Open Access

Cytokeratin and protein expression patterns in squamous cell carcinoma of the oral cavity provide evidence for two distinct pathogenetic pathways

  • Authors:
    • Gesche Frohwitter
    • Horst Buerger
    • Paul J. van Diest
    • Eberhard Korsching
    • Johannes Kleinheinz
    • Thomas Fillies
  • View Affiliations

  • Published online on: May 16, 2016     https://doi.org/10.3892/ol.2016.4588
  • Pages: 107-113
  • Copyright: © Frohwitter 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

Squamous cell carcinoma (SCC) of the oral cavity is a morphological heterogeneous disease. Various cytokeratin (CK) expression patterns with different prognostic values have been described, but little is known concerning the underlying biological cell mechanisms. Therefore, the present study investigated 193 cases of oral SCCs using immunohistochemistry for α/β/γ‑catenin, glucose transporter 1, caspase‑3, X‑linked inhibitor of apoptosis protein, hypoxia inducible factor‑1α, carbonic anhydrase 9, heat shock protein (hsp) 70, mast/stem cell growth factor receptor, p21, p27, p16, p53, B‑cell lymphoma 6, epidermal growth factor receptor, cyclin D1 and CK1, 5/6, 8/18, 10, 14 and 19. Expression patterns were analyzed with biomathematical permutation analysis. The present results revealed a significant association between the expression of low‑molecular weight CK8/18 and 19 and a high‑tumor grade, β and γ‑catenin expression, deregulated cell cycle proteins and a predominant localization of the tumor on the floor of the mouth. By contrast, expression of high‑molecular weight CK1, 5/6, 10 and 14 was significantly associated with the expression of p21 and hsp70. In conclusion, the current study presents evidence for the existence of two parallel pathogenetic pathways in oral SCCs, characterized by the expression of low‑ and high‑molecular weight CKs. Additional studies are required to demonstrate the extent that these results may be used to improve therapeutic regimens.

Introduction

Squamous cell carcinoma (SCC) represents the vast majority (90%) of malignant oral neoplasms (1). Recent figures on SCCs emphasise the growing incidence rate and the limited and often unsatisfying treatment options (2,3). Treatments are known to be unsatisfactory in terms of survival rates, as local and regional metastases occur frequently even in small tumors (4). The harm caused by agents, such as alcohol or tobacco, is important in cancerization, and may be responsible for the formation of secondary tumors and disease recurrence (5,6). A detailed knowledge concerning the molecular mechanisms in the pathogenesis and progression of SCCs, and its precursor lesions, is required to aid in the improvement of therapeutics and incidence rates.

Cytokeratins (CKs) are major intermediate filaments in squamous epithelium and are critical in cell stabilization, shape, intracellular signalling and transport (7,8). CK expression is a hallmark of tumor progression. Previous studies demonstrated that the expression of high-molecular weight CK8 and 18 was associated with dysplasia grades of tumor precursor lesions and an unfavorable prognosis for patients with SCCs (9,10). Other studies had similar observations (11,12). Loss of CK8 phosphorylation initiates an increased cell migration and tumor spread in SCCs, whereas loss of CK8 and 18 led to alterations in α6β4-integrin mediated signalling and decreased neoplastic progression (11,13,14). In addition, an increased expression of low-molecular weight CK19 was associated with high-grade dysplasia and squamous intraepithelial neoplasia and a decreased survival rate of patients with SCCs (9,15). CK19 expression is not observed in benign or hyperplastic regions of keratinized oral epithelium (10,15). Additionally, the expression of CKs interferes with a multitude of other intracellular regulation pathways, including numerous kinases, receptors and apoptotic proteins (16); therefore, the effects of CKs are highly complex.

The present study demonstrates the frequencies of low and high molecular weight CKs in SCCs of the oral cavity with respect to their anatomical sublocalization, and evaluates the coexpression of a multitude of proteins involved in various cellular regulatory pathways. The aim of the present study was to elucidate the complex interaction between CK expression patterns and alterations in other cellular pathways. The present results revealed the existence of two pathogenetic pathways in the evolution of oral SCC, which are associated with the preferential expression of low- and high-molecular weight CKs.

Patients and methods

Patients

All patients eligible for the current study presented with histologically confirmed oral SCC, and underwent surgery at the University Hospital Muenster (Münster, Germany) between January 1988 and December 2000. Data collection and evaluation was conducted in compliance with the current version of the Declaration of Helsinki and the International Conference for Harmonization of Good Clinical Practice (17,18). All data and specimens were evaluated after obtaining written informed consent from the patient and ethical approval from the Ethical Commission of the Medical Association Westfalen-Lippe and the Faculty of Medicine of the Westphalian Wilhelms-University Muenster. The tumor samples were formalin-fixed, archival paraffin-embedded tissues from 193 patients (154 men; 39 women) with primary oral SCC. The patients' mean age was 59 years (range, 31–90 years) (Table I). Information regarding clinicopathological details and treatment modalities of this tumor series is described in previous studies (9,10,1921). According to the Union for International Cancer Control guidelines (22), all the SCCs were classified post-surgery by the tumor-node-metastasis system (T1=96, T2=82, T3-4=15; N0=136, N>0=57). All patients attended a follow-up program with clinical evaluation for 4–181 months. As described in previous studies (9,10,1921), the time of survival was defined as the time between the date of surgery and the date of histopathologically proven tumor recurrence, metastatic disease, death associated with the disease or a tumor-free follow-up period of 60 months. Patients who missed regular attendance at follow-up were excluded from the present study.

Table I.

Clinicopathological characteristics of 193 patients with oral squamous cell carcinoma.

Table I.

Clinicopathological characteristics of 193 patients with oral squamous cell carcinoma.

CharacteristicValue
Age at diagnosis, years
  Mean  59
  Range31–90
Gender
  Female  39
  Male154
Tumor stage
  T1  96
  T2  82
  T3-T4  15
Lymph node
  Negative136
  Positive  57
Tumor grade
  G1  44
  G2126
  G3  23
Disease recurrence
  Positive  66
  Negative127
Localization
  Floor of mouth  76
  Tongue  49
  Other  68
Immunohistochemistry

To provide equal assessment conditions, all tumor samples were analyzed by tissue microarray (TMA) and immunohistochemistry. As previously described (23), all TMAs were used according to a standard procedure. For the TMA block construction, two punch biopsies (diameter, 0.6 mm) were extracted from formalin-fixed, paraffin-embedded tumor tissue (thickness, 4 µm) using a tissue microarray instrument (Beecher Instruments, Inc., Sun Prairie, WI, USA) and inserted into a novel acceptor block. The acceptor block underwent deparaffinization, using various concentrations of ethanol (70, 95 and 100%; Walter-CMP GmbH & Co. KG, Kiel, Germany) and rehydration. Endogenous peroxidase activity was blocked using methanol (with 0.3% hydrogen peroxide) (Walter-CMP GmbH & Co. KG) for 30 min. Antigen retrieval was preceded by a cooling time of 20 min at room temperature and incubation with primary antibodies for 30 min at room temperature (Table II). Catalyzed Signal Amplification System (Dako, Glostrup, Denmark) was used for CK1, 5/6, 8/18, 10, 14 and 19 immunohistochemistry, according to the manufacturer's protocol. The staining procedures for p53, p21, p27, p16, cyclin D1, epidermal growth factor receptor, mast/stem cell growth factor receptor (c-kit), B-cell lymphoma 6, α/β/γ-catenin, hypoxia inducible factor-1α (HIF-1α), glucose transporter 1 (GLUT1), carbonic anhydrase 9 (CAIX), caspase-3, heat shock protein (hsp) 70 and X-linked inhibitor of apoptosis protein (XIAP) were performed as previously described (24,25). Antigen detection was performed by a standardised avidin-biotin complex method using anti-rabbit and anti-mouse biotinylated antibodies [Dako REAL Detection Systems (LSAB+); catalog no. K5003; Dako] and a Biotin-Blocking System (Ready-to-Use, catalog no. X0590; Dako). Diaminobenzidine (included in the p16 and EGFR kits; Table II) or LSAB 2 System-AP (Dako) was used for visualization, along with counterstaining with hematoxylin for 45 sec, followed by dehydration in alcohol and xylene (Walter-CMP GmbH & Co. KG). During TMA analysis and immunohistochemistry, negative (omission of the primary antibody) and positive controls were performed.

Table II.

Primary antibodies used for immunohistochemistry in the present study.

Table II.

Primary antibodies used for immunohistochemistry in the present study.

AntibodySupplierCatalog no.Clone Mono/polyclonalSpeciesDilutionAntigen retrieval
p21Merck Millipore05–655CP74MonoMouse1:500Citrate buffer (pH6)
p27BD TL61024157/Kip1/p27MonoMouse1:1,000Citrate buffer (pH6)
p53DakoM7001DO-7MonoMouse1:100EDTA (pH8)
HIF-1αBD TL61095854/HIF-1αMonoMouse1:50EDTA (pH8)
GLUT1DakoM7211Clone A 35MonoMouse1:40EDTA (pH8)
CAIXAbcamab128883PolyRabbit1:1,000Citrate buffer (pH6)
XIAPBD TL61071628/hILP/XIAPMonoMouse1:50Citrate buffer (pH6)
Hsp 70Invitrogen33–3800MB-H1MonoMouse1:40Citrate buffer (pH6)
α-cateninBD TL6101945/a-cateninMonoMouse1:250EDTA (pH8)
β-cateninBD TL610153 14/beta-CateninMonoMouse1:1,000EDTA (pH8)
γ-cateninBD TL61025315/γ-cateninMonoMouse1:1,500EDTA (pH8)
BCL-6DakoM7211PG-B6pMonoMouse1:50Citrate buffer (pH6)
Caspase-3Invitrogen35–1600Z43191MonoMouse1:100Citrate buffer (pH6)
C-kitDakoA4502PolyRabbit1:200Citrate buffer (pH6)
CK1NovocastraNCL-Ck134βB4MonoMouse1:150Citrate buffer (pH6)
CK5/6DakoM7237D5/16 B4MonoMouse1:80Autoclave (10 min)
CK10DakoM7002DE-K10MonoMouse1:400Citrate buffer (pH6)
CK14Dianova GmbHDLN-06600LL002MonoMouse1:50Citrate buffer (pH6)
CK8/18Dianova GmbHDLN-08110K8.8/DC10MonoMouse1:40Autoclave (10 min), citrate buffer (pH)
CK19Dianova GmbHDLN-08330KS19.1MonoMouse1:80Citrate buffer (pH6)
Cyclin D1NovocastraNCL-L-cyclin D1-GMP2D11F11MonoMouse1:20EDTA (pH8)
EGFRDakoK1492pharmDX kitMonoMouse
p16CINtec9517E6H4MonoMouse Citrate buffer (pH6)

[i] Merck Millipore, Darmstadt, Germany; BD TL, BD Transduction Laboratories™, BD Biosciences, Frankling Lakes, NJ, USA; Dako, Glostrup, Denmark; Invitrogen™, Thermo Fisher Scientific, Inc., Waltham, MA, USA; Abcam, Cambridge, UK; Novocastra™, Leica Biosystems GmbH, Nussloch, Germany; Dianova GmbH, Hamburg, Germany; CINtec®, Roche AG, Basel Switzerland. EDTA and citrate buffers were purchased from Dako and Zytomed Systems GmbH (Berlin, Germany), respectively. CK, cytokeratin; HIF-1α, hypoxia inducible factor-1α; GLUT1, glucose transporter 1; XIAP, X-linked inhibitor of apoptosis protein; CAIX, carbonic anhydrase 9; Hsp, heat shock protein; C-kit, mast/stem cell growth factor receptor; BCL-6, B-cell lymphoma-6; EGFR, epidermal growth factor receptor.

Scoring of staining

Expression of CK1, 5/6, 8/18, 10, 14 and 19 was evaluated by the rate of positively stained cells in each core. The expression levels (%) were classified into three groups for CK19 (0%, no expression; 1–50%, moderate expression; >50%, high expression) and into two groups for CK5/6, 8/18, 1, 10 and 14 (0%, no expression; ≥1%, positive expression). The mean percentage value of two cores from one tumor was calculated. Cytoplasmic expression of hsp70, caspase-3 and XIAP was graded as negative or positive (intermediate to strong expression), irrespective of the relative number of stained tumor cells. The remaning molcecules were assessed as follows: CAIX and p21/27/16 (<1%, no expression; ≥1%, positive expression); HIF-1α and GLUT1 (<1%, no expression; ≥1–4%, low expression; ≥5%, high expression); BCL-6, α-catenin, cyclin D1, c-kit and EGFR (0–15%, no expression; 16–50%, low expression; 85–100%, positive expression); β/γ-catenin (0–15%, no expression; 16–50%, low expression; 51–100%, high expression); p53 (<5%, no expression; ≥5–50%, low expression; ≥50%, high expression). Moderate, intermediate, strong and high expressions were rendered as positive expression.

Statistical analysis

Statistical analysis was performed using χ2 analysis. Biomathematical analysis of immunohistochemical data was evaluated using permutation analysis, which analyses information from protein-expression patterns of TMA data and identifies synergistic or antagonistic effects amongst all evaluated proteins (26). Botstein et al (27) describe this method as preserving the original physiological information of the tumor tissue and revealing the different compounds of the tumor samples to the smallest detail (28,29). This combinatorial analysis calculates the ideal precedence of protein-expression coherence; therefore allowing the generation of an overview of differential regulation patterns in different tumor subgroups. A detailed description of this approach and its use in a clinical setting, using TMA data, have been previously described (24,29). Statistical analysis was performed on R version 3.1.3 software (www.r-project.org/), Fortran 95-based program ‘TMAinspiration’ (complex-systems.uni-muenster.de/tma_inspiration.html) and SPSS version 21.0 software (IBM SPSS, Armonk, NY, USA)

Results

CK expression patterns and tumor localization

The expression (%) of the 6 CKs and other biomarkers in the SCC tumors are presented in Table III. Representative images of immunohistochemical staining are presented in Fig. 1.

Table III.

Expression of CKs and other biomarkers in 193 samples of oral squamous cell carcinoma.

Table III.

Expression of CKs and other biomarkers in 193 samples of oral squamous cell carcinoma.

Expression, % of tumors

ProteinNegativePositive
CK153.9   0.5
CK5/6   1.7   98.3
CK8/1833.3   66.7
CK1062.8   37.2
CK14   2.6   97.4
CK1959.9   40.1
α-catenin34.6   65.4
β-catenin15.8   84.2
γ-catenin35.0   65.0
GLUT1   8.5   91.5
Caspase-374.2   25.8
XIAP80.5   19.5
CAIX73.4   26.6
Hsp 7089.3   10.7
C-kit86.4   13.6
p1679.4   20.6
p2128.8   71.2
p2779.8   20.2
p53   0.0100.0
BCL-678.7   21.3
EGFR24.9   75.1
Cyclin D150.6   49.4
HIF-1α42.2   57.8

[i] CK, cytokeratin; GLUT1, glucose transporter 1; XIAP, X-linked inhibitor of apoptosis protein; CAIX, carbonic anhydrase 9; Hsp, heat shock protein; C-kit, mast/stem cell growth factor receptor; BCL-6, B-cell lymphoma-6; EGFR, epidermal growth factor receptor; HIF-1α, hypoxia inducible factor-1α.

Global, but not individual, CK expression in oral SCCs was significantly different between the anatomical localization of the tumor in the floor of the mouth and other localizations (floor of mouth vs. tongue, P=1.6×10−4; floor of mouth vs. other localizations, P=1.3×10−4; Fig. 2A). SCCs located on the floor of the mouth revealed inverse regression lines in contrast to SCCs of other tumor subsites within the oral cavity, including the maxilla, tonsils and buccal region. Expression of CK8/18 and 19 was associated with SCCs of the floor of the mouth, whereas CK1, 10, 8/18 and 19 were equally expressed in all other subsites (Fig. 2A).

CK expression patterns and tumor grade

Significant differences could be observed in global CK expression patterns in association with tumor grade. Regression lines for grade 2 (G2) and 3 (G3) SCCs were similar, but regression lines for grade 1 (G1) SCCs exhibited an inverse behavior compared to G2 and G3 (Fig. 2B).

G1 carcinomas revealed a statistically significant inverse association with G2 and G3 carcinomas concerning the expression of CK19 (P=4.1×10−5 and P=6.9×10−5, respectively). Expression of CK14 and 1 was predominantly observed in G3 SCCs (P=5.1×10−3 and P=0.03, respectively).

CK expression patterns and cell cycle proteins and growth factors

Two patterns of cell cycle proteins expression were observed in association with CK expression. High-molecular weight CK14, 5/6, 1 and 10 exhibited similar regression lines compared with the regression lines exhibited by low-molecular weight CK8/18 and 19. In this molecular pattern p21, due to its extreme position in the regression approach, has the strongest impact in discriminating between the 6 CKs (Fig. 2C). By contrast, EGFR does not play a major regulative role.

CK expression patterns and factors involved in cell motility, apoptosis and cellular stress responses

Similar findings were observed in a second approach for factors involved in cell motility, apoptosis and cellular stress responses. As demonstrated in Fig. 2C, CK14, 5/6, 1 and 10 revealed a contrasting expression compared with CK8/18 and 19. Comparable results were observed for c-kit, α/β/γ-catenin, HIF-1α, GLUT1, CAIX, caspase-3, hsp 70 and XIAP (Fig. 2D). In this second pattern, the discriminating role was primarily performed by γ-catenin and hsp 70.

Discussion

SCC of the oral cavity is associated with a variety of risk factors, including smoking, alcohol abuse, tobacco chewing, oral hygiene and human papilloma virus (HPV) infection (3034). The current treatment protocols are based on a combination of surgery and radio/chemotherapy (35,36). Despite these complex, multidisciplinary treatment regimens, the long term results are unsatisfactory (1,37), the reasons for which are unclear. The underlying biology of SCCs remains poorly understood, and discerning the distinct molecular mechanisms underlying SCC may lead to improvements in treatment strategies.

CKs are the major intermediate filaments of squamous epithelium (38,39), and in different organ systems it has been demonstrated that alterations in CK expression patterns leads to the altered expression of numerous genes and proteins (911,40).

The present study demonstrates that previously described (9,10) prognostically relevant cytokeratin expression patterns are associated with different expression patterns of crucial cellular proteins. Low-molecular weight CK8/18 and 19 expression had opposing patterns to high-molecular weight CK1, 5/6, 10 and 14 with biomarkers involving cell cycle regulation (p21), hypoxic stress (HIF-1α and CAIX) or cellular adhesion (α/γ-catenin). This indicates that CK8/18 and 19 are expressed in poorly-differentiated hypoxic SCC with a higher degree of cell cycle deregulation, whereas CK1, 5/6, 10 and 14 appear to be expressed in well-differentiated cancers with lower hypoxia and cell cycle deregulation. These results are similar to the general observation that low-molecular weight CK8/18 and 19 cytokeratins, whose expression is a hallmark of glandular tissues, are not physiologically expressed in normal squamous epithelium, but may be expressed during carcinogenesis (11,12,41).

These results are similar to previous studies regarding the prognostic significance of these proteins (9,10,19,21). However, the interpretation of these findings is challenging, since it cannot be excluded that these alterations in expression of CKs and the chosen biomarkers are a reflection of tumor progression. By contrast, the present results may allow an alternative interpretation; in invasive breast cancer it was previously demonstrated that the expression of distinct high-molecular weight CKs defines a subgroup of poorly-differentiated breast cancer, representing a unique, independent pathway associated with poor prognosis, which has different responses to various treatment modalities (42,43). Therefore, the present results may be as a result of the existence of different pathways in the pathogenesis of oral SCCs. At present, two pathways characterized by the differential expression of CKs may be distinguished in the present study. In various other tumor entities, such parallel, independent pathogenetic pathways have been reported for colorectal cancer, ovarian cancer and SCCs of the female genital tract, including the vulva and cervix (24,4446).

In addition, the present authors consider that the results support the hypothesis that SCCs of the oral cavity may not be regarded as a homogeneous entity. In the present study, global expression patterns of high- and low-molecular weight CKs exhibit clear differences between G1 and G2/3 SCCs. It may be argued that the alteration in CK expression has been interpreted as a result of tumor dedifferentiation. However, the present authors hypothesize that this may not be true for several reasons. The expression of CK8/18 and 19 has been described in epithelial precursor lesions of oral SCC (10), and therefore does not support the hypothesis that CK8/18 and 19 expression is a late phenomena in carcinogenesis after invasion has occurred. It is also widely accepted that CK expression patterns appear to be highly conserved during tumor progression (8). As a consequence, the existence of CK8/18 and/or 19-positive oral SCC raises fundamental controversies concerning their formal pathogenesis (9,10,12,41). These carcinomas appear to represent an independent pathway rather than being the endstage of a stepwise dedifferentiation of CK8/18/19 negative SCCs. Therefore, it may be postulated that there is a low- and high-grade pathway. A similar concept has also been proposed for squamous intraepithelial neoplasms in the cervix and the vulva (47,48), which is associated with various HPV subtypes (44).

The oral cavity is a highly complex anatomical region, emerging from various branchial arches that are associated with an aggregation of mesenchyme, ectoderm and endoderm (4951); however, to what extent tumor localization has an effect on tumor biology and prognosis remains to be demonstrated. The present results reveal that differences appear to exist between SCCs on the floor of the mouth and SCCs of other anatomical subsites within the oral cavity, as observed by previous studies (5,21,52,53). In the present study, SCCs of the floor of the mouth had an increased expressed of CK8/18 and 19. The number of tumors investigated in the present study was too small to allow for a definite conclusion; however the present results reveal that complex interactions appear to exist between the expression of CKs and other major cellular proteins, as well as tumor grade and anatomical sublocalization of SCCs of the oral cavity.

In summary, the present study analyzed low- and high-molecular weight CK expression in association with the expression of various other major cellular proteins using a sophisticated biomathematical algorithm in a series of 193 SCCs of the oral cavity. The present study provided evidence of the existence of two pathogenetic pathways characterized by the expression of low- and high-molecular weight CKs in oral SCC. These results provide evidence for additional investigation concerning the pathways identified and provide improved understanding of oral tumor biology.

References

1 

Scully C and Bagan J: Oral squamous cell carcinoma overview. Oral Oncol. 45:301–308. 2009. View Article : Google Scholar : PubMed/NCBI

2 

American Cancer Society: Cancer Facts & Figures 2015. American Cancer Society. Altanta, GA: 2015.

3 

Cancer Research UK: Oral cancer incidence statistics. http://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/oral-cancer/incidence.s

4 

D'Cruz AK, Vaish R, Kapre N, Dandekar M, Gupta S, Hawaldar R, Agarwal JP, Pantvaidya G, Chaukar D, Deshmukh A, et al: Head and Neck Disease Management Group: Elective versus therapeutic neck dissection in node-negative oral cancer. N Engl J Med. 373:521–529. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Yamauchi K, Fujioka Y, Kogashiwa Y and Kohno N: Quantitative expression study of four cytokeratins and p63 in squamous cell carcinoma of the tongue: Suitability for sentinel node navigation surgery using one-step nucleic acid amplification. J Clin Pathol. 64:875–879. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Slaughter DP, Southwick HW and Smejkal W: Field cancerization in oral stratified squamous epithelium; clinical implications of multicentric origin. Cancer. 6:963–968. 1953. View Article : Google Scholar : PubMed/NCBI

7 

Chu PG and Weiss LM: Keratin expression in human tissues and neoplasms. Histopathology. 40:403–439. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Moll R, Franke WW, Schiller DL, Geiger B and Krepler R: The catalog of human cytokeratins: Patterns of expression in normal epithelia, tumors and cultured cells. Cell. 31:11–24. 1982. View Article : Google Scholar : PubMed/NCBI

9 

Fillies T, Werkmeister R, Packeisen J, Brandt B, Morin P, Weingart D, Joos U and Buerger H: Cytokeratin 8/18 expression indicates a poor prognosis in squamous cell carcinomas of the oral cavity. BMC Cancer. 6:102006. View Article : Google Scholar : PubMed/NCBI

10 

Fillies T, Jogschies M, Kleinheinz J, Brandt B, Joos U and Buerger H: Cytokeratin alteration in oral leukoplakia and oral squamous cell carcinoma. Oncol Rep. 18:639–643. 2007.PubMed/NCBI

11 

Alam H, Gangadaran P, Bhate AV, Chaukar DA, Sawant SS, Tiwari R, Bobade J, Kannan S, D'Cruz AK, Kane S and Vaidya MM: Loss of keratin 8 phosphorylation leads to increased tumor progression and correlates with clinico-pathological parameters of OSCC patients. PLoS One. 6:e277672011. View Article : Google Scholar : PubMed/NCBI

12 

Gires O, Mack B, Rauch J and Matthias C: CK8 correlates with malignancy in leukoplakia and carcinomas of the head and neck. Biochem Biophys Res Commun. 343:252–259. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Raul U, Sawant S, Dange P, Kalraiya R, Ingle A and Vaidya M: Implications of cytokeratin 8/18 filament formation in stratified epithelial cells: Induction of transformed phenotype. Int J Cancer. 111:662–668. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Alam H, Kundu ST, Dalal SN and Vaidya MM: Loss of keratins 8 and 18 leads to alterations in α6β4-integrin-mediated signalling and decreased neoplastic progression in an oral-tumour-derived cell line. J Cell Sci. 124:2096–2106. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Lindberg K and Rheinwald JG: Suprabasal 40 kd keratin (K19) expression as an immunohistologic marker of premalignancy in oral epithelium. Am J Pathol. 134:89–98. 1989.PubMed/NCBI

16 

Paramio JM and Jorcano JL: Beyond structure: Do intermediate filaments modulate cell signalling? Bioessays. 24:836–844. 2002. View Article : Google Scholar : PubMed/NCBI

17 

World Medical Association: World Medical Association Declaration of Helsinki. Ethical principles for medical research involving human subjects. Bull World Health Organ. 79:373–374. 2001.PubMed/NCBI

18 

Barton A: Handbook for good clinical research practice (GCP): Guidance forsimplementation. J Epidemiol Community Health. 61:5592007. View Article : Google Scholar

19 

Fillies T, Woltering M, Brandt B, Van Diest JP, Werkmeister R, Joos U and Buerger H: Cell cycle regulating proteins p21 and p27 in prognosis of oral squamous cell carcinomas. Oncol Rep. 17:355–359. 2007.PubMed/NCBI

20 

Fillies T, Werkmeister R, van Diest PJ, Brandt B, Joos U and Buerger H: HIF1-alpha overexpression indicates a good prognosis in early stage squamous cell carcinomas of the oral floor. BMC Cancer. 5:842005. View Article : Google Scholar : PubMed/NCBI

21 

Fillies T, Buerger H, Gaertner C, August C, Brandt B, Joos U and Werkmeister R: Catenin expression in T1/2 carcinomas of the floor of the mouth. Int J Oral Maxillofac Surg. 34:907–911. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Sobin LH, Gospodarowicz MK and Wittekind C: UICC TNM Classification of Malignant Tumours (7th). Wiley-Blackwell. Hoboken, NJ: 2009.

23 

Packeisen J, Buerger H, Krech R and Boecker W: Tissue microarrays: A new approach for quality control in immunohistochemistry. J Clin Pathol. 55:613–615. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Schymik B, Buerger H, Krämer A, Voss U, van der Groep P, Meinerz W, van Diest PJ and Korsching E: Is there ‘progression through grade’ in ductal invasive breast cancer? Breast Cancer Res Treat. 135:693–703. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Helms MW, Packeisen J, August C, Schittek B, Boecker W, Brandt BH and Buerger H: First evidence supporting a potential role for the BMP/SMAD pathway in the progression of oestrogen receptor-positive breast cancer. J Pathol. 206:366–376. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Packeisen J, Korsching E, Herbst H, Boecker W and Buerger H: Demystified…tissue microarray technology. Mol Pathol. 56:198–204. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Botstein D and Fink GR: Yeast: An experimental organism for 21st Century biology. Genetics. 189:695–704. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Vidal M, Cusick ME and Barabási AL: Interactome networks and human disease. Cell. 144:986–998. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Buerger H, Boecker F, Packeisen J, Agelopoulos K, Poos K, Nadler W and Korsching E: Analyzing the basic principles of tissue microarray data measuring the cooperative phenomena of marker proteins in invasive breast cancer. Open Access Bioinformatics. 5:1–21. 2013.

30 

Warnakulasuriya S: Global epidemiology of oral and oropharyngeal cancer. Oral Oncol. 45:309–316. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Petti S: Lifestyle risk factors for oral cancer. Oral Oncol. 45:340–350. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Moergel M, Kammerer P, Kasaj A, Armouti E, Alshihri A, Weyer V and Al-Nawas B: Chronic periodontitis and its possible association with oral squamous cell carcinoma - a retrospective case control study. Head Face Med. 9:392013. View Article : Google Scholar : PubMed/NCBI

33 

Feller L, Wood NH, Khammissa RA and Lemmer J: Human papillomavirus-mediated carcinogenesis and HPV-associated oral and oropharyngeal squamous cell carcinoma. Part 1: Human papillomavirus-mediated carcinogenesis. Head Face Med. 6:142010. View Article : Google Scholar : PubMed/NCBI

34 

Feller L, Wood NH, Khammissa RA and Lemmer J: Human papillomavirus-mediated carcinogenesis and HPV-associated oral and oropharyngeal squamous cell carcinoma. Part 2: Human papillomavirus associated oral and oropharyngeal squamous cell carcinoma. Head Face Med. 6:152010. View Article : Google Scholar : PubMed/NCBI

35 

AWMF and GAOOAM: Surgery Guidelines for Oncology, S3: Guideline for diagnostics and treatment of oral cancer. AWMF-Register-Number (007–100O, 2012L). 119:2012.(In German).

36 

Frerich B: Standard therapy of oral squamous epithelial carcinoma. Onkologe. 16:527–536. 2010.(In German). View Article : Google Scholar

37 

Institut RK and GEKID: Cancer in Germany 2007/2008. Journal. 137:2012.(In German).

38 

Steinert PM, Idler WW and Zimmerman SB: Self-assembly of bovine epidermal keratin filaments in vitro. J Mol Biol. 108:547–567. 1976. View Article : Google Scholar : PubMed/NCBI

39 

Romano V, Bosco P, Rocchi M, Costa G, Leube RE, Franke WW and Romeo G: Chromosomal assignments of human type I and type II cytokeratin genes to different chromosomes. Cytogenet Cell Genet. 48:148–151. 1988. View Article : Google Scholar : PubMed/NCBI

40 

Thiel UJ, Feltens R, Adryan B, Gieringer R, Brochhausen C, Schuon R, Fillies T, Grus F, Mann WJ and Brieger J: Analysis of differentially expressed proteins in oral squamous cell carcinoma by MALDI-TOF MS. J Oral Pathol Med. 40:369–379. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Zhong LP, Chen WT, Zhang CP and Zhang ZY: Increased CK19 expression correlated with pathologic differentiation grade and prognosis in oral squamous cell carcinoma patients. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 104:377–384. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Korsching E, Packeisen J, Agelopoulos K, Eisenacher M, Voss R, Isola J, van Diest PJ, Brandt B, Boecker W and Buerger H: Cytogenetic alterations and cytokeratin expression patterns in breast cancer: Integrating a new model of breast differentiation into cytogenetic pathways of breast carcinogenesis. Lab Invest. 82:1525–1533. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Korsching E, Jeffrey SS, Meinerz W, Decker T, Boecker W and Buerger H: Basal carcinoma of the breast revisited: An old entity with new interpretations. J Clin Pathol. 61:553–560. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Schiffman M, Castle PE, Jeronimo J, Rodriguez AC and Wacholder S: Human papillomavirus and cervical cancer. Lancet. 370:890–907. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Kurman RJ and Shih Ie M: Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer-shifting the paradigm. Hum Pathol. 42:918–931. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Rajagopalan H, Nowak MA, Vogelstein B and Lengauer C: The significance of unstable chromosomes in colorectal cancer. Nat Rev Cancer. 3:695–701. 2003. View Article : Google Scholar : PubMed/NCBI

47 

Lee BH, Roh S, Kim YI, Lee A and Kim SY: Difference of genome-wide copy number alterations between high-grade squamous intraepithelial lesions and squamous cell carcinomas of the uterine cervix. Korean J Pathol. 46:123–130. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Tornesello ML, Buonaguro L, Giorgi-Rossi P and Buonaguro FM: Viral and cellular biomarkers in the diagnosis of cervical intraepithelial neoplasia and cancer. Biomed Res Int. 2013:5196192013. View Article : Google Scholar : PubMed/NCBI

49 

Helms JA, Cordero D and Tapadia MD: New insights into craniofacial morphogenesis. Development. 132:851–861. 2005. View Article : Google Scholar : PubMed/NCBI

50 

Diewert VM: Development of human craniofacial morphology during the late embryonic and early fetal periods. Am J Orthod. 88:64–76. 1985. View Article : Google Scholar : PubMed/NCBI

51 

Moore KL and Persuad TVN: The Developing Human: Clinicall Oriented Embryology (8th). Saunders/Elsevier. Philadelphia, PA: 2007.

52 

Löfdahl HE, Du J, Näsman A, Andersson E, Rubio CA, Lu Y, Ramqvist T, Dalianis T, Lagergren J and Dahlstrand H: Prevalence of human papillomavirus (HPV) in oesophageal squamous cell carcinoma in relation to anatomical site of the tumour. PLoS One. 7:e465382012. View Article : Google Scholar : PubMed/NCBI

53 

Li R, Koch WM, Fakhry C and Gourin CG: Distinct epidemiologic characteristics of oral tongue cancer patients. Otolaryngol Head Neck Surg. 148:792–796. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 12 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Frohwitter G, Buerger H, van Diest PJ, Korsching E, Kleinheinz J and Fillies T: Cytokeratin and protein expression patterns in squamous cell carcinoma of the oral cavity provide evidence for two distinct pathogenetic pathways. Oncol Lett 12: 107-113, 2016
APA
Frohwitter, G., Buerger, H., van Diest, P.J., Korsching, E., Kleinheinz, J., & Fillies, T. (2016). Cytokeratin and protein expression patterns in squamous cell carcinoma of the oral cavity provide evidence for two distinct pathogenetic pathways. Oncology Letters, 12, 107-113. https://doi.org/10.3892/ol.2016.4588
MLA
Frohwitter, G., Buerger, H., van Diest, P. J., Korsching, E., Kleinheinz, J., Fillies, T."Cytokeratin and protein expression patterns in squamous cell carcinoma of the oral cavity provide evidence for two distinct pathogenetic pathways". Oncology Letters 12.1 (2016): 107-113.
Chicago
Frohwitter, G., Buerger, H., van Diest, P. J., Korsching, E., Kleinheinz, J., Fillies, T."Cytokeratin and protein expression patterns in squamous cell carcinoma of the oral cavity provide evidence for two distinct pathogenetic pathways". Oncology Letters 12, no. 1 (2016): 107-113. https://doi.org/10.3892/ol.2016.4588