Skip to main content
Erschienen in: Cancer Cell International 1/2023

Open Access 01.12.2023 | Research

Prognosis of Midkine and AT1R expression in resectable head and neck squamous cell carcinoma

verfasst von: Tai-Jan Chiu, Chang-Han Chen, Yi-Ju Chen, Yinshen Wee, Ching-Shuen Wang, Sheng‑Dean Luo

Erschienen in: Cancer Cell International | Ausgabe 1/2023

Abstract

Background

Research studies have demonstrated that Midkine (MDK) can influence the expression and activity of Renin-angiotensin system (RAS) components. Angiotensin II is involved in tumor growth and angiogenesis in different cancers. We previously observed Angiotensin II receptor blockers (ARBs) improve the survival rates of patients with oral cancers. These findings have prompted us to investigate whether MDK can influence the RAS pathway, mainly through its association with angiotensin II type 1 receptor (AT1R), which contributes to the observed poor prognosis in head and neck squamous cell carcinoma (HNSCC) patients.

Methods

MDK and AT1R expressions were examined in 150 HNSCC patients post-operation by immunohistochemical staining between 1 January 2010 and 31 December 2016. We tested the over-expression and silencing of MDK to evaluate the AT1R expression and functional biological assays in HNSCC cell lines HSC-3 and SAS.

Results

Positive expression of MDK is correlated with positive AT1R expression. MDK predicted poor NSCC patients’ survival. Silencing MDK could suppress AT1R and pAKT expression and reduce the growth, migration, and invasion of HNSCC cells. ARB also inhibits MDK stimulating HNSCC cell proliferation. Overexpression of MDK could upregulate AT1R and pAKT.

Conclusions

MDK is an independent prognostic factor of HNSCC post-operation, and AT1R regulates HNSCC cell growth, invasion, and migration. Positive MDK and AT1R expressions are highly correlated. Mechanistically, the interaction between MDK and AT1R is crucial for MDK-mediated cell viability, and inhibiting AT1R can effectively counteract or abolish these effects. Furthermore, MDK exerts a regulatory role in the expression of AT1R, as well as in the growth and motility of HNSCC cells. These findings highlight the involvement of the interaction between MDK, AT1R, and the pAkt signaling pathways in HNSCC cell viability growth.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12935-023-03060-z.

Publisher's Note

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

Introduction

More than one million new head and neck cancer patients are diagnosed annually, the seventh most common cancer globally [1]. In Taiwan, head and neck is the sixth most common cancer and the fourth cancer-related death in man. The dominant histological type of head and neck cancer is squamous cell carcinoma. Surgery is the most effective treatment for early and local advanced head and neck squamous cell carcinoma (HNSCC). The prognosis of tumor recurrence and overall survival for HNSCC patients post-operation depends on pathological features, including tumor size, lymph node status, AJCC tumor stage, and extra-nodal extension. Besides, patient-related factors were also correlated with the prognosis of HNSCC patients, such as age, sex, smoking, alcohol, betel nuts, and performance status. Despite progress in diagnosis and treatments, the outcome of HNSCC remains unsatisfactory, even in patients who received complete tumor resection. Identifying new treatment targets is important in HNSCC patients with poor prognostic factors.
The renin-angiotensin system (RAS) is essential in blood pressure control and electrolyte balance. Angiotensin I-converting enzyme inhibitors (ACEIs) and angiotensin II type 1 receptor blockers (ARBs) are RAS antagonists to inhibit the effect of angiotensin II. ACEI and ARBs are the most common in the treatment of chronic hypertension and congestive heart failure [2]. Growing evidence demonstrated that RAS promoted cell proliferation and neovascularization by angiotensin II signaling stimulation of vascular endothelial growth factor (VEGF)-mediated angiogenesis in malignancy [3]. The previous study showed that ACEI and ARBs might inhibit tumor development and progression [4] and a promising anti-tumor strategy. Our previous study also found that advanced HNSCC patients who received ARBs for more than 180 days could improve their overall survival after tumor resection [5]. The mechanisms of RAS inhibitors in patients with HNSCC remain unclear. Angiotensin II, a peptide hormone, has biological effectors in RAS. AT1R and AT2R are two types of angiotensin II play different roles in cardiovascular functions. The RAS was observed to activate angiotensin II and upregulate AT1R expression in some cancers. There is only few studies to discuss about AT1R in HNSCC.
Midkine (MDK), a retinoic acid-inducible heparin-binding growth factor, is a useful biomarker to predict HNSCC survival after surgery in our previous study [6]. MDK expression was upregulated in tumor tissue and was associated with lower recurrence-free and overall survival (OS) rates in this study. MDK plays a role in the multiple biological functions of cancer, such as promoting tumor cell proliferation, transformation, and epithelial-to-mesenchymal (EMT) transition [79]. One study also showed that MDK could regulate RAS in mice models [10]. The literature review shows the limited relationship between MDK and RAS in HNSCC. The purpose of this study will identify whether angiotensin receptors (AT1R) regulated head and neck cancer cell proliferation and metastases by MDK expression.

Material and methods

Patient population

This retrospective study enrolled 150 HNSCC patients who received tumor resection between 1 January 2010 and 31 December 2016 at Kaohsiung Chang Gung Memorial Hospital Medical Center in Taiwan. Patients with synchronous cancers or receiving preoperative chemotherapy, radiotherapy, or concurrent chemoradiotherapy (CCRT) were excluded. The pathological paraffin blocks and medical information of HNSCC patients were from the Kaohsiung Chang Gung Memorial Hospital biobank. The pathological TNM stage was according to the 7th American Joint Committee on Cancer (AJCC) staging system. Overall survival (OS) was counted from surgery to death due to all causes. Disease-free survival (DFS) was computed from the time of surgery to the recurrence or death of any reason without evidence of recurrence. The study was performed under the Declaration of Helsinki and was approved by the Human Research Ethics Committee of Chang Gung Memorial Hospital.

Immunohistochemical study

A pathologist reviewed the tissue sample from our hospital’s biobank to confirm the histologic type of squamous cell carcinoma. Immunohistochemistry was used to evaluate the levels of MDK and AT1R proteins from 150 HNSCC patients. The protocol of immunohistochemistry for MDK (Abcam Plc, Cambridge, UK) was according to our previous studies [6]. Immunohistochemistry staining for AT1R (A14201, 1:100, ABclonal, USA) was done using an immunoperoxidase technique as in Li et al. study [11]. Staining was performed on slides of formalin-fixed, paraffin-embedded tissue sections with primary antibodies against AT1R. Antibody assay without the primary antibody was used as the negative control. Two pathologists independently evaluated immunohistochemical staining for MDK and AT1R blinded to the clinical information. The scores of the expression of MDK and AT1R followed the previously published methods [1214]. Pathologists scored MDK in each specimen from 1 to 4 according to the percentage of positive cells: 1 for ≤ 5% of the cells, 2 for 6—35% of the cells, 3 for 36—70% of the cells, and 4 for ≧ 71% of the cells. In addition, we also assigned each specimen another score from 1 to 4 based on staining intensity: 1 for negative staining, 2 for weak staining, 3 for moderate staining, and 4 for intense staining. We calculated the MDK expression by multiplying the percentage and intensity scores. The strong MDK protein expression indicated a score of ≥ 4; otherwise, a score of < 4 was weak. The strong expression of AT1R was defined as at least staining≧35% of tumor cells, and < 35% was weak expression.

Western blotting

Cells of HNSCC cell lines were collected and lysed with RIPA buffer (Thermo SCIENTIFIC, Rockford, USA), protease inhibitor cocktail set III (MedChemExpress, HY-K0010), and phosphatase inhibitor cocktail (MedChemExpress, HY-K0021) on ice for 30 min. The clear lysate was harvested by centrifugation at 13,000 rpm for 30 min at 4 °C. The total protein concentration was measured, and equal amounts of protein were separated by SDS-PAGE and then transferred onto PVDF membranes. For blocking, membranes were incubated with 1% PBST containing with 5% non-fat milk for 60 min. Primary antibodies to detect MDK (Abcam Plc, Cambridge, UK), AT1R (ABclonal, USA), pAKT (cell signaling#4060), AKT (cell signaling#9272), and β-actin (Sigma #A5441) were added to the membranes and were then incubated overnight at 4 °C. Horseradish peroxidase-conjugated anti-mouse or anti-rabbit IgG secondary antibodies were added to the membranes and left for 1 h at room temperature the next day. X-ray films explored the proteins.

Cell lines, cell culture, and transfection

Human HNSCC cell lines CAL27 (RRID: CVCL_1107), SAS (RRID: CVCL_1675) and HSC-3 (RRID: CVCL_1228) were obtained from ATCC (American Type Culture Collection) and cultured in DMEM (Life Technologies, Inc., Carlsbad, USA) supplemented with 10% fetal bovine serum (FBS) (Life Technologies, Inc., Carlsbad, USA), 100 U/ml penicillin and 100 μg/ml streptomycin, 1% non-essential amino acid and 1% sodium pyruvate (Life Technologies, Inc., Carlsbad, USA). In a humidified atmosphere, we cultured cells at 37 °C, 5% CO2. Transfections of cells were carried out using Lipofectamine™ 3000 Transfection Reagent (Invitrogen) according to the manufacturer’s instructions. Cells were harvested after 24 h transfection for subsequent treatments. The human MDK-mediated shRNA sequences were: Oligo Sequence 1 CCGGCAAGACCAAAGCAAAGGCCAACTCGAGTTGGCCTTTGCTTTGGTCTTGTTTTTG; Oligo Sequence 2 CCGGCGACTGCAAGTACAAGTTTGACTCGAGTCAAACTTGTACTTGCAGTCGTTTTTG.

MTT assay

HNSCC cells were plated in a 96-well plate (3000 cells/well) and treated with or without irbesartan (IRB) at indicated dosages for 72 h. After treatment, 100 μl of 0.5 mg/ml MTT was added to each well and incubated at 37 °C for three hours. The medium was then removed, and 100 μl DMSO was added to each well to lyse cells. Plates were measured at 595 nm using Umax Kinetic Microplate Reader (Molecular Devices, Celifomie, USA). We purchased human MDK and IRB from Sigma (Sigma Aldrich, St Louis, MO, USA).

Migration and invasion assay

Transwell inserts (pore size: 8 μm) coated with or without Matrigel (BD Biosciences) were used to evaluate the migratory and invasive abilities of CAL27, SAS and HSC-3 cells. For migration assays, 1 × 104 cells in 100 μl of serum-free medium in the upper chamber and added 500 μl of medium in the lower chamber. For invasion assays, inserts coated 5% matrigel in PBS. 2 × 104 cells were added in 100 μl of serum-free medium in the upper chamber and 500 μl of DMEM medium in the lower chamber. After the cells were incubated for 20 h, they were fixed and stained with crystal violet for 15 min. The numbers of migratory and invasive cells were counted in five fields under a microscope. All groups of experiments were conducted in triplicate.

Statistical analysis

The SPSS 19 software was used to analyze the HNSCC patients’ data. To compare data between the two groups, we performed the Chi-square test and Fisher’s exact test. The Kaplan–Meier method was used for univariate analysis of DFS and OS, and a log-rank method tested the difference between survival curves. The significant parameters at the univariate level were assigned to the Cox regression model to analyze their relative prognostic importance. For HNSCC cell line experiments, a t-test was used for the statistical analysis. Every study was carried out independently at least twice, with three repeats each.

Results

Immunohistochemical expression of MDK and AT1R and its correlations with other clinicopathologic parameters in resectable HNSCC

The clinicopathologic factors of the 150 patients with HNSCC post-operation showed in Table 1. The median age of these patients was 51 years old (ranging from 29 to 75 years). One hundred and forty-two patients were male, and eight patients were female. The most common primary tumor site was oral (98 patients, 65.3%), followed by hypopharynx/larynx (28 patients, 18.7%) and oropharynx (24 patients, 16%). Before surgery, 55 patients (36.7%) were ECOG performance status 0, 68 patients (45.3%) were 1, and 27 patients (18%) were 2. The pathological AJCC tumor stage 1 accounted for eight patients (5.3%), stage II for 25 patients (16.7%), stage III for 26 patients (17.3%), and stage IVA-B for 91 patients (60.7%). The P16 positive rate was 12.7% (19 patients), and the extra-nodal extensions (ENE) rate was 30.7% (46 patients). Fifty-four patients (36%) had lymphovascular invasion (LVI), and 69 patients (46%) had perineural invasion (PNI). One hundred and twenty HNSCC patients in this study had a habit of cigarette smoking, 106 patients had alcohol consumption, and 99 patients had betel nuts chewing. Of these 150 HNSCC patients, 69 had medical records of diabetes mellitus, 50 had viral hepatitis, and 81 had hypertension.
Table 1
Clinical characteristics of HNSCC in this study
 
Patient Numbers
Percentage (%)
Gender
 Male
142
94.7
 Female
8
5.3
Age (Median: 51 ± 9.9 years old)
 ≦51
76
50.7
  > 51
74
49.3
Performance status (ECOG)
 0
55
36.7
 1
68
45.3
 2
27
18.0
Tumor site
 Oral
98
65.3
 Oropharynx
24
16.0
 Hypopharynx/larynx
28
18.7
Tumor stage
 T1
10
6.7
 T2
48
32.0
 T3
30
20.0
 T4
62
41.3
Lymph nodes
 N0
63
42.0
 N1
26
17.3
 N2
56
37.3
 N3
5
3.3
AJCCTMN stage
 I
8
5.3
 II
25
16.7
 III
26
17.3
 IV
91
60.7
P16
 Positive
19
12.7
 Negative
131
87.3
Extra-nodal extension
 Positive
46
30.7
 Negative
104
69.3
Viral hepatitis
 No
100
66.6
 Yes
50
33.3
Diabetes mellitus
 No
94
62.7
 Yes
56
37.3
Hypertension
 No
69
46.0
 Yes
81
54.0
Lyphovascular invasion
  
 No
96
64.0
 Yes
54
36.0
Perineural invasion
 No
81
54.0
 Yes
69
46.0
Adjuvant treatment
 No
38
25.3
 RT
37
24.7
 CCRT
75
50.0
Smoking
 No
30
20.0
 Yes
120
80.0
Alcohol
  
 No
44
29.3
 Yes
106
70.7
Betel nuts
 No
51
34.0
 Yes
99
66.0
Midkine
 Weak
88
58.7
 Strong
62
41.3
AT1R
 Weak
92
61.3
 Strong
58
38.7
Table 2 describes the correlation between the clinicopathological factors with immunohistochemical expression of AT1R and MDK (Fig. 1A). Strong AT1R expression was significantly associated with AJCC tumor stage (stage IVA/B, p = 0.001), hypertension (p = 0.004), and strong MDK expression (p < 0.001). Strong MDK expression was also associated with AJCC tumor stage (p = 0.004), lymph node metastases (p < 0.001), ENE (p < 0.001), and AT1R expression (p < 0.001).
Table 2
Correlation between expression of AT1R and MDK and clinicopathological factors of HNSCC
 
No. of patients
AT1R
P
MDK
P
Weak expression
Strong expression
Weak expression
Strong expression
Age
   
0.226
  
0.599
 ≦51
76
43 (56.6%)
33 (43.4%)
 
43 (56.6%)
33 (43.4%)
 
  > 51
74
49 (66.2%)
25 (33.8%)
 
45 (60.8%)
29 (39.2%)
 
Gender
   
0.944
  
0.141
 Male
142
87 (61.3%)
55 (38.7%)
 
81 (57.0%)
61 (43.0%)
 
 Female
8
5 (62.5%)
3 (37.5%)
 
7 (87.5%)
1 (12.5%)
 
ECOG PS
   
0.087
  
0.072
 0
55
40 (72.7%)
15 (27.3%)
 
37 (67.3%)
18 (32.7%)
 
 1
68
38 (55.9%)
30 (44.1%)
 
40 (58.8%)
28 (41.2%)
 
 2
27
14 (51.9%)
13 (48.1%)
 
11 (40.7%)
16 (59.3%)
 
Tumor site
   
0.182
  
0.249
 Oral cavity
98
64 (65.3%)
34 (37.4%)
 
62 (63.3%)
36 (36.7%)
 
 Oropharynx
24
15 (62.5%)
9 (37.5%)
 
13 (54.2%)
11 (45.8%)
 
 Hypopharynx/larynx
28
13 (46.4%)
15 (53.6%)
 
13 (46.4%)
15 (53.6%)
 
AJCC tumor stage
   
0.001**
  
0.004**
 I–III
59
45 (76.3%)
14 (23.7%)
 
43 (72.9%)
16 (27.1%)
 
 IVA–B
91
47 (51.6%)
44 (48.4%)
 
45 (49.5%)
46 (50.5%)
 
T stage
   
0.303
  
0.424
 1–3
88
57(64.8%)
31(35.2%)
 
54 (61.4%)
34 (38.6%)
 
 4A–B
62
35 (56.5%)
27 (43.5%)
 
34 (54.8%)
28 (45.2%)
 
N stage
   
0.423
  
 < 0.001**
 Negative
63
41(65.1%)
22(34.9%)
 
50 (79.4%)
13 (20.6%)
 
 Positive
87
51 (58,6%)
36 (41.4%)
 
38 (43.7%)
49 (56.3%)
 
P16 expression
   
0.405
  
0.324
 Negative
131
82 (62.6%)
49 (37.4%)
 
79 (60.3%)
52 (39.7%)
 
 Positive
19
10 (52.6%)
9 (47.4%)
 
9 (47.4%)
10 (52.6%)
 
ENE
   
0.001**
  
 < 0.001**
 Negative
104
73 (70.2%)
31 (29.8%)
 
75 (72.1%)
29 (27.9%)
 
 Positive
46
19 (41.3%)
27 (58.7%)
 
13 (28.3%)
33 (71.7%)
 
LVI 0.759
  
0.814
 Negative
96
58 (60.4%)
38 (39.6%)
 
57 (59.4%)
39 (40.6%)
 
 Positive
54
34 (63.0%)
20 (37.0%)
 
31 (57.4%)
23 (42.6%)
 
PNI 0.435
  
0.622
 Negative
81
52 (64.2%)
29 (35.8%)
 
49 (60.5%)
32 (39.5%)
 
 Positive
69
40 (58.0%)
29 (42.0%)
 
39 (56.5%)
30 (43.5%)
 
Alcohol drinking
   
0.065
  
0.127
 No
44
32 (72.7%)
12 (27.3%)
 
30 (68.2%)
14 (31.8%)
 
 Yes
106
60 (56.6%)
46 (43.4%)
 
58 (54.7%)
48 (45.3%)
 
Smoking
   
0.276
  
0.562
 No
30
21 (70.0%)
9 (30.0%)
 
19 (63.3%)
11 (36.7%)
 
 Yes
120
71(59.2%)
49 (40.8%)
 
69 (57.5%)
51(42.5%)
 
Betel nuts
   
0.651
  
0.075
 No
51
30 (58.8%)
21 (41.2%)
 
35 (68.6%)
16 (31.4%)
 
 Yes
99
62 (62.6%)
37 (37.4%)
 
53 (53.5%)
46 (46.5%)
 
DM
   
0.132
  
0.525
 Negative
94
62 (66.0%)
32 (34.0%)
 
57 (60.6%)
37 (39.4%)
 
 Positive
56
30 (53.6%)
26 (46.4%)
 
31 (55.4%)
25 (44.6%)
 
Hypertension
   
0.004**
  
0.242
 No
69
52 (75.4%)
17 (24.6%)
 
44 (63.8%)
25 (36.2%)
 
 Yes
81
40 (49.4%)
41 (50.6%)
 
44 (54.3%)
37 (45.7%)
 
Viral hepatitis
   
0.236
  
0.348
 No
100
58 (58.0%)
42 (42.0%)
 
56 (56.0%)
44 (44.0%)
 
 Yes
50
34 (68.0%)
16 (32.0%)
 
32 (64.0%)
18 (36.0%)
 
MDK
   
 < 0.001**
   
 Weak
88
66 (75.0%)
22 (25.0%)
    
 Strong
62
26 (41.9%)
36 (58.1%)
    
AT1R
      
 < 0.001**
 Weak
92
   
67 (72.8%)
25 (27.2%)
 
 Strong
58
   
21 (36.2%)
37 (63.8%)
 
Bold indicates statistically significant results

Survival analysis of HNSCC after tumor resection

The median follow-up duration of this study was 44 months (ranging from 2 to 105 months). After tumor resection, 78 patients experienced tumor recurrence, and 86 died during this period. The median DFS time was 26 months, and the 2-years DFS rate was 56.2%. The median OS duration was 46 months, and the 5-year OS rate was 43.6%. The univariate analyses demonstrated that ECOG performance status 1 or 2 (p < 0.001), T stage 4A/4B (p = 0.001), lymph node metastases (p = 0.005), AJCC tumor stage IVA/IVB (p < 0.001), positive LVI (p = 0.005, positive PNI (p < 0.001), positive extra-nodal extension (p < 0.001), the habitus of betel nuts chewing (p = 0.006), strong MDK (p = 0.001, Fig. 1B), and strong AT1R expression (p = 0.009, Fig. 1B) had poor shorter DFS (Table 3). In addition, our study also indicated that ECOG PS 0/1 (p < 0.001), T stage 4A/4B (p = 0.016), lymph nodes metastases (p = 0.003), AJCC tumor stage IVA/B (p < 0.001), positive LVI (p = 0.001), positive PNI (p < 0.001), ENE (p < 0.001), P16 positive (p = 0.015), cigarette smoking (p = 0.029), betel nuts chewing (p = 0.006), viral hepatitis (p = 0.017), strong AT1R(p = 0.001, Fig. 1E), and strong MDK expression (p < 0.001, Fig. 1) were associated with inferior OS.
Table 3
Correlation between the clinicopathological features and Disease-Free survival in HNSCC
Variable
Category
Univariate analysis
Multivariate analysis
HR
95% CI
p
Adjusted HR
95% CI
p
Age
≦51 vs > 51
0.932
0.59–1.43
0.724
0.742
0.46–1.21
0.228
Gender
Female vs male
0.814
0.30–2.23
0.689
0.871
0.28–2.69
0.810
Tumor site
Oral
Reference
Reference
 
Oropharynx
0.609
0.30–1.23
0.168
0.715
0.33–1.53
0.387
 
Hypopharynx/larynx
0.931
0.53–1.65
0.806
0.477
0.28–0.98
0.046
ECOG PS
0
Reference
Reference
 
1
4.129
2.23–7.63
 < 0.001
3.07
1.50–6.26
0.002
 
2
18.296
8.88–37.70
 < 0.001
9.478
3.90–23.06
0.001
T stage
1–3 vs 4A–B
0.478
0.25–0.71
0.001
   
N stage
Negative vs positive
0.515
0.32–0.83
0.005
   
AJCC stage
I–III vs IVA–B
0.404
0.25–0.67
 < 0.001
0.917
0.51–1.64
0.770
LVI
Negative vs positive
0.521
0.33–0.82
0.005
0.703
0.42–1.17
0.177
PNI
Negative vs positive
0.326
0.21–0.52
 < 0.001
0.507
0.29–0.88
0.017
ENE
Negative vs Positive
0.221
0.14–0.35
 < 0.001
0.298
0.15–0.58
0.001
P16 expression
Negative vs positive
1.725
0.83–3.59
0.145
1.385
0.61–3.16
0.438
Alcohol drinking
No vs yes
1.050
0.65–1.70
0.844
0.890
0.52–1.54
0.676
Smoking
No vs yes
0.742
0.42–1.32
0.313
1.222
0.67–2.45
0.519
Betel nuts
No vs yes
0.426
0.25–0.72
0.002
0.555
0.32–0.98
0.041
DM
No vs yes
1.005
0.63–1.40
0.983
1.228
0.73–2.07
0.439
Hypertension
No vs yes
1.047
0.67–1.63
0.841
1.227
0.74–2.03
0.425
Viral hepatitis
No vs yes
0.838
0.53–1.34
0.458
1.310
0.74–2.33
0.358
MDK
Weak vs strong
0.446
0.29–0.70
0.001
0.312
0.15–0.66
0.038
AT1R
Weak vs strong
0.552
0.35–0.86
0.009
0.755
0.44–1.29
0.303
Bold indicates statistically significant results
In a multivariate comparison, MDK expression remained independently associated with DFS, together with ECOG PS, PNI, ENE, and betel nuts chewing (Table 3). For OS, multivariate Cox regression analysis also showed that MDK, ECOG PS, LVI, and ENE were independent prognostic factors (Table 4).
Table 4
Overall survival associated with clinicopathological factors in post-operation HNSCC patients
Variable
Category
Univariate analysis
Multivariate analysis
HR
95% CI
p
Adjusted HR
95% CI
p
Age
≦51 vs > 51
1.008
0.66–1.54
0.970
0.683
0.43–1.10
0.113
Gender
Female vs male
0.305
0.08–1.24
0.097
0.510
0.12–2.25
0.373
Tumor site
Oral
Reference
Reference
 
Oropharynx
1.023
0.59–1.78
0.935
0.648
0.31–1.36
0.253
 
Hypopharynx/larynx
0.610
0.28–1.35
0.221
0.477
0.24–0.95
0.034
ECOG PS
0
Reference
Reference
 
1
4.355
2.42–7.83
 < 0.001
2.81
1.42–5.60
0.003
 
2
17.229
8.73–34.00
 < 0.001
7.662
3.16–18.58
 < 0.001
T stage
0–3 vs 4A–B
0.594
0.39–0.91
0.016
   
N stage
Negative vs positive
0.507
0.32–0.80
0.003
   
AJCC stage
I–III vs IVA–B
0.411
0.26–0.66
 < 0.001
1.035
0.59–1.83
0.905
LVI
Negative vs positive
0.469
0.31–0.72
0.001
0.449
0.28–0.72
0.001
PNI
Negative vs positive
0.279
0.18–0.44
 < 0.001
0.608
0.35–1.06
0.077
ENE
Negative vs positive
0.179
0.11–0.0.28
 < 0.001
0.241
0.13–0.46
 < 0.001
P16 expression
Negaive vs positive
2.814
1.23–6.46
0.015
2.452
0.98–6.13
0.055
Alcohol drinking
No vs yes
1.031
0.65–1.64
0.896
0.850
0.51–1.42
0.535
Smoking
No vs yes
0.494
0.26–0.93
0.029
0.862
0.44–1.68
0.519
Betel nuts
No vs yes
0.506
0.31–0.82
0.006
0.701
0.41–1.19
0.188
DM
No vs yes
0.754
0.49–1.16
0.196
1.093
0.68–1.75
0.712
Hypertension
No vs yes
0.794
0.52–1.22
0.289
0.736
0.45–1.20
0.216
Viral hepatitis
No vs yes
0.590
0.38–0.91
0.017
0.711
0.40–1.26
0.240
MDK
Weak vs strong
0.433
0.28–0.66
 < 0.001
0.475
0.28–0.82
0.007
AT1R
Weak vs strong
0.472
0.31–0.72
0.001
0.597
0.35–1.03
0.065
Bold indicates statistically significant results

Irbesartan (IRB) inhibited HNSCC cell proliferation

Irbesartan (IRB), an angiotensin II inhibitor and oral selective AT1R blocker, extensively treats high blood pressure. We initially surveyed the protein expressions of the MDK and AT1R by Western blotting in 5 HNSCC cell lines and confirmed both proteins were expressed in these cell lines, including HSC-3 and SAS (Fig. 2A). In SAS and HSC-3 cell lines, MDK could stimulate cell proliferation (Fig. 2B), and IRB could suppress HNSCC cells (Fig. 2C). In addition, IRB also inhibited the MDK-dependent growth of HNSCC cells (Fig. 2D).

MDK regulated AT1R expression and HNSCC cell growth and motility

Transfected with MDK shRNA or shControl in CAL27 and HSC-3 were analyzed by Western blotting (Fig. 3A). After being transfected with shMDK, the AT1R expression was significantly reduced (Fig. 3A). The MTT tests showed that depleted MDK significantly suppressed the cell proliferation of CAL27 and HSC-3 when compared with the shControls (Fig. 3B). We also performed invasion and migration assays in MDK-suppressed CAL27 and HSC-3 cells. As shown in Fig. 3C, the knock-down of MDK significantly inhibited cell migration and invasion compared to control.
Overexpressed MDK in the HNSCC cell lines, CAL27 and SAS, promoted AT1R expressions (Fig. 4A). The MTT test also showed increased HNSCC cell lines proliferation while MDK overexpressed (Fig. 4B), and the migration and invasion increased in Transwell assays (Fig. 4C).

MDK also affected pAkt expression in human HNSCC cells

As shown in Fig. 5A, pAKT expression was suppressed while knocked down MDK in CAL27 and HSC-3 cells. In contrast, pAKT expression was increased while MDK was overexpressed in HNSCC cell lines SAS and CAL27 (Fig. 5B).

Discussion

Our study showed a potentially new mechanism to improve survival in resectable HNSCC patients. AT1R and MDK expression was significantly correlated in HNSCC patient tissue samples. The positive expression of MDK and AT1R in HNSCC patients predicted poor DFS and OS. Silencing MDK in HNSCC cells decreased their proliferation, invasion, and migration. Inhibition of MDK also suppressed AT1R and p-AKT expression in our HNSCC cell lines. IRB could suppress the MDK-stimulating HNSCC cell growth. These findings suggested that AT1R could be targeted in the MDK-positive HNSCC.
Complete tumor resection is the mainstay of curative treatment of early and localized advanced HNSCC. Tumor recurrence often impacts patients’ survival due to lower remission and higher mortality. Adjuvant radiotherapy or concurrent chemoradiotherapy after operation in the pathological high-risk group is the standard management. High locoregional failure and poor disease-free duration suggested lacking effective therapy and even received maintenance treatments [1517]. Several studies have demonstrated that MDK is an effective biomarker for predicting the outcomes of HNSCC patients [18]. Our previous study also found that MDK expression was associated with lower disease-free and OS rates after surgery. Our current study also consistently results with positive MDK expression in HNSCC patients with prompt tumor recurrence. From the literature review, MDK seems a drugable target in different cancers [19], including oral squamous cell carcinoma [20]. However, no medication was available to block MDK expression directly in cancer patients. Our study provided inhibition of AT1R by IRB may reduce tumor progression in MDK-expressed HNSCC patients.
Growth factors promote cancer cell proliferation, invasion, and migration. MDK activates the AKT pathway to promote GBM and oral squamous cell carcinoma progression [20, 21]. In the current study, MDK expression was associated with advanced tumor stage, lymph node metastases, and extra-nodal extension. These findings demonstrated our previous study results and predict HNSCC patients’ poor prognoses. MDK is a secreted protein, and the concentration of MDK increased significantly in the MDK overexpressed HNSCC medium (Additional file 1: Fig. S1). However, the secreted MDK-affected HNSCC cell function mechanism needs more well-designed research. Currently, a novel finding in our study was that MDK and AT1R expression was highly correlated. All uncropped western blotting membranes were showed in Additional file 2.
Limited research explored the relationship between MDK and RAS in cancers. Akinori et al. showed that MDK protein enhanced ACE expression in mice with chronic kidney disease [10]. Their study showed that nephrectomy-induced MDK expression increases ACE activity and plasma angiotensin II levels. To our best knowledge, there is no literature to discuss how MDK affects AT1R expression in cancer. Our current study showed that MDK and AT1R expression was highly correlated in HNSCC patients, and MDK could regulate AT1R expression in HNSCC cells. However, there was no interaction between MDK and AT1R in HNSCC cells by Co-IP approach (data not shown), indicating that MDK might modulate the AT1R protein stability via other pathways, such as proteasome pathway or ubiquitin pathway. Further experiments will be designed and performed in the future. The AT1R and pAKT expressions were also down-regulated while shMDK was transfected into HNSCC cell lines. In contrast, the AT1R and pAKT upregulated while MDK was overexpressed.
Increasing evidence show that AT1R is involved in tumor growth, metastases, and angiogenesis in different animal models [22]. ACE synthesizes angiotensin II and stimulates tumor cell growth through AT1R. Selective AT1R blockade might be more effective than ACE inhibition [23]. Although recent research illustrated the activation of RAS and upregulation of AT1R in different tumor tissues [24, 25], there were no reports to analyze the AT1R expression in HNSCC. Our current study also showed AT1R was associated with advanced tumor stage, hypertension, MDK expression, and worse survival in HNSCC patients. IRB could inhibit HNSCC cell growth by suppressing AT1R under MDK stimulation. AT1R may play an important role in MDK enhancing HNSCC cell proliferation. This result could explain oral squamous cell carcinoma patients who received ARB improved overall survival in our retrospective study. Lin et al. also showed ARB had effects of anti-proliferation and anti-angiogenesis in nasopharyngeal cancer patients [26].
One study in breast cancer also found AT1R increases cell migration through the AKT pathway [27]. Recently, Zhang et al. reported that suppression of AT1R expression inhibited lung cancer cell proliferation and migration by regulating the AKT pathway [28]. It has been observed that either AT1R or MDK can activate the AKT pathway. However, it has not been shown whether MDK interacts with AT1R to impact the AKT signaling pathway involved in driving HNSCC cell viability, growth, and motility. In our study, we found that knockdown of MDK resulted in a reduction in the expression of both AT1R and pAkt. Furthermore, we also showed that the activity or function of MDK in promoting cell viability is dependent on the presence or activation of AT1R. These findings suggest that MDK modulates the RAS pathway through AT1R. In sum, these findings highlight the potential interaction between MDK, AT1R, and the pAkt signaling pathway, which appears to be involved in HNSCC cell viability, growth, and motility. The current study’s limitations included a retrospective study to enroll post-operative HNSCC patients. First, we wanted to evaluate the expressions of MDK and AT1R to affect DFS and OS in HNSCC patients post-operation. However, in HNSCC, the second primary tumor in a different location and repeated tumor resection may affect DFS and OS. Besides, most of our HNSCC patients were male (94.7%) and came from the oral cavity. In Taiwan, smoking, alcohol, and betel nuts are the essential risk factors for HNSCC patients, and most are male. Second, our study found that IRB could inhibit HNSCC cell proliferation, even in MDK stimulation. This finding needs carcinogen-induced HNSCC mouse models or xenograft models to help verification in the future. Third, our study found that MDK influences AT1R expression and affects proliferation, migration, and invasion in HNSCC cells. However, the mechanism of MDK regulating AT1R to control HNSCC cell functions is unclear.

Conclusion

Our study showed MDK and AT1R were important prognostic factors in resectable HNSCC patients. MDK and AT1R were highly correlated, and MDK affected AT1R and pAKT expressions in HNSCC. Suppression of AT1R by IRB decreased HNSCC cell proliferation even under MDK stimulation. Overall, these findings underscore the importance of the interplay between MDK, AT1R, and the pAkt signaling pathways in driving HNSCC cell viability, growth, and motility. More importantly, the blocking the AT1R pathway, possibly in combination with targeting MDK, could be a promising approach for the treatment of HNSCC.

Acknowledgements

We would like to thank The National Biobank Consortium of Taiwan and its cooperative institutions for providing the biological specimen and related clinical data (all are de-identified) for our research. “National Biobank Consortium of Taiwan” is supported by grants from Ministry of Health and Welfare and National Health Research Institutes, Taiwan.

Declarations

This cohort study was approved by the Institutional Review Board (IRB) of the Kaohsiung branches of Chang Gung Memorial Hospital with reference numbers 202101021B0. Patient consent was waived due to the study design and IRB regulations. Restrictions apply to the availability of these data. Data were obtained from Chang Gung Biobank and are available with the permission of the Institutional Review Board (IRB) of the Kaohsiung branches of Chang Gung Memorial Hospital.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Mody MD, Rocco JW, Yom SS, Haddad RI, Saba NF. Head and neck cancer. Lancet. 2021;398(10318):2289–99.CrossRefPubMed Mody MD, Rocco JW, Yom SS, Haddad RI, Saba NF. Head and neck cancer. Lancet. 2021;398(10318):2289–99.CrossRefPubMed
2.
Zurück zum Zitat Afsar B, Afsar RE, Ertuglu LA, Kuwabara M, Ortiz A, Covic A, Kanbay M. Renin-angiotensin system and cancer: epidemiology, cell signaling, genetics and epigenetics. Clin Transl Oncol. 2021;23(4):682–96.CrossRefPubMed Afsar B, Afsar RE, Ertuglu LA, Kuwabara M, Ortiz A, Covic A, Kanbay M. Renin-angiotensin system and cancer: epidemiology, cell signaling, genetics and epigenetics. Clin Transl Oncol. 2021;23(4):682–96.CrossRefPubMed
3.
Zurück zum Zitat Wegman-Ostrosky T, Soto-Reyes E, Vidal-Millan S, Sanchez-Corona J. The renin-angiotensin system meets the hallmarks of cancer. J Renin Angiotensin Aldosterone Syst. 2015;16(2):227–33.CrossRefPubMed Wegman-Ostrosky T, Soto-Reyes E, Vidal-Millan S, Sanchez-Corona J. The renin-angiotensin system meets the hallmarks of cancer. J Renin Angiotensin Aldosterone Syst. 2015;16(2):227–33.CrossRefPubMed
4.
Zurück zum Zitat Rosenthal T, Gavras I. Angiotensin inhibition and malignancies: a review. J Hum Hypertens. 2009;23(10):623–35.CrossRefPubMed Rosenthal T, Gavras I. Angiotensin inhibition and malignancies: a review. J Hum Hypertens. 2009;23(10):623–35.CrossRefPubMed
5.
Zurück zum Zitat Wu CN, Wu SC, Chen WC, Yang YH, Chin JC, Chien CY, Fang FM, Li SH, Luo SD, Chiu TJ. Angiotensin II receptor blockers and oral squamous cell carcinoma survival: a propensity-score-matched cohort study. PLoS ONE. 2021;16(12): e0260772.CrossRefPubMedPubMedCentral Wu CN, Wu SC, Chen WC, Yang YH, Chin JC, Chien CY, Fang FM, Li SH, Luo SD, Chiu TJ. Angiotensin II receptor blockers and oral squamous cell carcinoma survival: a propensity-score-matched cohort study. PLoS ONE. 2021;16(12): e0260772.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Chiu TJ, Chen YJ, Rau KM, Chen CH, Chien CY, Li SH, Tsai HT, Eng HL. Midkine neurite growth-promoting factor 2 expression as a potential prognostic marker of adjuvant therapy in head and neck squamous cell carcinoma. Biomarkers. 2013;18(8):687–98.CrossRefPubMed Chiu TJ, Chen YJ, Rau KM, Chen CH, Chien CY, Li SH, Tsai HT, Eng HL. Midkine neurite growth-promoting factor 2 expression as a potential prognostic marker of adjuvant therapy in head and neck squamous cell carcinoma. Biomarkers. 2013;18(8):687–98.CrossRefPubMed
7.
Zurück zum Zitat Erguven M, Bilir A, Yazihan N, Ermis E, Sabanci A, Aktas E, Aras Y, Alpman V. Decreased therapeutic effects of noscapine combined with imatinib mesylate on human glioblastoma in vitro and the effect of midkine. Cancer Cell Int. 2011;11(1):18.CrossRefPubMedPubMedCentral Erguven M, Bilir A, Yazihan N, Ermis E, Sabanci A, Aktas E, Aras Y, Alpman V. Decreased therapeutic effects of noscapine combined with imatinib mesylate on human glioblastoma in vitro and the effect of midkine. Cancer Cell Int. 2011;11(1):18.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Kadomatsu K, Kishida S, Tsubota S. The heparin-binding growth factor midkine: the biological activities and candidate receptors. J Biochem. 2013;153(6):511–21.CrossRefPubMed Kadomatsu K, Kishida S, Tsubota S. The heparin-binding growth factor midkine: the biological activities and candidate receptors. J Biochem. 2013;153(6):511–21.CrossRefPubMed
10.
Zurück zum Zitat Hobo A, Yuzawa Y, Kosugi T, Kato N, Asai N, Sato W, Maruyama S, Ito Y, Kobori H, Ikematsu S, et al. The growth factor midkine regulates the renin-angiotensin system in mice. J Clin Invest. 2009;119(6):1616–25.CrossRefPubMedPubMedCentral Hobo A, Yuzawa Y, Kosugi T, Kato N, Asai N, Sato W, Maruyama S, Ito Y, Kobori H, Ikematsu S, et al. The growth factor midkine regulates the renin-angiotensin system in mice. J Clin Invest. 2009;119(6):1616–25.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Li SH, Lu HI, Chang AY, Huang WT, Lin WC, Lee CC, Tien WY, Lan YC, Tsai HT, Chen CH. Angiotensin II type I receptor (AT1R) is an independent prognosticator of esophageal squamous cell carcinoma and promotes cells proliferation via mTOR activation. Oncotarget. 2016;7(41):67150–65.CrossRefPubMedPubMedCentral Li SH, Lu HI, Chang AY, Huang WT, Lin WC, Lee CC, Tien WY, Lan YC, Tsai HT, Chen CH. Angiotensin II type I receptor (AT1R) is an independent prognosticator of esophageal squamous cell carcinoma and promotes cells proliferation via mTOR activation. Oncotarget. 2016;7(41):67150–65.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Arrieta O, Pineda-Olvera B, Guevara-Salazar P, Hernandez-Pedro N, Morales-Espinosa D, Ceron-Lizarraga TL, Gonzalez-De la Rosa CH, Rembao D, Segura-Pacheco B, Sotelo J. Expression of AT1 and AT2 angiotensin receptors in astrocytomas is associated with poor prognosis. Br J Cancer. 2008;99(1):160–6.CrossRefPubMedPubMedCentral Arrieta O, Pineda-Olvera B, Guevara-Salazar P, Hernandez-Pedro N, Morales-Espinosa D, Ceron-Lizarraga TL, Gonzalez-De la Rosa CH, Rembao D, Segura-Pacheco B, Sotelo J. Expression of AT1 and AT2 angiotensin receptors in astrocytomas is associated with poor prognosis. Br J Cancer. 2008;99(1):160–6.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Ota K, Fujimori H, Ueda M, Jono H, Shinriki S, Ota T, Sueyoshi T, Taura M, Taguma A, Kai H, et al. Midkine expression is correlated with an adverse prognosis and is down-regulated by p53 in oral squamous cell carcinoma. Int J Oncol. 2010;37(4):797–804.PubMed Ota K, Fujimori H, Ueda M, Jono H, Shinriki S, Ota T, Sueyoshi T, Taura M, Taguma A, Kai H, et al. Midkine expression is correlated with an adverse prognosis and is down-regulated by p53 in oral squamous cell carcinoma. Int J Oncol. 2010;37(4):797–804.PubMed
14.
Zurück zum Zitat Takeda H, Kondo S. Differences between squamous cell carcinoma and keratoacanthoma in angiotensin type-1 receptor expression. Am J Pathol. 2001;158(5):1633–7.CrossRefPubMedPubMedCentral Takeda H, Kondo S. Differences between squamous cell carcinoma and keratoacanthoma in angiotensin type-1 receptor expression. Am J Pathol. 2001;158(5):1633–7.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Harrington K, Temam S, Mehanna H, D’Cruz A, Jain M, D’Onofrio I, Manikhas G, Horvath Z, Sun Y, Dietzsch S, et al. Postoperative adjuvant lapatinib and concurrent chemoradiotherapy followed by maintenance lapatinib monotherapy in high-risk patients with resected squamous cell carcinoma of the head and neck: a phase III, randomized, double-blind, Placebo-Controlled Study. J Clin Oncol. 2015;33(35):4202–9.CrossRefPubMed Harrington K, Temam S, Mehanna H, D’Cruz A, Jain M, D’Onofrio I, Manikhas G, Horvath Z, Sun Y, Dietzsch S, et al. Postoperative adjuvant lapatinib and concurrent chemoradiotherapy followed by maintenance lapatinib monotherapy in high-risk patients with resected squamous cell carcinoma of the head and neck: a phase III, randomized, double-blind, Placebo-Controlled Study. J Clin Oncol. 2015;33(35):4202–9.CrossRefPubMed
16.
Zurück zum Zitat Matuschek C, Bolke E, Belka C, Ganswindt U, Henke M, Stegmaier P, Bamberg M, Welz S, Debus J, Gioules A, et al. Feasibility of 6-month maintenance cetuximab after adjuvant concurrent chemoradiation plus cetuximab in squamous cell carcinoma of the head and neck. Strahlenther Onkol. 2013;189(8):625–31.CrossRefPubMed Matuschek C, Bolke E, Belka C, Ganswindt U, Henke M, Stegmaier P, Bamberg M, Welz S, Debus J, Gioules A, et al. Feasibility of 6-month maintenance cetuximab after adjuvant concurrent chemoradiation plus cetuximab in squamous cell carcinoma of the head and neck. Strahlenther Onkol. 2013;189(8):625–31.CrossRefPubMed
17.
Zurück zum Zitat Racadot S, Thennevet I, Ouldbey Y, Kaminsky MC, Bosset M, Martin L, Tao Y, Sire C, de Raucourt D, Alfonsi M, et al. Afatinib maintenance therapy following post-operative radiochemotherapy in head and neck squamous cell carcinoma: results from the phase III randomised double-blind placebo-controlled study BIB2992ORL (GORTEC 2010–02). Eur J Cancer. 2023;178:114–27.CrossRefPubMed Racadot S, Thennevet I, Ouldbey Y, Kaminsky MC, Bosset M, Martin L, Tao Y, Sire C, de Raucourt D, Alfonsi M, et al. Afatinib maintenance therapy following post-operative radiochemotherapy in head and neck squamous cell carcinoma: results from the phase III randomised double-blind placebo-controlled study BIB2992ORL (GORTEC 2010–02). Eur J Cancer. 2023;178:114–27.CrossRefPubMed
18.
Zurück zum Zitat Yamashita T, Shimada H, Tanaka S, Araki K, Tomifuji M, Mizokami D, Tanaka N, Kamide D, Miyagawa Y, Suzuki H, et al. Serum midkine as a biomarker for malignancy, prognosis, and chemosensitivity in head and neck squamous cell carcinoma. Cancer Med. 2016;5(3):415–25.CrossRefPubMedPubMedCentral Yamashita T, Shimada H, Tanaka S, Araki K, Tomifuji M, Mizokami D, Tanaka N, Kamide D, Miyagawa Y, Suzuki H, et al. Serum midkine as a biomarker for malignancy, prognosis, and chemosensitivity in head and neck squamous cell carcinoma. Cancer Med. 2016;5(3):415–25.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Erdogan S, Doganlar ZB, Doganlar O, Turkekul K, Serttas R. Inhibition of midkine suppresses prostate cancer CD133(+) stem cell growth and migration. Am J Med Sci. 2017;354(3):299–309.CrossRefPubMed Erdogan S, Doganlar ZB, Doganlar O, Turkekul K, Serttas R. Inhibition of midkine suppresses prostate cancer CD133(+) stem cell growth and migration. Am J Med Sci. 2017;354(3):299–309.CrossRefPubMed
20.
Zurück zum Zitat Masui M, Okui T, Shimo T, Takabatake K, Fukazawa T, Matsumoto K, Kurio N, Ibaragi S, Naomoto Y, Nagatsuka H, et al. Novel midkine inhibitor iMDK inhibits tumor growth and angiogenesis in oral squamous cell carcinoma. Anticancer Res. 2016;36(6):2775–81.PubMed Masui M, Okui T, Shimo T, Takabatake K, Fukazawa T, Matsumoto K, Kurio N, Ibaragi S, Naomoto Y, Nagatsuka H, et al. Novel midkine inhibitor iMDK inhibits tumor growth and angiogenesis in oral squamous cell carcinoma. Anticancer Res. 2016;36(6):2775–81.PubMed
21.
Zurück zum Zitat Hu B, Qin C, Li L, Wei L, Mo X, Fan H, Lei Y, Wei F, Zou D. Midkine promotes glioblastoma progression via PI3K-Akt signaling. Cancer Cell Int. 2021;21(1):509.CrossRefPubMedPubMedCentral Hu B, Qin C, Li L, Wei L, Mo X, Fan H, Lei Y, Wei F, Zou D. Midkine promotes glioblastoma progression via PI3K-Akt signaling. Cancer Cell Int. 2021;21(1):509.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Egami K, Murohara T, Shimada T, Sasaki K, Shintani S, Sugaya T, Ishii M, Akagi T, Ikeda H, Matsuishi T, et al. Role of host angiotensin II type 1 receptor in tumor angiogenesis and growth. J Clin Invest. 2003;112(1):67–75.CrossRefPubMedPubMedCentral Egami K, Murohara T, Shimada T, Sasaki K, Shintani S, Sugaya T, Ishii M, Akagi T, Ikeda H, Matsuishi T, et al. Role of host angiotensin II type 1 receptor in tumor angiogenesis and growth. J Clin Invest. 2003;112(1):67–75.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Fujimoto Y, Sasaki T, Tsuchida A, Chayama K. Angiotensin II type 1 receptor expression in human pancreatic cancer and growth inhibition by angiotensin II type 1 receptor antagonist. FEBS Lett. 2001;495(3):197–200.CrossRefPubMed Fujimoto Y, Sasaki T, Tsuchida A, Chayama K. Angiotensin II type 1 receptor expression in human pancreatic cancer and growth inhibition by angiotensin II type 1 receptor antagonist. FEBS Lett. 2001;495(3):197–200.CrossRefPubMed
24.
Zurück zum Zitat Deshayes F, Nahmias C. Angiotensin receptors: a new role in cancer? Trends Endocrinol Metab. 2005;16(7):293–9.CrossRefPubMed Deshayes F, Nahmias C. Angiotensin receptors: a new role in cancer? Trends Endocrinol Metab. 2005;16(7):293–9.CrossRefPubMed
25.
Zurück zum Zitat O’Rawe M, Kilmister EJ, Mantamadiotis T, Kaye AH, Tan ST, Wickremesekera AC. The renin-angiotensin system in the tumor microenvironment of glioblastoma. Cancers (Basel). 2021;13(16):4004.CrossRefPubMedPubMedCentral O’Rawe M, Kilmister EJ, Mantamadiotis T, Kaye AH, Tan ST, Wickremesekera AC. The renin-angiotensin system in the tumor microenvironment of glioblastoma. Cancers (Basel). 2021;13(16):4004.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Lin YT, Wang HC, Tsai MH, Su YY, Yang MY, Chien CY. Angiotensin II receptor blockers valsartan and losartan improve survival rate clinically and suppress tumor growth via apoptosis related to PI3K/AKT signaling in nasopharyngeal carcinoma. Cancer. 2021;127(10):1606–19.CrossRefPubMed Lin YT, Wang HC, Tsai MH, Su YY, Yang MY, Chien CY. Angiotensin II receptor blockers valsartan and losartan improve survival rate clinically and suppress tumor growth via apoptosis related to PI3K/AKT signaling in nasopharyngeal carcinoma. Cancer. 2021;127(10):1606–19.CrossRefPubMed
27.
Zurück zum Zitat Zhao Y, Wang H, Li X, Cao M, Lu H, Meng Q, Pang H, Li H, Nadolny C, Dong X, et al. Ang II-AT1R increases cell migration through PI3K/AKT and NF-kappaB pathways in breast cancer. J Cell Physiol. 2014;229(11):1855–62.CrossRefPubMed Zhao Y, Wang H, Li X, Cao M, Lu H, Meng Q, Pang H, Li H, Nadolny C, Dong X, et al. Ang II-AT1R increases cell migration through PI3K/AKT and NF-kappaB pathways in breast cancer. J Cell Physiol. 2014;229(11):1855–62.CrossRefPubMed
28.
Zurück zum Zitat Zhang S, Wang Y. Telmisartan inhibits NSCLC A549 cell proliferation and migration by regulating the PI3K/AKT signaling pathway. Oncol Lett. 2018;15(4):5859–64.PubMedPubMedCentral Zhang S, Wang Y. Telmisartan inhibits NSCLC A549 cell proliferation and migration by regulating the PI3K/AKT signaling pathway. Oncol Lett. 2018;15(4):5859–64.PubMedPubMedCentral
Metadaten
Titel
Prognosis of Midkine and AT1R expression in resectable head and neck squamous cell carcinoma
verfasst von
Tai-Jan Chiu
Chang-Han Chen
Yi-Ju Chen
Yinshen Wee
Ching-Shuen Wang
Sheng‑Dean Luo
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2023
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-023-03060-z

Weitere Artikel der Ausgabe 1/2023

Cancer Cell International 1/2023 Zur Ausgabe

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Alter verschlechtert Prognose bei Endometriumkarzinom

11.05.2024 Endometriumkarzinom Nachrichten

Ein höheres Alter bei der Diagnose eines Endometriumkarzinoms ist mit aggressiveren Tumorcharakteristika assoziiert, scheint aber auch unabhängig von bekannten Risikofaktoren die Prognose der Erkrankung zu verschlimmern.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.