Lymph node (LN) metastasis affects both the management and prognosis of head and neck squamous cell carcinoma (HNSCC). Here, we explored the relationship between lymphatic metastasis and CEA family member 5 (CEACAM5), including its possible regulatory role in HNSCC. The levels of CEACAM5 in tissues from patients with HNSCC, with and without LN metastases, were assessed by transcriptome sequencing. The associations between CEACAM5 and the N stage of LN metastasis in HNSCC were predicted through The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases and a pan-cancer analysis of CEACAM5 expression in 33 common human tumors was conducted. CEACAM5 levels were analyzed in tumor and normal tissue specimens from HNSCC patients and the correlation between CEACAM5 levels and prognosis was evaluated. The influence of CEACAM5 on cell proliferation, invasion, migration, and apoptosis was investigated in HNSCC cell lines, as were the downstream regulatory mechanisms. A mouse model of LN metastasis was constructed. CEACAM5 levels were significantly higher in HNSCC tissue without LN metastasis than in that with LN metastasis. Similar findings were obtained for the clinical specimens. CEACAM5 levels were associated with better clinical prognosis. CEACAM5 was found to inhibit the proliferation and migration and promote the apoptosis of HNSCC cells. A mouse xenograft model showed that CEACAM5 inhibited LN metastasis. In conclusions, CEACAM5 inhibited epithelial–mesenchymal transition (EMT) in HNSCC by reducing murine double minute 2 (MDM2) expression and thereby suppressing LN metastasis. CEACAM5 has potential as both a prognostic marker and a therapeutic target in HNSCC.

Head and neck squamous cell carcinoma (HNSCC) originates from mucosal epithelial cells in the mouth, pharynx, and larynx [1] and affects approximately 600000 people per year worldwide [2,3]. Oral cavity tumors and laryngeal cancer are often associated with excessive tobacco and alcohol consumption, while there is growing evidence that pharyngeal cancer is linked to human papillomavirus (HPV) infection, mainly HPV-16 [4]. Moreover, HPV-negative and HPV-positive HNSCC differ both genetically and in clinicopathological appearance [5]. More than 50% of patients with HNSCC have locally advanced lesions at first diagnosis and between 60 and 80% of them exhibit lymph node (LN) metastasis when starting treatment [6–9]. Despite the availability of a variety of comprehensive therapeutic options, the prognosis of HNSCC patients remains poor, with a five-year survival of approximately 60% for patients with laryngeal cancer and 25% for those with hypopharyngeal cancer [10–12]. Accordingly, novel strategies are needed for the effective prevention and treatment of LN metastasis.

RNA sequencing is widely used in both cancer diagnosis and research into therapeutic strategies for this disease [13,14], including the exploration of biomarkers and the characterization of cancer heterogeneity, drug resistance, and the immune microenvironment [15–17]. Single-cell sequencing has shown that the epithelial–mesenchymal transition (EMT) signature can independently predict LN metastasis, cancer grade, and adverse pathological features [18,19]. The CEA family member 5 (CEACAM5) family of proteins includes both membrane junction and secretory glycoproteins. Membrane junction glycoproteins are either embedded within membranes via glycosylphosphatidylinositol (GPI) anchors or attached to the cell surface through transmembrane domains. GPI-anchored family members include CEACAM5, CEACAM6, CEACAM7, and CEACAM8 [20]. CEACAM5 is involved in the regulation of cell differentiation, apoptosis, and cell polarity, and can thus affect tumor development [21]. This protein can also serve as a biomarker of gastrointestinal cancer [22]. Murine double minute 2 (MDM2) is a RING-domain E3 ubiquitin ligase that, when phosphorylated, translocates to the nucleus, where it regulates the expression of a variety of genes in both tumor and normal cells [23]. Interaction between the NH2-terminal domain of MDM2 and the α-helix in the NH2-terminal domain of p53 induces the degradation of the latter, thereby promoting carcinogenesis [24].

EMT involves the acquisition of mesenchymal features by epithelial cells and is normally active during tissue development and repair, as well as stem cell functions [25]. EMT reactivation is associated with tumorigenesis, cancer cell invasion and metastasis, and drug resistance [26–28]. This transformation in function is largely regulated by the transcription factors Twist, Slug, Snail, and ZEB1. In addition, EMT can inhibit cell senescence and apoptosis and increase the tolerance to radiotherapy and chemotherapy [29,30].

In the present study, CEACAM5 expression was observed to be significantly lower in both HNSCC samples and cell lines. Importantly, we found that CEACAM5 inhibits HNSCC lymphatic metastasis by negatively regulating MDM2 expression and thereby inhibiting EMT both in vivo and in vitro. These findings provide insights into the function of CEACAM5 in HNSCC.

Tissues and patient data

Fresh HNSCC and adjacent normal tissue specimens were collected from patients undergoing tumor resection in the First Affiliated Hospital of Chongqing Medical University between 2014 and 2022. The specimens included 57 paraffin-embedded tumor tissues that were used for histological analysis. Patients who had received preoperative antitumor therapy such as radiotherapy or chemotherapy were excluded. The study was carried out in accordance with the Declaration of Helsinki and was authorized by the Ethics Committee of the First Affiliated Hospital of Chongqing Medical University.

RNA extraction and quantitative reverse transcription-PCR

Total RNA was extracted from fresh frozen samples using an E.Z.N.A. Total RNA Kit I (Omega Bio-Tek, U.S.A.). The extracted RNA was reverse transcribed into cDNA (37°C for 15 min, 70°C for 5 min) using a PrimeScript RT Reagent Kit (Takara, Dalian, China). Quantitative-PCR (qPCR) was performed with the SYBR PrimeScript RT-PCR Kit (Takara). GAPDH served as the internal control and relative gene expression levels were calculated using the 2−ΔΔCt method. The sequences of the primers used for qPCR are shown in Additional file 1: Supplementary Table S1.

Protein extraction and western blotting

Total protein was isolated using a protein extraction kit (KGP250, KeyGen, Jiangsu, China). The lysates were centrifuged and the protein concentration in the supernatants was measured using the BCA method (P0010S, Beyotime, Shanghai, China). Equal amounts of protein were separated via 10% SDS-PAGE and electroblotted onto polyvinylidene fluoride (PVDF) membranes. After blocking, the membranes were incubated overnight at 4°C with primary antibodies targeting CEACAM5 (Abcam, U.S.A.), MDM2 (Cell Signaling Technology, U.S.A.), E-cadherin (Cell Signaling Technology), N-cadherin (Cell Signaling Technology), vimentin (Cell Signaling Technology), LYVE-1 (Abcam), and GAPDH (Abcam). After washing, the membranes were incubated with a secondary antibody for 1 h at room temperature. An enhanced chemiluminescence (ECL) kit (Thermo, Shanghai, China) was used for visualization. Detailed antibody information is provided in Additional file 2: Supplementary Table S2.

Immunohistochemistry

An immunohistochemistry (IHC) kit (SP-9000, Beijing, Zhongshan Jinqiao, China) was used to stain 4-μm-thick paraffin-embedded tumor sample sections. Specifically, after dewaxing and rehydration in fresh xylene and a graded alcohol series, antigen retrieval was performed with citrate buffer at 95°C for 15 min, followed by incubation with an endogenous peroxidase blocker for 15 min at 37°C. The sections were then incubated first with primary antibodies overnight at 4°C and then with HRP-conjugated streptavidin (100 μL) for 15 min, followed by counterstaining with hematoxylin and visualization with diaminobenzidine (DAB). Different fields of view of each section were randomly imaged under a light microscope at ×200 magnification. Gray levels were estimated in ImageJ and graded as follows: 0, negative; 1–3, weak; 4–5, medium; and 6–7, strong.

Cell culture

FaDu and SCC15 cells were obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). Cells were grown in Dulbecco’s Modified Eagle’s Medium (DMEM; Gibco, MA, U.S.A.) containing 10% fetal bovine serum (FBS; Gibco, Australia) and 1% penicillin/streptomycin (Beyotime, Shanghai, China) at 37°C with 5% CO2.

RNA interference and cell transfection

The siRNA used in the present study was obtained from GenePharma (Shanghai, China). Cells were seeded in 6-well plates (2.5 × 105 cells per well) and allowed to grow to between 60 and 70% confluence. The cells were then transfected with siRNA using Lipofectamine iMAX (Invitrogen, U.S.A.) and incubated in serum-free DMEM for 6 h. The medium was then replaced with DMEM supplemented with 10% FBS and the cells were cultured for a further 48–72 h. RNA and protein were extracted for the assessment of transfection efficiencies as well as to select the best sequence for use in subsequent experiments.

Lentiviral vectors and cell transduction

To knock down CEACAM5, short hairpin RNA (shRNA)-targeting human CEACAM5 was cloned into the hU6-MCS-Ubiquitin-firefly_Luciferase-IRES-puromycin lentiviral vector (GV344, Genechem, Shanghai, China). For CEACAM5 overexpression, full-length CEACAM5 cDNA was cloned into the Ubi-MCS-firefly_Luciferase-IRES-Puromycin lentiviral vector (GV260, Genechem). Specifically, cells were grown in 6-well plates (1 × 105 cells per well) to between 20 and 30% confluence. Lentivirus was transfected into the cells at a multiplicity of infection (MOI) of 10 using HiTransG A/P infection-enhancing solution. After 24 h, the cells were incubated in DMEM with 10% FBS and 2 μg/mL puromycin for 1 week, followed by selection.

Colony formation assay

Colony formation assays were used to assess the tumorigenic ability of the cells. Stably lentivirus-transduced FaDu and SCC15 cells were seeded in 6-well plates (1000 cells/well and 650 cells/well, respectively) in DMEM with 10% FBS and grown for approximately 2 weeks. The medium was replaced every 4 days. The cells were subsequently fixed in 4% paraformaldehyde and stained with 0.1% crystal violet for 20 min, and the number of colonies was counted.

Transwell migration and invasion assays

The migratory and invasive potential of the cells was assessed using 8-μm-pore Transwell inserts (Corning, U.S.A.) coated or not with Matrigel (BD Biosciences, San Jose, CA, U.S.A.). Specifically, 1 × 105 cells were suspended in 200 μL of serum-free medium and placed in the upper chamber of the inserts. The lower chamber contained 600 μL of DMEM supplemented with 15% FBS. After incubation for 24 h, cells remaining on the upper chamber of the membrane (unmigrated cells) were wiped off with a cotton swab. Cells that had crossed the membrane (migrated cells) were fixed in 4% paraformaldehyde and stained with 0.1% crystal violet for 20 min. The number of migrated cells was counted under a microscope.

Flow cytometry

For the analysis of apoptosis, an Annexin V-FITC/PI Detection Kit (SunGENE BioTEch, Tianjin, China) was used to label apoptotic cells. Cells were grown in 6-well plates (1 × 105/well) to 90% confluence, harvested, and subjected to flow cytometry (FCM; BD Biosciences, U.S.A.). For the cell cycle assay, stably transfected cells were fixed in 70% ice-cold ethanol overnight at 4°C, incubated with RNase A and propidium iodide, and then analyzed in a flow cytometer.

EdU proliferation assay

An EdU detection kit (RiBoBio, Guangzhou, China) was used for the cell proliferation assay. Briefly, cells were grown in 6-well plates to 70–80% confluence and then incubated with EdU (50 μM) at 37°C for 2 h. The cells were subsequently fixed in 4% paraformaldehyde, stained with Apollo for 30 min, and imaged under an inverted microscope.

Plasmid transfection and wound-healing assay

A total of 2.5 × 105 cells were seeded per well of a 6-well plate, allowed to grow to 60–70% confluence, and then transfected using a liposome transfection kit (NEOFECTTMDNA transfection reagent, Beijing, China). The medium was replaced after 24 h and the cells were grown to approximately 95% confluence. A 200-μL pipette tip was used to make four scratches in the cell monolayer. After being allowed to grow for 24 h in serum-free DMEM, the cells were imaged under an inverted microscope.

Generation of an animal model of inguinal LN metastasis

Experiments involving mice were performed at a specific pathogen-free facility at Chongqing Medical University. Male BALB/c nude mice aged between 4 and 6 weeks were purchased from HFK Bioscience Limited Company (Beijing, China). SCC15 cells (5 × 106 in 40 μL of PBS) transfected with the empty vector or the CEACAM5 overexpression plasmid were injected into the foot pads of the mice. After 35 days, the mice received intraperitoneal injections of 15 mg/mL D-Luciferin (Beyotime). Mice were monitored using in vivo bioluminescence imaging and, following euthanasia, the foot pad tumors and ipsilateral inguinal LNs were collected for further analysis.

Bioinformatics

Transcriptome sequencing was performed on 11 tumor tissues from HNSCC patients with or without LN metastasis [31]. Differentially expressed genes (DEGs) were identified using a threshold of P<0.05 and volcano plots were drawn using the Enhanced Volcano package in R. Protein–protein interaction (PPI) networks for genes related to CEACAM5 were compiled using the STRING database (https://cn.string-db.org/) [32] and visualized in Cytoscape [33]. GO enrichment analysis of the DEGs was performed through the Network Analyst database (https://www.networkanalyst.ca/) [34]. The HNSCC high-throughput sequencing datasets were downloaded from The Cancer Genome Atlas (TCGA) (https://portal.gdc.cancer.gov/) and Gene Expression Omnibus (GEO) databases (dataset GSE2379, platform GPL91).

Statistical analysis

GraphPad Prism version 8.0 (GraphPad Software, U.S.A.) and SPSS 20.0 (IBM Corp, NY, U.S.A.) were used for statistical analysis. T-tests were used for comparisons between two groups. One-way ANOVA was used for univariate comparisons among three or more groups. Multivariate between-group analysis was performed using two-way ANOVA. Pearson χ2 tests were used to assess correlations between CEACAM5 expression and clinicopathological features. Kaplan–Meier and logarithmic rank analyses were used to evaluate prognostic factors. All data were expressed as means ± standard deviation (SD). A P-value <0.05 was considered significant.

Low CEACAM5 expression in HNSCC is associated with LN (N) stage

To explore the role of CEACAM5 in human HNSCC, transcriptome sequencing was performed on six HNSCC tissues with LN metastases and five without. A total of 2294 DEGs between the two tissue types were identified (shown in the heatmap in Figure 1A and the volcano plot in Figure 1C). Expression profile microarray data for ten HNSCC cases with and four without LN metastases were obtained from the GEO database (depicted in the heatmap in Figure 1B). Analysis of the data indicated that CEACAM5 expression was lower in cases with LN metastases than in those without. We then conducted a pan-cancer analysis of CEACAM5 levels in 33 common human cancers together with an assessment of the disease risk ratio. We found that the levels of CEACAM5 were significantly higher in normal tissues (n=44) than in HNSCC tissues (n=520) and that the CEACAM5 risk ratio in HNSCC was less than 1 (P<0.05) (Figure 1D,E). In addition, CEACAM5 expression in HPV-positive HNSCC was higher than that in HPV-negative HNSCC (Figure 1D). These findings indicated that CEACAM5 may act as a tumor suppressor in HNSCC. The mRNA levels of CEACAM5 were markedly higher in normal tissues than in tumor tissues and were also higher at the N0 stage than at the N1 and N3 stages based on TCGA data (Figure 1F). Survival analysis based on the same dataset confirmed that high CEACAM5 levels were associated with better overall survival (OS), disease-free survival, and disease-specific survival (DSS) (Figure 1G–I). Combined, these findings suggested that CEACAM5 expression is significantly correlated with the N (LN) stage in HNSCC.

Low CEACAM5 expression in HNSCC is associated with lymph node (N) stage

Figure 1
Low CEACAM5 expression in HNSCC is associated with lymph node (N) stage

(A,C) Transcriptome sequencing was performed on HNSCC tissues and the results were visualized in a heatmap and volcano plot. CEACAM5 was identified as a DEG in HNSCC. L: HNSCC tissue with LN metastasis. NL: HNSCC tissue without LN metastasis. (B) Gene expression microarray data were downloaded from the GEO database and CEACAM5 was displayed in a heat map. The columns represent sample numbers and the rows represent genes. (D) CEACAM5 expression in 33 common human tumors and HPV correlation in HNSCC was analyzed based on data from TCGA database. (E) The risk ratio of CEACAM5 in 33 common human tumors was analyzed via a forest plot. (F) The CEACAM5 transcript levels were measured in 263 HNSCC and 44 normal tissues based on data from TCGA. Nχ represents lymph node (N) stage. (GI) Survival analysis relating to CEACAM5 in HNSCC based on data from TCGA. Abbreviations: OS, overall survival; PFS, progression-free survival; DSS, disease-specific survival. *P<0.05, **P<0.01, ***P<0.001.

Figure 1
Low CEACAM5 expression in HNSCC is associated with lymph node (N) stage

(A,C) Transcriptome sequencing was performed on HNSCC tissues and the results were visualized in a heatmap and volcano plot. CEACAM5 was identified as a DEG in HNSCC. L: HNSCC tissue with LN metastasis. NL: HNSCC tissue without LN metastasis. (B) Gene expression microarray data were downloaded from the GEO database and CEACAM5 was displayed in a heat map. The columns represent sample numbers and the rows represent genes. (D) CEACAM5 expression in 33 common human tumors and HPV correlation in HNSCC was analyzed based on data from TCGA database. (E) The risk ratio of CEACAM5 in 33 common human tumors was analyzed via a forest plot. (F) The CEACAM5 transcript levels were measured in 263 HNSCC and 44 normal tissues based on data from TCGA. Nχ represents lymph node (N) stage. (GI) Survival analysis relating to CEACAM5 in HNSCC based on data from TCGA. Abbreviations: OS, overall survival; PFS, progression-free survival; DSS, disease-specific survival. *P<0.05, **P<0.01, ***P<0.001.

Close modal

CEACAM5 expression is down-regulated in HNSCC tissue samples and is correlated with clinical prognosis

The sequencing datasets for 53 normal and 520 HNSCC tissues were downloaded from TCGA. Analysis of the data showed that CEACAM5 levels were significantly higher in normal tissue than in HNSCC tissue (Figure 2A). Next, we analyzed CEACAM5 mRNA levels in nine HNSCC specimens without and nine with LN metastases, as well as in control tissues. The results showed that CEACAM5 levels in the nonmetastasis group were higher than those in the metastasis group (Figure 2C). The CEACAM5 protein levels were consistent with the results of the mRNA analysis as determined by western blotting (WB) (Figure 2B,D) and further confirmed by IHC (Figure 2E–G). Subsequent univariate and multivariate Cox regression analyses indicated that CEACAM5 expression was a prognostic factor for HNSCC. The clinicopathological data are shown in Tables 1 and 2. Finally, survival analysis demonstrated that high CEACAM5 levels were linked with improved OS in patients with HNSCC (Figure 2H). Together, these findings indicated that CEACAM5 expression is down-regulated in HNSCC and that elevated CEACAM5 levels are associated with better clinical outcome.

CEACAM5 expression is down-regulated in HNSCC tissue samples and is correlated with prognosis

Figure 2
CEACAM5 expression is down-regulated in HNSCC tissue samples and is correlated with prognosis

(A) CEACAM5 expression was analyzed based on data from TCGA. (B) The protein expression of CEACAM5 in HNSCC tissues and adjacent normal tissues was analyzed by WB. NAT represents normal adjacent tissue, and T represents tumor tissue. (C) CEACAM5 expression in 18 HNSCC tissues with or without LN metastasis and adjacent normal tissues was analyzed by quantitative reverse transcription-PCR (qRT-PCR). LM(+) represents lymphatic metastasis, and LM(−) represents nonlymphatic metastasis. (D) CEACAM5 protein expression in HNSCC tissues with or without LN metastasis was assessed by WB. (E) The expression of CEACAM5 in 57 paraffin-embedded HNSCC tissue sections with or without lymphatic metastasis was analyzed via IHC. Scale bars: 200×, 200 μm; 400×, 100 μm. (F) Positive and negative controls. (G,H) Proportion distribution and survival curve relating to CEACAM5 expression in 57 HNSCC paraffin-embedded sections are shown. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.

Figure 2
CEACAM5 expression is down-regulated in HNSCC tissue samples and is correlated with prognosis

(A) CEACAM5 expression was analyzed based on data from TCGA. (B) The protein expression of CEACAM5 in HNSCC tissues and adjacent normal tissues was analyzed by WB. NAT represents normal adjacent tissue, and T represents tumor tissue. (C) CEACAM5 expression in 18 HNSCC tissues with or without LN metastasis and adjacent normal tissues was analyzed by quantitative reverse transcription-PCR (qRT-PCR). LM(+) represents lymphatic metastasis, and LM(−) represents nonlymphatic metastasis. (D) CEACAM5 protein expression in HNSCC tissues with or without LN metastasis was assessed by WB. (E) The expression of CEACAM5 in 57 paraffin-embedded HNSCC tissue sections with or without lymphatic metastasis was analyzed via IHC. Scale bars: 200×, 200 μm; 400×, 100 μm. (F) Positive and negative controls. (G,H) Proportion distribution and survival curve relating to CEACAM5 expression in 57 HNSCC paraffin-embedded sections are shown. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.

Close modal
Table 1
Correlation between CEACAM5 expression and clinicopathological characteristics of HNSCC patients
CharacteristicsNo. of cases (%)CEACAM5 expressionX2P-value
Low (N=33)High (N=24)
Age (y)      
<60 23 (40.4) 13 10 0.030 0.863 
≥60 34 (59.6) 20 14   
Gender      
Male 56 (98.2) 33 23 1.400 0.237 
Female 1 (1.8)   
Cigarette      
No 9 (15.8) 0.793 0.373 
Yes 48 (84.2) 29 19   
Alcohol      
No 17 (29.8) 0.244 0.621 
Yes 40 (70.2) 24 16   
T classification      
T1 3 (5.3) 3.186 0.364 
T2 9 (15.8)   
T3 26 (45.6) 15 11   
T4 19 (33.3) 13   
N classification      
N0 17 (29.8) 11 12.494 0.006** 
N1 8 (14.0)   
N2 27 (47.4) 21   
N3 5 (8.8)   
Pathologic differentiation      
Poor 14 (24.6) 10 1.508 0.470 
Moderate 29 (50.9) 15 14   
Well 14 (24.6)   
LN metastasis      
No 17 (29.8) 11 5.076 0.024* 
Yes 40 (70.2) 27 13   
Extranodal extension      
No 46 (80.7) 23 23 6.095 0.014* 
Yes 11 (19.3) 10   
CharacteristicsNo. of cases (%)CEACAM5 expressionX2P-value
Low (N=33)High (N=24)
Age (y)      
<60 23 (40.4) 13 10 0.030 0.863 
≥60 34 (59.6) 20 14   
Gender      
Male 56 (98.2) 33 23 1.400 0.237 
Female 1 (1.8)   
Cigarette      
No 9 (15.8) 0.793 0.373 
Yes 48 (84.2) 29 19   
Alcohol      
No 17 (29.8) 0.244 0.621 
Yes 40 (70.2) 24 16   
T classification      
T1 3 (5.3) 3.186 0.364 
T2 9 (15.8)   
T3 26 (45.6) 15 11   
T4 19 (33.3) 13   
N classification      
N0 17 (29.8) 11 12.494 0.006** 
N1 8 (14.0)   
N2 27 (47.4) 21   
N3 5 (8.8)   
Pathologic differentiation      
Poor 14 (24.6) 10 1.508 0.470 
Moderate 29 (50.9) 15 14   
Well 14 (24.6)   
LN metastasis      
No 17 (29.8) 11 5.076 0.024* 
Yes 40 (70.2) 27 13   
Extranodal extension      
No 46 (80.7) 23 23 6.095 0.014* 
Yes 11 (19.3) 10   

*P<0.05, **P<0.01

Table 2
Univariate and multivariate analyses of various prognostic parameters in patients with HNSCC Cox-regression analysis
VariablesUnivariate analysisMultivariate analysis
PHazard ratio95% confidence intervalPHazard ratio95% confidence interval
CEACAM5 0.036* 0.306 0.101–0.928 0.142 0.421 0.133–1.334 
Age 0.440 0.701 0.284–1.727    
Gender 0.654 0.048 0.286–5.275    
Cigarette 0.660 1.391 0.320–6.044    
Alcohol 0.752 0.856 0.325–2.253    
T classification 0.005** 3.052 1.407–6.620 0.012* 2.807 1.250–6.303 
N classification 0.026* 1.825 1.073–3.102 0.234 1.443 0.788–2.643 
Pathologic differentiation 0.122 0.597 0.310–1.149    
LN metastasis 0.173 2.356 0.686–8.090    
Extranodal extension 0.013* 3.203 1.285–7.989 0.832 1.124 0.381–3.317 
VariablesUnivariate analysisMultivariate analysis
PHazard ratio95% confidence intervalPHazard ratio95% confidence interval
CEACAM5 0.036* 0.306 0.101–0.928 0.142 0.421 0.133–1.334 
Age 0.440 0.701 0.284–1.727    
Gender 0.654 0.048 0.286–5.275    
Cigarette 0.660 1.391 0.320–6.044    
Alcohol 0.752 0.856 0.325–2.253    
T classification 0.005** 3.052 1.407–6.620 0.012* 2.807 1.250–6.303 
N classification 0.026* 1.825 1.073–3.102 0.234 1.443 0.788–2.643 
Pathologic differentiation 0.122 0.597 0.310–1.149    
LN metastasis 0.173 2.356 0.686–8.090    
Extranodal extension 0.013* 3.203 1.285–7.989 0.832 1.124 0.381–3.317 

*P<0.05, **P<0.01

CEACAM5 inhibits the proliferation, migration, and invasion and promotes the apoptosis of HNSCC cells in vitro

Cell culture experiments were employed to assess the role of CEACAM5 in HNSCC. First, FaDu cells were transfected with three siRNAs targeting different sites in CEACAM5 (si-CEACAM5#1, si-CEACAM5#2, and si-CEACAM5#3), and the silencing efficiency of the siRNAs was assessed by qRT-PCR and WB (Figure 3A,B). Si-CEACAM5#1 displayed the best knockdown efficiency and was used for subsequent studies. Next, we established stable CEACAM5 overexpression and knockdown FaDu and SCC15 cell lines using lentiviral infection. Overexpression and knockdown efficiencies were confirmed at both the mRNA and protein levels (Figure 3C–F). EdU cell proliferation assays were performed on stably transfected HNSCC cells in the logarithmic growth phase. The results showed that the proliferative ability of CEACAM5-silenced cells (both the FaDu and SCC15 lines) was higher than that of the respective controls, whereas the opposite was observed in CEACAM5-overexpressing cells (Figure 3H,I). In addition, CEACAM5-silenced cells formed more and larger colonies compared with control cells (Figure 4A). CEACAM5-silenced cells also displayed increased migratory and invasive capacity relative to that of control cells; however, this trend was reversed in CEACAM5-overexpressing cells (Figure 4B,C). Additionally, compared with the respective controls, CEACAM5 overexpression increased the cell apoptosis rate, whereas CEACAM5 depletion exerted the opposite effect (Figure 4D). These data indicated that CEACAM5 inhibits the proliferation, survival, and motility of HNSCC cells.

CEACAM5 inhibits the proliferative ability of HNSCC cells

Figure 3
CEACAM5 inhibits the proliferative ability of HNSCC cells

(A,B) The efficiencies of the different siRNAs in silencing CEACAM5 expression in FaDu cells were screened by qRT-PCR and WB. CEACAM5 knockdown and overexpression efficiencies in stably transduced FaDu and SCC15 cells were detected by qRT-PCR (C,D) and WB (E,F). (GI) Changes in the proliferative capacity of HNSCC cells after stable lentivirus-mediated transduction were investigated using an EdU proliferation assay. Scale bar: 100 μm. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001; Abbreviation: NS, not significant.

Figure 3
CEACAM5 inhibits the proliferative ability of HNSCC cells

(A,B) The efficiencies of the different siRNAs in silencing CEACAM5 expression in FaDu cells were screened by qRT-PCR and WB. CEACAM5 knockdown and overexpression efficiencies in stably transduced FaDu and SCC15 cells were detected by qRT-PCR (C,D) and WB (E,F). (GI) Changes in the proliferative capacity of HNSCC cells after stable lentivirus-mediated transduction were investigated using an EdU proliferation assay. Scale bar: 100 μm. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001; Abbreviation: NS, not significant.

Close modal

CEACAM5 inhibits proliferation, migration, and invasion and promotes apoptosis in HNSCC cells in vitro

Figure 4
CEACAM5 inhibits proliferation, migration, and invasion and promotes apoptosis in HNSCC cells in vitro

(A) A colony formation assay was used to assess the tumorigenicity of HNSCC cells. (B) The migratory and invasive abilities of FaDu and SCC15 cells were assessed by Transwell assay. (C) Flow cytometry was used to measure the rate of apoptosis. The relevant statistics are displayed in the right panel. Scale bar: 100 μm. *P<0.05, **P<0.01, ***P<0.001.

Figure 4
CEACAM5 inhibits proliferation, migration, and invasion and promotes apoptosis in HNSCC cells in vitro

(A) A colony formation assay was used to assess the tumorigenicity of HNSCC cells. (B) The migratory and invasive abilities of FaDu and SCC15 cells were assessed by Transwell assay. (C) Flow cytometry was used to measure the rate of apoptosis. The relevant statistics are displayed in the right panel. Scale bar: 100 μm. *P<0.05, **P<0.01, ***P<0.001.

Close modal

The overexpression of CEACAM5 inhibits lymphatic metastasis in vivo

To evaluate the role of CEACAM5 in lymphatic metastasis in vivo, we xenografted SCC15 cells transfected with the empty vector or the CEACAM5 overexpressing plasmid into the foot pads of nude mice. The sizes and weights of the primary tumors and inguinal metastatic LNs were smaller in the CEACAM5-overexpressing group relative to those of the empty vector group (Figure 5A–E,G–I). Additionally, CEACAM5 overexpression reduced the LN metastatic potential of SCC15 cells (Figure 5F). CEACAM5 overexpression in the tumors was confirmed by qRT-PCR and WB (Figure 5J,K). Lastly, immunohistochemical analysis of the foot pad tumors showed greater staining intensities and proportion of CEACAM5 in tumors formed of CEACAM5-overexpressing cells, in contrast with reduced staining of LYVE-1 (Figure 5L). These findings indicated that CEACAM5 inhibits lymphatic metastasis in vivo.

CEACAM5 overexpression inhibits lymphatic metastasis in vivo

Figure 5
CEACAM5 overexpression inhibits lymphatic metastasis in vivo

(A) Procedure and animal grouping in the in vivo experiment. (B) Representative images of inguinal LN metastasis in nude mice. (C,G) Foot pad tumors and inguinal LNs collected from nude mice. (D,E) Foot pad tumor volumes and weights, respectively. (H,I) LN volumes and weights, respectively. (F) Representative bioluminescence images of inguinal LN metastasis in nude mice overexpressing CEACAM5. (J,K) The expression of CEACAM5 in foot pad tumors was analyzed by qRT-PCR and western blot. (L) The expression of CEACAM5 and the lymphangiogenesis factor LYVE-1 in primary tumor tissues of nude mice were evaluated by IHC. (M) Images of the negative control. Scale bar: CEACAM5, 200 μm; LYVE-1, 100 μm. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.

Figure 5
CEACAM5 overexpression inhibits lymphatic metastasis in vivo

(A) Procedure and animal grouping in the in vivo experiment. (B) Representative images of inguinal LN metastasis in nude mice. (C,G) Foot pad tumors and inguinal LNs collected from nude mice. (D,E) Foot pad tumor volumes and weights, respectively. (H,I) LN volumes and weights, respectively. (F) Representative bioluminescence images of inguinal LN metastasis in nude mice overexpressing CEACAM5. (J,K) The expression of CEACAM5 in foot pad tumors was analyzed by qRT-PCR and western blot. (L) The expression of CEACAM5 and the lymphangiogenesis factor LYVE-1 in primary tumor tissues of nude mice were evaluated by IHC. (M) Images of the negative control. Scale bar: CEACAM5, 200 μm; LYVE-1, 100 μm. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.

Close modal

CEACAM5 inhibits EMT in HNSCC cells

Numerous transcription factors have been implicated in tumorigenic processes. Among them, the EMT process is believed to be involved in tumor invasion and early metastasis. Accordingly, we used Spearman’s correlations to assess the relationships between CEACAM5 and key EMT-related transcription factors in HNSCC based on high-throughput data from TCGA. The results showed that CEACAM5 was negatively associated with these transcription factors, especially Snail, Slug, and Twist (Figure 6A). In addition, qRT-PCR analysis of the stably transduced FaDu and SCC15 cells showed that, compared with the controls, the levels of the epithelial marker E-cadherin were reduced in CEACAM5-silenced cells, whereas those of key EMT-related transcription factors and the interstitial markers N-cadherin and vimentin were increased. In contrast, in CEACAM5-overexpressing cells, the mRNA levels of E-cadherin were up-regulated, while those of Slug, N-cadherin, and vimentin were suppressed, compared with that seen in cells transfected with the empty vector (Figure 6B,C). These findings were confirmed at the protein level (Figure 6D,E). These observations implied that CEACAM5 inhibits EMT in HNSCC cells.

CEACAM5 inhibits EMT in HNSCC cells

Figure 6
CEACAM5 inhibits EMT in HNSCC cells

(A) Spearman’s correlation between the expression of EMT-related transcription factors and that of CEACAM5 was analyzed based on data from TCGA. (B,C) The mRNA expression levels of EMT-associated transcription factors and markers in stably transfected HNSCC cells were measured by qRT-PCR. (D,E) The protein expression levels of epithelial and mesenchymal markers in HNSCC cells after CEACAM5 knockdown or overexpression were detected by western blot. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001; Abbreviation: NS, not significant.

Figure 6
CEACAM5 inhibits EMT in HNSCC cells

(A) Spearman’s correlation between the expression of EMT-related transcription factors and that of CEACAM5 was analyzed based on data from TCGA. (B,C) The mRNA expression levels of EMT-associated transcription factors and markers in stably transfected HNSCC cells were measured by qRT-PCR. (D,E) The protein expression levels of epithelial and mesenchymal markers in HNSCC cells after CEACAM5 knockdown or overexpression were detected by western blot. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001; Abbreviation: NS, not significant.

Close modal

CEACAM5 inhibits the expression of MDM2

It has been reported that the phosphorylation of MDM2 allows its entry into the nucleus where it regulates the expression of numerous genes [35,36]. Here, using the STRING database, we constructed a PPI network based on the HNSCC transcriptome-sequencing data (Figure 7H). CEACAM5 and MDM2 were seen to share many similarities in GO term enrichment in the Biological Process category (Figure 7A,B). In addition, we analyzed the expression of CEACAM5 and MDM2 in HNSCC based on data from TCGA and found that CEACAM5 levels were significantly higher in normal tissues than in tumor tissues, whereas the MDM2 levels showed the opposite trend (Figure 7C,E). This suggested that a negative regulatory relationship may exist between CEACAM5 and MDM2 (Figure 7D). To confirm this possibility, we next assayed the stably transfected FaDu and SCC15 cell lines for MDM2 expression using qRT-PCR, with the results showing that MDM2 expression was higher in CEACAM5-knockdown cells than in control cells. In contrast, MDM2 levels were lower in CEACAM5-overexpressing cells than in cells transfected with the empty vector (Figure 7F,G). These findings were confirmed by WB (Figure 7J). Furthermore, the MDM2 mRNA levels were lower in CEACAM5-overexpressing mouse xenograft tumors compared with the empty vector group (Figure 7I). These findings confirmed the existence of a negative regulatory relationship between CEACAM5 and MDM2.

CEACAM5 inhibits the expression of MDM2

Figure 7
CEACAM5 inhibits the expression of MDM2

(A,B) Gene ontology (GO) terms related to the biological process (BP) category were assigned to genes associated with CEACAM5 and MDM2 based on HNSCC transcriptome-sequencing data. (C,E) The expression of CEACAM5 and MDM2 in normal and HNSCC tissues was analyzed based on data from TCGA. (D) Correlation analysis between MDM2 and CEACAM5 in HNSCC tissues based on data from TCGA (n=455). (F,G) The expression of MDM2 in HNSCC cells after stable lentivirus-mediated transduction was assessed by qRT-PCR. (H) PPI analysis was performed on genes related to the roles of CEACAM5 and MDM2 in HNSCC. (I) The mRNA expression levels of MDM2 in xenograft-derived tumors in nude mice were measured by qRT-PCR. (J) The protein expression of MDM2 in HNSCC cells was assessed by WB. The relevant statistics are shown in the right panel. *P<0.05, **P<0.01, ***P<0.001.

Figure 7
CEACAM5 inhibits the expression of MDM2

(A,B) Gene ontology (GO) terms related to the biological process (BP) category were assigned to genes associated with CEACAM5 and MDM2 based on HNSCC transcriptome-sequencing data. (C,E) The expression of CEACAM5 and MDM2 in normal and HNSCC tissues was analyzed based on data from TCGA. (D) Correlation analysis between MDM2 and CEACAM5 in HNSCC tissues based on data from TCGA (n=455). (F,G) The expression of MDM2 in HNSCC cells after stable lentivirus-mediated transduction was assessed by qRT-PCR. (H) PPI analysis was performed on genes related to the roles of CEACAM5 and MDM2 in HNSCC. (I) The mRNA expression levels of MDM2 in xenograft-derived tumors in nude mice were measured by qRT-PCR. (J) The protein expression of MDM2 in HNSCC cells was assessed by WB. The relevant statistics are shown in the right panel. *P<0.05, **P<0.01, ***P<0.001.

Close modal

MDM2 knockdown reverses the effect of CEACAM5 silencing on EMT

To further verify that a regulatory relationship indeed exists between CEACAM5 and MDM2, we silenced MDM2 in CEACAM5-knockdown cells. After puromycin selection for 1 week, the sh-CEACAM5-NC + sh-MDM2-NC, sh-CEACAM5-NC + sh-MDM2, sh-CEACAM5 + sh-MDM2-NC, and sh-CEACAM5 + sh-MDM2 SCC15 cell lines were constructed. We found that the mRNA level of the epithelial marker E-cadherin in sh-CEACAM5-NC + sh-MDM2 cells was increased in comparison with that in sh-CEACAM5-NC + sh-MDM2-NC cells. Moreover, the E-cadherin level was reduced in sh-CEACAM5 + sh-MDM2-NC cells compared with that in sh-CEACAM5-NC + sh-MDM2 cells and was higher in sh-CEACAM5 + sh-MDM2 cells than in sh-CEACAM5 + sh-MDM2-NC cells. Meanwhile, the mRNA expression levels of the EMT-related transcription factors as well as those of the interstitial proteins N-cadherin and vimentin displayed the opposite trend to that of E-cadherin (Figure 8A). These results were confirmed at the protein level (Figure 8B). Thus, the EMT process in cells in which both CEACAM5 and MDM2 were silenced was reversed in comparison with that in cells where only CEACAM5 was silenced.

MDM2 knockdown reversed the effect of CEACAM5 silencing on the EMT

Figure 8
MDM2 knockdown reversed the effect of CEACAM5 silencing on the EMT

sh-MDM2-NC or sh-MDM2 was transfected into HNSCC cells after stable lentivirus-mediated transduction. (A) The mRNA levels of EMT-related transcription factors and biomarkers were measured by qRT-PCR. (B) The protein expression levels of MDM2 and EMT biomarkers were determined by WB. The relevant statistics are shown in the bottom panel. (C) Flow cytometry was used to detect the cell-cycle distribution of different cell lines. (D) The wound healing capacity of HNSCC cells transfected with sh-MDM2-NC or sh-MDM2 is shown. (E,F) Diagram showing the mechanism of the hypothesis postulated in the present study (generated by Figdraw). *P<0.05, **P<0.01, ***P<0.001; Abbreviation: NS, not significant.

Figure 8
MDM2 knockdown reversed the effect of CEACAM5 silencing on the EMT

sh-MDM2-NC or sh-MDM2 was transfected into HNSCC cells after stable lentivirus-mediated transduction. (A) The mRNA levels of EMT-related transcription factors and biomarkers were measured by qRT-PCR. (B) The protein expression levels of MDM2 and EMT biomarkers were determined by WB. The relevant statistics are shown in the bottom panel. (C) Flow cytometry was used to detect the cell-cycle distribution of different cell lines. (D) The wound healing capacity of HNSCC cells transfected with sh-MDM2-NC or sh-MDM2 is shown. (E,F) Diagram showing the mechanism of the hypothesis postulated in the present study (generated by Figdraw). *P<0.05, **P<0.01, ***P<0.001; Abbreviation: NS, not significant.

Close modal

Cell cycle assays subsequently showed that the proportion of sh-CEACAM5-NC + sh-MDM2 cells in the S phase was reduced in comparison with that of sh-CEACAM5-NC + sh-MDM2-NC cells. Additionally, the proportion of sh-CEACAM5 + sh-MDM2-NC cells in the S phase was higher than that for sh-CEACAM5-NC + sh-MDM2 cells and was lower in sh-CEACAM5 + sh-MDM2 cells than in sh-CEACAM5 + sh-MDM2-NC cells. Finally, wound-healing assays showed that the migratory ability of sh-CEACAM5-NC + sh-MDM2 cells was reduced compared with that of sh-CEACAM5-NC + sh-MDM2-NC cells. Meanwhile, the migratory potential of sh-CEACAM5 + sh-MDM2-NC cells was greater than that of sh-CEACAM5-NC + sh-MDM2 cells and was lower in sh-CEACAM5 + sh-MDM2 cells than in sh-CEACAM5 + sh-MDM2-NC cells. In conclusion, the depletion of both MDM2 and CEACAM5 counteracted the inhibitory effect of CEACAM5 silencing on the EMT, resulting in EMT inhibition in HNSCC cells.

Cancer-related morbidity and mortality are increasing rapidly worldwide [37,38]. While excessive smoking is associated with an increased incidence of HNSCC in developing countries, HPV infection is increasingly recognized as an important factor contributing to the increased incidence of oropharyngeal tumors in nonsmokers in developed countries [39]. The therapeutic modality of HPV-positive and HPV-negative HNSCC is essentially similar, including traditional surgical and chemoradiotherapy modalities, as well as new surgical techniques such as transoral robotic surgery and laser microsurgery [40]. The specific treatment plan depends on the site and stage of tumor development. OS for patients with HNSCC remains poor despite the advances in its diagnosis and treatment [41], while LN metastasis and extranodal extension are both important predictors of worse prognosis in HNSCC [42–44]. These observations underline the urgent need for further exploration of the mechanisms underlying HNSCC occurrence and progression, as well as identify novel targets and prognostic markers and develop effective strategies for the diagnosis and treatment of this cancer.

CEA was originally isolated from the blood of patients with colorectal cancer by Gold and Freedman [45]. The expression of this protein has since been observed to be up-regulated in many other human tumors, including pancreatic, lung, liver, gallbladder, breast, and bladder cancers, among others [46–48]. Accordingly, studies have mostly focused on its role as an oncogene in promoting cancer progression. In the present study, transcriptome sequencing in HNSCC tissues from patients with and without LN metastasis and analysis of high-throughput data downloaded from TCGA and GEO databases indicated that CEACAM5 expression was significantly reduced in HNSCC tumors compared with that in normal tissues. Generally, HPV-positive cases are more sensitive to chemoradiotherapy and, thus, have a better prognosis and a higher survival rate compared with HPV-negative cases [49]. However, in the present study, CEACAM5 expression was found to be higher in HPV-positive HNSCC than in HPV-negative HNSCC, and its expression level was also significantly lower in HNSCC tissues than in normal tissues, with a disease risk ratio of less than 1. This suggests that alterations in CEACAM5 expression are more likely to be a protective manifestation against this disease. Subsequently, survival analysis confirmed that elevated CEACAM5 expression was linked with improved OS, progression-free survival, DSS, and lower N-stage classification in patients with HNSCC.

p53 has been described as the guardian of the genome and controls multiple cellular processes, including DNA repair, cell-cycle arrest, apoptosis, autophagy, senescence, and metabolism through numerous mechanisms [50,51]. Ubiquitination is a reversible post-translational modification that modulates protein degradation and signal transduction through the binding of ubiquitin to protein substrates. This process mainly involves the E1-E2-E3 enzyme cascade and requires the activity of E1 ubiquitin-activating enzymes, E2 ubiquitin-conjugating enzymes, and E3 ubiquitin ligases. In addition, the regulation of NF-κB signaling by ubiquitination may be a potential therapeutic target for HNSCC [52]. MDM2 is an E3 ubiquitin ligase that can modulate p53 levels by promoting its degradation by the proteasome [53,54] or by binding to and stabilizing Slug mRNA independently of its ubiquitination activity, thereby stimulating cell invasion and metastasis [55–57]. Here, by analyzing transcriptome sequencing data to obtain DEGs and employing the STRING database to construct a PPI network, we found that CEACAM5 and MDM2 may have a negative regulatory relationship. Through the Network Analyst database, we assigned GO terms to genes associated with CEACAM5 and MDM2 expression and found similar enrichment patterns. In addition, analysis of TCGA dataset indicated that CEACAM5 and MDM2 expression exhibited opposing trends in HNSCC. Combined, these data suggested that CEACAM5 may regulate EMT processes by suppressing the expression of MDM2.

Malignant tumors induce lymphatic dilation (lymphangiogenesis) in the primary tumor and the drainage sentinel area by releasing lymphatic growth factors such as VEGF-C and LYVE1, thereby promoting LN metastasis [58–61]. Moreover, EMT is known to drive the metastasis of cervical cancer cells to LNs [62]. Here, we investigated the effect of CEACAM5 on LN metastasis in vivo using bioluminescence imaging and used IHC to assess the expression of LYVE1 in foot pad tumors derived from xenografted SCC15 cells in mice. The obtained results indicated that CEACAM5 overexpression abrogated the lymphatic metastasis of SCC15 cells. Despite our important findings, our study had several limitations. First, the number of samples used for transcriptomic analysis was small. Second, although our study indicated that CEACAM5 inhibits MDM2 expression in HNSCC, further investigation is required to clarify the specific underlying mechanisms.

In conclusion, this is the first study to show that CEACAM5 expression is down-regulated in HNSCC and that CEACAM5 expression is correlated with survival outcomes in HNSCC patients. CEACAM5 was also found to inhibit EMT in HNSCC cells by suppressing the expression of MDM2 and, ultimately, also LN metastasis. These findings contribute to the understanding of the molecular mechanism involved in HNSCC development.

  • Background: OS in patients with HNSCC remains poor despite the advances in its diagnosis and treatment. Additionally, both LN metastasis and extranodal extension are important predictors of the prognosis of this cancer.

  • Results: Using in vivo and in vitro experiments as well as bioinformatics analysis, we found that CEACAM5 suppresses LN metastasis by suppressing EMT via inhibiting the expression of its downstream target MDM2.

  • Significance: This is the first study to suggest that CEACAM5 may play a role in inhibiting LN metastasis and also provides a potential therapeutic target and prognostic marker for HNSCC.

All data generated or analyzed during the present study are included in this published article.

The authors declare that there are no competing interests associated with the manuscript.

The present study was supported by the (National Natural Science Foundation of China) [grant number 82173303] and the (China Postdoctoral Science Foundation) [grant number 2021XM3029].

Xudong Wang: Resources, Data curation, Software, Investigation, Visualization, Methodology, Writing—original draft. Yanshi Li: Software, Supervision. Min Pan: Validation, Writing—review & editing. Tao Lu: Software, Methodology. Min Wang: Formal Analysis. Zhihai Wang: Software. Chuan Liu: Methodology. Guohua Hu: Resources, Supervision, Project administration, Writing—review & editing.

Not applicable.

Clinical specimens were obtained from patients undergoing hypophargiectomy at the First Affiliated Hospital of Chongqing Medical University. Informed consent was obtained from each patient. Meanwhile, all mice were suffocated with 1% sodium pentobarbital plus a high concentration of carbon dioxide. All experiments in the present study were conducted in accordance with the Code of Ethics of the World Medical Association (Declaration of Helsinki). All protocols for clinical specimens and in vivo experiments in mice were approved by the Ethics Committee of the First Affiliated Hospital of Chongqing Medical University (2022-K184), and were conducted in accordance with the UK Animals (Scientific Procedures) Act and the National Institutes of Health guidelines.

Thanks to Molecular Medicine and Cancer Research Center of Chongqing Medical University for providing experimental equipment and experimental platform.

BP

biological process

ANOVA

analysis of variance

PBS

phosphate buffered saline

SDS-PAGE

sodium dodecyl sulfate polyacrylamide gel electrophoresis

CEACAM5

carcinoembryonic antigen-related cell adhesion molecule 5

DEG

differentially expressed gene

EMT

epithelial–mesenchymal transition

FBS

fetal bovine serum

GEO

Gene Expression Omnibus

GPI

glycosylphosphatidylinositol

HNSCC

head and neck squamous cell carcinoma

HPV

human papillomavirus

IHC

immunohistochemistry

L

lymphatic metastasis

LM (+)

lymphatic metastasis

LM (−)

non-lymphatic metastasis

LN

lymph node

LYVE-1

lymphatic vessel endothelial hyaluronan receptor 1

MDM2

murine double minute 2

MOI

multiplicity of infection

NAT

normal adjacent tissue

NC

negative control

NL

no lymphatic metastasis

PPI

protein–protein interaction

qRT-PCR

quantitative reverse transcription-PCR

shRNA

short hairpin RNA

siRNA

small interfering RNA

T

tumor tissue

TCGA

The Cancer Genome Atlas

WB

western blotting

1.
Kang
H.
,
Kiess
A.
and
Chung
C.H.
(
2015
)
Emerging biomarkers in head and neck cancer in the era of genomics
.
Nat. Rev. Clin. Oncol.
12
,
11
26
[PubMed]
2.
Panvongsa
W.
,
Pegtel
D.M.
and
Voortman
J.
(
2022
)
More than a bubble: extracellular vesicle microRNAs in head and neck squamous cell carcinoma
.
Cancers
14
,
1160
,
[PubMed]
3.
Ferlay
J.
,
Colombet
M.
,
Soerjomataram
I.
,
Mathers
C.
,
Parkin
D.M.
,
Pineros
M.
et al.
(
2019
)
Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods
.
Int. J. Cancer
144
,
1941
1953
[PubMed]
4.
Johnson
D.E.
,
Burtness
B.
,
Leemans
C.R.
,
Lui
V.W.Y.
,
Bauman
J.E.
and
Grandis
J.R.
(
2020
)
Head and neck squamous cell carcinoma
.
Nat. Rev. Dis. Primers
6
,
92
,
5.
Seiwert
T.Y.
,
Zuo
Z.X.
,
Keck
M.K.
,
Khattri
A.
,
Pedamallu
C.S.
,
Stricker
T.
et al.
(
2015
)
Integrative and comparative genomic analysis of HPV-positive and HPV-negative head and neck squamous cell carcinomas
.
Clin. Cancer Res.
21
,
632
641
[PubMed]
6.
Zhang
X.-m.
,
Han
W.-x.
,
Wang
H.-y.
and
He
Q.
(
2017
)
Correlation between lymphatic endothelial markers and lymph node status or N-staging of colorectal cancer
.
World J. Surg. Oncol.
15
,
204
,
7.
Xing
Y.
,
Zhang
J.
,
Lin
H.
,
Gold
K.A.
,
Sturgis
E.M.
,
Garden
A.S.
et al.
(
2016
)
Relation between the level of lymph node metastasis and survival in locally advanced head and neck squamous cell carcinoma
.
Cancer
122
,
534
545
[PubMed]
8.
Shah
J.P.
,
Shaha
A.R.
,
Spiro
R.H.
and
Strong
E.W.
(
1976
)
Carcinoma of the hypopharynx
.
Am. J. Surg.
132
,
439
443
[PubMed]
9.
Amit
M.
,
Liu
C.
,
Gleber-Netto
F.O.
,
Kini
S.
,
Tam
S.
,
Benov
A.
et al.
(
2021
)
Inclusion of extranodal extension in the lymph node classification of cutaneous squamous cell carcinoma of the head and neck
.
Cancer
127
,
1238
1245
[PubMed]
10.
Licitra
L.
,
Locati
L.D.
and
Bossi
P.
(
2004
)
Head and neck cancer
.
Ann. Oncol.
15
,
267
273
11.
Karam
S.D.
and
Raben
D.
(
2019
)
Radioimmunotherapy for the treatment of head and neck cancer
.
Lancet Oncol.
20
,
E404
E416
[PubMed]
12.
Budach
V.
and
Tinhofer
I.
(
2019
)
Novel prognostic clinical factors and biomarkers for outcome prediction in head and neck cancer: a systematic review
.
Lancet Oncol.
20
,
E313
E326
[PubMed]
13.
Mroz
E.A.
,
Tward
A.M.
,
Hammon
R.J.
,
Ren
Y.
and
Rocco
J.W.
(
2015
)
Intra-tumor genetic heterogeneity and mortality in head and neck cancer: analysis of data from The Cancer Genome Atlas
.
PLoS Med.
12
,
e1001786
[PubMed]
14.
Byron
S.A.
,
Van Keuren-Jensen
K.R.
,
Engelthaler
D.M.
,
Carpten
J.D.
and
Craig
D.W.
(
2016
)
Translating RNA sequencing into clinical diagnostics: opportunities and challenges
.
Nat. Rev. Genet.
17
,
257
271
[PubMed]
15.
Peng
J.
,
Sun
B.-F.
,
Chen
C.-Y.
,
Zhou
J.-Y.
,
Chen
Y.-S.
,
Chen
H.
et al.
(
2019
)
Single-cell RNA-seq highlights intra-tumoral heterogeneity and malignant progression in pancreatic ductal adenocarcinoma
.
Cell Res.
29
,
725
738
[PubMed]
16.
Papalexi
E.
and
Satija
R.
(
2018
)
Single-cell RNA sequencing to explore immune cell heterogeneity
.
Nat. Rev. Immunol.
18
,
35
45
[PubMed]
17.
Hong
M.
,
Tao
S.
,
Zhang
L.
,
Diao
L.-T.
,
Huang
X.
,
Huang
S.
et al.
(
2020
)
RNA sequencing: new technologies and applications in cancer research
.
J. Hematol. Oncol.
13
,
166
,
18.
Zheng
Y.
and
Yang
X.
(
2022
)
Application and prospect of single-cell sequencing in cancer metastasis
.
Fut. Oncol.
18
,
2723
2736
19.
Puram
S.V.
,
Tirosh
I.
,
Parikh
A.S.
,
Patel
A.P.
,
Yizhak
K.
,
Gillespie
S.
et al.
(
2017
)
Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer
.
Cell
171
,
1611
+
[PubMed]
20.
Blumenthal
R.D.
,
Hansen
H.J.
and
Goldenberg
D.M.
(
2005
)
Inhibition of adhesion, invasion, and metastasis by antibodies targeting CEACAM6 (NCA-90) and CEACAM5 (Carcinoembryonic antigen)
.
Cancer Res.
65
,
8809
8817
[PubMed]
21.
Saeland
E.
,
Belo
A.I.
,
Mongera
S.
,
van Die
I.
,
Meijer
G.A.
and
van Kooyk
Y.
(
2012
)
Differential glycosylation of MUC1 and CEACAM5 between normal mucosa and tumour tissue of colon cancer patients
.
Int. J. Cancer
131
,
117
128
[PubMed]
22.
Blumenthal
R.D.
,
Leon
E.
,
Hansen
H.J.
and
Goldenberg
D.M.
(
2007
)
Expression patterns of CEACAM5 and CEACAM6 in primary and metastatic cancers
.
BMC Cancer
7
,
2
,
[PubMed]
23.
Wade
M.
,
Li
Y.-C.
and
Wahl
G.M.
(
2013
)
MDM2 MDMX and p53 in oncogenesis and cancer therapy
.
Nat. Rev. Cancer
13
,
83
96
[PubMed]
24.
Jung
C.-H.
,
Kim
J.
,
Park
J.K.
,
Hwang
S.-G.
,
Moon
S.-K.
,
Kim
W.-J.
et al.
(
2013
)
Mdm2 increases cellular invasiveness by binding to and stabilizing the Slug mRNA
.
Cancer Lett.
335
,
270
277
[PubMed]
25.
Lamouille
S.
,
Xu
J.
and
Derynck
R.
(
2014
)
Molecular mechanisms of epithelial-mesenchymal transition
.
Nat. Rev. Mol. Cell Biol.
15
,
178
196
[PubMed]
26.
Wang
X.
,
Xu
X.
,
Peng
C.
,
Qin
Y.
,
Gao
T.
,
Jing
J.
et al.
(
2019
)
BRAF(V600E)-induced KRT19 expression in thyroid cancer promotes lymph node metastasis via EMT
.
Oncol. Lett.
18
,
927
935
[PubMed]
,
27.
Pastushenko
I.
and
Blanpain
C.
(
2019
)
EMT transition states during tumor progression and metastasis
.
Trends Cell Biol.
29
,
212
226
[PubMed]
28.
Karlsson
M.C.
,
Gonzalez
S.F.
,
Welin
J.
and
Fuxe
J.
(
2017
)
Epithelial-mesenchymal transition in cancer metastasis through the lymphatic system
.
Mol. Oncol.
11
,
781
791
[PubMed]
29.
Liu
K.
,
Xue
B.
,
Bai
G.
and
Zhang
W.
(
2020
)
F-box protein FBXO31 modulates apoptosis and epithelial-mesenchymal transition of cervical cancer via inactivation of the PI3K/AKT-mediated MDM2/p53 axis
.
Life Sci.
259
,
118277
,
30.
De Craene
B.
and
Berx
G.
(
2013
)
Regulatory networks defining EMT during cancer initiation and progression
.
Nat. Rev. Cancer
13
,
97
110
[PubMed]
31.
Li
Y.
,
Lu
T.
and
Hu
G.
(
2020
)
Gene sequencing and expression of Raf-1 in lymphatic metastasis of hypopharyngeal carcinoma
.
Cancer Biomark.
28
,
181
191
[PubMed]
32.
Szklarczyk
D.
,
Gable
A.L.
,
Nastou
K.C.
,
Lyon
D.
,
Kirsch
R.
,
Pyysalo
S.
et al.
(
2021
)
The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets
.
Nucleic. Acids. Res.
49
,
D605
D612
[PubMed]
33.
Shannon
P.
,
Markiel
A.
,
Ozier
O.
,
Baliga
N.S.
,
Wang
J.T.
,
Ramage
D.
et al.
(
2003
)
Cytoscape: a software environment for integrated models of biomolecular interaction networks
.
Genome Res.
13
,
2498
2504
[PubMed]
34.
Zhou
G.
,
Soufan
O.
,
Ewald
J.
,
Hancock
R.E.W.
,
Basu
N.
and
Xia
J.
(
2019
)
NetworkAnalyst 3.0: a visual analytics platform for comprehensive gene expression profiling and meta-analysis
.
Nucleic. Acids. Res.
47
,
W234
W241
[PubMed]
35.
Chibaya
L.
,
Karim
B.
,
Zhang
H.
and
Jones
S.N.
(
2021
)
Mdm2 phosphorylation by Akt regulates the p53 response to oxidative stress to promote cell proliferation and tumorigenesis
.
Proc. Natl. Acad. Sci. U.S.A.
118
,
e2003193118
,
[PubMed]
36.
Carr
M.I.
,
Roderick
J.E.
,
Gannon
H.S.
,
Kelliher
M.A.
and
Jones
S.N.
(
2016
)
Mdm2 phosphorylation regulates its stability and has contrasting effects on oncogene and radiation-induced tumorigenesis
.
Cell Reports
16
,
2618
2629
[PubMed]
37.
Sung
H.
,
Ferlay
J.
,
Siegel
R.L.
,
Laversanne
M.
,
Soerjomataram
I.
,
Jemal
A.
et al.
(
2021
)
Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J. Clin.
71
,
209
249
[PubMed]
38.
Hirshoren
N.
,
Danne
J.
,
Dixon
B.J.
,
Magarey
M.
,
Kleid
S.
,
Webb
A.
et al.
(
2017
)
Prognostic markers in metastatic cutaneous squamous cell carcinoma of the head and neck
.
Head Neck-J. Sci. Spec. Head Nec.
39
,
772
778
39.
Lawrence
M.S.
,
Sougnez
C.
,
Lichtenstein
L.
,
Cibulskisl
K.
,
Lander
E.
,
Gabriel
S.B.
et al.
(
2015
)
Comprehensive genomic characterization of head and neck squamous cell carcinomas
.
Nature (London)
517
,
576
582
40.
Nichols
D.S.
,
Zhao
J.
,
Boyce
B.J.
,
Amdur
R.
,
Mendenhall
W.M.
,
Danan
D.
et al.
(
2021
)
HPV/p16-positive oropharyngeal cancer treated with transoral robotic surgery: The roles of margins, extra-nodal extension and adjuvant treatment
.
Am. J. Otolaryngol.
42
,
102793
,
[PubMed]
41.
Hsieh
R.W.
,
Borson
S.
,
Tsagianni
A.
and
Zandberg
D.P.
(
2021
)
Immunotherapy in recurrent/metastatic squamous cell carcinoma of the head and neck
.
Front. Oncol.
11
,
705614
,
42.
Matsumoto
F.
,
Mori
T.
,
Matsumura
S.
,
Matsumoto
Y.
,
Fukasawa
M.
,
Teshima
M.
et al.
(
2017
)
Prognostic significance of surgical extranodal extension in head and neck squamous cell carcinoma patients
.
Jpn. J. Clin. Oncol.
47
,
699
704
[PubMed]
43.
Lydiatt
W.M.
,
Patel
S.G.
,
O'Sullivan
B.
,
Brandwein
M.S.
,
Ridge
J.A.
,
Migliacci
J.C.
et al.
(
2017
)
Head and neck cancers-major changes in the American Joint Committee on Cancer Eighth Edition Cancer Staging Manual
.
CA Cancer J. Clin.
67
,
122
137
[PubMed]
44.
Melkane
A.E.
,
Mamelle
G.
,
Wycisk
G.
,
Temam
S.
,
Janot
F.
,
Casiraghi
O.
et al.
(
2012
)
Sentinel node biopsy in early oral squamous cell carcinomas: a 10-year experience
.
Laryngoscope
122
,
1782
1788
[PubMed]
45.
Chan
C.H.F.
and
Stanners
C.P.
(
2007
)
Recent advances in the tumour biology of the GPI-anchored carcinoembryonic antigen family members CEACAM5 and CEACAM6
.
Curr. Oncol. (Toronto, Ont.)
14
,
70
73
[PubMed]
46.
Wang
X.-M.
,
Zhang
Z.
,
Pan
L.-H.
,
Cao
X.-C.
and
Xiao
C.
(
2019
)
KRT19 and CEACAM5 mRNA-marked circulated tumor cells indicate unfavorable prognosis of breast cancer patients
.
Breast Cancer Res. Treat.
174
,
375
385
[PubMed]
47.
DeLucia
D.C.
,
Cardillo
T.M.
,
Ang
L.
,
Labrecque
M.P.
,
Zhang
A.
,
Hopkins
J.E.
et al.
(
2021
)
Regulation of CEACAM5 and therapeutic efficacy of an anti-CEACAM5-SN38 antibody-drug conjugate in neuroendocrine prostate cancer
.
Clin. Cancer Res.
27
,
759
774
[PubMed]
48.
Baek
D.-S.
,
Kim
Y.-J.
,
Vergara
S.
,
Conard
A.
,
Adams
C.
,
Calero
G.
et al.
(
2022
)
A highly-specific fully-human antibody and CAR-T cells targeting CD66e/ CEACAM5 are cytotoxic for CD66e-expressing cancer cells in vitro and in vivo
.
Cancer Lett.
525
,
97
107
[PubMed]
49.
Ochoa
I.S.
,
O'Regan
E.
,
Toner
M.
,
Kay
E.
,
Faul
P.
,
O'Keane
C.
et al.
(
2022
)
The role of HPV in determining treatment, survival, and prognosis of head and neck squamous cell carcinoma
.
Cancers
14
,
4321
,
[PubMed]
,
50.
Koo
N.
,
Sharma
A.K.
and
Narayan
S.
(
2022
)
Therapeutics targeting p53-MDM2 interaction to induce cancer cell death
.
Int. J. Mol. Sci.
23
,
5005
,
51.
Cheng
F.
,
Dou
J.
,
Zhang
Y.
,
Wang
X.
,
Wei
H.
,
Zhang
Z.
et al.
(
2021
)
Urolithin A inhibits epithelial-mesenchymal transition in lung cancer cells via P53-MDM2-snail pathway
.
Onco. Targets Ther.
14
,
3199
3208
[PubMed]
52.
Morgan
E.L.
,
Chen
Z.
and
Van Waes
C.
(
2020
)
Regulation of NF kappa B signalling by ubiquitination: a potential therapeutic target in head and neck squamous cell carcinoma?
Cancers
12
,
2877
,
53.
Sun
C.
,
Li
M.
,
Zhang
L.
,
Sun
F.
,
Chen
H.
,
Xu
Y.
et al.
(
2022
)
IDO1 plays a tumor-promoting role via MDM2-mediated suppression of the p53 pathway in diffuse large B-cell lymphoma
.
Cell Death Dis.
13
,
572
,
54.
Chene
P.
(
2003
)
Inhibiting the p53-MDM2 interaction: An important target for cancer therapy
.
Nat. Rev. Cancer
3
,
102
109
[PubMed]
55.
Zhou
G.
,
Zhai
Y.
,
Cui
Y.
,
Zhang
X.
,
Dong
X.
,
Yang
H.
et al.
(
2007
)
MDM2 promoter SNP309 is associated with risk of occurrence and advanced lymph node metastasis of nasopharyngeal carcinoma in Chinese population
.
Clin. Cancer Res.
13
,
2627
2633
[PubMed]
56.
Zhang
F.
,
Xu
Y.
,
Ye
W.
,
Jiang
J.
and
Wu
C.
(
2020
)
Circular RNA S-7 promotes ovarian cancer EMT via sponging miR-641 to up-regulate ZEB1 and MDM2
.
Biosci. Rep.
40
,
BSR20200825
,
57.
Wang
S.-P.
,
Wang
W.-L.
,
Chang
Y.-L.
,
Wu
C.-T.
,
Chao
Y.-C.
,
Kao
S.-H.
et al.
(
2009
)
p53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug
.
Nat. Cell Biol.
11
,
694
704
[PubMed]
58.
Tyagi
A.
(
2022
)
Underlying facets of cancer metastasis
.
Cancers
14
,
2989
,
59.
Stacker
S.A.
,
Williams
S.P.
,
Karnezis
T.
,
Shayan
R.
,
Fox
S.B.
and
Achen
M.G.
(
2014
)
Lymphangiogenesis and lymphatic vessel remodelling in cancer
.
Nat. Rev. Cancer
14
,
159
172
[PubMed]
60.
Sainz-Jaspeado
M.
and
Claesson-Welsh
L.
(
2018
)
Cytokines regulating lymphangiogenesis
.
Curr. Opin. Immunol.
53
,
58
63
[PubMed]
61.
Karaman
S.
and
Detmar
M.
(
2014
)
Mechanisms of lymphatic metastasis
.
J. Clin. Invest.
124
,
922
928
[PubMed]
62.
Liao
Y.
,
Huang
J.
,
Liu
P.
,
Zhang
C.
,
Liu
J.
,
Xia
M.
et al.
(
2022
)
Downregulation of LNMAS orchestrates partial EMT and immune escape from macrophage phagocytosis to promote lymph node metastasis of cervical cancer
.
Oncogene
41
,
1931
1943
[PubMed]
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY-NC-ND).

Supplementary data