Open Access

Expression patterns of uPAR, TF and EGFR and their potential as targets for molecular imaging in oropharyngeal squamous cell carcinoma

  • Authors:
    • Anders Christensen
    • Christian Grønhøj
    • Jakob Schmidt Jensen
    • Giedrius Lelkaitis
    • Katalin Kiss
    • Karina Juhl
    • Birgitte Wittenborg Charabi
    • Jann Mortensen
    • Andreas Kjær
    • Christian Von Buchwald
  • View Affiliations

  • Published online on: June 30, 2022     https://doi.org/10.3892/or.2022.8359
  • Article Number: 147
  • Copyright: © Christensen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The clinical introduction of molecular imaging for the management of oropharyngeal squamous cell carcinoma (OPSCC) relies on the identification of relevant cancer‑specific biomarkers. The application of three membrane‑bound receptors, namely urokinase‑type plasminogen activator receptor (uPAR), tissue factor (TF) and EGFR have been previously explored for targeted imaging and therapeutic strategies in a broad range of solid cancers. The present study aimed to investigate the expression patterns of uPAR, EGFR and TF by immunohistochemistry (IHC) to evaluate their potential for targeted imaging and prognostic value in OPSCC. In a retrospective cohort of 93 patients with primary OPSCC, who were balanced into the 45 human papillomavirus (HPV)‑positive and 48 HPV‑negative groups, the IHC‑determined expression profiles of uPAR, TF and EGFR in large biopsy or tumor resection specimens were analyzed. Using the follow‑up data, overall survival (OS) and recurrence‑free survival were measured. Specifically, associations between survival outcome, biomarker expression and clinicopathological factors were examined using Cox proportional hazards model and log‑rank test following Kaplan‑Meier statistics. After comparing the expression pattern of biomarkers within the tumor compartment with that in the adjacent normal tissues, uPAR and TF exhibited a highly tumor‑specific expression pattern, whereas EGFR showed a homogeneous expression within the tumor compartment as well as a consistent expression in the normal mucosal epithelium and salivary gland tissues. The positive expression rate of uPAR, TF and EGFR in the tumors was 98.9, 76.3 and 98.9%, respectively. No statistically significant association between biomarker expression and survival outcome could be detected. Higher uPAR expression levels had a trend towards reduced OS according to results from univariate analysis (P=0.07; hazard ratio=2.01; 95% CI=0.92‑4.37). Taken together, these results suggest that uPAR, TF and EGFR may be suitable targets for molecular imaging and therapy in OPSCC. In particular, uPAR may be an attractive target owing to their high positive expression rates in tumors and a highly tumor‑specific expression pattern.

Introduction

Head and neck squamous cell carcinoma (HNSCC) is the sixth most frequent cancer globally, with an incidence of 890,000 new cases in 2018 (1). For oropharyngeal squamous cell carcinoma (OPSCC), a rising incidence rate has been reported over recent decades (2). This rising incidence has been observed in particular in the sub-group of patients with disease related to human papilloma virus (HPV) infection (2). Classically, treatment of OPSCC mainly involves chemo- and/or radiotherapy (3). However, a primary surgical treatment strategy has been recently established for early-stage primary disease and recurrent cases (3). Molecular imaging as a form of applied health technology is a field that is developing rapidly. Optimization of existing modalities or translation of novel modalities is currently in progress, with the aim of improving imaging for the staging and treatment of cancer (4). In particular, targeted optical imaging has been explored intensively over the past decade, since this modality allows for the real-time intraoperative visualization of tumor extension and margins (5). This in turn may aid in addressing the major challenge of inadequate tumor margins across a range of solid cancers that are treated surgically (5). For surgically treated OPSCC, the reported pooled rate of tumor-positive margins is 8% (6). Identification of molecular targets that are highly expressed in cancer and absent or minimally expressed in adjacent healthy tissues is key to the efficacy of molecular imaging (7). To evaluate potential targets for imaging purposes, studies investigating the exact expression pattern within the tumor compartment and the demarcation boundary with the non-cancerous tissue at the margins in appropriately-sized cohorts of patients are required (8).

Key parameter for the applicability of a target are tumor specificity and the expression rate in most if not all types of cancer. The present study investigated the expression pattern of urokinase-type plasminogen activator receptor (uPAR; also known as CD87), EGFR and tissue factor (TF; also known as CD142) in OPSCC. These are membrane-bound receptors that are considered to be candidate targets for imaging purposes in several solid cancers, including HNSCC (911) For uPAR and EGFR, numerous clinical studies with targeted probes for positron emission tomography and optical imaging are currently in progress (ClinicalTrials.gov Identifiers: NCT02945826, NCT02965001, NCT02960724, NCT02755675 and NCT03134846).

The plasminogen activator (PA) system serves a central role in cancer invasion and metastasis, which is mediated through pericellular proteolytic activity to degrade the extracellular matrix at the invasive front of a tumor. uPAR is expressed by both cancer cells and tumor-associated stromal cells. High expression levels in tumors and limited expression of uPAR in non-cancerous tissues have been previously reported in various cancers, including oral cavity squamous cell carcinoma (OSCC) (1215) In addition, higher expression levels of uPAR have been associated with poorer survival and more aggressive disease in numerous types of cancer including breast, bladder and colorectal cancer (1618).

The role of EGFR in HNSCC has been extensively explored, primarily for antibody-based targeted therapy. However, it also emerged as a candidate for targeted imaging over the past decade (19,20) EGFR is closely associated with tumor growth, since it is expressed by tumor cells in addition to various normal tissues in the human body (21).

An established relationship between cancer and thrombosis and venous thromboembolic events has been extensively reported. TF form part of the extrinsic coagulation cascade and it has been reported to be highly expressed in several solid tumors (22). In the tumor microenvironment, TF participates in multiple oncogenic signaling pathways that stimulate tissue remodeling and tumor angiogenesis in the invasive process; TF also modulates the immune response within the tumor compartment and assists tumor cells to escape the host immune system (23). Preclinical data has been reported to support the rationale for targeting TF using imaging and therapy (11,24).

Therefore, the present study aimed to investigate the expression patterns of uPAR, EGFR and TF by immunohistochemistry (IHC) to evaluate their potential for targeted imaging and possible prognostic value in OPSCC.

Materials and methods

Patients and tumor characteristics

From an established retrospective database of patients with primary OPSCC diagnosed between January 2000 and January 2012 in the department of Otolaryngology, Head & Neck surgery & Audiology at Rigshospitalet (Copenhagen, Denmark), a balanced cohort of 48 (52%) HPV-positive and 46 (48%) HPV-negative patients were randomly assembled. The inclusion criteria were primary OPSCC with preserved large biopsies or diagnostic resection specimens being available for IHC expression analysis. Exclusion criteria were insufficient tumor tissue for IHC analysis or incomplete dataset in the database. All specimens were collected prior to non-surgical treatment if this was indicated. Because HPV is a well-known powerful prognostic marker of OPSCC outcome, a balanced selection based on HPV status was performed. Both p16 and HPV-DNA status was available. The dataset in this current study has been part of previous publications and assays for the analysis of p16 and HPV-DNA have previously been described (25). Briefly, for p16 detection by IHC, the anti-P16 clone E6HA OptiView DAB IHC Detection Kit (Roche Diagnostics); for HPV-DNA analysis, DNA was isolated using the QIAamp DNA FFPE Tissue Kit (Qiagen, Inc.) and PCR was performed using General Primers GP5+/GP6+. The 8th version of the TNM Union For International Cancer Control (UICC8) staging manual was applied (26). The recorded clinicopathological characteristics and treatment modalities are provided in Table I. The present study was approved by the Ethical Committee of the Capital Region of Denmark (protocol H-15016322; Copenhagen, Denmark) and was conducted in accordance with The Declaration of Helsinki.

Table I.

Clinicopathological characteristics of the patients with oropharyngeal squamous cell carcinoma.

Table I.

Clinicopathological characteristics of the patients with oropharyngeal squamous cell carcinoma.

Clinicopathological characteristicN (%) or median (range)
Sex
  Female24 (26)
  Male69 (74)
Age, years60 (46–84)
Tumor location
  Pharynx wall8 (9)
  Palatine tonsils70 (75)
  Lingual tonsils12 (13)
  Soft palate3 (3%)
HPV-DNA
  Positive45 (48)
  Negative48 (52)
Smoking status
  Never smoker16 (17)
  Previous38 (41)
  Current smoker39 (42)
Tumor differentiation
  Non-keratinizing23 (25)
  High3 (3)
  Modrate37 (40)
  Low30 (32)
T stage
  T116 (17)
  T245 (48)
  T314 (15)
  T418 (19)
N stage
  N024 (26)
  N143 (46)
  N2a/2b/2c22 (24)
  N34 (4)
UICC8 TMN stage
  S136 (39)
  S220 (22)
  S316 (17)
  S421 (23)
Treatment
  RT33 (35)
  RCT46 (49)
  Surgery8 (9)
  Surgery + RT1 (2)
  Surgery + RCT1 (2)
  Palliation4 (4)
Recurrence
  No74 (80)
  Yes19 (20)
  Local6 (6)
  Regional6 (6)
  Locoregional1 (2)
  Distant6 (6)
Second primary
  No79 (85)
  Yes14 (15)

[i] HPV, human papillomavirus; RCT, radiochemotherapy; RT, radiotherapy.

IHC

At the time of collection, fresh tissue was fixed in 10% buffered formalin at room temperature for 24 h and then embedded in paraffin. The paraffin-embedded tissue blocks were sliced into 4-µm thick serial sections. Deparaffinization was performed with xylene for 15 min followed by dehydration using an ethanol gradient (from 99 to 70%, then demineralized water). IHC staining was performed on a Ventana Benchmark Ultra semi-automated autostainer (Roche Diagnostics). All antibody protocols have been previously optimized on positive and negative control tissues. The following antibodies were used: anti-cytokeratin (CK) AE1/AE3+8/18 (Pan Cytokeratin Plus; cat. no. 162; 1:200; Biocare Medical, LLC), mouse anti-human uPAR R2 (1:20,000; in-house antibody; Finsen Laboratory), anti-EGFR (Confirm anti-EGFR 5B7; cat. no. 790-4347; ready-to-use; Ventana Medical Systems, Inc.; Roche Diagnostics) and anti-TF (anti-TF; cat. no. 4509; 1:150; American Diagnostica, Inc.). Antigen retrieval for uPAR was performed with proteinase K (1:50 in Tris-HCl; cat. no. S3004; DAKO, Agilent Technologies, Inc.) for 8 min at room temperature, followed by 100°C heating with Cell Conditioning 1 buffer (CC1; Ventana Medical Systems, Inc.; Roche Diagnostics) for 16 min. For TF, EGFR and CK, heat-induced epitope retrieval was performed at 100°C in CC1 buffer for 32 min. The blocking step was performed using Peroxidase Blocking Solution (cat. no. S2023; DAKO, Agilent Technologies, Inc.) for 8 min at room temperature, followed by 2% BSA (cat. no. A7906; Sigma-Aldrich; Merck KGaA) blocking for 10 min at room temperature. For uPAR and TF, EnVision mouse HRP-conjugated EnVision FLEX+ secondary antibodies were used at room temperature for 40 min (cat. no. K8002; ready-to-use; DAKO; Agilent Technologies, Inc.), followed by detection with DAB and DAB+ Chromogen Solution (cat. no. K4065; diluted according to manufacturer's instructions; DAKO, Agilent Technologies, Inc.). For EGFR and CK, ultraView Universal DAB Detection Kit combined with ultraView Kit+ amplification (cat. nos. 760-500 and 760-080, respectively; Roche Diagnostics) and OptiView DAB IHC Detection Kit (cat. no. 760-700; Roche Diagnostics), respectively, were applied for detection.

In addition, separate sections were also stained with H&E for a 60 min protocol including deparaffinization, dehydration as described above, then stained in hematoxylin (cat. no. 854183; Capital Region, Hospital Farmacy) for five minutes, rinsed and stained with eosin (cat. no. 854653; Capital Region, Hospital Farmacy) for three minutes at room temperature. Digital images of all IHC slides were acquired using a Axio Scan.Z1 (Carl Zeiss AG) brightfield microscope slide scanner.

Expression analysis and scoring system

Five slides from each individual tumor were reviewed and scored by two experienced head and neck pathologists (KK and GL) blinded to the clinical dataset. In case of any discrepant assessment of IHC target expression, individual slides would be reviewed together to produce a consensus score. For each tumor, the extent of tumor compartmentalization and differentiation was evaluated using H&E and CK staining images. For uPAR, TF and EGFR respectively, IHC staining was semi-quantitatively assessed for intensity (I-score) and proportional expression (P-score; that is, the proportion of cells with positive biomarker expression) within the tumor compartment by assessment of the whole tumor section at lowest magnification. The I-score was stratified as 0–3 (none, weak, moderate and strong intensity, respectively) whereas the P-score was stratified as 0–3 (0–10, 11–50, 51–75 and 76–100%, respectively). The P-score was estimated based on the observed target expression on tumor cells, stromal cells and inflammatory cells within the tumor compartment. By adding the I-score and P-score, a composite semi-quantitative score (PI-score) was calculated, as proposed by Allred et al (27). The PI-score is a 7-point system stratified from 0 to 6. If present, IHC expression in adjacent non-cancerous tissues would also be qualitatively characterized. To estimate the expression rate of uPAR, TF and EGFR within the cohort, expression would be considered positive if the PI-score was >1. To assess the possible prognostic value of the three targets, the PI-score was dichotomized into high and low expression based on the mean value for each target for association analyses of associations. PI-scores <3 was used for the dichotomization of uPAR and TF, whereas PI-scores <4 was used for EGFR.

Statistical analysis

For associations between biomarker expression and each of the clinicopathological variables, Pearson's χ2 test or Fisher's exact test (for low number of events) was applied; for age comparison, an unpaired t-test was performed. For survival statistics, overall survival (OS) was defined as time from the date of diagnosis until death of any cause. Recurrence-free survival (RFS) was defined as time from the date of diagnosis until the date of recurrence confirmed by biopsy, death by any cause. Kaplan-Meier curves were applied for survival plots and 5-year survival estimates, where log-rank test was used for comparison between the groups. Using the Cox proportional hazards model, univariate and multivariate estimates of hazard ratios (HRs) were calculated for OS and RFS following adjustment for sex, age, smoking status, HPV status, tumor location, T-site, T-stage, N-stage, UICC8 TNM-stage, tumor differentiation and biomarker expression. P<0.05 was considered to indicate a statistically significant difference. Data analysis was performed in the SAS software package, version 6.1 (SAS Institute, Inc.) and R statistics (version 3.6.1).

Results

Basal patient characteristics

In this cohort of 93 patients, the median age at diagnosis was 60 years (range, 40–84 years), where 69 of the patients (74%) were men. The median follow-up calculated from the day of diagnosis was 4.8 years (range, 0.03-14.2 years). The tumor subsites were palatine tonsils (75%), base of the tongue (13%), pharyngeal wall (9%) and soft palate (3%). At follow up, January 2020, 51 patients were alive, and 42 patients were deceased, 25 (27%) of whom succumbed to OPSCC. During follow-up, 19 (20%) patients had recurrence confirmed by biopsy after primary treatment. The median time to recurrence was 9 months.

Target expression analysis

The positive expression rates in tumors based on the PI-score (PI>1) for uPAR, TF and EGFR were calculated to be 98.9, 76.3 and 98.9%, respectively. For the I-score, positive staining (I>0) for uPAR, TF and EFGR was present in 100, 89.3 and 100% of the specimens. The exact extension of the tumor compartment, and the margin demarcation from adjacent non-cancerous tissues were evaluated on the H&E and CK stainings and then compared to the topographic distribution of the IHC stainings for each of the three biomarkers. For uPAR a tumor-specific staining pattern was observed, with enhanced expression within the tumor compartment but limited or absent expression in the adjacent non-cancerous tissues (Fig. 1). In the tumor compartment, membranous uPAR staining was seen on tumor cells as well as on stromal cells. Tumor-associated stromal uPAR expression was also observed on macrophages, neutrophils, fibroblasts. At the mucosal margin, there was a sharp demarcation between carcinoma and adjacent epithelium, with the absence of uPAR expression in the unaffected epithelial lining. At the deep margins, uPAR staining delineated the tumor compartment. In addition, in the majority of cases, a narrow rim of expression was present in the stroma at the invasive border. Intense uPAR staining was frequently observed at the invasive front of the tumor and in the microscopic tumor buds separated from the primary tumor. In 10 (11%) of the specimens, uPAR positivity was also found on neutrophilic granulocytes, where pronounced inflammatory infiltration or abscess formation was discovered outside the tumor compartment.

TF demonstrated a tumor-specific expression pattern when positive, though in some cases adjacent normal epithelium also demonstrated weak positivity. As a general pattern, more intense TF staining was typically seen at the invasive front at the deep margin (Fig. 1).

For EGFR, strong and homogeneous expression was observed in the tumor compartment, but high expression could also be observed in the normal mucosal epithelium (Fig. 1). EGFR expression in the salivary tissue was consistently positive in the ductal epithelium (Fig. 1).

Survival and recurrence analysis

A Cox proportional hazards univariate and multivariate analysis was performed for RFS and OS (Table II). Age, HPV status and UICC8 TNM stage were identified to be independent prognostic factors for OS according to the univariate analysis, but only age and HPV status maintained significance in the multivariate analysis (Table II). Only increase in TNM staging for more advanced stage 3 and 4 disease was associated with RFS according to univariate analysis. For the three biomarkers, high uPAR PI-scores had a non-significant trend towards reduced OS according to univariate analysis only (Table II). In addition, Kaplan-Meier survival curves were generated with univariate log-rank tests performed to assess the association of the PI-scores of the three biomarkers with the OS and RFS (Fig. 2). For the entire cohort, the 5-year OS and RFS was 59 and 76%, respectively. For HPV-positive and HPV-negative patients, the respective 5-year OS was 77 and 41%, whereas the respective 5-year RFS was 81 and 67%. For patients with a high and low PI-scores for uPAR, the 5-year OS was 57 and 68%, respectively, whereas the RFS was 74 and 79% for patients with high and low PI-scores, respectively (Fig. 2). In addition, separate tests were performed for the P-score and I-score but no statistically significant association with OS or RFS could be detected (data not shown).

Table II.

Cox proportional hazards model on survival outcome from univariate and multivariate statistics.

Table II.

Cox proportional hazards model on survival outcome from univariate and multivariate statistics.

Overall survivalRecurrence-free survival


UnivariateMultivariateUnivariateMultivariate




Clinicopathological characteristicHR (CI)P-valueHR (CI)P-valueHR (CI)P-valueHR (CI)P-value
Sex
  FemaleRef. Ref. Ref. Ref.
  Male1.03 (0.51-2.05)0.941.16 (0.46-2.91)0.750.91 (0.33-2.54)0.860.75 (0.18-3.05)0.69
Age
  Risk per 1-year increase in age1.06 (1.03-1.10)<0.011.08 (1.03-1.13)<0.011.05 (1.00-1.10)0.051.05 (0.98-1.13)0.14
Tumor location
  Palatine and lingual tonsilsRef. Ref. Ref. Ref.
  Other1.57 (0.70-3.54)0.281.51 (0.48-4.79)0.480.41 (0.06-3.09)0.390.51 (0.05-5.20)0.57
Human papillomavirus status
  PositiveRef. Ref. Ref. Ref.
  Negative4.23 (2.08-8.63)<0.015.48 (1.11-27.05)0.041.73 (0.70-4.32)0.241.31 (0.17-10.21)0.8
Smoking status
  Never smokerRef. Ref. Ref. Ref.
  Previous or current smoker2.29 (0.82-6.43)0.110.95 (0.24-3.81)0.941.96 (0.45-8.48)0.375.21 (0.44-62.31)0.19
T-stage
  T1Ref. Ref. Ref. Ref.
  T20.80 (0.33-1.91)0.612.11 (0.60-7.38)0.240.87 (0.23-3.29)0.840.99 (0.21-4.76)0.99
  T30.26 (0.05-1.23)0.090.73 (0.12-4.23)0.720.67 (0.11-4.01)0.660.68 (0.06-7.09)0.75
  T43.17 (1.28-7.85)0.012.64 (0.66-10.56)0.173.54 (0.87-14.38)0.0810.27 (0.50-210.62)0.13
N-stage
  N0Ref. Ref. Ref. Ref.
  N10.72 (0.33-1.57)0.410.93 (0.31-2.78)0.91.19 (0.37-3.86)0.781.23 (0.22-6.81)0.81
  N21.42 (0.63-3.19)0.390.73 (0.20-2.66)0.641.58 (0.42-5.87)0.51.56 (0.24-10.07)0.64
  N33.72 (1.02-13.56)0.052.57 (0.42-15.74)0.313.22 (0.36-28.83)0.31.79 (0.09-33.93)0.7
UICC8 –TNM stage
  1Ref. Ref. Ref. Ref.
  20.60 (0.19-1.91)0.390.19 (0.03-1.15)0.071.00 (0.24-4.17)10.73 (0.10-5.25)0.75
  33.21 (1.35-7.62)<0.011.68 (0.36-7.84)0.513.71 (1.17-11.70)0.032.64 (0.28-24.56)0.4
  44.45 (2.03-9.76)<0.010.95 (0.12-7.44)0.962.17 (0.58-8.12)0.250.15 (0.00-7.73)0.35
Urokinase-type plasminogen activator receptor
  LowRef. Ref. Ref. Ref.
  High2.01 (0.92-4.37)0.070.87 (0.32-2.31)0.771.20 (0.43-3.34)0.720.88 (0.27-2.91)0.83
Tissue factor
  LowRef. Ref. Ref. Ref.
  High0.75 (0.38-1.50)0.421.17 (0.50-2.76)0.721.60 (0.64-3.97)0.312.39 (0.67-8.51)0.18
EGFR
  LowRef. Ref. Ref. Ref.
  High1.37 (0.72-2.61)0.341.55 (0.73-3.28)0.251.33 (0.52-3.37)0.552.11 (0.68-6.53)0.19

[i] HR(CI), hazard ratio (95% confidence interval); Ref, reference (equal to 1).

Biomarker association analysis

Possible associations between clinicopathological parameters and high or low PI-scores of uPAR, EGFR and TF were next investigated by univariate analysis (Table III). No statistically significant relationships were be detected.

Table III.

Univariate analysis of the level of biomarker expression and clinicopathological characteristics. Biomarker expression based on PI-score.

Table III.

Univariate analysis of the level of biomarker expression and clinicopathological characteristics. Biomarker expression based on PI-score.

Urokinase-type plasminogen activator receptorTissue factorEGFR



Clinicopathological characteristicHigh, n (%)Low, n (%)P-valueHigh, n (%)Low, n (%)P-valueHigh, n (%)Low, n (%)P-value
Sex
  Male51 (55)18 (19) 22 (24)47 (51) 42 (45)27 (29)
  Female17 (18)7 (8)0.776 (6)18 (19)0.5315 (16)9 (10)0.89
Age 0.29 0.44 0.37
Tumor location
  Pharynx wall1 (1)7 (7) 8 (9)0 (0) 3 (3)5 (5)
  Palatine tonsils18 (19)52 (56) 48 (52)22 (24) 26 (28)44 (47)
  Lingual tonsils4 (4)8 (9) 7 (8)5 (5) 7 (7)5 (5)
  Soft palate2 (2)1 (1)0.312 (2)1 (1)0.230 (0)3 (3)0.27
Human papilloma virus-DNA
  Positive35 (49)15 (16) 16 (17)32 (34) 28 (30)20 (22)
  Negative35 (38)10 (11)0.3312 (13)33 (35)0.4829 (31)16 (17)0.55
Smoking status
  Never27 (29)8 (9) 14 (15)21 (23) 21 (23)14 (15)
  Ever41 (44)17 (18)0.514 (15)44 (47)0.1139 (63)22 (24)0.84
Tumor differentiation
  G1-G217 (18)9 (10) 6 (6)20 (22) 12 (13)14 (15)
  G3-G451 (55)16 (17)0.2922 (24)45 (48)0.3645 (48)22 (24)0.06
T-stage
  T1-T242 (45)19 (20) 17 (18)44 (47) 61 (66)26 (28)
  T3-T326 (28)6 (6)0.211 (12)21 (23)0.5222 (24)11 (28)0.28
N-stage
  N016 (17)8 (9) 6 (6)18 (19) 15 (16)9 (10)
  N+52 (56)17 (18)0.4122 (24)47 (51)0.5342 (45)27 (29)0.89
TNM stage
  S1-S239 (42)17 (18) 21 (23)35 (38) 34 (37)22 (24)
  S3-S429 (31)9 (32)0.357 (8)30 (32)0.0623 (25)14 (15)0.89
Recurrence
  No54 (58)20 (22) 20 (22)54 (58) 45 (48)29 (31)
  Yes14 (15)5 (5)0.958 (9)11 (12)0.212 (13)7 (8)0.85

[i] Pearson's χ2 or Fisher's exact test (for low numbers) was used, except for age (as a continuous variable), in which an unpaired t-test was performed.

Discussion

The present study investigated the expression patterns and applicability of three candidate receptor targets in OPSCC for targeted molecular imaging based on IHC staining in large biopsy or tumor resection specimens. To the best of our knowledge, such topographic data on uPAR and TF expression for this large sub-site type of HNSCC has not been previously reported. Expression of EGFR in OPSCC has been previously described in a number of other studies (2830) An important finding from the present study was that uPAR in particular appeared to be a promising target in OPSCC due to its highly tumor-specific expression profile with limited expression in the non-cancerous regions adjacent to the tumor compartment. Although a tumor-specific expression pattern could also be observed for TF, the positive expression rate in the whole cohort was lower (76%). EGFR exhibited a high positive expression rate and a homogeneous expression pattern within the tumor compartment, equivalent to high receptor densities, rendering it a potential marker for targeted strategies. However, EGFR did not show a tumor-specific expression pattern, instead having regular expression levels in the adjacent normal epithelium and salivary gland tissues. Therefore, EGFR may be a less attractive target for molecular imaging. Our group previously performed a similar study of the expression of uPAR, TF and EFGR in OSCC (31); in comparison, the microscopic expression patterns and tumor-specificity of these three biomarkers appears very similar to the findings in OPSCC. This may indicate that individual targeted molecular strategies against either uPAR, TF or EGFR may be applied in OSCC and OPSCC, and possibly in HNSCC in general, which expands the utility of individual-targeted imaging agents or therapeutics.

It has been reported previously that targeted intraoperative optical imaging allows for a lower detection threshold for tumor deposits in the µm-range (10). Therefore, it is highly likely that this technology can be used to guide resection with adequate margins using biomarkers with tumor-specific expression patterns. In a clinical study using optical imaging in HNSCC by coupling with optically labeled antibodies against EGFR, Gao et al (32) previously demonstrated the potential use of targeted optical imaging for margin evaluation in the post-operative histopathological work-up. In addition, this previous study also reported tracer uptake in non-cancerous tissues outside the tumor compartment, such as the salivary gland tissues (32). The challenge of EGFR expression in normal tissues compared with targeted tumor imaging has been discussed by several previous studies, which underlines the need for exploring novel biomarkers in different types of cancer that exhibit highly tumor-specific expression (3335)

The identification of biomarkers for targeting purposes with a high positive expression rate is of importance for ensuring applicability and reproducibility in highly heterogenous populations. Van Oosten et al (8) previously suggested a lower expression rate of 80% for qualifying a given biomarker as clinically relevant for a targeted imaging strategy. Collecting preoperative biopsies for assessing biomarker expression in individual patients, which is already performed during immunotherapeutic procedures, is another alternative to imaging (36). However, it is a less feasible strategy if the targeted imaging of biomarkers with lower positive expression rates is anticipated. Baart et al (33) proposed the simultaneous use of several targeting agents against different biomarkers to achieve maximal agent-to-target binding in the same patient.

The role of the PA system in HNSCC has been previous reported, especially for OSCC and to a lesser extent for laryngeal SCC (37,38) However, it has not been previously described for OPSCC (37,38). In addition, little is known about TF in HNSCC, though a tumor-specific expression pattern in OSCC, very similar to the expression pattern found in this study, has been reported previously by our group (31). Since the mucosal lining in both the oral cavity and the pharynx is non-keratinizing squamous epithelium, it is likely that the same biomarker expression profiles are present in both locations (39). Depending on the IHC scoring system used, the positive expression rate of uPAR in OSCC has been reported to be between 39 and 100% (38,40) which is in line with a positivity rate of 98.9% in the present study. The positive expression rate for TF in OSCC was reported to be 58% (31), which is lower compared with the positive expression rate of 76.9% in OPSCC seen in the current study.

The present study did not detect an association between the expression levels of uPAR, TF and EFGR with HPV status, where the microscopic distribution of these three biomarkers did not reveal any differences between sections with and without HPV. It is well documented that HPV-positive and -negative tumors are two distinctly different diseases in terms of pathogenesis and prognosis, such that the carcinogenesis process in the presence of HPV infection can interact with the immune (41,42) Both uPAR and TF have been reported to serve a role in immunological regulation and responses in the tumor microenvironment (17,23) However, the expression of only three receptors within a highly complex biological system was studied in the present study, which may explain why no relationship with HPV status could be observed.

In the present cohort, HPV-positive patients had significantly more favorable survival outcome. In the survival analysis, the level of uPAR, TF and EGFR expression did not significantly associate with any of the survival outcomes tested. Only high uPAR expression had a non-significant trend towards reduced OS. Other previous studies of OSCC and other types of cancer, like breast or bladder cancer, have found that high uPAR or uPA expression significantly associated with reduced survival and more aggressive histopathological tumor physiology (43,44) A possible reason for the lack of prognostic significance in terms of uPAR in OPSCC expression in the present study may be the small sample size and the inherent lack of granularity and accuracy in the IHC-based scoring. Nevertheless, these methodological shortcomings should equally influence all three molecular targets. Therefore, they indicate that of the three markers tested, uPAR is the most prognostic. However, the present study was limited by a relatively small sample size to perform robust association analyses on survival statistics.

In conclusion, results from the present study suggest that uPAR, TF and EGFR are suitable targets for molecular imaging and therapy in OPSCC. uPAR in particular may be an attractive target owing to its highly tumor-specific expression pattern and association with prognosis. For TF, preselection of patients may be necessary due to a lower positivity rate.

Acknowledgements

Not applicable.

Funding

Funding was obtained from University of Copenhagen, The Agnes & Poul Friis Foundation and The Haboe Foundation.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

AC and CG designed the study in collaboration with KK, GL, KJ, BWC, JM, AK and CvB. CG and JSJ collected tissue specimens and established the dataset. AC and CG have assessed the authenticity of the raw data. KK, GL and KJ conducted the immunohistochemistry. KK and GL performed the histopathological scoring. AC, CG and JSJ performed the statistical data analysis. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Approval for this study was obtained from the Ethical Committee of the Capital Region of Denmark (protocol H-15016322; Copenhagen, Denmark). The Dataset was anonymized prior to analysis. The Ethical Committee waived the need to obtain consent to participate from the patients due to the circumstance that a great proportion of the cohort had succumbed to disease or were severely ill, where revealing that information about this project would lead to more distress in the patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

EGFR

epidermal growth factor receptor

HNSCC

head and neck squamous cell carcinoma

IHC

immunohisto-chemistry

OPSCC

oropharyngeal squamous cell carcinoma

OS

overall survival

RFS

recurrence-free survival

TF

tissue factor

uPAR

urokinase-type plasminogen activator receptor

References

1 

Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, Znaor A and Bray F: Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J cancer. 144:1941–1953. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Mehanna H, Beech T, Nicholson T, El-Hariry I, McConkey C, Paleri V and Roberts S: Prevalence of human papillomavirus in oropharyngeal and nonoropharyngeal head and neck cancer-systematic review and meta-analysis of trends by time and region. Head Neck. 35:747–755. 2013. View Article : Google Scholar : PubMed/NCBI

3 

De Virgilio A, Costantino A, Mercante G, Pellini R, Ferreli F, Malvezzi L, Colombo G, Cugini G, Petruzzi G and Spriano G: Transoral robotic surgery and intensity-modulated radiotherapy in the treatment of the oropharyngeal carcinoma: A systematic review and meta-analysis. Eur Arch Otorhinolaryngol. 278:1321–1335. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Visgauss JD, Eward WC and Brigman BE: Innovations in intraoperative tumor visualization. Orthop Clin North Am. 47:253–264. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Nguyen QT and Tsien RY: Fluorescence-guided surgery with live molecular navigation-a new cutting edge. Nat Rev Cancer. 13:653–662. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Gorphe P and Simon C: A systematic review and meta-analysis of margins in transoral surgery for oropharyngeal carcinoma. Oral Oncol. 98:69–77. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Boonstra MC, De Geus SWL, Prevoo HAJM, Hawinkels LJAC, van de Velde CJH, Kuppen PJK, Vahrmeijer AL and Sier CFM: Selecting targets for tumor imaging: An overview of cancer–associated membrane proteins. Biomark Cancer. 8:119–133. 2016. View Article : Google Scholar : PubMed/NCBI

8 

van Oosten M, Crane LMA, Bart J, van Leeuwen FW and van Dam GM: Selecting potential targetable biomarkers for imaging purposes in colorectal cancer using target selection criteria (TASC): A novel target identification tool. Transl Oncol. 4:71–82. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Boonstra MC, van Driel PBAA, van Willigen DM, Stammes MA, Prevoo HAJM, Tummers QRJG, Mazar AP, Beekman FJ, Kuppen PJK, van de Velde CJH, et al: uPAR-targeted multimodal tracer for pre- and intraoperative imaging in cancer surgery. Oncotarget. 6:14260–14273. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Christensen A, Juhl K, Persson M, Charabi BW, Mortensen J, Kiss K, Lelkaitis G, Rubek N, von Buchwald C and Kjær A: uPAR-targeted optical near-infrared (NIR) fluorescence imaging and PET for image-guided surgery in head and neck cancer: Proof-of-concept in orthotopic xenograft model. Oncotarget. 8:15407–15419. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Nielsen CH, Jeppesen TE, Kristensen LK, Jensen MM, Ali HHE, Madsen J, Wiinberg B, Petersen LC and Kjaer A: PET imaging of tissue factor in pancreatic cancer using 64Cu-labeled active site-inhibited factor VII. J Nucl Med. 57:1112–1119. 2016. View Article : Google Scholar : PubMed/NCBI

12 

de Geus SWL, Baart VM, Boonstra MC, Kuppen PJK, Prevoo HA, Mazar AP, Bonsing BA, Morreau H, van de Velde CJH, Vahrmeijer AL and Sier CF: Prognostic impact of urokinase plasminogen activator receptor expression in pancreatic cancer: Malignant versus stromal cells. Biomark Insights. 12:11772719177154432017. View Article : Google Scholar : PubMed/NCBI

13 

Boonstra MC, Verspaget HW, Ganesh S, Kubben FJGM, Vahrmeijer AL, van de Velde CJH, Kuppen PJK, Quax PHA and Sier CFM: Clinical applications of the urokinase receptor (uPAR) for cancer patients. Curr Pharm Des. 17:1890–1910. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Noh H, Hong S and Huang S: Role of urokinase receptor in tumor progression and development. Theranostics. 3:487–495. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Hundsdorfer B, Zeilhofer HF, Bock KP, Dettmar P, Schmitt M, Kolk A, Pautke C and Horch HH: Tumour-associated urokinase-type plasminogen activator (uPA) and its inhibitor PAI-1 in normal and neoplastic tissues of patients with squamous cell cancer of the oral cavity-clinical relevance and prognostic value. J Craniomaxillofac Surg. 33:191–196. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Bharadwaj AG, Holloway RW, Miller VA and Waisman DM: Plasmin and plasminogen system in the tumor microenvironment: Implications for cancer diagnosis, prognosis, and therapy. Cancers (Basel). 13:18382021. View Article : Google Scholar : PubMed/NCBI

17 

Madunić J: The urokinase plasminogen activator system in human cancers: An overview of its prognostic and predictive role. Thromb Haemost. 118:2020–2036. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Serpa MS, Mafra RP, Queiroz SIML, da Silva LP, de Souza LB and Pinto LP: Expression of urokinase-type plasminogen activator and its receptor in squamous cell carcinoma of the oral tongue. Braz Oral Res. 32:e932018. View Article : Google Scholar : PubMed/NCBI

19 

van Keulen S, van den Berg NS, Nishio N, Birkeland A, Zhou Q, Lu G, Wang HW, Middendorf L, Forouzanfar T, Martin BA, et al: Rapid, non-invasive fluorescence margin assessment: Optical specimen mapping in oral squamous cell carcinoma. Oral Oncol. 88:58–65. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Weele EJT, Van Scheltinga AGT, Linssen MD, Nagengast WB, Lindner I, Jorritsma-Smit A, de Vries EGE, Kosterink JGW and Hooge MN: Development, preclinical safety, formulation, and stability of clinical grade bevacizumab-800CW, a new near infrared fluorescent imaging agent for first in human use. Eur J Pharm Biopharm. 104:226–234. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Zaryouh H, De Pauw I, Baysal H, Peeters M, Vermorken JB, Lardon F and Wouters A: Recent insights in the PI3K/Akt pathway as a promising therapeutic target in combination with EGFR-targeting agents to treat head and neck squamous cell carcinoma. Med Res Rev. 42:112–155. 2022. View Article : Google Scholar : PubMed/NCBI

22 

Beleva E and Grudeva-Popova J: From Virchow's triad to metastasis: circulating hemostatic factors as predictors of risk for metastasis in solid tumors. J BUON. 18:25–33. 2013.PubMed/NCBI

23 

Han X, Guo B, Li Y and Zhu B: Tissue factor in tumor microenvironment: A systematic review. J Hematol Oncol. 7:542014. View Article : Google Scholar : PubMed/NCBI

24 

Theunissen JW, Cai AG, Bhatti MM, Cooper AB, Avery AD, Dorfman R, Guelman S, Levashova Z and Migone TS: Treating tissue factor-positive cancers with antibody-drug conjugates that do not affect blood clotting. Mol Cancer Ther. 17:2412–2426. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Zamani M, Grønhøj C, Jensen DH, Carlander AF, Agander T, Kiss K, Olsen C, Baandrup L, Nielsen FC, Andersen E, et al: The current epidemic of HPV-associated oropharyngeal cancer: An 18-year Danish population-based study with 2,169 patients. Eur J Cancer. 134:52–59. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Brierley JD, Gospodarowicz MK and Wittekind C: TNM classification of malignant tumours. 8th edition. Wiley Blackwell; 2017

27 

Allred DC, Harvey JM, Berardo M and Clark GM: Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol. 11:155–168. 1998.PubMed/NCBI

28 

Bossi P, Resteghini C, Paielli N, Licitra L, Pilotti S and Perrone F: Prognostic and predictive value of EGFR in head and neck squamous cell carcinoma. Oncotarget. 7:74362–74379. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Preuss SF, Weinell A, Molitor M, Semrau R, Stenner M, Drebber U, Wedemeyer I, Hoffmann TK, Guntinas-Lichius O and Klussmann JP: Survivin and epidermal growth factor receptor expression in surgically treated oropharyngeal squamous cell carcinoma. Head Neck. 30:1318–1324. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Chandarana SP, Lee JS, Chanowski EJP, Sacco AG, Bradford CR, Wolf GT, Prince ME, Moyer JS, Eisbruch A, Worden FP, et al: Prevalence and predictive role of p16 and epidermal growth factor receptor in surgically treated oropharyngeal and oral cavity cancer. Head Neck. 35:1083–1090. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Christensen A, Kiss K, Lelkaitis G, Juhl K, Persson M, Charabi BW, Mortensen J, Forman JL, Sørensen AL, Jensen DH, et al: Urokinase-type plasminogen activator receptor (uPAR), tissue factor (TF) and epidermal growth factor receptor (EGFR): Tumor expression patterns and prognostic value in oral cancer. BMC Cancer. 17:5722017. View Article : Google Scholar : PubMed/NCBI

32 

Gao RW, Teraphongphom NT, van den Berg NS, Martin BA, Oberhelman NJ, Divi V, Kaplan MJ, Hong SS, Lu G, Ertsey R, et al: Determination of tumor margins with surgical specimen mapping using near-infrared fluorescence. Cancer Res. 78:5144–5154. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Baart VM, van Duijn C, van Egmond SL, Dijckmeester WA, Jansen JC, Vahrmeijer AL, Sier CFM and Cohen D: EGFR and αvβ6 as promising targets for molecular imaging of cutaneous and mucosal squamous cell carcinoma of the head and neck region. Cancers (Basel). 12:14742020. View Article : Google Scholar : PubMed/NCBI

34 

de Boer E, Warram JM, Tucker MD, Hartman YE, Moore LS, de Jong JS, Chung TK, Korb ML, Zinn KR, van Dam GM, et al: In vivo fluorescence immunohistochemistry: Localization of fluorescently labeled cetuximab in squamous cell carcinomas. Sci Rep. 5:101692015. View Article : Google Scholar : PubMed/NCBI

35 

Voskuil FJ, de Jongh SJ, Hooghiemstra WTR, Linssen MD, Steinkamp PJ, de Visscher SAHJ, Schepman KP, Elias SG, Meersma GJ, Jonker PKC, et al: Fluorescence-guided imaging for resection margin evaluation in head and neck cancer patients using cetuximab-800CW: A quantitative dose-escalation study. Theranostics. 10:3994–4005. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Cramer JD, Burtness B and Ferris RL: Immunotherapy for head and neck cancer: Recent advances and future directions. Oral Oncol. 99:1044602019. View Article : Google Scholar : PubMed/NCBI

37 

Wojtukiewicz MZ, Sierko E, Zacharski LR, Rózanska-Kudelska M and Zimnoch L: Occurrence of components of fibrinolytic pathways in situ in laryngeal cancer. Semin Thromb Hemost. 29:317–320. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Bacchiocchi R, Rubini C, Pierpaoli E, Borghetti G, Procacci P, Nocini PF, Santarelli A, Rocchetti R, Ciavarella D, Lo Muzio L and Fazioli F: Prognostic value analysis of urokinase-type plasminogen activator receptor in oral squamous cell carcinoma: An immunohistochemical study. BMC Cancer. 8:2202008. View Article : Google Scholar : PubMed/NCBI

39 

van der Waal I: Potentially malignant disorders of the oral and oropharyngeal mucosa; terminology, classification and present concepts of management. Oral Oncol. 45:317–323. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Curino A, Patel V, Nielsen BS, Iskander AJ, Ensley JF, Yoo GH, Holsinger FC, Myers JN, El-Nagaar A, Kellman RM, et al: Detection of plasminogen activators in oral cancer by laser capture microdissection combined with zymography. Oral Oncol. 40:1026–1032. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Lechien JR, Seminerio I, Descamps G, Mat Q, Mouawad F, Hans S, Julieron M, Dequanter D, Vanderhaegen T, Journe F and Saussez S: Impact of HPV infection on the immune system in oropharyngeal and non-oropharyngeal squamous cell carcinoma: A systematic review. Cells. 8:10612019. View Article : Google Scholar : PubMed/NCBI

42 

Subbarayan RS, Arnold L, Gomez JP and Thomas SM: The role of the innate and adaptive immune response in HPV-associated oropharyngeal squamous cell carcinoma. Laryngoscope Investig Otolaryngol. 4:508–512. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Gonias SL and Zampieri C: Plasminogen receptors in human malignancies: Effects on prognosis and feasibility as targets for drug development. Curr Drug Targets. 21:647–656. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Magnussen S, Rikardsen OG, Hadler-Olsen E, Uhlin-Hansen L, Steigen SE and Svineng G: Urokinase plasminogen activator receptor (uPAR) and plasminogen activator inhibitor-1 (PAI-1) are potential predictive biomarkers in early stage oral squamous cell carcinomas (OSCC). PLoS One. 9:e1018952014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2022
Volume 48 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Christensen A, Grønhøj C, Jensen JS, Lelkaitis G, Kiss K, Juhl K, Charabi BW, Mortensen J, Kjær A, Von Buchwald C, Von Buchwald C, et al: Expression patterns of uPAR, TF and EGFR and their potential as targets for molecular imaging in oropharyngeal squamous cell carcinoma. Oncol Rep 48: 147, 2022
APA
Christensen, A., Grønhøj, C., Jensen, J.S., Lelkaitis, G., Kiss, K., Juhl, K. ... Von Buchwald, C. (2022). Expression patterns of uPAR, TF and EGFR and their potential as targets for molecular imaging in oropharyngeal squamous cell carcinoma. Oncology Reports, 48, 147. https://doi.org/10.3892/or.2022.8359
MLA
Christensen, A., Grønhøj, C., Jensen, J. S., Lelkaitis, G., Kiss, K., Juhl, K., Charabi, B. W., Mortensen, J., Kjær, A., Von Buchwald, C."Expression patterns of uPAR, TF and EGFR and their potential as targets for molecular imaging in oropharyngeal squamous cell carcinoma". Oncology Reports 48.2 (2022): 147.
Chicago
Christensen, A., Grønhøj, C., Jensen, J. S., Lelkaitis, G., Kiss, K., Juhl, K., Charabi, B. W., Mortensen, J., Kjær, A., Von Buchwald, C."Expression patterns of uPAR, TF and EGFR and their potential as targets for molecular imaging in oropharyngeal squamous cell carcinoma". Oncology Reports 48, no. 2 (2022): 147. https://doi.org/10.3892/or.2022.8359