You are on page 1of 8

Oral Oncology 50 (2014) 840–847

Contents lists available at ScienceDirect

Oral Oncology
journal homepage: www.elsevier.com/locate/oraloncology

Detection and typing of Human Papillomaviruses (HPV) in malignant,


dysplastic, nondysplastic and normal oral epithelium by nested
Polymerase Chain Reaction, immunohistochemistry and transitional
electron microscopy in patients of Northern Greece
E. Blioumi a,⇑, D. Chatzidimitriou b, Ch. Pazartzi c, Th. Katopodi d, G. Tzimagiorgis e,
E.-N. Emmanouil-Nikoloussi f, A. Markoloulos a, C. Kalekou a, N. Lazaridis g, E. Diza h, D. Antoniades a
a
Department of Stomatology, School of Dentistry, Aristotle University of Thessaloniki, Greece
b
B’ Department of Microbiology, School of Medicine, Aristotle University of Thessaloniki, Greece
c
Department of Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Greece
d
Department of Biology, School of Medicine, Aristotle University of Thessaloniki, Greece
e
Department of Biochemistry, School of Medicine, Aristotle University of Thessaloniki, Greece
f
Department of Histology, Embryology and Anthropology, School of Medicine, Aristotle University of Thessaloniki, Greece
g
Department of Orofacial Surgery, School of Dentistry, Aristotle University of Thessaloniki, Greece
h
Department of Microbiology, AHEPA University General Hospital of Thessaloniki, Greece

a r t i c l e i n f o s u m m a r y

Article history: Objectives: To evaluate the role of HPV in oral carcinogenesis, we examined the prevalence of HPV in
Received 24 February 2014 malignant, potentially malignant and normal oral epithelium and studied the relation of HPV prevalence
Received in revised form 24 May 2014 with other factors obtained from the patient’s records.
Accepted 12 June 2014
Materials and methods: Our material consisted of 291 tissue specimens from 258 individuals. From every
Available online 17 July 2014
individual formalin fixed and paraffin embedded tissues were examined by nested Polymerase Chain
Reaction (NPCR) for the detection of HPV DNA and by immunohistochemistry (IHC) for the in situ detec-
Keywords:
tion of HPV L1 protein. Positive PCR products were sequenced in order to type HPVs. Also 33 fresh tissues
Oral cancer
Human Papillomavirus (HPV)
were obtained, fixed and used to detect HPV particles by transitional electron microscopy (TEM).
Oral dysplasia Results: HPV was detected in 32.9% of the tissue specimens by NPCR, in 4.7% by immunohistochemistry
Polymerase Chain Reaction and in 28.1% by TEM. In detail, by nested PCR HPV L1 DNA was detected in 40% of normal tissues, 40% of
Immunohistochemistry fibromas, 35.8% of non-dysplastic leukoplakias, 31.6% of dysplastic leukoplakias and 22.2% of oral squa-
Electron microscopy mous cell carcinomas. The HPV viral load of 96.5% of the samples was very low (1 viral copy per 102–104
cells). HPV16 prevails in all histological groups in 89–100%.
Conclusion: We conclude that HPV does not seem, from the specific sample examined, to play a substan-
tial role in oral carcinogenesis. However, it cannot be excluded that HPV could be involved in oral
carcinogenesis only in cases with high viral load or at early stages of carcinogenesis possibly through
the hit-and-run mechanism.
Ó 2014 Elsevier Ltd. All rights reserved.

Introduction Specifically in Greece, 399 new oral cancers and 125 deaths come
up every year [2]. Although more than 40 different types of neo-
Oral cancer is the twelfth most frequent malignancy in the plasms grow up in the oral cavity, squamous cell carcinomas
world, representing almost 2.5% of malignant tumors [1]. The prev- account for 85–94% of all oral malignancies [3–6]. The etiology of
alence and incidence of oral cancer in 2002 worldwide was esti- oral cancer seems to be multifactorial. A number of extrinsic
mated about 741,000 and 274,289 cases, respectively [1]. factors, such as smoking, alcohol consumption, tobacco chewing,
ultraviolet radiation and several viruses, seem to play role in oral
⇑ Corresponding author. Present address: Theophrastou 3, P.S. 55132, Thessalo- carcinogenesis. Intrinsic agents, including oncogenes, tumor
niki, Greece. Tel.: +30 2310 452576; fax: +30 2313 304467, mobile: +30 suppression genes, hormones, immunosuppresion and enzymatic
6942577911. mechanisms also seem to be involved in the initiation and
E-mail address: eblioumi@yahoo.com (E. Blioumi).

http://dx.doi.org/10.1016/j.oraloncology.2014.06.011
1368-8375/Ó 2014 Elsevier Ltd. All rights reserved.
E. Blioumi et al. / Oral Oncology 50 (2014) 840–847 841

progression of oral carcinomas [3,7]. Moreover, many of the squa- was used mostly for the evaluation of HPV prevalence and the sta-
mous cell carcinomas develop on the ground of potentially malig- tistic analysis, whereas the other two techniques offered informa-
nant disorders (PMDs) [7]. Oral leukoplakia is the most frequent tion about the inter-epithelial and inter-cellular location of HPVs.
PMD, representing 85% of these lesions [4].
HPV is a small, epitheliotropic, DNA virus, which belongs to a Nested Polymerase Chain Reaction
large family of viruses, the Papillomaviridae, containing more than
100 different human types [8–11]. PV genome consists of 7900 Three to five adjacent 10 lm sections of each paraffin embed-
base pairs of closed-circular double-stranded DNA, containing 8 ded tissue were collected in sterile 1.5 mL microcentrifuge tube.
open reading frames. Some of HPV types, such as 16, 18, 31, 33, DNA was extracted with the Qiagen QIAamp DNA extraction
35, 39, 45, 51, 52, 56, 58, 59, 66, are carcinogenic to humans and method. To avoid cross-contamination at any step of the proce-
are characterized as ‘high risk types’. Moreover high risk types dure, paraffin-embedded compounds were cut between samples
are 26, 30, 34, 53, 67, 68, 69, 70, 73, 82 [8]. and subsequently subjected to DNA extraction and PCR analysis.
The first time that a relationship between HPV and head and DNA’s concentration and purity were estimated using 260 nm
neck cancer was mentioned was in 1980 [12–15]. Since then, the and 280 nm spectrophotometry (Eppendorf Centrifuge).
findings of HPV participation in the onset and the progression of Strict procedures were followed to avoid false-positive reac-
oral carcinomas differ among the researchers. Specifically, the tions due to contamination. DNA extraction and PCR reactions
prevalence of HPV in oral carcinomas varies between 0% and were carried out in separate rooms, sterilized with UV radiation
100% [16–26], and in the normal oral epithelium varies between both before and after reactions for 10 min. All surfaces were decon-
0% and 81% [27–35]. Most of the reports have selectively studied taminated with a special solution (DNA AWAY, Molecular Bioprod-
a certain type of oral lesions. To evaluate the role of HPV in oral ucts). Reagents were prepared and aliquoted, filtered pipette tips
carcinogenesis, we comprehensively investigated HPV infection and one-use gloves were used. Every PCR reaction was applied to
in 258 specimen of oral mucosa, representing the whole spectrum 10 samples, one positive control (DNA extracted from cervical can-
of normal, hyperplastic, potentially malignant and malignant cer) and one or more negative controls (distilled sterile water).
lesions, using a combination of three different techniques: To check the integrity of the extracted DNA and the absence of
nested-PCR, immunohistochemistry and transitional electronic PCR inhibitors a 110 bp segment of human b-globin gene was
microscopy, and studied the relation of HPV prevalence with other amplified by PCO3/PCO4 primers [37,38].
factors obtained from the patient’s records, such as histologic Detection of HPV sequences was performed by two sets of con-
diagnosis, gender, age, anatomic location, smoking and alcohol sensus primers, MY09/MY11 and GP5+/GP6+, which amplify a
drinking. 450 bp and an internal 150 bp region, respectively, in the highly
conserved L1 HPV gene. In the first amplification step, each reac-
tion of 50 ll contained in final concentrations: 1X PCR buffer,
Material
2 mM MgCl2, 200 lM of each dNTP, 1 lM of each primer MY09
(50 -CGTCCMARRGGAWACTGATC-30 ) and MY11 (50 -GCMCAGGGW-
Our material consisted of 291 tissue specimens from 258 indi-
CATAAYAATGG-30 , M: A + C, R: A + G, W: A + T, Y: C + T), 1.5 units
viduals. From every individual, formalin fixed and paraffin embed-
Taq DNA polymerase and 20 ll of DNA (600–1000 mg). The mix-
ded tissues (FFPE) were examined by nested Polymerase Chain
ture was subjected to a 5 min denaturation at 94 °C, then 40 cycles
Reaction and immunohistochemistry. Fresh tissues were obtained
of amplification at 94 °C for 1 min, 55 °C for 1 min and 72 °C for
and examined by transitional electron microscopy from 33 of the
1 min, in addition to a 5-min extension at 72 °C [12].
above patients.
2 ll of the above PCR product were used in the second amplifi-
The FFPE tissues consisted of 63 oral squamous cell carcinomas
cation step of the PCR reaction. Each reaction of 50 ll contained in
(OSSC), 57 oral leukoplakias with dysplasia (OLD), 53 leukoplakias
final concentrations: 1X PCR buffer, 3.5 mM MgCl2, 200 lM of each
without dysplasia (OL), 50 fibromas with normal epithelium (F)
dNTP, 0,5 lM of each primer GP5+ (50 -TTTGTTACTGTGGTAGATAC-
and 35 normal mucosas (N). The pathologic tissues were taken
TAC-30 ) and GP6+ (50 -GAAAAATAAACTGTAAATCATATTC-30 ), 1.25
from the routine files (1970–2009) of the Department of Stomatol-
units Taq DNA polymerase and 2 ll of the first PCR product. The
ogy, School of Dentistry, Aristotle University of Thessaloniki,
mixture was subjected to a 5 min denaturation at 95 °C, then
Greece, whereas the normal tissues were taken from the normal
40 cycles of amplification at 94 °C for 1 min, 40 °C for 2 min and
excision borders of healthy subjects during fibroma, epulis, muco-
72 °C for 2 min, in addition to a 3-min extension at 72 °C [11,37].
cele biopsies or third molar extractions in the Department of Sto-
After amplification, the reaction products were electrophoresed
matology or Maxillofacial Surgery (2008–2009).
on 2% agarose and visualized by ethidium bromide.
The 33 fresh oral specimens were collected during the biopsy of
To type the HPV, the final nested PCR product was further puri-
squamous cell carcinomas or hyperplastic or dysplastic lesions.
fied with PureLink™ purification procedure (Invitrogen Corpora-
They consisted of 15 OSSCs, 8 OLDs, 9 OLs and 1 fibroma. For each
tion, UK) and got sequenced with the Sanger method in
case, patient’s gender and age, smoking or alcohol drinking, lesion’s
automatic analyzer (CEQ 8000, Beckman Coulter, GENOTYPOS,
location, histological diagnosis and histological grading of dyspla-
Athens, Greece). Chromograms were analyzed with Chromas pro-
sia or cancer differentiation, according to Smith and Pindborg’s cri-
gram. The DNA sequences were aligned and compared to
teria [36], were recorded.
sequences of known HPV types, found in NCBI Genbank (NCBI,
National Institute of Health, Bethesda, MD, USA), with the use of
Methods NCBI Blast program (http://blast.ncbi.hlm.nih.gov/Blast.cgi).
HPV positive specimens were subjected to a type-specific PCR
Three different techniques were applied on the specimens for the amplification of a 120 bp segment of the HPV16 E6 gene.
collected: nested Polymerase Chain Reaction was applied on 258 Each reaction of 50 ll contained in final concentrations: 1X PCR
tissue specimens for the detection of HPV DNA, immunohisto- buffer, 1.5 mM MgCl2, 200 lM of each dNTP, 1 lM of each primer
chemistry was applied on 258 tissue specimens for the in situ F (50 -TCAAAAGCCACTGTGTCCTG-30 ) and R (50 -CGTGTTCTTGAT-
detection of HPV L1 protein and transitional electron microscopy GATCTGCA-30 ), 2 units Taq DNA polymerase and 10 ll of sample
was applied on the 33 fresh tissue specimens for detection of viral DNA (300–800 mg). The mixture was subjected to a 2 min denatur-
particles. Nested-PCR, the most sensitive and specific technique, ation at 95 °C, then 40 cycles of amplification at 94 °C for 1 min,
842 E. Blioumi et al. / Oral Oncology 50 (2014) 840–847

53 °C for 1 min and 72 °C for 2 min, in addition to a 7-min exten- method or Monte Carlo method [39]. To compare percentages,
sion at 72 °C [34]. we used Fisher’s Exact Test [40]. Pearson’s linear correlation coef-
The sensitivity of the above PCR protocols was determined ficient was used to correlate quantity parameters. P values lower
using serially diluted DNA extracted from cervical cancer. than 0.05 were considered significant.

Immunohistochemistry
Results
Three adjacent histological sections of each sample were incu-
bated in 60 °C for 1–2 h, dewaxed in xelene and rehydrated 258 Samples were derived from the buccal mucosa (30.2%), the
through alcohol to water. The antigens were retrieved by dipping tongue (24.0%), the gingival and alveolar mucosa (19.4%), the lips
the sections in citrate buffer solution and warming them in micro- (14%), the floor of the mouth (8.5%) and the palate (3.5%). No sam-
waves for 10 min at the highest temperature. The sections were ples were selected from the posterior oropharyngeal sites since
washed with Tris Buffer Solution (TBS) and incubated with 0.3% patients with such lesions are mostly referred to ENT or oral and
hydrogen peroxide in methanol for 30 min in dark to quench the maxillofacial surgeons. 127 samples were derived from male
endoxegous peroxidase activity. Non-specific background staining patients and 131 from female patients. The age of patients ranged
was reduced by incubating the sections with fetal bovine serum from 12 to 94 years, with a mean age of 53.1 years (mean devia-
(FBS) for 1 h. The sections were then incubated overnight at 4 °C tion: 16.6 years). 57.8% of samples were derived from smokers or
with the antibodies; the first section, with an antibody against ex-smokers (>1 cigarette per day), and 15.5% of samples were
HPV L1 capsid protein (Polyclonal rabbit Anti-papillomavirus anti- derived from alcohol consumers (>1 portion per day). Cancerous
body (Predilute Antibody, Ready-to-use, 08-0075, Zymed Laborato- lesions were classified into well-differentiated (37), moderately
ries, Invitrogen Immunodetection, USA) in 7 mg/ml concentration, differentiated (21) and poorly differentiated (5), according to Smith
the second section with a negative antibody (N-universal Negative and Pindborg’s criteria. Dysplastic lesions were classified into mild
control rabbit, code N1699, DakoCytomation, Denmark) for nega- epithelial dysplasia (34 patients), moderate dysplasia (6), severe
tive control and the third section with monoclonal mouse antibody dysplasia (10) and in situ carcinoma (7).
against cytokeratin (Monoclonal mouse anti-human cytokeratin, The results of the different techniques to demonstrate HPV
MNF clone, code M 0821, DakoCytomation, Denmark) for positive revealed that 90 out of 258 patients were HPV positive (35.9%)
control of the quality and the antigenicity of the tissue. The sec- by at least one technique. All techniques proved to be reliable as
tions were then washed with TBS and incubated with for 60 min all control specimens gave consistent results irrespective of the
with the secondary antibody and peroxidase (ChemMate™ Dako technique employed.
Envision/HRP, Rabbit/Mouse, DakoCytomation, Denmark) in room B-globin was detected in all tissues studied. NPCR analysis
temperature. Finally, the sections were incubated with DAB (diam- detected the presence of HPV genome in 85/258 specimen
inobenzidine) diluted 1:50 in hydrogen peroxide for 1.5–2 min, (32.9%). Of the positive specimens, 3.5% were identified by the first
stained with haematoxylin, dehydrated and mounted. The tech- step and 96.5% by the second step of NPCR assay (Fig. 1). By the
nique was further controlled by staining specimen with known first step, HPV was detected in 2 carcinomas with moderate differ-
HPV (oral wart, cervical epithelial dysplasia) and by staining a entiation and 1 in leukoplakia with mild dysplasia. Two samples
specimen without any history of HPV infection (normal minor sal- were derived from the tongue and one from the buccal mucosa.
ivary gland). All specimens were read by a trained pathologist HPV DNA was detected in 22.2% of OSCCs, 31.6% of dysplastic
without information on the origin of the specimen so as to avoid leukoplakias, 35.8% of non-dysplastic leukoplakias, 40% of fibromas
bias results. and 40% of normal oral epithelia. Statistical analysis with X2 test in
Positive staining was registered the localized dark brown inter- the whole sample revealed a negative, weak and statistically signif-
nuclear or para-nuclear staining of the epithelium, whereas dis- icant relation between age and HPV DNA detection (p = 0.017). HPV
perse cytoplasmic staining was considered as non-specific. DNA was detected more frequently in younger individuals
(653 years old). No significant relation was detected between
Transitional electron microscopy HPV prevalence and gender (p = 1.000), histological diagnosis
(p = 0.247), histological grading (p = 0.469), location (p = 0.317),
Immediately after excision, a 3 mm-diameter specimen was cut smoking (p = 0.182) or alcohol drinking p = 0.133). In oral carcino-
into slices of less than 1 mm thickness and fixed by immersion in mas, a statistically significant relation was detected between HPV
3% gluteraldehyde in phosphate buffer (PH:7.4) for 1 h at 4 °C. L1 DNA positivity and male gender (p = 0.023) and location on
After brief washing with phosphate buffer solution, the specimens the tongue (p = 0.059, a = 0.1). In oral leukoplakias with dysplasia,
were postfixed with 2% osmium tetroxide for 1–1.5 h at 4 °C. They HPV detection was significantly higher in female patients
were then blocked with 1% uranyl acetate for 12–14 h, dehydrated (p = 0.012) and non-alcohol drinkers (p = 0.022). In normal sam-
through a graded ethanol series, and embedded in EPON 812 (Ser- ples, fibromas and benign leukoplakias, no significant relation
na Electrophoresis GmbH, Heidelberg). Ultra thin sections were cut was found between HPV and the above parameters (Table 1).
using a diamond knife on a Leica EM UCG, stained with Reynold’s The typing of the HPV positive samples indicated HPV16 in
stain and examined with a JEOL Electron Microscope, JEM-2000 88.6%, HPV13 in 3%, HPV56 in 3%, double infection of HPV13/16
FX, at 80 kV. in 3%, double infection of HPV16/56 in 1% and HPV66 in 1.5%.
Viral particles were detected as spherical particles 50–60 nm in Totally, 97.1% of positive samples contained high risk HPV types
diameter, sparsely distributed or forming clusters, localized in the (HPV16, 56, 66). The distribution of HPV types in every patient
nucleus or in the cytoplasm or in the narrow extracellular space group is shown in table 2. Double infections were detected by
between the epithelial cells of oral mucosa. the combination of the results of sequencing and the results of
HPV16 E6 PCR. E6 was detected in 69.7% of HPV16 L1 positive spec-
Statistical analysis imens. No significant relation was found between histological diag-
nosis and HPV16 E6 detection (p = 0.988).
Statistical analysis was performed by X2 test for the study of Both MY09/MY11, GP5+/GP6+ and HPV16 E6 protocols detected
correlation between categorical quality parameters using SPSS a minimum of 0.78 ng of total (human and HPV) DNA in a 50 ll
15.0 software package. P-value was calculated either with Exact reaction, having a sensitivity of 1 HPV copy per 102 cells. Nested
E. Blioumi et al. / Oral Oncology 50 (2014) 840–847 843

Figure 1. PCR products of biopsy specimens. (A) PCR products with external MY09/MY11 primers of HPV L1 DNA after staining with ethidium bromide and photography
under UV illumination. DNA band of 450 bp indicated the presence of HPV DNA. Lanes 1–6: oral carcinoma tissues, NCTL: negative control, POS: positive control, Lad: 100 bp
DNA Ladder (molecular weight marker). (B) PCR products with internal GP5+/GP6+ primers of HPV L1 DNA. DNA band of 150 bp indicated the presence of HPV DNA. Lanes 1–
10: oral carcinoma tissues, NCTL: negative control, POS: positive control, Lad: 100 bp DNA Ladder (molecular weight marker).

Table 1
HPV prevalence with nested PCR and study of the associations of HPV DNA with histological diagnosis, gender, age, location, smoking and alcohol drinking in the total sample
(n = 258).

Parameters Categories HPV () HPV (+) Total Association p


Diagnosis Normal 21 14 (40.0%) 35
Fibromas 30 20 (40.0%) 50
Leukoplakias 34 19 (35.8%) 53
Dysplasias 39 18 (31.6%) 57
Carcinomas 49 14 (22.2%) 63
173 85 258 Nonsignificant p = 0.247
Gender Male 85 41 (32.3%) 127
Female 88 43 (32.8%) 131
173 85 258 Nonsignificant p = 1.000
Age 653 73 50 (40.7%) 123
>53 100 35 (25.9%) 135
173 85 258 Significant p = 0.017
Location Tongue 39 23 (37.1%) 62
Floor of mouth 17 5 (22.7%) 22
Buccal 52 26 (33.3%) 78
Alveolar mucosa. 30 20 (40.0%) 50
Lips 28 8 (22.2%) 36
Palate 7 2 (22.2%) 9
Oropharyngeal 0 1 (100%) 1
173 85 258 Nonsignificant p = 0.317
Smoking No 105 44 (29.5%) 149
Yes 68 41 (37.6%) 109
173 85 258 Nonsignificant p = 0.182
Alcohol No 144 77 (34.8%) 221
Yes 29 8 (21.6%) 37
173 85 258 Nonsignificant p = 0.133
Histological grading No dysplasia 85 53 (38.4%) 138
Mild dysplasia 23 11 (32.4%) 34
Moderate dysplasia 3 3 (50.0%) 6
Severe Dysplasia 8 2 (20.0%) 10
In situ carcinoma 5 2 (28.6%) 7
Well-differentiated 29 8 (21.6%) 37
Moderately-differentiated 16 5 (23.8%) 21
Poorly-differentiated 4 1 (20.0%) 5
173 85 258 Nonsignificant p = 0.469

PCR protocol detected a minimum of 0.78 pg of total DNA, namely HPV L1 antigen was detected in 12 out of 258 samples (4.7%) by
NPCR was 100 times more sensitive than the above three PCR pro- immunohistochemistry. Positive stain was localized inter- or para-
tocols with a sensitivity of 1 viral copy per 104 cells. nuclearly, in few epithelial neoplastic or dysplastic cells, often
844 E. Blioumi et al. / Oral Oncology 50 (2014) 840–847

Table 2
Distribution of HPV types in patients’ groups after sequencing.

Group HPV types


HPV16 HPV13 HPV56 HPV66 HPV13/16 HPV16/56 Total
Normal 11 1 12
Fibromas 15 1 1 2 1 20
Benign leukoplakias 17 1 18
Dysplasias 8 1 9
Carcinomas 11 11
Total 62 2 1 1 2 2 70

separate in intermediate (spinous) layer and seldom in clusters, lesions. The disturbed tissue has higher risk of progression into a
rete ridges, or in cells with koilocytic appearance (Fig. 2). HPV L1 malignant phenotype, with the later stages being independent of
antigen was detected in 0% of normal mucosa, fibromas and leuko- HPV [47,48,52,53].
plakias without dysplasia, and 1.8% of dysplastic leukoplakias and HPV antigens or particles were detected, disperse or in clusters,
17.5% of OSCCs. in few and isolated oral epithelial cells, which may indicate a local-
By transitional electron microscopy, viral particles were ized colonization of HPV in oral epithelium, without any clinical
observed in 9 out of 33 specimens. Specifically, no particles were significance for malignant transformation [54]. Also, para-nuclear
found in fibromas and non-dysplastic leukoplakias, whereas they detection of HPV antigens or particles could be attributed to frag-
were detected in 37.5% of dysplastic leukoplakias and 40% in mentation of infected nuclei and release of virus particles and
OSCCs. HPV particles were spherical, 50 nm in diameter and were virus- associated proteins, whereas cytoplasmic stain may indicate
localized mainly in the nucleus of epithelial cells, or in the cyto- non-specific reactivity of cytoplasmic substances with HPV anti-
plasm next to the nuclear membrane. Particles were detected more bodies [14].
frequently in the spinous layer and more seldom in the basal and Our findings indicate that HPV is detected in normal and benign
prickle cell layer, forming clusters in isolated cells (Fig. 3). lesions only by PCR, whereas in 16.7% of dysplastic lesions and
47.7% of malignant lesions it is detected by at least two different
techniques. This means that in normal and benign lesions HPV
Discussion infection may have a very low concentration and may be latent
(NPCR), while in some dysplastic and malignant lesions HPV can
Reports on HPV involvement in oral carcinogenesis are conflict- be found in higher concentration, either as DNA (MY-PCR, IHC or
ing, with percentages ranging from 0% [18,41–43] to 100% [17,20]. TEM), or as full viral particle in productive infection (IHC, TEM)
The observed discrepancies can be attributed to differences in epi- or with both of these forms at the same time (IHC/NPCR, TEM/
demiologic data, sample collection techniques, sample preserva- NPCR). Transitional electron microscopy seems to be more sensi-
tion techniques, HPV detection methods and protocols. In our tive than immunohistochemistry perhaps due to its capacity to
study, three different techniques were used to demonstrate HPV detect immature viral particles, indetectable by IHC [55].
in oral specimens. All techniques proved to be reliable as all control In our study, HPV DNA was detected in 40% in both ‘normal epi-
specimens gave consistent results irrespective of the technique thelium’ groups, which lies in accordance with some studies
employed. [30,34,48,56,57]. However, other groups have not detected any
The study revealed that HPV was detected in 34.9% of the tissue HPV DNA in normal mucosa samples [11,28,29,58–60]. Very high
specimens by at least one of the three methods used. The majority sensitivity of NPCR, examination of all epithelial layers and speci-
of the positive samples were found with the NPCR technique men collection from every site of oral cavity may explain the high
(32.9%), less were observed by electron microscopy (28.1%) and percentage of positive normal samples.
very few by immunohistochemistry (4.7%). The sensitivity of NPCR Our finding of a significant relation between HPV infection of
was roughly estimated to reach 1 viral copy per 104 epithelial cells, oral carcinomas and the male gender lies in agreement with many
about 100 times more than MY, GP+ and TS-PCR’s sensitivity previous studies [34,51,61–63] and may be explained by the
(1/102). These findings agree with the results of Gillison and Keert increased number of sexual partners and orogenital sexual con-
[44] and Remmerbach [45]. The fact that 96.5% of positive NPCR tacts of men. Moreover, our result that there was a significant rela-
samples were detected by the second step, means that viral load tion of HPV detection with tongue carcinomas coincides with the
ranged between 1 copy per 102 and 104 epithelial cells. According findings of other studies [15,48,64,65]. The majority of studies
to the clonal expansion theory [46,47], if viral load is less than 1 involving oral lesions do not separate oral cavity from oropharynx,
copy per cell – as it is for the majority of our positive specimens although the relation of HPV to oropharyngeal cancer is more sig-
– HPV does not seem to play any role in oral carcinogenesis. Higher nificant that to oral cancer [66]. Since our study is focused on oral
viral load (>1 copy per 102 cells) was detected only in 1.8% of dys- cavity, we tried to compare our findings with studies including
plastic and 3.2% of cancerous lesions by the use of MY-PCR. similar sample location.
An unusual finding of our study was the decreasing percentage In the total sample HPV DNA was detected more frequently in
of HPV DNA positive samples during the transition of normal epi- younger individuals (653 years old). The higher prevalence of
thelium to malignancy. In detail, by NPCR HPV L1 DNA was HPV in the cervix of younger women, the increased sexual activity
detected in 40% of normal tissues, 40% of fibromas, 35.8% of non- of young individuals, the increased number of sexual partners and
dysplastic leukoplakias, 31.6% of dysplastic leukoplakias and the more frequent oral-genital and oral- anal contacts in compari-
22.2% of oral carcinomas. Our results agree partly with some stud- son to older people may explain that finding [67]. No significant
ies [30,31,48,49], although there are many studies reporting con- relation found between HPV detection and histologic diagnosis
tradictory results [11,50,51]. Our findings can be explained by may indicate that in most cases HPV colonizes oral epithelium
the ‘hit-and-run mechanism’. According to that theory, HPV may without playing a role in malignant progression.
play a role in the initial stages of carcinogenesis, inactivating p53 HPV16 was the prevailing viral type, which was detected in
or pRb and predisposing the cells to accumulate additional genetic 94.3% of positive samples, either in single infections or in coinfec-
E. Blioumi et al. / Oral Oncology 50 (2014) 840–847 845

Figure 3. Transitional Electronic Microscopy in oral specimens. (A) Mild dysplasia


from the floor of the mouth. HPV particles (50–60 nm) are detected inside the
nucleous of oral squamous epithelial cell (TEM, X30.000). (B) Mild dysplasia from
the floor of the mouth. HPV particles are detected in the cytoplasm just next to the
nuclear membrane of oral squamous epithelial cell (TEM, X30.000). (C) Oral
squamous cell carcinoma of the alveolar mucosa, moderately differentiated. HPV
particles are found just out of the nuclear membrane of an epithelial cell of the
spinal layer (TEM, X25.000).
Figure 2. Immunohistochemical stain of HPV antigens in oral squamous cell
carcinoma from the tongue. Papillomavirus structural antigens are identified by incidental colonization of oral epithelium by the low-risk viral type
positive brown reaction (arrows) inside the nucleous (A, B), and next to the and, hence, a solitary tumorigenic effect of the high-risk type [11].
nucleous (C, D) in few isolated epithelial cells. These cells may be koilocytes (A, B) In conclusion, the decreasing HPV prevalence during the transi-
or form clusters (D) (40).
tion from normality to malignancy, the very low viral load of the
majority of the specimens and the sparse detection of viral anti-
tions. Most studies agree with the high prevalence of HPV16 in gens and particles indicate that HPV does not seem, from the spe-
normal tissues [8,27,31,48,57], leukoplakias [29,68] and oral carci- cific sample examined, to play a substantial role in oral
nomas [31,66,69,70]. High-high risk infection pattern may repre- carcinogenesis. It cannot be excluded that HPV could be involved
sent a synergistic oncogenic action of the coinfecting types. in oral carcinogenesis only in cases with high viral load or at early
Conversely, the low–high-risk infections may be indicative of an stages of carcinogenesis through the hit-and-run mechanism.
846 E. Blioumi et al. / Oral Oncology 50 (2014) 840–847

Conflict of interest [29] Nielsen H, Norrild B, Vedtofte P, Praetorius F, Reibel J, Holmstrup P. Human
papillomavirus in oral premalignant lesions. Oral Oncol, Eur J Cancer
1996;32B(4):264–70.
None declared. [30] Sugiyama M, Bhawal U, Dohmen T, Ono S, Miyauchi M, Ishikawa T, et al.
Detection of human papillomavirus-16 and HPV-18 in normal, dysplastic and
References malignant oral epithelium. Oral Surg Oral Med Oral Pathol Oral Radiol
Endodontal 2003;95:594–600.
[31] Herrero R, Castellsague X, Pawlita M, Lissowska J, Kee F, Balaram P. Human
[1] Parkin DM, Bray F, Ferlay J, et al. Global cancer statistics, 2002. CA Cancer J Clin
Papillomavirus and oral cancer: the international agency for research on
2005;55:74–108.
cancer multicenter study. J Natl Cancer Inst 2003;95:1772–83.
[2] Globocan 2002, International Agency for Research on Cancer, World Health
[32] Lawton G, Thomas S, Schonrock J, Monsour F, Frazer I. Human. Human
Organization, pqo9 rbarg rso http://www-dep.iarc.fr, gleqolgmi9 a
papillomaviruses in normal oral mucosa: a comparison of methods for sample
emgle9 qxrg1 22.11.09.
collection. J Oral Pathol Med 1992;21:265–9.
[3] Aggelopoulos A, Papamikolaou A, Papanikolaou S, Aggelopoulou E. Modern
[33] Llamas-Martinez S, Esparza-Gomez G, Campo-Trapero J, Cancela-Rodriguez P,
oral and maxillofacial pathology. Medical editions Litsas. Athens; 2000. p. 259–
Basconew-Martinez A, Moreno-Lopez LA, et al. Genotypic determination by
66.
PCR-RFLP of human papillomavirus in normal oral mucosa, oral leukoplakia
[4] Neville BW, Damm DD, Allen CM, Bouquot JE. Oral and maxillofacial pathology.
and oral squamous cell carcinoma samples in Madrid (Spain). Anticancer Res
2nd ed. W.B. Saunders Company; 2002.
2008;28:3733–42.
[5] Scully C. Oral and maxillofacial surgery. Elsevier Science; 2004. p. 31.
[34] Zhang Z, Sdek P, Cao J, Chen W. Human papillomavirus type 16 and 18 DNA in
[6] Barnes L, Eveson JW, Reichart P, Sidransky D, editors. World health
oral squamous cell carcinoma and normal mucosa. Int J Oral Maxillofac Surg
organization. Classification of tumours. Pathology and genetics of head and
2004;33:71–4.
neck tumours. IARC Press Lyon; 2005. p. 163–81.
[35] Woods K, Shillitoe E, Spitz M, Schantz S, Adler-Storthz K. Analysis of human
[7] Warnakulasuriya S, Johnson NW, van der Waal I. Nomenclature and
papillomavirus in oral squamous cell carcinomas. J Oral Pathol Med
classification of potentially malignant disorders of the oral mucosa. J Oral
1993;22:101–8.
Pathol Med 2007;36(10):575–80.
[36] Elamin F, Steingrimsdottir H, Wanakulasuriya S, Johnson N, Tavassoli M.
[8] IARC. IARC monographs on the evaluation of carcinogenic risks to humans, vol.
Prevalence of human papillomavirus infection in premalignant and malignant
90, Human Papillomaviruses, Lyon; 2007.
lesions of the oral cavity in UK subjects: a novel method of detection. Oral
[9] Zuckerman A, Banatvala J, Pattison J, Griffiths P, Schoub B. Principles and
Oncol 1998;34:191–7.
Practice of clinical virology. 5th ed. John Wiley & Sons, Ltd.; 2004.
[37] Tachezy R, Klozar J, Salakova M, Smith E, Turek L, Betka J, et al. HPV and other
[10] de Villiers E, Fauquet C, Broker T, Bernard H, Zur Hausen H. Classification of
risk factors of oral cavity/oropharyngeal cancer in the Czech Republic. Oral Dis
Papillomaviruses. Virology 2004;324:17–27.
2005;11:181–5.
[11] Bouda M, Gorgoulis V, Kastrinakis N, Giannoudis A, Tsoli E, Danassi-Afentaki D,
[38] Saiki RK, Scharf S, Faloona F, Mullis KB, Horn GT, Erlich HA, et al. Enzymatic
et al. ‘High risk’ HPV types are frequently detected in potentially malignant
amplification of b-globin genomic sequences and restriction site analysis for
and malignant oral lesions, but not in normal mucosa. Mod Pathol
diagnosis of sickle cell anemia. Science 1985;230(4732):1350–4.
2000;13(6):644–53.
[39] Mehta C, Patel R. SPSS exact tests 7.0 for windows. Chicago: SPSS Inc.; 1996.
[12] Syrjänen K, Pyrhönen S, Syrjänen S, Lamberg M. Immunohistichemical
[40] Zar J. Biostatistical analysis. New Jersey: Prentice-Hall International Inc.; 1996.
demonstration of human papilloma virus (HPV) antigens in oral squamous
[41] Al-Qahtani, Brousseau V, Paczeny D, Domanowski G, Hamid Q, Hier M, et al.
cell lesions. Br J Oral Pathol 1983;21:147–53.
Koilocytosis in oral squamous cell carcinoma: what does it mean? J Otolaryng
[13] Syrjänen K, Syrjänen S, Lamberg M, Pyrhönen S, Nuutinen T. Morphological
2007;36(1):26–31.
and immunohistochemical evidence suggesting human papillomavirus (HPV)
[42] Young S, Min K. In situ DNA hybridization analysis of oral papillomas,
involvement in oral squamous cell carcinogenesis. Int J Oral Surg 1983;12:418–24.
leukoplakias, and carcinomas for human papillomavirus. Oral Surg Oral Med
[14] Loning T, Reichart P, Staquet M, Becker J, Thivolet J. Occurrence of
Oral Pathol 1991;71:726–9.
papillomavirus structural antigens in oral papillomas and leukoplakias. J
[43] Zeuss M, Miller C, White D. In situ hydrization analysis of human
Oral Pathol 1984;13:155–65.
papillomavirus DNA in oral mucosal lesions. Oral Surg Oral Med Oral Pathol
[15] de Villiers EM, Weidauer H, Otto H, Zur Hausen H. Papillomavirus DNA in
1991;71:714–20.
human tongue carcinomas. Int J Cancer 1985;36:575–8.
[44] Gillison ML, Keert S. Human papillomavirus-associated head and neck
[16] Ibrahim S, Bertelsen B, Kalvenes M, Idris A, Vasstrand E, Nilsen R, et al.
squamous cell carcinoma: mounting evidence for an etiologic role for
Expression of keratin 13, 14 and 19 in oral squamous cell carcinomas from
human papillomavirus in a subset of head and neck cancers. Curr Opin
Sudanese snuff dippers: lack of association with human papillomavirus
Oncol 2001;13(3):183–8.
infection. APMIS 1998;106(10):959–69.
[45] Remmerbach T, Brickmann U, Hemprich A, Chekol M, Kuhndel K, Liebert U.
[17] Uobe K, Masuno K, Fang Y-R, Li L-J, Wen Y-M, Ueda Y, et al. Detection of HPV in
PCR detection of human papillomavirus of the mucosa: comparison between
Japanese and Chinese oral carcinomas by in situ PCR. Oral Oncol 2001;37:
MY09/11 and GP5+/6+ primer sets. J Clin Virol 2004;30:302–8.
146–52.
[46] Ha P, Pai S, Westra W, Gillison M, Tong B, Sidransky D, et al. Real-time
[18] Rivero E. HPV in oral squamous cell carcinomas of a Brazilian population:
quantitative PCR demonstrates low prevalence of human papillomavirus type
amplification by PCR. Bras Oral Res 2006;20(1):21–4.
16 in premalignant and malignant lesions of the oral cavity. Clin Cancer Res
[19] Gonzalez J, Gutierrez R, Keszler A, Colacino M, Alonio L, Teyssie A, et al. Human
2002;8:1203–9.
papillomavirus in oral lesions. Medicina (Buenos Aires) 2007;67:363–8.
[47] van Houten V, Snijders P, van den Breckel M, Kummer A, Meijer C, Leeuwen B,
[20] Koyama K, Uobe K, Tanaka A. Highly sensitive detection of HPV-DNA in
et al. Biological evidence that human papillomaviruses are etiologically
paraffin sections of human oral carcinomas. J Oral Pathol Med 2007;36:18–24.
involved in a subgroup of head and neck squamous cell carcinomas. Int J
[21] Sugiyama M, Bhawal U, Kawamura M, Ishioka Y, Shigeishi H, Higashikawa K,
Cancer 2001;93:232–5.
et al. Human papillomavirus-16 in oral squamous cell carcinoma: clinical
[48] D’ Costa J, Saranath D, Dedhia P, Sanghvi V, Mehta AR. Detection of HPV-16
correlates and 5-year survival. Br J Oral Maxillofac Surg 2007;45:116–22.
genome in human oral cancers and potentially malignant lesions from India.
[22] Zarei M, Moradie A, Hamkar R, Mohammadalizadeh S, Chamani G, Alizadeh N,
Oral Oncol 1998;34:413–20.
et al. Detection of human papillomavirus DNA sequences in oral lesions using
[49] Holladay E, Gerald W. Viral gene detection in oral neoplasms using the
polymerase chain reaction. Acta Med Iran 2007;45(2):177–82.
polymerase chain reaction. Am J Clin Pathol 1993;100:36–40.
[23] Montaldo C, Mastinu A, Quartuccio M, Piras V, Denotti G, Pisano E, et al.
[50] Ostwald C, Rutsatz K, Schweber J, Schmidt W, Gundlach K, Barten M. Human
Detection and genotyping of human papillomavirus DNA in samples from
papillomavirus 6/11, 16 and 18 in oral carcinomas and benign oral lesions.
healthy Sardinian patients: a preliminary study. J Oral Pathol Med 2007;36:
Med Microbiol Immunol 2003;192:145–8.
482–7.
[51] Miller CS, White DK. Human papillomavirus expression in oral mucosa,
[24] Chuang A, Chuang T, Chang S, Zhou S, Begum S, Westra W, et al. Presence of
premalignant conditions, and squamous cell carcinoma. A retrospective
HPV DNA in convalescent salivary rinses is an adverse prognostic marker in
review of the literature. Oral Surg Oral Med Oral Pathol Oral Radiol
head and neck squamous cell carcinoma. Oral Oncol 2008;44:915–9.
Endodontal 1996;82:57–68.
[25] Llamas-Martinez S, Esparza-Gomez G, Campo-Trapero J, Cancela-Rodriguez P,
[52] Evander M, Edlund K, Gustafsson A, et al. Human papillomavirus infection is
Bascones-Martinez A, Moreno-Lopez L, et al. Genotypic determination by PCR-
transient in young women: a population based cohort study. J Infect Dis
RFLP of human papillomavirus in normal oral mucosa, oral leukoplakia and
1995;171:1026–30.
oral squamous cell carcinoma samples in Madrid (Spain). Anticancer Res
[53] Syrjänen S. Human papillomavirus (HPV) in head and neck cancer. J Clin Virol
2008;28(6A):3733–41.
2005;32S:S59–66.
[26] Szarka K, Tar I, Feher E, Gll T, Kis A, Toth E, et al. Progressive increase of human
[54] Boy S, van Rensburg E, Engelbrecht S, Dreyer L, van Heerden M, van Heerden
papillomavirus carriage rates in potentially malignant and malignant oral
W. HPV detection in primary intra-oral squamous cell carcinomas –
disorders with increasing malignant potential. Oral Microbiol Immunol
commensal, aitiological agent or contamination? J Oral Pathol Med
2009;24:314–8.
2006;35:86–90.
[27] Terai M, Hashimoto K, Yoda K, Sata T. High prevalence of human
[55] Sato S, Okagaki T, Clark B, Teiggs L, Fukushima M, Ostrow R, et al. Sensitivity of
papillomaviruses in the normal oral cavity of adults. Oral Microbiol Immunol
koilocytosis, immunocytochemistry, and electron microscopy as compared to
1999;14:201–5.
DNA hybridization in detecting human papillomavirus in cervical and vaginal
[28] Fouret P, Martin F, Flahault A, Saint-Guily J. Human papillomavirus infection in
condyloma and intraepithelial neoplasia. Int J Gynecol Pathol 1987;5:
the malignant and premalignant head and neck epithelium. Diagn Mol Pathol
297–307.
1995;4(2):122–7.
E. Blioumi et al. / Oral Oncology 50 (2014) 840–847 847

[56] Jenison S, Valentine J, Koutsky L, Christianen A, Beckmann A, Galloway D, et al. [64] Shindoh M, Sawada Y, Kohgo T, Amemiya A, Fujinaga K. Detection of human
Evidence of prevalent genital-type human papillomavirus infections in adults papilloma virus DNA sequences in tongue squamous cell carcinoma utilizing
and children. J Infect Dis 1990;162(1):60–9. the polymerase chain method. Int J Cancer 1992;50:167–71.
[57] Kojima A, Maeda H, Kurahashi N, Sakagami G, Kubo K, Yoshimoto H, et al. [65] Chang J, Lin M, Chiang C. High-risk human papillomaviruses may have an
Human papillomaviruses in the normal cavity of children in Japan. Oral Oncol important role in non-oral habits-associated oral squamous cell carcinomas in
2003;39:821–8. Taiwan. Am J Clin Pathol 2003;120:909–16.
[58] Ostwald C, Muller P, Barten M, Rutsatz K, Sonnenburg M, Milde-Langosch K, [66] Gillison ML, Koch WM, Capone RB, Spafford M, Westra W, Wu L, et al. Evidence
et al. Human papillomavirus DANN in oral squamous cell carcinomas and for a causal association between human papillomavirus and a subset of head
normal mucosa. J Oral Pathol Med 1994;23:220–5. and neck cancers. J Natl Cancer Inst 2000;92:709–20.
[59] Eike A, Buchwald C, Rolighed J, Lindeberg H. Human papillomavirus (HPV) is [67] Smith E, Ritchie J, Summersgill K, Klussmann J, Lee J, Wang D, et al. Age, sexual
rarely present in normal oral and nasal mucosa. Clin Otolaryngol Allied Sci behavior and human papillomavirus infection in oral cavity and oropharyngeal
1995;20(2):171–3. cancers. Int J Cancer 2004;108:766–72.
[60] Van Rensburg E, Engelbrecht S, Van Heerden W, Raubennheimer E, Schoub B. [68] Palefsky J, Silverman S, Abdel-Salaam M, et al. Association between
Human papillomavirus DNA in oral squamous cell carcinomas from an African proliferative verrucous leukoplakia and infection with human papillomavirus
population sample. Anticancer Res 1996;16:969–74. type 16. J Oral Pathol Med 1995;24:193.
[61] Ritchie J, Smith E, Summersgill K, Hoffman H, Wang D, Klussmann J, et al. [69] Nemes J, Deli L, Nemes Z, Marton I. Expression of p16INK4A, p53, and Rb
Human papillomavirus infection as a prognostic factor in carcinomas of the proteins are independent from the presence of human papillomavirus genes in
oral cavity and oropharynx. Int J Cancer 2003;104:336–44. oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol
[62] Cruz I, Snijders R, Steenberg R, Meijer C, Snow G, Walboomers J, et al. Age- Endodontol 2006;102:344–52.
dependence of human papillomavirus DNA presence in oral squamous cell [70] Shindoh M, Chiba I, Yasuda M, Saito T, Funaoka K, Kohgo T, et al. Detection of
carcinomas. Oral Oncol, Eur J Cancer 1996;32B(1):55–62. human papillomavirus DNA sequences in oral squamous cell carcinomas and
[63] Smith E, Hoffman H, Summersgill K, Kirchner L, Turek L, Haugen T. Human their relation to p53 and proliferating cell nuclear antigen expression. Cancer
papillomavirus and risk of oral cancer. Am Laryng Rhinol Otal Soc 1998; 1995;76:1513–21.
108(7):1098–103.

You might also like