopia Company (Guangzhou, China).

Statistical analysis

Normally distributed Continuous variables were compared by Student’s t-test. Categorical variables were compared with Chi-square test. Disease free survival (DFS) was defined as the interval from the surgery to recurrence, while overall survival (OS) was defined as the interval from surgery to the HCC-associated death. The univariate, multivariate and Kaplan–Meier method was used to analyze the survival rates [34], and the equivalences of the survival curves were compared among groups by log-rank test. A P-value < 0.05 would be regarded as statistical significance in each test. All analyses were performed using SPSS v 13 software (Chicago, IL, USA).

Results

The expression of c-Myc protein and mRNA in HCC and adjacent tissues

The expression of c-Myc in normal liver tissues was negative (0/20). However, c-Myc protein was expressed in 78.5% (66/84) in HCC tissues and in 82.1% (69/84) in adjacent tissues, and these rates were significantly different from that of normal liver tissues (P <  0.001). In adjacent tissues and HCC tissues, the high expression (++ to +++) rates for c-Myc were 65.5% (55/84) and 59.5% (50/84), respectively, and there were no statistical difference between them (P = 0.426) (Table 1, Fig. 1).

Table 1 The expression of c-Myc in HCC
Fig. 1
figure 1

Expression of c-Myc. Immunohistochemical staining in different tissues is shown. Normal liver tissues (a), HCC adjacent tissues (b), HCC tissues (c) (original magnification × 400), and expression of c-Myc mRNA (d)

Similarly, we observed that the levels of c-myc mRNA in HCC tissues and in adjacent tissues were higher than that in normal liver tissues (P = 0.002 and P <  0.001, respectively). Interestingly, we found the level of c-myc mRNA in HCC tissues was remarkably lower than those in the corresponding adjacent tissue (P <  0.001). HCC tissues were divided into two groups in terms of TNM stage: Early stage HCC (TNM stage I and tumor size less than 10 cm, n = 17), and advanced HCC (TNM stage II and III, n = 12). There was no striking difference in c-myc mRNA expression between the early stage HCC and advanced HCC (Fig. 1).

Correlation between 15 clinicopathological characteristics and c-Myc protein expression in HCC and adjacent tissues

To better understand the importance of the prognostic value of c-Myc protein expression, we first analyzed the relationships between 15 clinicopathological characteristics and c-Myc protein expression in cancer tissues (Table 2). The results showed that the expression of c-Myc protein in tumor tissues was not associated with age, sex, serum HBsAg positivity, serum alanine aminotransferase (ALT), and AFP level. There was also no relation between c-Myc expression and tumor differentiation, TNM stage, completeness of tumor encapsulation, and postoperative complications. However, the level of c-Myc protein in HCC tissues was positively correlated with cirrhosis (P = 0.042), but negatively correlated with platelet (PLT) count (P = 0.036), tumor number (P = 0.038), portal vein tumor thrombosis (PVTT) (P = 0.019), tumor size (P = 0.031), and recurrence (P = 0.002).

Table 2 Correlation between c-Myc and the clinicopathological characteristics in HCC tissues and adjacent tissues

In tissues adjacent to HCC, high expression of c-Myc was only associated with complete tumor encapsulation (P = 0.006).

Independent prognostic factors of HCC

In order to discern the factors related to DFS and OS, c-Myc and clinicopathological factors were assessed by univariate analysis and the Multivariate analysis. The univariate analysis indicated that significant prognostic factors for DFS were PVTT, tumor number, tumor encapsulation, blood transfusion, blood loss, operative time, TNM stage, and c-Myc expression. The significant prognostic factors for OS were PVTT, tumor number, resection margin, tumor size, tumor differentiation, TNM stage, blood transfusion, blood loss, operative time and c-Myc expression (all P <  0.05) (Table 3). Multivariate analysis indicated that blood transfusion, TNM stage and c-Myc expression were significantly independent prognostic factors for both DFS and OS (all P <  0.05). (Table 4).

Table 3 Prognostic factors for DFS and OS by univariate analysis
Table 4 Prognostic factors for disease-free and overall survival by the multivariate Cox proportional hazards regression model

Low expression of c-Myc predicts short-term recurrence and the poor outcomes in patients with HCC.

All patients were divided into 2 groups in terms of their c-Myc expression profiles: high-expression (n = 50) and low-expression (n = 34). Analysis showed that c-Myc expression level was positively correlated with DFS and OS. The 1-, 3-, and 5-year DFS rates in the high c-Myc expression group were significantly higher than those of low expression group (70.0%, 54.9%, 45.9% vs. 17.6%, 11.8%, 11.8%, respectively, P <  0.001) (Fig. 2a). In addition, the 1-, 3-, and 5-year OS rates of the high expression group were still markedly higher than those of low expression group (92.0%, 72.0%, 62.0% vs. 64.7%, 20.6%, 14.7%, respectively, P <  0.001) (Fig. 2b).

Fig. 2
figure 2

Kaplan-Meier curves are shown for time to disease recurrence (a, P < 0.001, log-rank test) and overall survival (b, P < 0.001, log-rank test) among patients with high or low intratumoral c-Myc expression

In the current study, the total recurrence rate was 67.9% (57/84); however, 43 cases of recurrence happened in the first postoperative year. The 1-, 3-, and 5-year OS rates of the in patients with HCC with a recurrence in first year (62.8%, 20.9%, 9.3%, respectively) were obviously lower than that of patients with a recurrence after 1 years (100%, 50.0%, 35.7%, respectively, P <  0.001), or those without recurrence (100%,100%, 100%, P <  0.001). There was also a marked difference rates between patients with a recurrence after 1 year and those without a recurrence (P <  0.001, Fig. 3).

Fig. 3
figure 3

Kaplan-Meier curves are shown for time to overall survival among patients with recurrence in different times (P < 0.001, log-rank test)

We then analyzed the association between c-Myc expression and recurrence time, and found that the proportion of patients with low c-Myc expression with short term recurrence (62.8%) was higher than that in patients with a long term recurrence (14.3%, P = 0.002) or without recurrence (14.8%, P < 0.001). However, there was no apparent difference between patients with a long term recurrence and patients with no recurrence (P = 0.964) (Table 5).

Table 5 The correlationship between c-Myc and recurrence

Combining c-Myc expression with TGF-β1 or/and ELF reveals improved prognostic accuracy for HCC

As described in our previous study [30], high level of ELF protein (a Smad3/4 adaptor protein) was observed in normal liver tissues and low level of ELF protein in HCC tissues (Fig. 4a, b). In contrast, high level of TGF-β1 protein was found in human HCC tissues and low level of TGF-β1 protein in normal liver tissues (Fig. 4c, d). Then, we demonstrated a significant negative correlation between c-Myc and TGF-β1 (r = − 0.228, P = 0.037, Table 6), and a significant positive correlation between c-Myc and ELF (r = 0.217, P = 0.048, Table 7). We also analyzed the prognostic value of combined TGF-β1 and c-Myc levels for HCC. We divided patients into the following four groups: TGF-β1 high expression/c-Myc high expression (ThMh), TGF-β1 low expression/c-Myc high expression (TlMh), TGF-β1 high expression/c-Myc low expression (ThMl), TGF-β1 low expression /c-Myc low expression (TlMl). Analysis exhibited that the TlMh group had the best OS. The DFS and the TlMl groups had the second best rates, followed by the ThMh group, and the worst prognosis was seen in the ThMl group.

Fig. 4
figure 4

Expression of ELF and TGF-β1 immunohistochemical staining in different tissues is shown. Normal liver tissues (a,c), HCC tissues (b,d) (original magnification × 400)

Table 6 The correlationship between c-Myc and TGF-β1 in HCC
Table 7 The correlationship between c-Myc and ELF in HCC

The 1-, 3-, and 5-year DFS rates of the TlMh group (88.9%, 81.5%, 70.2%, respectively) were markedly higher than those of ThMh group (47.8%, 21.7%, 17.4%, respectively, P < 0.001) and the ThMl group (11.5%, 3.8%, 3.8%,respectively, P < 0.001). The 1-, 3-, and 5-year OS rates of the TlMh group (96.3%, 92.6%, 85.2%,respectively) were obviously higher than those of the ThMh group (87.0%, 47.8%, 34.8%,respectively, P < 0.001) and the ThMl group (57.7%, 11.5%, 7.7%,respectively, P < 0.001) (Fig. 5a, b). Furthermore, the 1-, 3- and 5-year DFS rates of the ThMl group were remarkably lower than those of the ThMh group (P = 0.004) and TlMl group (42.9%, 42.9%, 42.9%, respectively, P = 0.004). The 1-, 3-, and 5-year OS rates of the ThMl group were markedly lower than those of the ThMh group (P = 0.001) and TlMl group (42.9%, 42.9%, 2.9%, respectively, P = 0.012). Moreover, there was a significant difference of OS between the TlMh group and TlMl group (P = 0.007) (Fig. 5a, b). Overall, the results showed that the combination of TGF-β1 elevation and c-Myc reduction in HCC tissues seems to be predictive of poorest prognosis.

Fig. 5
figure 5

The combination of c-Myc and TGF-β1 was found to enhance prognostic accuracy for HCC. Disease-free survival curves (a, P < 0.001, log-rank test) and overall survival curves (b, P < 0.001, log-rank test)

Second, we divided patients into another four groups in order to analyze the prognostic value of combined ELF and c-Myc levels for patients with HCC: ELF high expression/c-Myc high expression (EhMh), ELF low expression/c-Myc high expression (ElMh), ELF high expression/c-Myc low expression (EhMl), ELF low expression/c-Myc low expression (ElMl). The analysis indicated that the EhMh group had the best OS and DFS rates, the ElMh group the second best, followed by the EhMl group, and the ElMl group had the worst prognosis.

The 1-, 3-, and 5-year DFS rates of EhMh group (89.3%, 71.4%, 64.3%, respectively) were markedly higher than those of the ElMh group (45.5%, 31.8%, 22.7%, respectively, P < 0.001) and the ElMl group (4.5%, 0%, 0%, respectively, P < 0.001). The 1-, 3-, and 5-year OS rates of the EhMh group (100%,85.7%, 78.6%, respectively) were significantly higher than those of the EhMl group (66.7%, 41.7%, 33.3%,respectively, P < 0.001) and the ElMl group (63.6%, 9.1%, and 4.5%,respectively, P < 0.001) (Fig. 6a, b). The 1-, 3-, and 5-year DFS rates of the ElMl group were remarkably lower than those of the ElMh group (P = 0.003) and the EhMl group (41.7%, 33.3%, and 33.3%, respectively, P = 0.010). The 1-, 3-, and 5-year OS rates of the ElMl group were markedly lower than those of the ElMh group (81.8%, 54.5%, 40.9%, respectively, P = 0.002). Moreover, there was a significant difference in OS between the EhMh and ElMh group (P = 0.003) (Fig. 6a, b). Overall, the results indicated that the combination of elevation of both ELF and c-Myc in HCC tissues seems to be predictive of the best prognosis.

Fig. 6
figure 6

The combination of c-Myc and ELF was found to enhance prognostic accuracy for HCC. Disease-free survival curves (a, P < 0.001, log-rank test) and overall survival curves (b, P < 0.001, log-rank test)

Finally, we observed the predictive effect of c-Myc expression combined with TGF-β1 and ELF for outcomes in patients with HCC through analysis of the following eight groups: TGF-β1 high/ELF high/c-Myc high expression (ThEhMh), TGF-β1/ELF low/c-Myc high expression (ThElMh), TGF-β1 high/ELF high/c-Myc low expression (ThEhMl), TGF-β1 high/ELF low/c-Myc low expression (ThElMl), TGF-β1 low/ELF high/c-Myc high expression (TlEhMh), TGF-β1 low/ELF high/c-Myc low expression (TlEhMl), TGF-β1 low/ELF low/c-Myc high expression(TlElMh), TGF-β1 low/ELF low/c-Myc low expression (TlElMl).

The 1-, 3-, and 5-year OS rates of the TlEhMh group (100%, 94.7%, 89.5%, respectively) were the best, and significantly higher than those of the ThEhMh (100.0%, 66.7%, 55.6%, respectively, P = 0.026), ThElMh (78.6%, 35.7%, 21.4%, respectively, P < 0.001), ThEhMl (57.1%, 14.3%, 14.3%, respectively, P < 0.001), TlElMl (100%, 0%, 0%, respectively, P < 0.001), and ThElMl groups (60.0%, 10.0%, 5.0%, P < 0.001). Similarly, the 1-, 3-, and 5-year OS rates of the TlElMh group (87.5%, 87.5%, 75.0%, respectively) were also remarkably higher than those of the ThElMh (P = 0.020), ThEhMl (P < 0.001), TlElMl (P = 0.029), and ThElMl groups (P < 0.001). The 1-, 3-, and 5-year OS rates of the TlEhMl group (80.0%, 80.0%, 60.0% respectively) were also higher than those of the ThElMl group (P = 0.011). The 1-, 3-, and 5-year OS rates of the ThEhMh group were also significantly higher than those of the ThEhMl (P = 0.038), and ThElMl group (P = 0.001). However, there was no significant difference of OS between the other two groups (P > 0.05).

The 1-, 3-, and 5-year DFS rates of the TlEhMh group (94.7%, 84.2%, 78.9%, respectively) were significantly higher than those of the ThEhMh (77.8%, 44.4%, 33.3%, P = 0.003), ThEhMl (28.6%, 14.3%, 14.3%, respectively, P < 0.001), ThElMh (28.6%, 7.1%,7.1%, respectively, P < 0.001), ThElMl (5.0%, 0.0%, 0.0%, respectively, P < 0.001), and TlElMl groups (0%, 0%, 0%, respectively, P < 0.001). The 1-, 3-, and 5-year DFS rates of the TlElMh group (75.0%, 75.0%, 50.0%, respectively) were also significantly higher than those of the ThEhMl (P = 0.024), ThElMh (P = 0.009), ThElMl (P < 0.001), and TlElMl (P = 0.029) groups. Likewise, the 1-, 3-, and 5-year DFS rates of the TlEhMl group (60.0%, 60.0%, 60.0%, respectively) were also significantly higher than those of the ThElMl (P = 0.007) and TlElMl group (P = 0.057). The 1-, 3-, and 5-year DFS rates of the ThEhMh group were also significantly higher than those of the ThEhMl (P = 0.037), ThElMh (P = 0.021), ThElMl (P < 0.001), and TlElMl (P = 0.004) groups. However, there was no obvious difference of DFS between the other two groups (P > 0.05) (Fig. 7).

Fig. 7
figure 7

The combination of c-Myc, TGF-β1 and ELF was found to enhance prognostic accuracy for HCC. Disease-free survival curves (a, P < 0.001, log-rank test) and overall survival curves (b, P < 0.001, log-rank test)

Discussion

Undoubtedly, c-Myc plays an important role in the occurrence and development of HCC. For example, overexpression of the c-Myc gene has been frequently observed in human HCC tissues. And c-Myc has induced the occurrence of HCC in animal models. Accordingly, c-Myc inhibition results in a loss of the neoplastic properties [18,19,20,21]. However, c-Myc also exhibits onco-suppressive activity, such as apoptosis and cell senescence [12, 17, 22, 23]. Kaposi-Novak et al. [27], Akita et al. [28] and Murphy et al. [29] have shown that when c-Myc was at a low level, oncogenic properties were increased and proliferation was induced, once c-Myc exceeded a threshold level, the upregulation of pro-apoptotic factors p53, p21, and p27 and cell death from apoptosis were detected.

There are few studies about the level of c-Myc protein in human HCC and adjacent tissues, or about the prognostic value of c-Myc protein in HCC. Cui et al. [35] found that there was higher c-Myc expression in HCC tissues with tumor recurrence within 1 year after tumor resection (15 cases), compared with that in HCC tissues without tumor recurrence (15 cases), and made a conclusion that high levels of c-Myc protein was related to a high recurrence rate. However, Yuen et al. [36] drew a completely different conclusion by analyzing the data of 150 patients with HCC. They found that the expression of c-Myc protein was negatively correlated with the grade of tumor differentiation and the levels of mutated p53.

Our results showed that there was low expression of c-Myc protein and mRNA in normal liver tissue, while there was high expression in HCC and adjacent tissues. We also observed a phenomenon reported by Yuen et al. [36] that there was the higher expression of c-Myc protein and mRNA in adjacent tissues as compared to those of HCC tissues. Furthermore, we found that c-Myc protein in HCC tissues was negatively correlated with PVTT, tumor number, tumor size, and recurrence, whereas there were no correlation between clinicopathological characteristics and c-Myc expression in adjacent tissues except complete tumor encapsulation.

We further analyzed the clinical significance of c-Myc protein in the outcomes of patients with HCC, and found that the expression of c-Myc protein was a significant and independent prognostic factor for DFS and OS, and the predictive accuracy was better than that of TNM stage, another independent prognostic factor for HCC. The results showed that low expression of c-Myc protein predicts recurrence and the poor outcomes in patients with HCC. The 1-, 3-, and 5-year DFS and OS rates (70.0%, 54.9%, 45.9% and 92.0%, 72.0%, 62.0%, respectively) in patients with HCC with high c-Myc expression were remarkably higher than those with low c-Myc expression (17.6%, 11.8%, 11.8% and 64.7%, 20.6%, 14.7%).

It is well known that the recurrence of HCC after resection is intimately associated with a decreased median survival. Moreover, the recurrence within the first postoperative year predicted a worse prognosis [37]. We observed that low c-Myc expression in HCC tissues was also intimately associated with short-term recurrence and a lower OS rate, compared to those with the long-term recurrence and no recurrence. The proportion of low c-Myc expression in patients with HCC with recurrence at ≤1 year was 62.8%, which was remarkably higher than that in patients with HCC with a recurrence after 1 year and no recurrence (14.3% and 14.8%, respectively).

Overexpression of TGF-β1 is usually found in HCC tissues, and it correlates with carcinogenesis and progression of HCC. Disruption of TGF-β signaling is recognized as the key mechanism through which the function of TGF-β is changed from tumor suppression to tumor promotion. Santoni-Rugiu et al. [33] indicated that c-Myc induced HCC carcinogenesis was strengthened by TGF-β1 overexpression. Our previous studies [30, 32] showed high-expression of TGF-β1 in rat HCC tissues, and abnormality of the TGF-β1 signaling pathway might promote the carcinogenesis of HCC, also indicated that a high level of TGF-β1 foreboded a poor prognosis of in patients with HCC after curative resection. The loss or down-regulation of ELF (a Smad3/4 adaptor protein) might be one of reasons for disruption of TGF-β1 signaling, which similarly predicted a poor OS and DFS rate for patients with HCC. Interestingly, their combination had a better prognostic value, compared with either one alone.

In this study, we analyzed the correlation of c-Myc with TGF-β1 and ELF, and found that c-Myc was negatively associated with TGF-β1 and positively with ELF. Intriguingly, the combination of c-Myc with ELF or/and TGF-β1 increased predictive accuracy for outcomes in patients with HCC who underwent resection, and in particular the combination of all three.

Our results showed that patients with HCC with the best prognosis had low TGF-β1 and high c-Myc expression (3-year OS and DFS rates of 92.6% and 81.5%, respectively), while the worst prognosis was seen in patients who have high TGF-β1 and low c-Myc expression (3-year OS and DFS rates of 11.5% and 3.8%, respectively). The second best was in patients of low TGF-β1 and low c-Myc expression (3-year OS and DFS rates of 42.9% and 42.9%, respectively), and the third was in patients of high TGF-β1 and high c-Myc expression (3-year OS and DFS rates of 47.8% and 21.7%, respectively). So we can conclude that high TGF-β1 or low c-Myc expression both predict poor prognosis in patients with HCC, and this combination is associated with the poorest outcomes. In addition, the prognostic value of TGF-β1 may be better than that of c-Myc for patients with HCC with hepatectomy.

As for ELF and c-Myc, we found that patients with HCC with high ELF expression and high c-Myc expression had the best prognosis (3-year OS and DFS rates of 85.7% and 71.4%, respectively), while the worst prognosis was seen in those with low ELF and low c-Myc expression (3-year OS and DFS rates of 9.1% and 0%, respectively). The second best was in patients of low ELF and high c-Myc expression (3-year OS and DFS rates of 54.5% and 31.8% respectively), and the third was in patients with high ELF and low c-Myc expression (3-year OS and DFS rates of 41.7% and 33.3%, respectively). So we can conclude that low ELF expression or low c-Myc expression both predict a poor prognosis, and the worst outcomes are seen with low levels of both. In addition, the prognostic value of c-Myc may be better than that of ELF for patients with HCC with curative resection.

When all three markers were examined together, the worst outcomes were seen in patients with any combination of two or more of high TGF-β1, low ELF, and low c-Myc expression. The sequence of prognostic value of the three proteins for patients with HCC was TGF-β1 > c-Myc > ELF. The prognosis of patients of low TGF-β1, high ELF, and high c-Myc expression was the best (3-year OS and DFS rates of 94.7% and 84.2%, respectively). The second best was seen in patients of low TGF-β1, low ELF, and high c-Myc expression (3-year OS and DFS rates of 87.5% and 75.0%, respectively), the third was in patients of low TGF-β1, high ELF, and low c-Myc expression (3-year OS and DFS rates of 80.0% and 60.0%, respectively), and the fourth was in patients of high TGF-β1, high ELF, and high c-Myc expression (3-year OS and DFS rates of 66.7% and 44.4%, respectively).

Based on our results, we speculate that c-Myc protein may play an important role in the process of HCC carcinogenesis, rather than sustaining the growth of the tumor cells. It may be one of the contributory mechanisms of HCC progression, i.e., the apoptosis of HCC cells is jeopardized by the diminished expression of c-Myc and accompanying uncoordinated control of cellular growth. Low expression of c-Myc predicts short-term recurrence and a poor outcome in patients with HCC, though there is c-Myc expression in most HCC tissues, whereas there is no c-Myc expression in normal liver tissues. c-Myc combined with TGF-β1 or/and ELF can more accurately predict the prognosis in patients with HCC, given the disruption of the TGF-β1 signaling pathway is another important mechanism of HCC development. The combination of TGF-β1, ELF, and c-Myc expression is a valuable predictive method for outcomes in patients with HCC, which can usefully guide the follow-up and the further treatment for patients with HCC after curative resection. For instance, close follow-up should be performed to detect early tumor recurrence for the patients with any two or all of high TGF-β1, low ELF, and low c-Myc expression in tumor tissues. Accordingly, the personalized comprehensive therapies should be provided for this kind of patients, such as TACE, chemotherapy, sorafenib and percutaneous ablation.

Certainly, this study still has several limitations and unanswered questions. First, our present study is a retrospective, single-institution study with a relatively small number of patients. A prospective and well-designed study with larger number of patients with HCC with radical surgery is needed. Second, why does the level of c-Myc expression in HCC tissues were lower than those in adjacent tissues? Why was the level of c-Myc in HCC tissues not correlated with TNM stage? Whether c-Myc plays the important roles on carcinogenesis and early stage of HCC? Thus, it is worthwhile to perform further studies to verify our findings and test their clinical application for patients with HCC.

Conclusion

Our study demonstrates that low expression of c-Myc protein predicts poor outcomes in patients with HCC with hepatectomy. The combination of TGF-β1, ELF and c-Myc can be used to accurately predict outcomes in patients with HCC. From a diagnostic point of view, our results prompt that the detection of c-Myc alone or combining with ELF/ TGF-β1 in tumor tissues could be a new prognostic marker in patients with HCC. We can depend on them to provide the personalized comprehensive therapies for patients with HCC, especially patients with any two or all of low c-Myc, high TGF-β1, and low ELF expression in tumor tissues. Further studies should be conducted to confirm our hypothesis that c-Myc proteins play an important role on hepato-carcinogenesis and early stage of HCC, and to elucidate the critical mechanisms of c-Myc in HCC.

Abbreviations

AFP:

Alpha-fetoprotein

CI:

Confidence interval

CT:

Computed tomography

DFS:

Disease Free survival

ELF:

Embryonic Liver Fodrin

HBsAg:

Hepatitis B surface antigen

HCC:

Hepatocellular cancer

IRS:

Immunoreactivity score

MRI:

Magnetic resonance imaging

OS:

Overall survival

PLT:

Platelet

PVTT:

portal vein tumor thrombi. HR: Hazard ratio

qRT-PCR:

Real -time Quantitative Polymerase Chain Reaction

TACE:

Transcatheter arterial chemoembolization

TGF-β:

Transforming growth factor β

TNM:

Tumor-node-metastasis

References

  1. Yao H, Bian X, Mao L, Zi X, Yan X, Qiu Y. Preoperative enteral nutritional support in patients undergoing hepatectomy for hepatocellular carcinoma: a strengthening the reporting of observational studies in epidemiology article. Medicine (Baltimore). 2015;94:e2006.

    Article  CAS  Google Scholar 

  2. Ji F, Liang Y, Fu SJ, Guo ZY, Shu M, Shen SL, et al. A novel and accurate predictor of survival for patients with hepatocellular carcinoma after surgical resection: the neutrophil to lymphocyte ratio (NLR) combined with the aspartate aminotransferase/platelet count ratio index (APRI). BMC Cancer 2016; 22. 16:137.

  3. Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the global burden of disease study 2010. Lancet. 2012;380:2095–128.

    Article  PubMed  Google Scholar 

  4. El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epide¬miology and molecular carcinogenesis. Gastroenterology. 2007;132:2557–76.

    Article  CAS  PubMed  Google Scholar 

  5. Zhi X, Lin L, Yang S, Bhuvaneshwar K, Wang H, Gusev Y, et al. βII-spectrin (SPTBN1) suppresses progression of hepatocellular carcinoma and Wnt signaling via regulation of Wnt inhibitor Kallistatin. Hepatology. 2015;61:598–612.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pelengaris S, Khan M. The many faces of c-MYC. Arch Biochem Biophys. 2003;416:129–36.

    Article  CAS  PubMed  Google Scholar 

  7. Hoffman B, Liebermann DA. Apoptotic signaling by c-MYC. Oncogene. 2008;27:6462–72.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  9. Shiraha H, Yamamoto K, Namba M. Human hepatocyte carcinogenesis. (Review) Int J oncol. 2013;42:1133–8.

    Article  CAS  PubMed  Google Scholar 

  10. Cairo S, Wang Y, de Reyniès A, Duroure K, Dahan J, Redon MJ, et al. Stem cell-like micro-RNA signature driven by Myc in aggressive liver cancer. Proc Natl Acad Sci U S A. 2010;107:20471–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Dang CV. C-Myc target genes involved in cell growth, apoptosis, and metabolism. Mol Cell Biol. 1999;19:1–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sakamuro D, Eviner V, Elliott KJ, Showe L, White E, Prendergast GC. C-Myc induces apoptosis in epithelial cells by both p53-dependent and p53-independent mechanisms. Oncogene. 1995;11:2411–8.

    CAS  PubMed  Google Scholar 

  13. Cook JR, Goldman B, Tubbs RR, Rimsza L, Leblanc M, Stiff P, et al. Clinical significance of MYC expression and/or "high-grade" morphology in non-Burkitt, diffuse aggressive B-cell lymphomas: a SWOG S9704 correlative study. Am J SurgPathol. 2014;38:494–501.

    Article  Google Scholar 

  14. Nevzorova YA, Hu W, Cubero FJ, Haas U, Freimuth J, Tacke F, et al. Overexpression of c-myc in hepatocytes promotes activation of hepatic stellate cells and facilitates the onset of liver fibrosis. Biochim Biophys Acta. 2013;1832:1765–75.

    Article  CAS  PubMed  Google Scholar 

  15. Lin CP, Liu CR, Lee CN, Chan TS, Liu HE. Targeting c-Myc as a novel approach for hepatocellular carcinoma. World J Hepatol. 2010;2:16–20.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Frau M, Biasi F, Feo F, Pascale RM. Prognostic markers and putative therapeutic targets for hepatocellular carcinoma. Mol Asp Med. 2010;31:179–93.

    Article  CAS  Google Scholar 

  17. Zimonjic DB, Popescu NC. Role of DLC1 tumor suppressor gene and MYC oncogene in pathogenesis of human hepatocellular carcinoma: potential prospects for combined targeted therapeutics. (Review) Int J Oncol. 2012;41:393–406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ersahin T, Ozturk M, Cetin‐Atalay R. Molecular Biology of Liver Cancer. Rev Cell Biol Mol Med.2015;1:206–43

  19. Wang Y, Wu MC, sham JS, Zhang W, Wu WQ, guan XY. Prognostic significance of cmyc and AIB1 amplification in hepatocellular carcinoma.A broad survey using high-throughput tissue microarray. Cancer. 2002;95:2346–52.

    Article  CAS  PubMed  Google Scholar 

  20. Srivastava J, Siddiq A, Gredler R, Shen XN, Rajasekaran D, Robertson CL, et al. Astrocyte elevated gene-1 (AEG-1) and c-Myc cooperate to promote Hepatocarcinogenesis. Hepatology. 2015;61:915–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Srivastava J, Siddiq A, Gredler R, Shen XN, Rajasekaran D, Robertson CL, et al. MYC inactiva¬tion uncovers pluripotent differentiation and tumour dormancy in hepatocellular cancer. Nature. 2004;431:1112–7.

    Article  Google Scholar 

  22. Bouchard C, Lee S, Paulus-Hock V, Loddenkemper C, Eilers M, Schmitt CA. FoxO transcription factors suppress Myc-driven lymphomagenesis via direct activation of Arf. Genes Dev. 2007;21:2775–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Finch A, Prescott J, Shchors K, Hunt A, Soucek L, Dansen TB, et al. Bcl-xL gain of function and p19 ARF loss of function cooperate oncogenically with Myc in vivo by distinctmechanisms. Cancer Cell. 2006;10:113–20.

    Article  CAS  PubMed  Google Scholar 

  24. Pelengaris S, Khan M, Evan GI. Suppression of Myc-induced apoptosis in beta cells exposes multiple oncogenic properties of Myc and triggers carcinogenic progression. Cell. 2002;109:321–34.

    Article  CAS  PubMed  Google Scholar 

  25. Murakami H, Sanderson ND, Nagy P, Marino PA, Merlino G, Thorgeirsson SS. Transgenic mouse model for synergistic effects of nuclear oncogenes and growth factors in tumorigenesis: interaction of c-myc and transforming growth factor alpha in hepatic oncogenesis. Cancer Res. 1993;53:1719–23.

    CAS  PubMed  Google Scholar 

  26. Lee JS, Chu IS, Mikaelyan A, Calvisi DF, Heo J, Reddy JK, et al. Application of comparative functional genomics to identify best-fit mouse models to study human cancer. Nat Genet. 2004;36:1306–11.

    Article  CAS  PubMed  Google Scholar 

  27. Kaposi-Novak P, Libbrecht L, Woo HG, Lee YH, Sears NC, Coulouarn C, et al. Central role of c-Myc during malignant conversion in human hepatocarcinogenesis. Cancer Res. 2009;69:2775–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Akita H, Marquardt JU, Durkin ME, Kitade M, Seo D, Conner EA, et al. MYC activates stem-like cell potential in Hepatocarcinoma by a p53-dependent mechanism. Cancer Res. 2014;74:5903–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Murphy DJ, Junttila MR, Pouyet L, Karnezis A, Shchors K, Bui DA, et al. Distinct thresholds govern Myc’s biological output in vivo. Cancer Cell. 2008;14:447–57.

    Article  CAS  PubMed  Google Scholar 

  30. Ji F, Fu SJ, Shen SL, Zhang LJ, Cao QH, Li SQ, et al. The prognostic value of combined TGF-β1 and ELF in hepatocellular carcinoma. BMC Cancer. 2015;15:116.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Hua YP, Yin XY, Peng BG, Li SQ, Lai JM, Liang HZ, et al. Mechanisms and influence of octreotide-induced regulation of somatostatin receptor 2 on hepatocellular carcinoma. Chemotherapy. 2009;55:312–20.

    Article  CAS  PubMed  Google Scholar 

  32. Hua YP, Li SQ, Lai JM, Liang LJ, Peng BG, Liang HZ, et al. Changes in TGF-β/Smads signaling pathway in rats with chemical hepatocarcinogenesis. J South Med Univ. 2008;28:1848–52.

    CAS  Google Scholar 

  33. Santoni-Rugiu E, Jensen MR, Factor VM, Thorgeirsson SS. Acceleration of c-myc-induced hepatocarcinogenesis by co-expression of transforming growth factor (TGF)-alpha in transgenic mice is associated with TGF-beta1 signaling disruption. Am J Pathol. 1999;154:1693–700.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Jin J, Zhu P, Liao Y, Li J, Liao W, He S. Elevated preoperative aspartate aminotransferase to lymphocyte ratio index as an independent prognostic factor for patients with hepatocellular carcinoma after hepatic resection. Oncotarget. 2015;6:19217–27.

    PubMed  PubMed Central  Google Scholar 

  35. Cui J, Dong BW, Liang P, Yu XL, Yu DJ. Effect of c-myc, Ki-67, MMP-2 and VEGF expression on prognosis of hepatocellular carcinoma patients undergoing tumor resection. World J Gastroenterol. 2004;10:1533–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yuen MF, Wu PC, Lai VC, Lau JY, Lai CL. Expression of c-Myc, c-Fos, and c-Jun in hepatocellular carcinoma. Cancer. 2001;91:106–12.

    Article  CAS  PubMed  Google Scholar 

  37. Tabrizian P, Jibara G, Shrager B, Schwartz M, Roayaie S. Recurrence of hepatocellular Cancer after resection: patterns, treatments, and prognosis. Ann Surg. 2015;261:947–55.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Professor Mushfiquddin Khan (Department of Pediatrics, Medical University of South Carolina) for serious revision and providing insightful comments on this study.

Funding

This study was supported by grants from Science and Technology Program of Guangzhou (201707010387), the Science and Technology Project of Guangdong Province (No. 2014A020212626) and the Scientific Research Foundation for Returned Overseas Chinese Scholars, State Education Ministry (2015, no. 311).

Availability of data and materials

All data generated or analyzed during this study are included in this article. Also the data sets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Author information

Authors and Affiliations

Authors

Contributions

FJ, ZHZ, YZ and YPH were the main authors of the manuscript. They were involved in the conception, design and coordination of the study as well as in data analysis, interpretation of results and drafting the manuscript. YPH was in-charge of all experimental procedures. SLS, LJZ, QHC, SQL, BGP, and LJL participated in the experimental procedures and revised critically the content of the manuscript. All authors contributed to the interpretation of data and critically revised the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Yun-Peng Hua.

Ethics declarations

Ethics approval and consent to participate

This study was retrospective study and approved by the Ethics Review Committee of the First Affiliated Hospital of Sun Yat-sen University. All participants provided written informed consent to participate.

Competing interests

The authors declare that they have no competing interests, and they consented to publish.

Publisher’s Note

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

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ji, F., Zhang, Zh., Zhang, Y. et al. Low expression of c-Myc protein predicts poor outcomes in patients with hepatocellular carcinoma after resection. BMC Cancer 18, 460 (2018). https://doi.org/10.1186/s12885-018-4379-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-018-4379-5

Keywords