molecular pathogenesis of cervical carcinoma160985/fulltext01.pdfpathology presented at uppsala...

77
Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine 1080 Molecular Pathogenesis of Cervical Carcinoma Analysis of Clonality, HPV16 Sequence Variations and Loss of Heterozygosity BY XINRONG HU ACTA UNIVERSITATIS UPSALIENSIS UPPSALA 2001

Upload: others

Post on 09-Jul-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

Comprehensive Summaries of Uppsala Dissertationsfrom the Faculty of Medicine 1080

Molecular Pathogenesis of

Cervical CarcinomaAnalysis of Clonality, HPV16 Sequence Variations

and Loss of Heterozygosity

BY

XINRONG HU

ACTA UNIVERSITATIS UPSALIENSISUPPSALA 2001

Page 2: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

1

Dissertation for the Degree of Doctor of Philosophy (Faculty of Medicine) inPathology presented at Uppsala University in 2001

Abstract

Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma: Analysis ofClonality, HPV16 Sequence Variations and Loss of Heterozygosity. ActaUniversitatis Upsaliensis. Comprehensive Summaries of Uppsala Dissertations fromthe Faculty of Medicine 1080. 76pp. Uppsala. ISBN 91-554-5127-6

A previous model of morphological pathogenesis assumed that cervical carcinomais of monoclonal origin and progresses through multiple steps from normal epitheliumvia CINS into invasive carcinomas. The aim of this study was to investigate themolecular mechanism of pathogenesis of cervical neoplasia.

In the clonality study, we found that 75% (6/8) of informative cases of cervicalcarcinoma had identical patterns of loss of heterozygosity (LOH) in the multiplesynchronous lesions, while the remaining cases had different LOU patterns. In anextensively studied "golden case", the multiple carcinoma and cervical intraepithelialneoplasia (CIN) lesions could be divided into several different clonal groups by theX-chromosome inactivation patterns, HPV 16 mutations and LOH patterns. Thebiggest clonal family included one CIN II, one CIN III and four carcinoma samples,while four other monoclonal families of carcinoma did not include CIN lesions. Theseresults suggested that cervical carcinoma can be either monoclonal or polygonal andcontains clones developing either directly or via multiple steps. In the study of HPVtypes and HPV16 variations, the results confirmed that specific HPV types are thecause of cervical carcinoma but failed to support the previous opinion that HPV16 E6variants are more malignant than the prototype. We established a novel classificationcalled oncogene lineage of HPV16, and found that additional variations of HPV 16oncogenes might be a weak further risk factor for cervical carcinoma. In the study ofLOH, we found that interstitial deletion of two common regions of chromosome 3p,i.e., 3p2l.1-3p2l.3, and 3p22, was an early event in the development of cervicalcarcinoma. The results showed that the hMLH1 gene, located in 3p22 and showingLOH in 43% of the studied cases, was not involved in the development of cervicalcarcinoma because neither the expression level of protein nor the gene sequence wasaltered in these cases.

In summary, a suggested model of molecular pathogenesis of cervical carcinoma isas follows. Specific types of HPV infect one or more committed stem cells in thebasal layer of the epithelium. Fully efficient LOH events turn one (monoclonal origin)or more (polyclonal origin) HPV-infected stem cells into carcinoma cells without CINsteps. Less efficient LOH events would lead to CIN steps where some other unknownfactors require to be added to facilitate the formation of carcinoma. In the absence ofLOH events no carcinoma develops from the HPV-infected stem cells.

Keywords: cervical carcinoma, CIN, HPV, LOH, X-chromosome inactivation,clonality, molecular pathogenesisXinrong Hu, Department of Genetics and Pathology, Rudbeck Laboratory, UppsalaUniversity, SE-751 85, Uppsala, Sweden © Xinrong HuISSN 0282-7476ISBN 91-554-5127-6

Page 3: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

2

Contents

Abstract ………………………………………………………………………… 1

Contents ………………………………………………………………………… 2

Publications and Manuscript included …………………………………… 5

Abbreviations …………………………………………………………………… 6

1 Introduction ……………………………………………………… 7

1.1 Morphological Biology …………………………………… 71.1.1 Anatomy and histology of cervix ……………………………………… 7

1.1.2 Cell kinetics in the transitional zone …………………………………… 7

1.1.3 Benign tumors …………………………………………………………… 8

1.1.4 Cervical intraepithelial neoplasia (CIN) ………………………………… 8

1.1.5 Cervical invasive carcinoma …………………………………………… 9

1.1.6 Assumed morphological pathogenesis ………………………………… 10

1.2 Etiology ………………………………………………………………… 11

1.2.1 Human Papillomavirus (HPV) ………………………………………… 11

1.2.1.1 Epidemiology ………………………………………………………… 12

1.2.1.2 Organization of HPV ……………………………………………… 12

1.2.1.3 HPV detection and typing …………………………………………… 13

1.2.1.4 HPV types and infection of the human tissue ………………………… 13

1.2.1.5 Prevalence of the presence of HPV DNA in cervical neoplasia ……… 14

1.2.1.6 HPV is the main cause of cervical neoplasia ………………………… 15

1.2.2 Cigarette smoking ……………………………………………… 15

1.2.3 Hormonal contraception …………………………………………… 16

1.2.4 Inheritance ……………………………………………………………… 16

1.3 Molecular Biology ……………………………………… 17

1.3.1 Clonality of cervical neoplasms ……………………………………… 17

1.3.1.1 Strategies of clonality analysis ……………………………………… 17

1.3.1.2 Polymorphism of inactivation of X-chromosome linked androgen

receptor gene ……………………………………………………… 17

1.3.1.3 Clonality features of cervical neoplasms ………………………… 19

1.3.1.4 Clonality status of some other common human tumors ………… 21

1.3.2 HPV16 ……………………………………………………………… 22

1.3.2.1 The structure of the HPV16 genome and the function of HPV16 genes 22

Page 4: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

3

1.3.2.2 HPV16 variants in progression of cervical carcinoma ………………… 25

1.3.2.3 Physical stage of HPV16 ……………………………………………… 26

1.3.3 Loss of heterozygosity (LOH) ………………………………………… 28

1.3.3.1 LOH on chromosome 3p ……………………………………………… 28

1.3.3.2 LOH on chromosome 6p ……………………………………………… 29

1.3.3.3 LOH on chromosomes 4, 5p, 11, 17, 18 and 19 ………………………. 29

1.3.4 Point mutations and gene polymorphisms …………………………… 30

1.3.4.1 Point mutations in TP53 ……………………………………………… 30

1.3.4.2 Susceptibility of patients with TP53 codon 72 polymorphism to HPV

infection ……………………………………………………………… 31

1.3.4.3 Susceptibility of patients with different HLA class II genotypes

to HPV infection ……………………………………………………… 31

1.3.5 Assumed molecular pathogenesis …………………………………… 32

2 Specific aims of study ………………………………………………… 33

3 Experimental Design ………………………………………………… 33

3.1 Cross-sectional case control study system ……………………… 33

3.2 Synchronous lesion study system …………………………………… 34

4 Materials and Methods ……………………………………………… 34

4.1 Patients ……………………………………………………………… 34

4.1.1 Paper 1 ……………………………………………………………… 34

4.1.2 Paper 2 ………………………………………………………………… 35

4.1.3 Paper 3 …………………………………………………………… 35

4.1.4 Paper 4 ……………………………………………………………… 35

4.1.5 Paper 5 ……………………………………………………………… 36

4.1.6 Paper 6 …………………………………………………………… 36

4.1.7 Paper 1-6 …………………………………………………………… 36

4.2 Microdissection (Paper 1 –6) ……………………………………… 37

4.3 Immunohistochemistry (Paper 5) …………………………………… 37

4.4 DNA preparation (Paper 1-6) and purification (Paper 6) …………… 37

4.5 Methylation sensitive Hpa II restriction enzyme digestion (Paper 6) … 38

4.6 Polymerase chain reaction (PCR) (Paper 1-6) ………………………… 38

4.7 Polyacrylamide gel electrophoresis with single-strand conformation

polymorphism (SSCP) and silver staining for HPV typing (Paper 5) … 39

4.8 Fragment analysis (Paper 4 and 6) …………………………………… 39

4.9 DNA sequence analysis (Paper 1-6) ………………………………… 40

Page 5: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

4

5 Results and Discussions ………………………………………… 40

5.1 Paper 1 …………………………………………………………… 40

5.1.1 Results …………………………………………………………… 40

5.1.2 Discussion ………………………………………………………… 42

5.2 Paper 2 …………………………………………………………… 43

5.2.1 Results …………………………………………………………… 43

5.2.2 Discussion ………………………………………………………… 44

5.3 Paper 3 …………………………………………………………… 46

5.3.1 Results ……………………………………………………………… 46

5.3.2 Discussion ………………………………………………………… 47

5.4 Paper 4 …………………………………………………………… 47

5.4.1 Results …………………………………………………………… 47

5.4.2 Discussion ………………………………………………………… 48

5.5 Paper 5 ……………………………………………………………… 49

5.5.1 Results ……………………………………………………………… 49

5.5.2 Discussion ………………………………………………………… 49

5.6. Paper 6 ……………………………………………………………… 50

5.6.1 Results ……………………………………………………………… 50

5.6.2 Discussion ………………………………………………………… 52

6 General discussion and summary of conclusions ………………… 53

7 Acknowledgements ………………………………………… 57

8 References ………………………………………………………… 59

Page 6: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

5

Publications and Manuscript included

This dissertation is based on the following original publications and manuscript,

which are referred to in the text by their Arabic numerals:

1. Xinrong Hu, Zhongmin Guo, Pang Tianyun, Fredrik Pontén, Erik Wilander, Sonja

Anderson and Jan Pontén. HPV Typing and HPV16 E6-Sequence Variations in

Synchronous Lesions of Cervical Squamous-Cell Carcinoma from Swedish Patients.

Int. J. Cancer 1999; 83:34-37.

2. Xinrong Hu, Pang Tianyun, Zhongmin Guo, Natalia Mazurenko, Fjodor Kisseljov,

Jan Pontén and Monica Nistér. Human Papillomavirus Type 16 E6 Gene Variations in

Invasive Cervical Squamous Cell Carcinoma and Cancer in Situ from Russian

Patients. Br. J. Cancer 2001; 84(6): 791-795.

3. Xinrong Hu, Pang Tianyun, Zhongmin Guo, Jan Pontén, Monica Nistér and Gijs

Afink. Oncogene Lineage of Human Papillomavirus Type 16 E6, E7 and E5 in Pre-

invasive and Invasive Cervical Squamous Cell Carcinoma. J. Pathol. 2001, in press.

4. Zhongmin Guo, Xinrong Hu, Gijs Afink, Fredrik Pontén, Erik Wilander and Jan

Pontén. Comparison of Chromosome 3p Deletion between Cervical Pre-cancers

Synchronous with and without Invasive Cancer. Int. J. Cancer 2000; 86:518-523.

5. Xinrong Hu, Zhongmin Guo, Pang Tianyun, Qing Li, Gijs Afink and Jan Pontén.

Immunohistochemical and DNA Sequencing Analysis on Human Mismatch Repair

Gene MLH1 in Cervical Squamous Cell Carcinoma with LOH of this Gene.

Anticancer Research 2000; 20:171-176.

6. Xinrong Hu, Pang Tianyun, Anna Asplund, Jan Pontén and Monica Nistér.

Clonality Analysis of Synchronous lesions of Cervical Carcinoma based on X-

chromosome Inactivation Polymorphism, HPV16 Genome Mutations and Loss of

Heterozygosity. Submitted.

Page 7: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

6

Abbreviations

CIN: cervical intraepithelial neoplasia

MPU: minimum proliferative unit

FIGO: international Federation of Gynaecology and Obstetrics

HPV: human papillomavirus

Nt: nucleotide

LTR: long terminal region

LOH: loss of heterozygosity

HLA: human Leukocyte Antigen

PGK: phosphoglucokinase

G6PD: glucose-6-phosphate dehydrogenase

FHIT: fragile histidine triad

PCR: polymerase chain reaction

SSCP: single strand conformational polymorphism

Page 8: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

7

1 Introduction

Perhaps what ordinary people nowadays know about cervical cancer may be the

following. It is one of the most common malignancies in women; It can be sexually

transmitted by men (with HPV); Vaginal cytology can be used to detect it; Cervical

conization can be used to treat and prevent it; And unproperly treated, patients will be

killed by it. But cervical carcinoma is much more if you set your sight into it.

1.1 Morphological Biology

1.1.1 Anatomy and histology of cervix

Cervix is a hollow cone. On the hollow surface, the cervical os is lined with

squamous epithelium and endocervix with columnar epithelium. The former consists

of several layers of squamous cells with short cubic cells in the basal layer. The latter

contains only one layer of columnar cell. The border area between the squamous

epithelium and columnar epithelium is called transitional zone (1) or transformation

zone (2). On the average, the transitional zone is populated by a 3-4 cell deep layer of

epithelium of squamous cells presumed to be deriving from ectocervical squamous

epithelium. It is the meeting ground of infection, regeneration, squamous metaplasia

and neoplasia (1-8).

1.1.2 Cell kinetics in the transitional zone

In the basal layer, there are a limited number of committed stem cells in the

transition zone (9). They are reversibly sleeping in G0 and capable of self-renewal.

Normally, the committed stem cells take the way of asymmetrical division by giving

rise to two daughter cells. One of the daughters inherits all natures of the mother and

stays in the basal layer serving as the resource of stem cells, while another is able to

divide and differentiate into more mature epithelial cells and is called precursor cell

of the epithelium. It is postulated that the transitional zone is built up by a number of

minimum proliferative unit (MPU)s (9). Each MPU contains a precursor cell that

proceeds through several rounds of DNA replication and division and differentiates

into mature epithelial cells, moving outwards to form a cell lineage. Different MPUs

Page 9: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

8

stand together unidentifiable by morphology. Older MPUs will finally die out by

shedding off and is replaced by the new ones. When the epithelium is injured, the

committed stem cells are responsible for repairing it by regeneration of MPUs, in

which process the structures and functions of the epithelial cells and epithelium

recover completely (10).

If the MPUs proliferate faster than they die, a lesion of hyperplasia will arise (11).

The lesion protrudes above the cervical surface and the cells of the hyperplastic lesion

look normal. It is often the result of a responsive proliferation to inflammation. When

inflammation is eliminated, it will disappear. In some cases, the committed stem cells

in the glands may produce precursor cells that differentiate into squamous cells that

occupy the glands partly or fully. This is the course we all know as metaplasia (11,

12). The metaplasia has normal-looking cells and can be cured as long as the

stimulation is removed. Neither the hyperplasia nor the metaplasias represent any true

tumors.

Challenged by some factors such as HPV, the committed stem cells become tumor

stem cells. It is suggested that, in this way, tumors on the cervix are initiated.

Neoplasia of the cervical squamous epithelium is classified as papilloma/condyloma,

cervical intraepithelial neoplasia (CIN) and invasive carcinoma.

1.1.3 Benign tumors

Benign tumors of the cervix are of the type squamous cell papilloma, also called

condyloma (13). The cells of a condyloma do not show conspicuous atypia but often

show obvious koilocytosis as a sign of HPV synthesis (14-18). The condylomas are

divided into two types: exophytic and flat. Exophytic condylomas grow outward with

an appearance of nipples (15-17, 19). It is believed that an exophytic condyloma

never develops into dysplasia. Flat condylomas grow deep wards with a slightly

raised plaque on the cervical surface (20, 21). At a low frequency, flat condylomas

may develop into dysplasia (21-26).

1.1.4 Cervical Intraepithelial Neoplasia (CIN)

CIN is a malignant tumor growing within the epithelium. It has been considered a

pre-cancerous lesion. In CIN lesions, the number of layers of tumor cells increase and

the order of the cell stratum is confused. Similar to the cells of invasive carcinoma,

Page 10: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

9

the cells of CIN typically show atypia: bizarre cell body, enlarged bulk of cells,

enlarged nuclei with thick membrane and strongly stained pieces of chromatin, high

frequency of mitotic figures and abnormal mitotic figures. CIN lesions are often

demarcated from the adjacent normal epithelium. CIN I occupies one third of the

epithelial stratum, CIN II two thirds, and CIN III the whole epithelial layer. All CIN

lesions have not yet broken through the basal membrane and never kill the patient

because they do not metastasis (27, 28). When CIN cells break through the basal

membrane, they become invasive carcinoma and invade into stroma and neighboring

tissues.

According to the clinical and epidemiological surveys, the prevalence of CIN

attains its peak around the age at 35, with about 3,500/100,000 women in Sweden

(29). Although the reported percentages show a wide range, in the absence of

intervention, roughly one-third of CIN lesions disappear spontaneously, one-third

persist and one-third progress (30). Bos et al. screened a population in Denmark

(1966-82) and found that 24% of CINs would progress, 39% regress, and 38%

remain, if all pre-clinical lesions in women aged 25-50 were included (31). In a

Swedish series of 555 women with CIN I followed up for an average of 39 months,

progression to CIN II or III took place in 16% of the cases, whereas 62% regressed

and 22% persisted (32). In another study, 894 women with CIN II were followed for

78 months. Here, progression to CIN III was seen in 30%, regression in 54% and

persistence in 16% (33). The time from discovery of CIN I and II to the observation

of CIN III was around 3.5-4.5 years. The mechanisms that determine the fate of CIN

lesions have not been clarified at all.

The non-invasive tumor of cervix, i.e., intra epithelial neoplaia, was once

categorized into four types: mild dysplasia, moderate dysplasia, severe dysplasia and

carcinoma in situ (30). Nowadays these subdivisions have to a large extent been

replaced by the simplified scheme of CIN. CIN I corresponds to mild dysplasia, CIN

II to moderate dysplasia, and CIN III to severe dysplasia and carcinoma in situ.

1.1.5 Cervical invasive carcinoma

The incidences of cervical carcinoma show a highly global variation, ranging from

4.2 to 54.6/100, 000 (34). Weighted age-specific incidence of invasive carcinoma

from European countries shows that the peak of incidence is around 45 years of age

(30). Cervical invasive carcinoma includes adenocarcinoma and squamous cell

Page 11: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

10

carcinoma. Since squamous cell carcinoma accounts for over 85% of cervical

invasive carcinoma and is thought to have relationship with CIN, in my dissertation

only squamous cell carcinoma is considered.

Unlike CIN, invasive carcinoma can metastasize and cause death of the patients

(35, 36). Based on the differentiation of the carcinoma cells, invasive carcinoma is

graded into three degrees: well, moderate and low differentiation. A lower

differentiated invasive carcinoma might easier give rise to metastases and have poorer

prognosis, but some contradictions have been encountered.

Based on invasion and metastasis, the FIGO (the standard of the International

Federation of Gynecology and Obstetrics subdivides the cervical carcinoma cases into

stage I to IV) stages (37) have been used to predict the prognosis of the patients.

Stage I: growth is strictly limited to the cervix. Stage II: carcinoma extends beyond

the cervix but does not reach the pelvic wall and/or involves the vagina but not the

lower third of vagina. Stage III: carcinoma reaches the pelvic wall and/or involves the

lower third of the vagina. Stage IV: carcinoma involves the bladder, rectum or far

organs.

1.1.6 Assumed morphological pathogenesis

Invasive carcinoma often coexists with one or more CIN lesions in the same

cervix. The scene of CIN III breaking through the basal membrane and invading into

stroma can be seen under the microscope some time. The risk of subsequent cervical

carcinoma increases in the patients with CINs. Introduction of cytological screening

and consequent removal of CIN lesions diminishes the incidence of cervical cancer.

CIN occurs with a peak incidence in women about 10 years younger than those with

invasive carcinoma. The CIN lesions commonly are demarcated from surrounding

normal epithelium and show uniform morphology within the lesion, which suggests

that the neoplastic expansion of cells is “locked” in a fixed scheme of differentiation

and expand sideways at the expense of normal cells. Due to these phenomena, a

model of morphological carcinogenesis of cervical carcinoma has been assumed. It is

said that cervical carcinoma originates form a single stem cell and develops via

different degrees of CIN, i.e. putative pre-cancerous lesions, into invasive carcinoma.

However, these arguments are not entirely convincing. Morphological coexistence as

well as subsequent development of carcinoma in patients with CINs could be

explained by a common etiologic process acting over the whole cervical mucosa

Page 12: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

11

rather than by progression from one lesion to the other. Treatment of CINs by

conization or laser will not selectively remove CINs but also most of the target

cervical mucosa susceptible to future development of carcinoma in a putative “pre-

cancerous” but morphologically normal field.

The question if cervical carcinoma originates from a single stem cell and develops

via CINs into invasive carcinoma, or the invasive carcinoma and CINs in the same

cervix originate and develop independently, is crucial for understanding the cause of

cervical carcinoma, the mechanism of carcinogenesis, and the rational design of

treatment and prevention. Nevertheless, morphology limits itself when it comes to

resolving these problems. Molecular studies, especially the clonality analysis has

proven to be a powerful way to address these issues. For example, if invasive

carcinoma and CIN are monoclonal, it will favor the interpretation that cervical

carcinoma originates from a single stem cell and takes multiple steps of

carcinogenesis. Thus, genetic deficiency is likely the initial cause, or more efforts

should be made to search for cellular genetic markers, and it is rational to remove

CINs and expect to have prevented progression to more severe forms of the neoplasm.

Otherwise, if CIN and cervical carcinoma are polyclonal lesions, this will favor the

interpretation that invasive carcinoma and CINs originate and develop independently.

Based on this, the common field factors such as HPV should be considered the cause,

and elimination of the field factors is the best way to treat and prevent this cancer.

1.2 Etiology

Epidemiological investigations provide some proposed risk factors for cervical

carcinoma. Some of the proposed risk factors included are early age at first

intercourse, large number of sexual partners, a male partner himself with many sexual

partners, tobacco smoking, diet poor in fruit, vegetables and some micronutrients

(vitamin C, beta-carotene, folate), use of oral contraceptives, infection by HPV,

presence of HPV DNA in cervical specimens, infection by genital herpes virus, and

presence of certain host cellular genotypes (30). Among those proposed risk factors,

HPV has been proven to be an etiological agent by both the clinical and experimental

evidences. So HPV is one of the main subjects focused on in my review and study.

1.2.1 Human Papillomavirus (HPV)

Page 13: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

12

1.2.1.1 Epidemiology

Early epidemiological data have long suggested that cervical neoplasia is a

venereally transmitted infectious disease. Brinton found that the risk of cervical

neoplasia is increased among women who initiate sexual intercourse at early ages,

who have great numbers of sexual partners and who have non-related diseases (38).

Later on, epidemiologists have studied the prevalence of HPV infection in normal

control women (39-41) and women with neoplasms (42). They have also studied the

shared risk factors of HPV and cervical neoplasia (40, 42-44), and whether HPV

infection predicts the development, recurrence, or pathological progression of the

cervical neplasia (45-47). If HPV is the venereally transmissible cause, positivity of

HPV should have a higher prevalence in the cases with neoplasia than in the normal

control individuals, HPV should share many of the same risk factors as cervical

neoplasia, and HPV infection should be a predictor of the neoplasia. Most although

not all studies of these kinds support that HPV is the cause of cervical neoplasia. The

results from different studies varied a lot and proven to be strikingly dependent on

methods of cervical tissue sampling and DNA detection (48-51).

1.2.1.2 Organization of HPV

The virion consists of a small capsid and a one-coding DNA strand. The capsid is

about 55nm in diameter consisting only of protein. Analysis of electron micrographs

reveals 12 five-coordinated and 60 six-coordinated capsomers arranged on a T= 7

surface lattice (52). After negative staining the capsomers appear as hollow cylinders

of equal height and width, which are connected at their base by fibrous structures

(53). The HPV capsid consists of two structural proteins. One protein with a Mr in the

range of 53, 000-59, 000 (54-56) represents 80% of the total viral protein. A minor

component has an average Mr of 70, 000. The two proteins are coded for by two open

reading frames of the HPV DNA.

Mature HPV particles contain the viral genome in the form of a double-stranded

circular DNA molecule. For the majority of HPVs, the size of the genome is about 7.9

kb (55, 57-59). Some variations have been observed in the range of ±10% (60). The

GC content of the HPV genome is rather low. The average value is 42.6%, the

extreme cases being HPV16 (36.5%) (61). There is no clear GC distribution pattern

Page 14: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

13

throughout the genome with the exception of the non-coding regions that show

slightly lower GC values as a rule.

HPV genomes reveal a well-conserved general organization. All putative open

reading frames (ORFs) are restricted to one DNA strand. The second, presumably

non-coding strand contains only short ORFs, which are conserved neither with regard

to localization nor composition. This is in line with all known data on viral

transcription, which uniformly point to a single sense strand. Most major ORFs

occupy similar positions relative to each other and there is no substantial difference in

the length of homologous ORFs, which can be used to align the genomes of different

HPVs. The individual frames are classified as “early” (E) or “late” (L) genes in

analogy with other DNA viruses, where genes are turned on according to a specific

time schedule in the course of a productive infection. The so-called early genes are

expressed shortly after infection and prior to the onset of DNA replication. Products

of these genes mediate specific functions controlling replication and expression of

viral DNA. In the case of tumor viruses, early gene products are also involved in

transformation of the host cell. The late genes code for structural proteins of viral

particles and are activated during the final stages of the viral cycle. Up to six early

genes and two late genes can be detected in HPV (62).

1.2.1.3 HPV detection and typing

Epidemiologists often use at least four different techniques of collecting

exfoliated cervical cells for HPV testing: swab, scrape, cytobrush and lavage.

Cervical lavage has been shown to be the most sensitive non-tissue method to harvest

cells for molecular identification of HPV (48). Pathologists would either use cervical

tissue. Once cervical samples are collected, multiple methods of light microscopy,

electron microscopy, immunohistochemistry and molecular biology can be used to

identify HPV. Molecular biology is now commonly used. Southern blot (48, 63, 64),

Northern blot (50), Dot (Spot) blot (63, 65), Filter In Situ (66) , and In Situ (63, 64,

67) hybridization are applied for HPV detection and typing. Recently PCR has been

developed to amplify DNA sequences shared by many of the 20 or so different types

of genital HPVs (51). However, no “gold standard” has yet been established (51, 68-

70).

1.2.1.4 HPV types and infection of the human tissue

Page 15: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

14

A new type of HPV is defined by having less than 90% homology with other types

when comparing the DNA sequence of the complete L1 coding region (71). HPVs

reveal a remarkable plurality of different genotypes. Until now, 85 HPV types have

been identified and fully characterized. Thus far, all identified types appear to be

strictly epitheliotropic (72). They infect the skin (73, 74) or epithelium of external

genital and anal regions (75), oral cavity (76), larynx (77, 78), lung (79), and

esophagus (80-82). No evidence for HPV infections has yet been found in the gastric,

ileojejunal, or colon mucosa. They may cause warts, verruciform, benign tumors and

malignant tumors. Only a limited number of the approximately 40 types infect the

anogenital tract. This recalls the different biological activities of individual HPV

types (72).

1.2.1.5 Prevalence of HPV DNA in cervical neoplasia

Most early investigators have observed an elevated prevalence of HPV in women

with existing cervical neoplasia compared with control women. Because the

prevalence of HPV among cytologically normal women has ranged unbelievably from

5% (83) to 80% (84), the estimated magnitude of the increase varied substantially,

primarily. The impression is that misclassification of HPV status may have limited

the first generation of epidemicological studies. With new and better methods for the

HPV assay, recent case-control data have firmly established that a great majority of

women with existent cervical neoplasia concurrently has detectable HPV DNA. In

contrast, the control women have a much lower percentage of HPV DNA (85-89). In

India, 98% of cases with cervical neoplasia and 18% of normal control women are

HPV positive (89); in Taiwan, 92% of patients with cervical neoplasia and 9% of

normal control women are HPV positive (86).

The relation between specific HPV types and different degrees of cervical

neoplasia has been investigated. Although up to 40 congenital types of HPV have

been identified, the distribution of the types of HPV to the cervical neoplasia is

uneven. HPV6, 11, 42, 43, and 44 mainly distribute in condyloma and CIN I (90-94);

HPV 30, 31, 33, 35, 39, 40, 49, 52, 59 and 68 are often seen in CIN II and III (91, 92,

95, 96); and HPV16, 18, 45 and 56 are the major types of HPV seen in CIN III and

invasive carcinoma (90-94, 97), with about 50% of invasive squamous cell

carcinoma infected by HPV16 (98). The HPVs, such as HPV6 and 11, mainly present

Page 16: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

15

in low grade cervical neoplasia are defined as low risk types of HPV. The HPVs, such

as HPV31, 33, 59, often seen in moderate grade of cervical lesions are classified into

intermediate risk types of HPV. The high-risk types of HPV, such as HPV16 and 18,

are the ones found to be present in the high-grade lesions of cervical neoplasia.

HPV infections in normal cervix are often transient. Many HPV infections

disappear in about two years (71, 99). Most cases of HPV infected women have been

found with only one type of HPV present, but a low percentage of the cases has

double or multiple types of HPV (47, 100). Fewer cases of cervical carcinoma or high

grade CIN lesions than cases with normal cervix or cervix with low grade CIN lesions

can be found with double or multiple types of HPV infection (47). This indicates that

the immunogenicity of the original infection of HPV may be efficient to prevent

against infection of other types of HPV in many cases.

1.2.1.6 HPV is the main cause of cervical neoplasia

So far no specific oncogenes or tumor suppressor genes have been strongly

associated with cervical neoplasia. HPV considered the major cause of cervical

neoplasia has drawn most attention from the scientists who engage in studies of this

issue. Due to the plenty of efforts, data accumulate to prove that HPV is the cause of

cervical neoplasia. The evidence include the following. Cervical neoplasia is a

venereally transmitted infectious disease. The prevalence of presence of HPV in

patients with cervical neoplasia is higher than in normal control women. HPV shares

many of the risk factors with cervical neoplasia. Specific types of HPV infection

predict the development of the cervical neoplasm (47, 71, 99). Experimental data

reinforce the conclusion. The proteins of the viral oncogenes E6 and E7 of some types

of HPV can degrade p53 and pRb and transform epithelial cells in vitro (101-104).

However, only a minority of women with HPV infection develop any cervical lesions

(71, 105). This leads to an adjustment of the conclusion suggesting that HPV is the

necessary but insufficient cause of cervical neoplasia. More than HPV infection is

needed to cause cervical neoplasia. In other words, there must be some further risk

factors or cofactors that take part in the process of cervical carcinogenesis.

1.2.2 Cigarette smoking

Page 17: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

16

Although studies show no direct association between smoking and cervical

neoplasia (106), there are evidence that cigarette smoking increases the risk (87, 107-

111). Some observations have also suggested that there may be an interaction

between HPV and smoking in the causation of cervical neoplasia (112, 113).

However, Reeves reported that smoking is significantly associated with cervical

carcinoma (52% of cases vs. 27% controls) but is not associated with HPV infection

(110). The confusion about the possible relation between smoking and cervical

neoplasia and between smoking and HPV infection indicates that smoking might be a

weak factor to impact the development of cervical neoplasia and the infection of

HPV. It is difficult to control the investigative settings of epidemiological studies to

get consistent results. Seemingly, cigarette smoking at the most is a minor cofactor of

some specific types of HPV in certain populations.

1.2.3 Hormonal contraception

It has been shown that long-term use of hormonal contraceptives tends to increase

the risk of cervical neoplasia (111, 112, 114-116) or increase the HPV infection (117).

By contrast, women using barrier contraceptives and spermicidal foams or jellies

seem to be at lower risk for this neoplasm (118). However, Pike reported that

hormonal contraceptive reduces the risk of cervical neoplasm (119). So, similarly

with smoking, hormonal contraception might be another weak cofactor of HPV

infection in the development of cervical neoplasia.

1.2.4 Inheritance

Since only a small number of women infected by HPV (considered the main cause

of cervical carcinoma) develop any lesions, it prompts people to search for further

risk factors. Genetic factors are an attractive field although so far no specific

oncogenes or tumor suppressor genes have been found. Magnusson et al. found a

significant family clustering of Swedish cases among biological, but not adoptive,

relatives of patients with cervical tumors, which provided an epidemiological

evidence of a genetic predisposition to cervical carcinoma (120). Several other

articles with similar conclusion were also published (121-124). They explained that

genetic susceptibility to HPV infection appears to be important in determining the

individual risk to develop this virally induced cancer.

Page 18: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

17

1.3 Molecular Biology

1.3.1 Clonality of cervical neoplasms

It is generally believed that a single cell accumulates a randomly occurring set of

somatic mutations and then alone is responsible for developing a tumor because it is

unlikely for a number of cells to catch the same set of genetic events simultaneously.

Human tumors are therefore thought to be monoclonal and the monoclonality has

been taken to serve as a hall-mark of neoplastic proliferation and a diagnostic criteria

for some tumors. Clonality analysis will be a key issue to understand the cause and

mechanism of carcinogenesis of cervical neoplasia.

1.3.1.1 Strategies of clonality analysis

Currently the strategies of clonality analysis of tumors include X-chromosome

inactivation based analysis, somatic mutation detection, genetic rearrangement based

assay of lymphocytes and viral integration analysis. They are applied in different

situations. The former two are used for clonality analysis of most types of tumors. In

X-chromosome inactivation analysis, the restriction fragment length polymorphism

(RFLPs) of the G6PD (glucose-6-phosphate dehydrogenase) gene, PGK

(phosphoglycerokinase) gene, HPRT (hypoxanthine phosphoribosyltransferase) gene

(125-128) and microsatellite repeat polymorphism of the AR (androgen receptor)

gene (129) have been used. For somatic mutation analysis, genetic deletions,

chromosomal translocations and specific point mutations are often detected. The

immunoglobulin assay is used for analysis of B- lymphocyte tumors and T-cell

receptor gene assay is used for T-cell tumors. Genetic EBV termini, HIV and HPV

integration sites have been used to analyze the clonality status of the tumors harboring

the corresponding virus.

1.3.1.2 Polymorphism of inactivation of X-chromosome linked androgen receptor

gene

Because of the fact that up to 90% of the cases turn out to be informative for the

androgen receptor gene, the polymorphism of this gene is used the broadest for the

Page 19: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

18

clonality analysis of tumors. Thus, here is introduced the working principle of this

analysis.

One X-chromosome, the one from the mother or the one from the father in an

embryonic stem cell is randomly inactivated by methylation of CpG sites at a stage

with about 50 cells in the female embryo to avoid double dosage of the genes on X-

chromosomes. The inactivation pattern of the X-chromosome in a stem cell is stably

inherited by subsequent descendants forming a cell lineage (130-132). Therefore, a

female body including cervical epithelium develops into a mosaic of Xm- and Xp-

inactivated cells. Cervical carcinoma cells derived from a committed epithelial stem

cell with a certain Xm- or Xp-inactivation pattern are equipped with the same pattern

as their progenitor, and this pattern serves as the clonality sign of the carcinoma.

Located at the first exon of the human androgen receptor gene, a highly polymorphic

microsatellite consisting of a short tandem repeat, [CAG]n (n=11-13), is about 100bp

away from HpaII and HhaI (methylation sensitive) restriction sites. This

polymorphism of the microsatellite allows the use of enzyme cleavage of DNA and

PCR-fragments in an assay to identify the X-chromosome inactivation pattern of the

cells tested (133, 134). Figure 1 shows how the analysis works and how the

interpretation is done. However, the interpretation of the clonality information for a

XX Embryonic stem cell

XX XX Random inactivation

XX XX.XX XX Mosaic of cell lineage

Enzymatic digestion

Bands of PCR product

Monoclonal Polyclonal Monoclonal

Page 20: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

19

Fig. 1. Technique of X-chromosome inactivation analysis. Before digestion with

methylation sensitive restriction enzyme HpaII, the informative sample gives two

bands. After digestion, the active allele of an X-chromosome linked gene is destroyed.

Only the inactivated allele is amplified and will give a band. One band indicates the

presence of one type of cell population in the analyzed sample (monoclonality). Two

bands indicate more than one type of cell populations in the analyzed sample

(polyclonality).

few samples with the same X-chromosome inactivation pattern in an individual

requires further markers.

To get reliable and reproducible results, one has to take notice of some facts (135).

In larger cell populations, the average numbers of paternally- and maternally-

inherited inactivated X-chromosomes is close to 50:50 due to the random inactivation

of the X-chromosomes, but in small cell populations, it would be possible to find

skewing towards one allele to an extent that meets the criteria for clonal derivation.

This kind of skewing is different from tissue to tissue in the same patient and

probably from case to case. So in every case, the tumor sample must be compared

with matched controls from the same patient to test the heterozygosity for the marker.

Fine microdissection to separate the tumor cells and normal cells as much as possible

is one of the requirements. The sample size is also a limiting factor. The lower the cell

number, the higher is the probability to detect a monoclonal pattern based on patch

size mosaic in normal cells. More than 100 cells each in multiple microdissected

samples from different tumor areas and from controls are required. The sample

number from the same patient is one among other impact factors. As many samples as

possible should be taken to represent the whole situation of the tumor. Good

purification of DNA samples and optimal enzymatic digestion of the samples are

always important factors.

1.3.1.3 Clonality features of cervical neoplasms

Evidence shows that cervical neoplasia can be monoclonal or polyclonal. More

cases with monoclonal origin than cases with polyclonal origin have been reported.

This actually depends on the type of cervical neoplasm analyzed. The lower grade of

the cervical neoplasia (toward CIN I) has higher frequency of polyclonality.

Page 21: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

20

With inactivation polymorphism of the X-chromosome linked androgen receptor

gene as the clonality marker, some efforts on clonality analysis of cervical neoplasia

have been made. Park et al. demonstrated that 100% of CIN III (25/25) and 68% of

CIN II (54/79) are monoclonal, while 32% of CIN II (25/79) are polyclonal (136).

Enomoto et al. found that 100% CIN III (30/30) are monoclonal and found 1 case of

CIN II to be polyclonal. Six cases of cervical invasive carcinoma were found to be

monoclonal and their synchronous CIN lesions shared the clonality composition with

the corresponding invasive carcinoma (137). In another study, Enomoto et al. found

all 13 cases of cervical squamous cell carcinoma and 6 cases of adeneocarcinoma to

be monoclonal (138). Based on the finding of a 100% monoclonality in cervical

carcinoma (12/12) including two early invasive carcinomas, Guo et al. pointed out

that genetic events are critical in the transition of pre-malignant epithelia to invasive

cancer in cervical carcinogenesis and that monoclonality of human tumors is not a

late event during the process of clonal competition or selection (139). In a series of

studies, however, Guo et al. found that among 22 cases of cervical invasive carcinoma

and synchronous CIN lesions two thirds of these cases were monoclonal, while the

remaining CINs were polyclonal (140). Later on, they found that 25% of cervical

invasive carcinoma ((2/8) were polyclonal (141). In line with these results, Ko et al.

reported that 33.3% of cervical invasive carcinoma cases (6/18) are polyclonal (142).

These reports indicate that the pathogenesis of cervical carcinoma is probably even

more complicated than that of many other types of cancers involving selection of sub-

clones of different clonal composition. In a study with 64 cases of cervical neoplasms,

Chuaqui et al. showed that invasive carcinoma and CIN III had high percentages of

cases with monoclonality and identical LOH patterns on chromosome 6q, while CIN I

and II had low percentages of cases with monoclonality and identical LOH patterns

on chromosome 6q, suggesting that CINs develop into invasive carcinoma through

progression from polyclonal lesions to monoclonal lesions (Table 1) (143).

PGK is also an X-chromosome linked gene. Sawada et al. used the X-

chromosome inactivation polymorphism of PGK to analyze the clonality of 25 cases

of cervical invasive and metastatic carcinoma. They found that all analyzed cases

were monoclonal and the same allele of the PGK gene was inactivated in the primary

invasive carcinoma and the metastatic carcinoma in each case (144).

LOH is another commonly used clonality marker (145). However, LOH patterns

are not always identical in all cases in other studies. A distinct type of cervical

carcinoma, adeno-squamous cell carcinoma, contains an adenocarcinoma component

Page 22: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

21

Table 1. Monoclonal patterns as seen in analysis of the AR inactivationand identical LOH patterns on chromosome 6q in cervical neoplasms(ref. 143). AR: X-chromosome linked androgen receptor geneinactivation.

ICC (%) CIN III (%) CIN II (%) CIN I (%)

AR 92 93 20 0

6q 55 40 37 10

and a squamous cell carcinoma component in the same case. Kersemaekers et al.

performed LOH analysis on nine chromosomes, i.e., chromosomes1, 2, 3, 6, 11, 15,

17, 18, and X, in two cases of adeno-squamous cell carcinoma. Many genetic

alterations were the same in each component in both cases, which indicates that the

adeno-squamous cell carcinoma most likely has one cell origin. But the presence of

some differences of genetic changes associated with the component in one cases favor

a diversion of different development (146). With the combination of LOH analysis

and X-chromosome inactivation analysis, Guo et al. found two of eight cases of

cervical invasive carcinoma to be of polyclonal origin (141).

Park et al. even associated the clonality status with HPV types in cervical

neoplasia (136). All cases of 24 high-grade CIN and 47 out of 71 cases of low-grade

CIN were found to be monoclonal and contain HPV types 16, 18, 33, 35, 45, 56, 58 or

65. In contrast, 22 of 71 cases of low-grade CIN were polyclonal and contained other

types of HPV. Their findings suggest that the histopathological entity termed low-

grade CIN consists of two different types of lesions that are biologically distinct.

1.3.1.4 Clonality status of some other common human tumors

Most cases of other kinds of human tumors show monoclonality (147-151). This

favors the theory that genetic defects cause most tumors in a model of multiple steps.

Simultaneously randomly occurring multiple genetic events in multiple cells are very

rare. One progenitor cell that undergoes a series of genetic changes required for the

initiation of lesions will give rise to a monoclonal neoplasia. So monoclonality is

believed to be a hallmark of neoplastic proliferation. In some tumors, clonality

Page 23: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

22

analysis has been used as a tool to diagnose the tumor, to exclude hyperplasia or

responsive proliferation, and to monitor the response to treatment (148, 152, 153).

However, there are a significant number of reported cases of different kinds of tumors

with polyclonality. In 11 cases of multiple squamous cell carcinomas of the

aerodigestive tract the p53 mutations and LOH patterns were stable during tumor

progression, and different p53 mutations or LOH patterns in multiple tumor areas

were observed in each case (154). In a study of bilateral breast carcinoma, four of 12

cases were scored as being of polyclonal origin (155). Yamamoto et al. found that two

of eight cases of multiple human hepatocellular carcinomas were monoclonal (had

intrahepatic metastases) and six of eight were polyclonal (showed independent

multicentric occurrence) (156). Sporadic medullary thyroid carcinoma has often been

found to result from a mutational event occurring at the single cell level and therefore

should be monoclonal, however, 10 of 11 such cases showed polyclonal patterns

(157). Polyclonal origin in some cases can also be seen in other tumors such as

“Kaposi” sarcoma (158), benign breast tumors (159, 160), and multifocal urothelial

papillary tumors (161).

While the origin of tumors, whether from one cell or many, has been a source of

fascination for experimental oncologists for many years, in recent years there has

been an explosion of information. Although these results have apparently confirmed

the monoclonal origin of tumors, there are some studies in which this conclusion just

cannot be made. The potential impact of such considerations as the patch size of a

normal clonal cell population and clonal evolution on the determination of clonality

has largely been ignored, with the result that a number of these determinations are

confounded. It is clear, for many reasons, that more efforts should be put into the

techniques of analysis (162).

1.3.2 HPV16

HPV16 is the most commonly seen type of HPV in cervical squamous cell

neoplasia, so this chapter mainly describes the features of HPV16.

1.3.2.1 The structure of the HPV16 genome and the function of HPV16 genes

The DNA sequence of the HPV16 genome contains 7905bp (61). HPV16 has a

long terminal region (LTR) and eight genes including six early genes and two late

Page 24: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

23

genes. The six early genes are E6 (495bp, nt 65-559), E7 (315bp, nt 544-858), E1

(1955bp, nt 859-2813), E2 (1128bp, nt 2725-3852), E4 (287bp, nt 3332-3619), and

E5 (296bp, nt 3804-4100). The two late genes are L2 (1523bp, nt 4134-5657) and L1

(1630bp, nt 5527-7155). The LTR is 815bp long located between E6 and L1. Fig.2

shows the structure of the HPV16 genome. E6, E7 and E5 are viral oncogenes

encoding proteins with growth-stimulatory and transforming properties.

The E6 protein is an approximately 150 amino acid protein that is localized to the

nuclear matrix, as well as to non-nuclear membrane (163). E6 is not normally capable

of inducing transformation by itself, but has been shown to induce immortalization of

primary human keratinocytes in conjunction with E7 (164), as well as to promote

anchorage-independent growth of rat cells (165). Binding and degrading cellular p53,

E6 intervenes with the cell cycle and functions as an anti-apoptosis factor (104). E6

can also destabilize chromosomes (166), enhance foreign DNA integration and

mutagenicity (167) and activate telomerase (168).

The E7 protein is a 98 –amino acid protein that is located in the cytoplasm of cells

(169). Expression of E7 alone in epithelial cells is sufficient for transformation, but

E7-mediated transformation is much more efficient when co-expressed with the HPV

E6 protein (170). E7 can bind and inactivate Rb protein and Rb-related pocket

E6

E7

E1

E2

E4L2

L1

LTR

Fig. 2. The genome of HPV16

Page 25: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

24

proteins to deregulate the cell cycle (101, 171). To intervene with the cell cycle, E7

also inhibits cyclin-dependent kinase inhibitors (172) and activates cyclins E and A

(173). Like E6 protein, E7 is able to enhance foreign DNA integration and

mutagenicity (174).

The E5 protein is a small hydrophobic protein located in cytoplasmic and plasma

membranes (175, 176). It complexes with a variety of other trans-membrane proteins,

such as the epidermal growth factor receptor, platelet-derived growth factor β-

receptor, and colony-stimulating factor-1 receptor (175). The E5 protein also binds to

adenosine triphosphatase (ATPase) in membrane (177). The HPV16 E5 protein

possesses weak transforming activity and induces a protein kinase-mediated, PKC-

independent, activation of membrane-associated protein kinase (178, 179). Its

transient induction in mouse 3T3 cells or immortalized human keratinocytes results in

suppression of the cyclin-dependent kinase inhibitor p21 and in the induction of c-jun

expression (180), and enhances endothelin-1-induced keratinocyte growth (181). The

expression pattern of E5 is less well established, but like E6 and E7, it is also

correlated with abnormal cervical cytology and can be detected in all stages of

development of cervical carcinoma (182, 183).

The E2 gene codes for two proteins with a transcriptional regulatory function. The

E2 protein has a transcriptional activation domain in the N-terminus and a DNA

binding domain in its C-terminus. The E2 gene can be expressed in two forms, a

complete protein and a short protein with only the DNA binding domain (184). Both

E2 and E1 proteins are necessary and sufficient in vivo for efficient viral DNA

replication. The ability of E2 and E1 to complex with each other appears to be

essential for efficient viral DNA replication (185, 186). E1 may play a role to

maintain HPV in episomal form, while E2 may regulate expression of E6 and E7

(163). Mutation of E2 increases immortalization capacity (187). The E4 protein

localizes to cytoplasmic inclusion granules and results in the collapse of the epithelial

cell intermediate filament network (188, 189). E4 expression is abundant in CIN and

condyloma, but the significance of this is unknown. L1 protein is the major capsid

protein and L2 is the minor one. Both L1 and L2 constitute the viral coat protein that

surrounds the viral DNA genome to assemble a complete infectious HPV particle

(190). LTR is a region of the genome adjacent to the E6 open reading frame. It does

not encode any protein but rather contains promoters and enhancers that influence

viral gene transcription (191).

Page 26: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

25

1.3.2.2 HPV16 variants in progression of cervical carcinoma

HPV variants are defined as those HPV DNA sequences differing from each other

by less than 2% of nucleotides (71). Variations refer to any change of nucleotides

compared to the “prototype” nucleotides at the same positions. Based on the

variations in E6, L1 and LTR, HPV16 has been classified into six classes: E-350T, E-

350G, As, AA, Af1, and Af2. The geographical distribution of HPV16 variant classes

by continents is shown in table 2 (192). This classification can not reflect the

Table 2. Distribution of HPV16 variant classes by continent (Ref. 192)

No. (%) of specimens from:

Variantclass

Europe NorthAmerica

CentralSouthAmerica

SoutheastAsia

Africa Total no.

E-350T 20(40.0) 16(53.3) 56(24.6) 19(54.3) 5(7.8) 116E-350G 22(44.0) 12(40.0) 119(52.2) 2(5.7) 1(1.6) 156As 1(2.0) 1(3.3) 0(0.0) 9(25.7) 0(0.0) 11AA 7(14.0) 0(0.0) 45(19.7) 2(5.7) 0(0.0) 54Af1 0(0.0) 0(0.0) 5(2.2) 0(0.0) 39(60.9) 44Af2 0(0.0) 1(3.3) 3(1.3) 3(8.6) 19(29.7) 26

Total 50 30 228 35 64 407

biological function of HPV16 variants. When studying the clinical significance of

HPV16 variants, some people classified the HPV16 sequence into “prototype” and

variants groups, or “prototype”-like and “non-prototype”-like groups. Some other

studies use more precise categories.

The HPV16 sequence has frequent variations in the whole genome, particularly in

E6 (47, 71, 100, 192-195). The variants are reported to have different biological and

biochemical properties. In prospective studies, women with HPV16 “non-prototype”-

like variants were 6.5 times more likely to develop CIN II-III than those with

“prototype”-like variants (71). Londesborough et al. found that only 1 of 16 women

infected with the HPV16 prototype developed CIN or invasive cancer; in contrast,

Page 27: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

26

10/12 women infected with HPV16 E6 variants had persistent infection which was

associated with development of CIN and invasive cancer (47). Zehbe et al. concluded

that sequence variation in HPV16 E6 predicted risk of progression from CIN III,

because 15/16 cases of ICC contained variant E6 in contrast to CIN III where only

11/25 had variant E6 (196). Alvare-Salas et al. showed that variants of HPV16 E6

correlated positively with clinical aggressiveness (193). Stöppler et al. described that

variants of HPV16 E6 protein differed in the abilities to suppress keratinocyte

differentiation and to induce p53 degradation in vitro (197). HPV16 E6 variants

represent a significant risk factor is common in both Western and Japanese women

despite the different distribution of each variant in them (198). Variants of HPV16 E7

could affect the properties of E7, including the binding to pRb (199).

However, the results are not geographically uniform. In women living in the

South London area, the E6 variants were identified in only 3/95 (3%) individuals, two

of whom had normal histology and one had a CIN II lesion. Wild type E6 sequence

was identified in the remaining 92/95 (97%) individuals. These data suggest that the

E6 variant does not play a major role in the pathogenesis of HPV16 related cervical

disease (200). Fujinaga et al. found that E7 variants were distributed uniformly among

CIN lesions and invasive carcinoma and the variant proteins had transforming

potential similar to the “prototype” E7 (201). Terry et al. reported that there were no

co-relation between HPV16 E2 variants and the grades of cervical neoplasia (202).

There was no significant correlation between HPV16 variant incidence and disease

progression or viral persistence and no significant correlation with any HLA allele

(203).

1.3.2.3 Physical stage of HPV16

Generally, HPV16 DNA is a circular molecule and remains episomal, i.e., it

remains separate from the host chromosome. When the circular molecule breaks at a

point it may linearize to integrate into the chromosome by two end-points. HPV16

can be in an episomal or an integrated form inside the infected cells. Theoretically the

two different physical stages would represent different biological status because the

breaking point must destroy the integrity of the viral genome and the integrated viral

genome must be out of control by itself and be controlled by or interact with the

cellular DNA elements. The E1/E2 gene region is the common place to be broken

when HPV16 linearizes to integrate into the host chromosome. Since E2 protein can

Page 28: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

27

regulate the expression of E6 and E7, loss of E2 function may release the control of

E6 and E7 expression. High expression of E6 and E7 increases the immortalizing

capacity of HPV16. It is believed that one of the important events contributing to the

development of cervical neoplasia is viral integration, with subsequent loss of the E2

expression, and enhanced E6 and E7 expression. The integration status of the viral

DNA may also be important because the viral genome may integrate into a site that

results in alteration of human gene expression or its regulation (163). Additional

evidence for contributing effects of E2 loss of function comes from the studies in

vivo. In cells transfected with HPV16, the viral DNA was integrated in the genome of

the host cell, and these cells acquired a growth advantage that could not be explained

by the known effects of other retained viral genes, E6 and E7 (204). Later on, it was

demonstrated that the reintroduction of a normal E2 protein in Hela cells, suppressed

the growth of these cells (184). For this effect to take place, both the transactivating

and DNA binding domains of this E2 protein have to be intact (184).

Although HPV16 integration into host DNA is found in all cases of cervical

carcinoma (98), their metastases and derived cell lines (205), many cases of cervical

carcinoma contain both episomal form and integrated form of HPV (206-209). The

deficient function of the genes in the integrated form of HPV16 can be compensated

for easily by the corresponding function of the genes in the episomal form of HPV. In

addition, some cases of cervical carcinoma retain only episomal form of HPV16

(209). Hall et al. established a cervical carcinoma cell line that harbors episomal

copies of HPV16 DNA of approximately10 kb. Restriction enzyme and two-

dimensional gel analysis confirmed that HPV16 DNA was extra-chromosomal with

both monomeric and multimeric forms present. HPV16 was maintained as episomes

during passage both in culture and after subcutaneous growth in nude mice. The10 kb

viral genome, consisting of a full-length copy of HPV16 and a partial duplication of

the long terminal region and the L1 open reading frame, exhibited transforming

activity comparable to prototype HPV16 (210). These studies suggest that HPV16 can

transform target cells by mechanisms that do not require viral integration. In contrast,

many CIN cases contain an integrated form of HPV16 (211, 212).

The consequences of HPV16 integration on the cellular genome seem not to be

important because there is a lack of homology between the locations of integration

sites. Several integration sites and their corresponding normal target sequences have

been cloned and studied (213-216). Sequence comparison does not detect any

significant homology among different cellular target regions. This lack of homology

Page 29: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

28

suggests that the integration of HPV16 is a random event. Taken together, HPV16

integration seems not as important in the carcinogenesis of cervical neoplasia as

people ever thought it was.

1.3.3 Loss of heterozygosity (LOH)

The LOH means that a particular DNA region has been lost, and therefore, if this

loss is recurrent in a significant number of cases of a particular tumor type we can

conclude that this chromosome region plays a role in that tumor. LOH is generally

thought of as an intermediate step in the inactivation of tumor suppressor genes, such

as p53 or pRb, which requires the inactivation of both alleles in order to display their

phenotype. However, in most cases, the LOH reflects a hemizygous situation and its

effect might be a dose effect rather than total inactivation of a tumor suppressor gene.

Analysis of LOH has been broadly applied to cervical neoplasms. Like many other

tumors, cervical neoplasms have extensive deletions in multiple chromosomes.

1.3.3.1 LOH on chromosome 3p

Chromosome 3p is one of the most frequently deleted chromosome regions in

cervical neoplasms. A broad region, 3p12-24 was identified as a target for LOH. The

data show that the prevalence of LOH in 3p varies in different studies, ranging from

35% to 100% depending on the number of markers and the different 3p loci selected

(217-221). The LOH in 3p might be an indicator of tumor progression and involve

tumor suppressor genes located at the region. Larson et al. reported that the frequency

of LOH in 3p increases from 25% in FIGO stage I cases to 100% in stage IV cases

(220). Some studies of CIN cases found that LOH in 3p occurred in about 30% of the

patients (222-224). These results suggest that the LOH at many loci is just a neutral

sign of evolution of cervical neoplasia, only those that occur during progression from

the early to the late stage lesions are non-random genetic events. Several studies agree

that within the region 3p14-22 there are two sub-regions where the deletions

concentrate, and frequently both are altered in the same tumor. These two regions are

3p14.2 and 3p21 (218-221), where tumor suppressor genes might harbor.

Recently, the FHIT gene has been identified within the chromosome region

3p14.2. A high incidence of LOH was also described at this region and FHIT is

altered in several other types of human tumors, such as lung (225), breast (226), and

Page 30: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

29

esophageal carcinoma (227). The FHIT gene comprises the FRA3B fragile site at

which HPV16 spontaneously integrated (228). Although aberrant FHIT transcripts

were detected in 11 (63%) cervical carcinoma cell lines and in 35 out of 74 (47%)

primary cervical carcinomas of different types and stages, similar aberrant transcripts

were detected in normal tissues in 12/31 (39%) of the cases (229). A similar

phenomenon that both tumor and normal tissues contained aberrant transcripts could

also be detected in cases with some other types of solid tumors, and thus the

functional importance of the FHIT gene as a tumor suppressor gene has been

challenged.

In the 3p21 region the affected gene is not known. This region contains the

hMLH1 gene. The hMLH1 gene is one of the DNA mismatch repair genes which are

indispensable for DNA replication fidelity including the stabilization of short

nucleotide repeats (230-232). When the functions of the DNA mismatch repair system

are deficient, carcinogenesis might occur with the phenotype of microsatellite

instability in some types of tumors such as colon cancer (233), breast cancer (234)

and so on. Two mechanisms can inactivate those DNA mismatch repair genes, i.e.,

hypermethylation and mutation. Although the LOH at an hMLH1 locus can be

detected in up to 22% of cervical carcinomas (235) and the microsatellite instability

frequency is about 15% in cervical carcinoma (236), it is not clear if alterations of

hMLH1 expression and mutation in this gene occur in cervical carcinoma.

1.3.3.2 LOH on chromosome 6p

In cervical carcinoma studies have shown a high incidence of LOH at

chromosome 6p21.3-25 where HLA is located (237-240). As seen in in vitro

functional studies, chromosome 6 transferred into breast and ovarian cancer cells can

successfully suppress the tumorigenesis and metastatic ability of the target cells,

indicating potential location of tumor suppressor genes and cell scenescene genes on

this chromosome (241, 242). However, it remains to be determined if allelic losses of

chromosome 6 represent an early genetic event and play a role in the progression of

cervical pre-cancer.

1.3.3.3 LOH on chromosomes 4, 5p, 11, 17, 18 and 19

Page 31: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

30

LOH on chromosomes 4, 5p, 11, 17, 18 and 19 can be seen in cervical neoplasia

(237, 238, 240). The introduction of chromosome 4 into Hela cells conferred a

senescence phenotype, which suggests that this chromosome might be important in

cervical carcinoma (243). Tumor suppressor genes might harbor at two regions, 4p16

and 4q21-35, frequently showing LOH in cervical neoplasms (240, 244). Two

different regions with LOH have been identified on chromosome11. The

chromosome11p15 region showed LOH in 12 out of 43 cervical carcinomas (28%)

(238) and LOH at 11q23 has a strong correlation with the presence of an invasive

carcinoma in FIGO stage I and II (240). Chromosome 17 was the first to attract

attention in cervical carcinoma because it contains the TP53 gene in the 17p3.3 band,

which is frequently mutated in many types of tumors. The frequency of LOH it

17p13.3 is 24%, and often it dose not affect the TP53 locus, thus, suggesting that the

candidate tumor suppressor gene in this region must be different from TP53 (245).

Although LOH occurs frequently on many chromosomes in cervical carcinoma and

some regions with a high frequency of LOH have been identified, no tumor

suppressor genes causally related to cervical neoplasia have been found so far.

1.3.4 Point mutations and gene polymorphisms

In contrast to loss of genetic material, recurrent amplifications and gain at

chromosomes can be seen in cervical carcinoma. Microsatellite instability is another

type of genetic change seen in cervical neoplasms, but the incidences of those

changes are relatively low. This chapter is going to briefly review some point

mutations and polymorphisms, which might be involved in cervical carcinoma.

1.3.4.1 Point mutations in TP53

Some studies have attempted to detect mutations in genes well known to have

point mutations in other tumors, including TP53, RB, H-RAS, and other cell cycle

genes. TP53 has been shown with a rather low frequency (less than 10%) of mutation

(246, 247), while others have reproted even much lower frequency of mutations in

cervical carcinoma. TP53 appears to be more frequently mutated in HPV-negative

tumors, but this negativity has been questioned since nowadays all tumors are

considered HPV-positive (98). This has lent support to the interpretation that the

mechanism implicated in these tumors is likely to be a consequence of the interaction

Page 32: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

31

of the HPV E6 viral protein with the p53 protein, and by which they could achieve a

biological situation resembling a partial p53 defect.

1.3.4.2 Susceptibility of patients with TP53 codon 72 polymorphism to HPV

infection

Storey et al. first reported the effect of p53 polymorphism on the susceptibility to

HPV infection (248). The E6 oncoprotein derived from tumor-associated HPVs binds

to and induces the degradation of the cellular tumor-suppressor protein p53. A

common polymorphism that occurs in the p53 amino-acid sequence results in the

presence of either a proline or an arginine at position 72. They found that the arginine

form of p53 is significantly more susceptible than the proline form to be degraded by

E6 mediation. Moreover, allelic analysis of patients with HPV-associated tumors

revealed a striking overrepresentation of homozygous arginine-72 p53 compared with

the normal population, which indicated that individuals homozygous for arginine 72

are about seven times more susceptible to HPV-associated tumorigenesis than

heterozygotes. The arginine-encoding allele therefore represents a significant risk

factor in the development of HPV-associated carcinoma. Although both forms of p53

variants are morphologically wild type and do not differ in their ability to bind to

DNA in a sequence-specific manner, in another study, they showed that there are a

number of differences between the p53 variants in their abilities to bind components

of the transcriptional machinery, to activate transcription, to induce apoptosis, and to

repress the transformation of primary cells. This may have implications for the

development of cancers which harbor wild-type p53 sequences and possibly for the

ability of such tumors to respond to therapy, depending on their p53 genotype (249).

There are some other supportive data (250-258). However, also more objecting results

have been reported (247, 248, 259-269). This far, this argument is still open.

1.3.4.3 Susceptibility of patients with different HLA class II genotypes to HPV

infection

An association between HLA type and cervical neoplasia or HPV infection has

been reported. In one study the likelihood of developing invasive cervical carcinoma

was increased about 7-fold among female Caucasian carriers of HLA DQw3

compared with women who lacked this antigen (270). HLA DQA1 *0311 increases

Page 33: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

32

the risk for CIN III and invasive carcinoma whereas DQA1 *0501 protects the

women from development of cervical neoplasia (271). The DQB1 ’03 is associated

with CIN III probably through being permissive for HPV infection (272). A higher

proportion of women without HPV infection carry DQB1 profiles than women with

HPV infection (273). Cuzick et al. showed that HLA DQB1 *0501 is protective

against development of cervical carcinoma in HPV16 positive patients while DRB1

*1301 has no protective effect (274). Allen et al. associated HLA 5 DQ-DR

haplotypes with development of cervical carcinoma in individuals infected with

HPV18, suggestive of a deficiency in antigen presentation by the HLA molecules

encoded by these haplotypes (275). Probably, HLA class II polymorphism is involved

in genetic susceptibility to cervical carcinoma and HPV infection (122). It can be

explained that viral infection is immunologically influenced. However, such a risk

figure was not confirmed in some other reports (276-278).

1.3.5 Assumed molecular pathogenesis

There are some assumed models for the pathogenesis of cervical carcinoma.

Many of them are similar in principle. Recently, Walboomers et al. suggested a model

of molecular pathogenesis of cervical neoplasia (279). They think of cervical

carcinogenesis as a multi-step process. First, high-risk HPV infects normal squamous

epithelium. In most cases this will not lead to a lesion or at worst give rise to a

regressing low grade CIN (CIN I). Both phenomena involve viral clearance. Second,

only persistent infections with high-risk HPV will lead to a high-grade lesion (CIN

II), subset of which may undergo malignant transformation. Third, at the transition of

CIN II to CIN III deregulated expression of the viral oncogenes E6 and E7 takes

place, resulting in genetic instability. Subsequently, activation of the telomere-

lengthening enzyme, telomerase occurs, as a result of which cells obtains a replication

capacity. Fourth, ultimately, successive allelic losses occur at different chromosomal

locations which, followed by a clonal outgrowth results in an invasive carcinoma.

This model sounds reasonable, but the process is too arbitrary to claim a strict and

fixed hierarchy for CINI, CIN II and CIN III and to stress high-risk HPV as the only

actively functioning factor during the process of carcinogenesis in different individual

patients. Actually, most of the cases with persistent high-risk HPV infection can stay

long in the stages of morphologically normal epithelium or different grades of CINs.

Some moderate-risk HPV can also cause cervical carcinoma. Even high grades of

Page 34: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

33

CINs do not have to progress, most of them can indeed regress. The model neglects

the fact that the neoplasia probably originates from one or more progenitor cells (stem

cells) but not from the common normal epithelial cells. This means that the model

ignores that the cellular genetic background can play a role during the carcinogenesis.

2 Specific Aims of the Study

(1) To investigate if all studied cases of cervical carcinoma are monoclonal.

(2) To investigate if all isolated CIN lesions and all cases of CIN are monoclonal.

(3) To investigate if cervical carcinoma and synchronous CIN lesions share the

clonality status.

(4) To determine if all lesion areas and areas of normal epithelium in the same cervix

have the same type of HPV or HPV16 variant.

(5) To determine the biological significance of HPV16 variants.

(6) To find out if all areas of lesions in the same case have LOH and have the same

pattern of LOH.

(7) To find out if there are specific chromosome loci with LOH in early step of

cervical neoplasia.

(8) To determine if the hMLH1 gene, which shows LOH in 43% of the cases is

involved in cervical carcinoma.

(9) Finally, to suggest potential models for the molecular pathogenesis of cervical

carcinoma.

3 Experimental Design

3.1 Cross-sectional case control study system

CIN shares some cytological features with cervical invasive carcinoma, but the two

lesions differ in histology and biological behavior. Invasive carcinoma can invade

into neighboring tissues or organs and metastasize to other parts of the body. CIN

grows on the basal membrane and does not invade or metastasize. A proportion of

CINs can break the basal membrane and become invasive carcinomas, but a higher

proportion of CINs would persist or regress. So CIN is different from but related to

invasive carcinoma. If one takes a suitable number of CIN cases as a group and

invasive carcinoma cases as another group to compare the variables from the two

Page 35: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

34

groups with each other with a statistics power, an event occurring at a higher

frequency in invasive carcinoma than in CIN would be said to be a factor involved in

the evolution process from CIN to invasive carcinoma. Contrarily, if the frequency is

the same, the event might not be involved in. Thus, this study system consists of two

group of cases, CIN groups and invasive carcinoma groups. For this kind of study,

random collection of enough number of studied cases is essential to draw a safe

conclusion.

3.2 Synchronous lesion study system

Some cases of cervical squamous cell carcinoma contain one or multiple

independent CIN lesions in the same cervix, which are called synchronous lesions of

cervical carcinoma. Since invasive carcinoma and CIN lesions are present in the same

cervix, they share many basic backgrounds of genetic factors and environment.

Comparing the variables between CIN lesions and invasive carcinoma in such cases

would make it possible to obtain the best precise and correct results. With such kinds

of results, one can often draw reliable and sound conclusions. Therefore, the use of

synchronous lesions of cervical carcinoma to conduct the study is called synchronous

lesion study system. Sometimes the statistics power in such a stusy can be

unimportant.

4 Materials and Methods

4.1 Patients

4.1.1 Paper 1

Fifteen cases of cervical squamous cell carcinoma from Sweden were studied. The

extirpated uteri with one or more synchronous CIN lesions were serially selected from

1996 to 1998 in the Uppsala province without any other selection. Tissue from 10

cases (L1-L10) was fixed by neutral formalin. Specimens from 5 cases (H1-H5) were

fresh-frozen. The mean age for those cases was 45 years (32-62 years). Twelve

squamous cell carcinoma cases were moderate, two poor and one high in

differentiation. The FIGO stage for the cases belonged to Ia to IIb.

Page 36: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

35

4.1.2 Paper 2

One hundred and four cases, including 55 cases of primary invasive squamous

cell carcinoma and 49 cases of CIN, from patients undergoing radical hysterectomy at

Blokhin Cancer Research Center, Moscow, Russian in the period from 1994 to 1998

were studied. The age range was 30 - 80 (mean 43.8) for the patients with ICC and 31

- 43 (mean 37.2) for those with CIN. The FIGO stage and degree of differentiation

(high, moderate or low) were recorded at the Blokhin Cancer Research Center. Tumor

samples were snap-frozen in liquid nitrogen. Part of each sample was transferred on

dry ice to the Department of Genetics and Pathology, Uppsala University, Sweden.

4.1.3 Paper 3

Sixty-one HPV16 positive cases, among which 30 were CIN III and 31 were

invasive squamous cell carcinomas, from 141 Swedish patients undergoing radical

hysterectomy or cervical conization at the Uppsala University Hospital in the period

from 1996 to 1999 were studied. The age range was 24-69 years (mean 40.9 years)

for those with CIN and 26-83 years (mean 48.5 years) for ICC patients. The FIGO

stages for ICC patients were from Ia to IIa. Of the 61 cases, nine were freshly-frozen

specimens and the remaining 52 were neutral formalin-fixed paraffin-embedded

specimens.

4.1.4 Paper 4

Thirty cases of CIN or cervical invasive carcinoma were collected from neutral

formalin-fixed and paraffin-embedded archival material at the Department of

Genetics and Pathology, Uppsala University Hospital, Sweden. Among the cases, 20

were CIN without invasive carcinoma, 10 were invasive carcinoma with one or more

synchronous CIN lesions in the same samples. The morphological diagnoses for the

morphology of the cases were confirmed by two experienced pathologists

independently.

Page 37: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

36

4.1.5 Paper 5

Eleven women with invasive cervical squamous cell carcinoma came from

Sweden. Their age ranged from 26 to 63 (mean 43.7), and the FIGO stage was Ia or

Ib. Ten cases carried high-risk HPV and the remaining one case had no detectable

HPV DNA. The paired tumor and normal samples from the same patient were from

the Department of Genetics and Pathology, Uppsala University Hospital, Sweden.

The cases, including seven cases with and four cases without LOH at the hMLH1

locus were subjected to immunohistochemical staining. 8/11 cases, seven with and

one without LOH of the hMLH1 gene, were used for DNA sequence analysis of the

hMLH1 gene. The invasive carcinoma was synchronous with CIN lesions in all but

one case.

4.1.6 Paper 6

One case (H2) was used. The patient was a Swedish woman when at the age of 33

had her uterus with cervical carcinoma removed. Macroscopically, tumor grew within

the cervix and around the external os with no involvement of uterus body or vagina.

The histopathological diagnosis in this case was cervical invasive squamous cell

carcinoma (moderate differentiation) with invasion of local vessels and metastases to

local lymph nodes. One month before the surgical procedure, the patient had been

found with cervical malignancy by vaginal cytology and latter on the diagnoses had

been confirmed by biopsy. HPV routine test showed HPV16 positively. Before this

HPV test, the HPV infectious situation was not known. In two previous vaginal

cytological examinations 11 and 8 years ago, no abnormality was found. The entire

fresh cervix was cut from external os to endocervix into six parts designated A, B, C,

D, E and F, respectively. Parts A, C and D were used for histopathological routine

examination, while B, D and F were frozen at –80°C for research.

4.1.7 Papers 1-6

All projects had received official institutional and local ethical approval from

Uppsala, Sweden and Moscow, Russia.

Page 38: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

37

4.2 Microdissection (Paper 1–6)

Serial cryosections (6 µm) were prepared from the freshly-frozen tissues and

stained shortly with Mayer´s hematoxylin. Ordinary sections were made from

paraffin-embedded tissues and deparaffinized in xylene then stained with Mayer´s

hematoxylin. According to the purpose and design of the experiments, only one

sample were picked up each case or multiple samples were picked up from different

areas and different lesions in the individual cervix.

In paper 6, the microdissection was used most extensively. Separate

microdissections of invasive cancer nest, CINs, normal epithelium, gland and stroma

in one section for each tissue block were performed. All lesions were sharply

demarcated from stroma or adjacent normal epithelium. Admixture of normal

epithelial or stroma cells was insignificant as judged by careful examination under the

microscope. The blade of the scalpel was changed after each microdissection. The

microdissected pieces were transferred to Eppendorf tubes containing 50µl 1 x PCR

buffer II (PE, Roche Molecular System, NJ). Each sample contained 500-1000 cells.

4.3 Immunohistochemistry (Paper 5)

Slides were deparaffinized in xylene, washed in gradients of ethanol and water,

then placed in a Tissue-Tek® Slide Staining Holder, the empty slots of which were

filled with blank slides. Slides were microwaved in citrate buffer pH 6.0 and treated

by 250 ml of working strength Antigen Retrieval solution according to instructions by

the manufacturers. The slides were allowed to cool for 20 minutes and then treated

with 0.3% hydrogen peroxide in methanol for 30 min. to suppress endogenous

peroxide. The anti-hMLH1 antiserum (Labora) diluted 1:100 was applied to the

sections and incubated overnight at 4oC. The slides were then incubated with

biotinylated IgG and with ABC each for 30 min, at room temperature. As a positive

control, normal colon mucosa known to display hMLH1 protein immunoreactivity

was used. Negative controls were obtained by replacing the primary antibody with a

non-immune rabbit serum.

4.4 DNA preparation (Papers 1-6) and purification (Paper 6)

Page 39: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

38

Lysis of cells by proteinase K (500 µg/ml) at 56°C over night was interrupted by

incubation of the sample at 95°C for 10 min. The roughly prepared DNA from each

sample was ready to be used for PCR-sequencing of HPV16 and PCR-genescanning

detection of LOH with microsatellite markers. A portion of roughly prepared DNA

sample (20µl) in paper 6 was further purified for X-chromosome inactivation

analysis. 50µl of 95% EtOH was added to 20µl roughly prepared DNA sample and

the sample was incubated at –70°C for 5 hours. After centrifugation at 13,000 rpm for

30 min, the precipitate was washed with 250µl of 70% EtOH and spinned at 13,000

rpm for 20 min and then air-dried.

4.5 Methylation sensitive Hpa II restriction enzyme digestion (Paper 6)

The pellet gained at step 4.4 was dissolved in 20µl reaction buffer for

methylation-sensitive restriction enzyme HpaII analysis (Promege, Madison, USA)

and halved into two tubes. One portion was added 15 Units of HpaII and incubated at

37°C over night. The reaction was terminated by heat inactivation at 95°C for 5 min.

Another portion of DNA, as a control, was not exposed to HpaII digestion. The non-

HpaII digested and the HpaII digested DNA portions were then ready to be used for

PCR amplification of androgen receptor gene.

4.6 Polymerase chain reaction (PCR) (Papers 1-6)

Information on the sequences of PCR primers for the genes of HPV16 and HPV

typing, the androgen receptor gene, the hMLH1 gene, and the microsatellite DNA

sequences have been described in the different papers. The PCR reaction mixture and

the PCR program for each PCR primer pair have also been described in

corresponding papers. The PCR was performed on a RoboCycler Gradient 96

(STRATAGENE) or GeneAmp PCR Systems 9600 (PE, Norwalk, CT).

An example shows the PCR procedure. To amplify HPV16 E6, 30µl volume (1 x

PCR bufferII, 3.0mM MgCl2, 200µM of each deoxynucleotide, 0.5U Taq DNA

polymerase, 0.5µM of each sense and anti-sense primer, and 1µl DNA solution) was

used. 35 cycles (1min at 95°C, 2min at 55°C and 3min at 72°C) with 10min initial

denaturation at 95°C and 7min final elongation at 72°C were used.

Page 40: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

39

To avoid contamination, we prepared a PCR master mix in an isolated room under

a hood, where UV-light was used to destroy any potential contaminating DNA or

PCR product at the working area before and after this manipulation and then added

template DNA under similar working conditions in another separate room.

4.7 Polyacrylamide gel electrophoresis with single-strand conformation

polymorphism (SSCP) and silver staining for HPV typing (Paper 5)

After PCR amplification, part of the PCR product from primer pair GP5+/GP6+

were electrophoresed in agarose gel and stained by ethidium bromide to roughly

check if the samples contained specific fragments. The samples with specific

fragments were subjected to SSCP on PhastSystem with semi-automated

electrophoresis in pre-formed 12.5% homogeneous polyacrylamide Phast gel and

native buffer strips (Pharmacia, Uppsala, Sweden). A pre-run at 400 V, 10.0 mA, 4.0

W, 10°C, was executed until it paused at 99 accumulated Volt hours (Avh). Then 1µl

of a denatured solution, consisting of 99% formamide, 0.05% bromphenol blue and

0.05% xylene cyanol, were blended in a 1.5-ml Eppendorf tube, centrifuged briefly,

de-natured for 5 min at 95°C in an Eppendorf thermomixer and cooled rapidly on ice.

A 0.8 -µl sample was pipetted onto a sample comb fitting the holders of the

PhastSystem intrument, and the comb was inserted into the holders. Electrophoresis

was carried out up to 230 Avh. The gel was stained with 0.5% silver nitrate in the

development chamber of the PhastSystem according to standard procedure. To

determine the HPV type of the samples, a reference picture made by Zehbe et al.

(280) was used. The 12 most common HPV types present in cervix could be clearly

distinguished.

4.8 Fragment analysis (Papers 4 and 6)

The inactivation pattern of the X-chromosome linked androgen receptor gene and

LOH of microsatellite loci were subjected to fragment analysis. An example presents

the procedure of fragment analysis. 1.5µl of the final PCR product of the androgen

receptor gene or a microsatellite locus was mixed with 0.5µl loading buffer, 0.5µl

internal size standard (GENESCAN-350 ROX, Perkin Elmer) and 2.5µl formamid,

and denatured at 95°C for 5min followed immediately by cooling on ice before

Page 41: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

40

loading on a 4.5% acrylamid gel. Electrophoresis was performed on an ABI Prism

377 sequencer (Perkin Elmer). Size and quantification of allelic fragments and LOH

were analyzed and determined by GeneScan Analysis 2.0.2 (PE Applied Biosystems).

A 60% or higher allelic signal reduction between any two alleles after HpaII digestion

was considered a homogenous pattern representing a sample with paternally or

maternally inactivated X-chromosome. The same criterion of allelic reduction was

adopted for judgement of LOH.

4.9 DNA sequence analysis (Papers 1-6)

The whole genome of HPV16 and 19 exons of the hMLH1 gene were subjected to

DNA sequence analysis. An example represents the DNA sequencing procedure. The

PCR amplicons of HPV16 E6 were electrophoretically separated on 1.5 % agarose gel

and stained with ethidium bromide. Desired bands were cut out with subsequent

purification on GenElute Minus EtBr Spin Columns (SUPELCO, Bellefonte, PA).

The purified PCR products were quantified and then applied to enzymatic extension

reactions for DNA sequencing using the Cycle Sequencing Ready Reaction Kit (Big-

Dye terminator reagent [PE Applied Biosystems] containing dye-labeled terminators)

in GeneAmp PCR Systems 9600 (PE, Norwalk, CT). The same forward and reverse

primers as for the PCR amplification of HPV16 were used separately in cycle

sequencing. The extension products were purified by Ethanol/Sodium Acetate

precipitation, then electrophoresed on an ABI Prism 377 sequencer. The sequence and

variations were analyzed and determined by the FacturaTM and Sequence Navigator

version 2.0 (PE Applied Biosystems).

With respect to all analyses performed, the test for each sample was repeated at

least once starting from DNA-PCR amplification with the same result.

5 Results and Discussions

5.1 Paper 1

5.1.1 Results

Page 42: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

41

One hundred and twenty-two samples from 15 cases of cervical squamous cell

carcinoma with synchronous CIN lesions were analyzed with HPV typing. By

comparison with the reference picture (Fig. 3) (280), the HPV types in the samples

were interpreted from the SSCP patterns (Fig. 4). 13/15 cases were HPV-positive.

Fig. 4. Representative SSCP patterns of HPV types. Lane 1 and 7: Molecular weight

marker. Lane 2 and 3: HPV16. Lane 4 and 5: HPV33. Lane 6: HPV18

HPV16 was the most common type (10/15). HPV31 was found in 2/15 and HPV18 in

1/15 cases. There was concordance between different synchronous lesions with regard

Page 43: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

42

to type of HPV or absence of HPV. HPV found in normal squamous epithelium was

always of the same type as that of any positive lesions in the same patient.

All 10 of the cases with HPV16 positivity were analyzed for E6 variation. By

comparison with the “prototype” of the HPV16 E6 sequence (61), three variations

were found (Fig. 5). One variation present only in case L10, was A→G at nt 131,

Fig. 5: Electropherograms of HPV16 E6 sequence variations. Upper line: Prototype

sequence from case H2. Lower line: Variations. Left column: A → G variation at nt

131 in case L10. Middle column: A → G variation at nt 276 in case L5. Left column:

T → G at nt 350 in case L9.

which altered arg→gly in codon 10. The second, only present in case L5, was A→G

at nt 276 which changed asn→ser in codon 58. The third variant, T→G at nt 350

which changed leu→val in codon 83, was common, seen in 5/10 cases. The “

prototype”, as well as the variant E6, was identical in the different lesions within any

individual patient. Multiple samples from the lesions and also the normal epithelium

were consistently positive either for the “prototype” or for the variant E6.

5.1.2 Discussion

The studies of single lesions of different grades of CINs and invasive carcinoma

have for instance shown that benign neoplasms, CIN I and CIN II are predominantly

associated with low-risk HPV but only rarely with high-risk HPV. CIN III and

invasive carcinoma are predominantly associated with high-risk HPV (90, 91). It is

Page 44: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

43

important to elucidate if the types of HPV would be different in normal epithelium,

different grades of CIN lesions and invasive carcinoma in the same case. In our cases

of cervical carcinoma with synchronous CIN lesions, one important result of the

present study is the regular persistence of one type only of HPV in simultaneously

present normal epithelium, CINs and invasive carcinoma. High-risk HPV, as

expected, dominated invasive carcinoma and CIN III. However, they were equally

prevalent in concomitantly present lower grades of CIN and normal epithelium. This

observation could be extended to the HPV16 E6 variants. Homogeneity in HPV type

was also seen for these lesions where multiple samples were compared. The results

strongly indicate that the original infection of HPV persists during progression from

normal via CIN lesions to invasive carcinoma, thus that HPV is a necessary and

inefficient cause of cervical neoplasia. The results also indicate that multiple types of

HPV infection may be very rare, or new types of HPV may for immunological or

other reasons have difficulty in establishing themselves.

Zehbe et al. concluded that the HPV16 E6 variants (most of the reported cases are

all E6 variant (nt 350 G) are particularly strongly associated with progression from

CIN to invasive carcinoma (195, 196). However, our results do not support the

previous conclusions since “prototype” HPV16 E6 was also common in invasive

carcinoma (50%) and, more importantly, the E6 variants were present in normal

epithelium, CIN lesions and invasive carcinoma in the same case. This indicates that

HPV16 E6 variants are not the strong risk factor to drive the CIN lesions into invasive

carcinoma either.

5.2 Paper 2

5.2.1 Results

Thirty-two cases of cervical invasive carcinoma and 18 cases of CIN were

analyzed for HPV16 E6 sequence variations. The E6 variants were classified as three

groups, A, B and M. A and B were identical except the polymorphic nt 350, which

was either guanine (A) or thymidine (B). Group M was a mixture of variants with

sequence departures from group A and/or B at other sites than nt 350. Group A

dominated both in CIN (10/18) and ICC (19/32). Group B, which corresponded to the

international “prototype,” occurred in 4/18 cases of CIN and 5/32 cases of ICC. The

third group (M) was represented in 4/18 cases of CIN and 8/32 cases of ICC. There

Page 45: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

44

was no statistically significant difference in the distribution of groups A, B or M

between CIN and ICC.

However, group B was mainly associated with cancers detected in early clinical

stages. Group A, the most common one, was represented in early as well as advanced

stages. The M group was not seen in FIGO stage I. Differences between the groups

were statistically significant indicating that the order of clinical malignancy between

the groups would be M > A > B.

Fourteen out of 32 cases of invasive carcinoma with synchronous lesions or

multiple samples were available. Among these cases, 10 contained a single HPV16

E6 isolate. The E6 Variant in these cases was concordant from different parts of each

ICC case. When CIN was also present (N3, M 2, M 21), all samples from these

precursors had the same variant as the simultaneously found invasive cancer. Normal

squamous epithelium sampled in case M19 and N3 showed the same variants as in the

invasive cancers.

Four cases (M4, M12, M13, M23) bore double/multiple variants. M4 had A and B

variants in CIN II lesions and invasive carcinoma. M12 contained both A and B

variant in carcinoma. M13 presented the B variant in 3/3 CIN II samples, and in

invasive cancer, one sample showed the A variant and another sample had an A

variant with an additional missense variation from T to G at nt 310. In M23, the

normal epithelium showed the B variant, the two CIN II samples showed either A or

B variant, the invasive carcinoma had either A (two samples) or B (two samples), and

one of the invasive cancer areas contained a B variant with additionally changed C to

T at nt 374 which created a stop signal.

5.2.2 Discussion

HPV16 E6 variants are reported to have different biological and biochemical

properties and the “variants” are more malignant than the “protorype” (47, 193, 195-

197). However, the rate, the type and the biological and clinical significance of the

variants of HPV16 E6 have not been geographically uniform. Besides, the definition

of “variant” has been a source of confusion in previous studies, because it has been

based on all departures from the original “prototype” once isolated randomly from a

case of cervical invasive carcinoma.

Page 46: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

45

We found that the variations at different nucleotide sites differ principally from

each other. Nt 350 behaves as a polymorphic site where roughly two thirds of the

isolates have a G and the remainder a T. The other sites register as classical variants

with one or occasionally two departures from a predominant configuration. This

classification permits a logical division into two homogenous groups (A and B)

supplemented by one heterogeneous group M. Failure to have noted this is the major

reason for a confused literature based on lumping together all “pure non -T” at nt 350

with all other departures from the sequence of the “prototype”. When our groups A, B

and M are applied, the distribution of the three groups in invasive cancer and CIN is

identical. This contrasts with claims that E6 variants at nt 350 have a higher

prevalence in invasive cancer than in CIN III (195, 196). Therefore our findings do

not support the conclusion that “variants” of E6 are more likely to cause progression

to invasive cancer than “prototype” E6. Even if the previous scheme (195, 196) which

classified E6 variants as two groups, “prototype” and “variants” group, is applied on

our cases, no difference in the distribution of the E6 groups can be seen between CIN

and invasive carcinoma. Since this investigation was done on samples from Russian

patients, our results might reflect the fact that the human genetic susceptibility

probably determined by HLA or TP53 polymorphisms of the Russian population to

different HPV16 variants differs from that of other ethnic populations (122, 248).

Since A and B variants of HPV16 E6 are so commonly distributed in cervical

neoplasms, they might be stable polymorphic variants having been mutated very long

ago and undergone natural selection. It is unlikely that the variation of A variant and

the variation of B variant would be substituted for each other. The four cases

containing both A and B may be the result of double infections because it should not

be difficult for the patients to pick up the E6 variants prevalent in the infection pool.

The two additional variants in cases M13 and M23 probably represent mutations of

the B variants within the host because the additional variants are very rare in nature.

The reason for such mutations of HPV16 E6 is unclear. Since cervical neoplasms

have pronounced genomic instability with allelic losses, mutated variants of E6 would

arise in vivo within cancer cells subject to genomic instability. The positive

association of the M group with clinical malignancy would then be an

epiphenomenon explained by an influence of the cancer cells on a residing viral

genome, rather than the reverse conventional hypothesis that different E6 variants

have a different potential to drive progression to invasive cervical carcinoma.

Page 47: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

46

5.3. Paper 3

5.3.1 Results

Sixty-one cases of HPV16 positive CIN III (30 cases) and cervical invasive

carcinoma (squamous cell type, 31 cases) out of 141 Swedish cases of cervical

neoplasms were selected. The nucleotide (nt) sequences of the full open reading

frames of HPV16 E6 (477 base pairs), E7 (315 base pairs) and E5 (297 base pairs)

from these 61 cases were determined, and they were compared with the HPV16

reference DNA sequence (61). Nucleotides at any position different from the

reference nucleotide at the corresponding position were recorded as variations. The

variations covered nearly the whole length of the open reading frame of each gene: E6

showed 13 variations with 9 being missense and 4 silent, E7 had 3 variations with

only 1 being missense and 2 silent, and E5 had 9 variations with 5 being missense and

4 silent. There was no evidence of premature stop codons or deletions. The highest

variation rate for the whole sequence length of the combined E6, E7 and E5 in a

single case was 1.5%, indicating that the analyzed HPV types should be regarded as

variants.

The nucleotide variations were not randomly distributed. There were clearly three

prevalent variation sites: nt 350 in E6 (41/61 T, 20/61 G), and in E5 nt 3979 (16/61 A,

45/61 C) and nt 4042 (14/61 A, 47/61 G). All these variations were missense

variations. Clustering of the prevalent variations in the order E6 nt 350, E5 nt 3979,

E5 nt 4042, gave five lineages: Va (T-A-A), Vb (T-C-G), Vc (T-A-G), Vd (G-A-A),

and Ve (G-C-G), accounting for 21.3%, 41.0%, 3.3%, 1.6%, and 32.8% of the cases,

respectively. The 3 major lineages combined (Va, Vb, and Ve) accounted for 95.1%

of the cases.

The distribution of the lineages showed no statistically significant difference

between CIN and invasive carcinoma. However, additional missense variations

occurred in 3 cases of CIN and 11 cases of invasive carcinoma, and a simple

correlation of the proportion of additional missense variations shows a statistically

significant difference of 3.7 times more additional missense variations in invasive

carcinoma than in CIN (P <0.025, chi-square test). If we analyze the number of

additional missense variations for the different oncogenes lineage, we do not detect

any significant correlation for most of the lineages, with the exception of Vb. In the

Page 48: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

47

Vb lineage we can see a strong prevalence of additional missense variations in ICC.

The Vc and Vd lineages were found in too few cases to allow us to assess any

difference in additional missense variations between CIN and ICC.

5.3.2 Discussion

Some of the previous studies on sequence variations of HPV16 often considered

only one of the oncogenes of the virus. This may be another source of confusion and

reason for inconsistent conclusions such as that E6 variants are more malignant than

the prototype (195, 196, 200, 203). Since genetic experiments have assigned

oncogenic activity to the viral genes E6, E7 and E5, and their co-operative action may

make a major contribution to the development of cervical neoplasia, we hypothesize

that HPV16 has different oncogene lineages at genetic level with different biological

functions. Clustering of the three prevalent variation sites in E5 and E6 theoretically

gives eight lineages, but only five lineages were identified with the three major ones

accounting for 95%. This indicated that the lineages have undergone natural selection

and become stable. The additional missense variations appear more frequently in

invasive carcinoma than in CIN. The Vb group, one of the oncogene lineages, has

more additional missense variation in invasive carcinoma than in CIN. This suggests

that the HPV16 oncogene lineages may represent different groups of HPV16 variants

with different biological functions and therefore present a further risk factor

contributing to the development of cervical carcinoma.

5.4 Paper 4

5.4.1 Results

Twenty cases of CIN and 10 cases of invasive carcinoma synchronous CIN lesions

were analyzed with 11 microsatellite markers covering chromosome 3p from 3p24 to

3p13. No LOH at the tested 3p loci was found in normal epithelium, squamous

metaplasia and CIN I lesions. Allelic losses at one or more 3p markers were recorded

in 33% (3/9) of CIN II and 36% (5/14) of CIN III lesions in the CIN cases. A

statistically significant increase of the percentage of LOH was observed in the

synchronous CIN lesions of invasive carcinoma, showing 71% (5/7) in CIN II and

76% (13/17) in CIN III.

Page 49: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

48

Patterns of LOH were compared between multiple synchronous CIN lesions and

corresponding invasive carcinoma in 10 cases. 75% (6/8) of informative cases had

identical LOH patterns between multiple CIN lesions and invasive carcinoma. The

remaining one case showed LOH in the invasive carcinoma but not in the two co-

existing CIN III lesions. In another remaining case, LOH was detected in the invasive

carcinoma and the CIN II lesion but not in the CIN I lesion.

Two common interstitially deleted regions with incidence of LOH higher than 50%

of the cases of invasive carcinoma were identified. The six cases with identical LOH

patterns showed a common deletion in the region between D3S1076 and D3S663 that

maps to 3p21.1-3p21.3, and the region between D3S1260 and D3S1611 that maps to

3p22. In contrast to the complex and extensive pattern of allelic deletions in invasive

carcinoma, LOH in CIN cases was more restricted to small regions on 3p. The

frequently deleted loci among the CIN cases were at D3S1260 (25%) and the locus

between markers D3S1076 and D3S1289 (33% and 27%, respectively).

5.4.2 Discussion

LOH in tumors represents a genotype of recurrent genetic deletions, some

occurring in the early stage of carcinogenesis, others during the process of tumor

development or in the late stage of the tumor (281). Two regions 3p22-21-3 and

3p21.1 showing frequent LOH in both groups of the cases studied promise to contain

novel potential tumor suppressor loci, which may play a role in early transition of

CIN into invasive carcinoma. Most cases of cervical neoplasms were reported to be of

monoclonal origin (136-138). Identical LOH patterns of these microsatellite markers

between synchronous multiple CIN lesions and invasive carcinoma in 75% of the

informative cases suggest that different cervical CIN lesions and invasive carcinoma

are genetically linked and most likely originate from a single progenitor cell.

Monoclonal origin of cervical neoplasms favors the probability that the genetic

defects are the causes of this neoplasm. However, no oncogenes or tumor suppressor

genes strongly related to cervical neoplasia have been found. The two cases (25%)

showing different LOH patterns between the synchronous CIN lesions and invasive

carcinoma in this study indicate that some of the cervical carcinomas and the

synchronous CIN lesions can originate independently from multiple progenitor cells.

This is consistent with other reports (142) and supports that HPV as a field factor is

the main cause of cervical neoplasia.

Page 50: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

49

5.5 Paper 5

5.5.1 Results

Seven cases with LOH within the hMLH1 gene and four without were stained

with the hMLH1 antibody. All cases were positive in CIN cells, carcinoma cells and

normal epithelial cells. The staining was strongest in the order: CIN > carcinoma >

normal cells.

The hMLH1 gene sequences of the carcinoma were compared with the ones of

their matching normal tissues and the wild type sequence of the hMLH1 gene from

the Gene bank database (282). No mutation was found in any exon in samples from

the normal epithelium, CIN or cancer regardless of the presence of LOH or not. Exon-

intron boundaries had no changes but in the exon 13 and 15 regions. For exon 15

fragments, 3 bp TTT in all sequenced samples of the cases substituted for the AAA of

the wild type sequence at a position 42 bp from the 3´end of the exon. For exon 13

fragments, change was seen at a nt 12bp away from 3´end of the exon in only one

case. No alteration was found in the remaining cases. Case 4 was represented by 6

samples, which were 1 stroma, 1 normal epithelium, 3 CIN III and 1 invasive

carcinoma. LOH of the hMLH1 gene was present in all CIN and invasive carcinoma

samples from this case, but not in stroma and normal epithelium (223). In this study

of the same case, stroma and epithelium had double signals, G (wild type) plus A,

while all CIN and invasive carcinoma had only A.

5.5.2 Discussion

The hMLH1 gene is one of the DNA mismatch repair genes. It has been mapped

to chromosome 3p21-23 (283). This gene covers about one million base pairs and

contains 19 exons (282, 284, 285). Inactivation of the gene by mutation and/or

hypermethylation causes microsatellite instability in sporadic and familial colorectal

tumors (282, 284, 285). Microsatellite instability is also found in some female-

specific tumors or corresponding cell lines such as breast cancer, ovarian cancer,

endometrial cancer and cervical cancer (236, 286, 287). Using a microsatellite marker

D3S1260 which is located within one of the introns of the hMLH1 gene, 22% - 43%

of cervical carcinoma cases were found with LOH in this gene. However, our results

Page 51: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

50

from the analysis of immunohistochemistry and DNA sequencing showed no

alteration of the hMLH1 gene or of the gene expression in the cases of cervical

carcinoma with LOH in this gene, indicating that the hMLH1 gene is not involved in

the development of cervical neoplasia.

One mutation within an intron of the hMLH1 gene was found in one case. All CIN

and invasive carcinoma lesions lost the wild type allele, while the normal epithelium

and stroma retained two alleles, which matched the finding of LOH analysis and

indicates that the multiple CIN lesions and invasive carcinoma shared the clonal

origin in this case.

5.6 Paper 6

5.6.1 Results

Twenty-four samples named H2-1 to H2-24 from one “golden” case of cervical

carcinoma with multiple synchronous CIN lesions were analyzed with respect to

clonality with the X-chromosome inactivation marker, HPV genome sequencing and

microsatellite markers.

The X-chromosome inactivation patterns were informative in this case. After

HpaII digestion, three common X-chromosome inactivation patterns were seen. The

microdissected samples with X-chromosome inactivation patterns are presented in

Table 3.

H2-9, a CIN II sample, was special. It seemed to have three alleles of the

androgen receptor gene, one was a, the second was b and the extra allele was the

shortest named –a. After HpaII digestion, b was cut down by 63% and a by 25% of –

a.

DNA sequence variations or mutations of the HPV16 genome were analyzed as an

assistant clonality marker. The entire genome of HPV16 with 7905 bp was PCR-

sequenced for 20 out of 24 samples (the other four were gland and stroma and other

HPV16 negative samples). Any nucleotide found at any position to be different from

that of the reference HPV16 sequence (61) is here described as variation (probably

occurring before infecting a specific host) or mutation (probably occurring inside a

specific host). Three common variations in all HPV16 positive samples and, in

Page 52: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

51

addition, two mutations in only some of the samples were found in this case (Table

3). If an HPV16 variant is defined as an HPV16 sequence with at least one differing

Table 3. X-chromosome inactivation pattern, HPV16 variations and mutations, and

LOH of the microdissected samples. Normal: normal squamous epithelium. Sup.

Inv.: superficially invasive carcinoma. Xc. pattern: X-chromosome inactivation

patterns (a, b, ab, -aa). The a: only short allele remains (inactivation), indicating

Page 53: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

52

single type of X-chromosome inactivation pattern in the cell population. The b: only

long allele remains, indicating another single type of X-chromosome inactivation

pattern in the cell population. The ab: two alleles retained, indicating two types of X-

chromosome inactivation patterns in the cell population. The –aa: an additional

allele remains (-a is shorter than a) in the cell population. -: the same nucleotide as

the reference nucleotide at this position. ng: negative for HPV. q57q: left q is

reference amino acid, 57 is codon position and right q is amino acid found at this

codon with a nucleotide variation, thus, q57q is a silent variation, while the other

three are missense variations or mutations. s: short allele remains. l: long allele

remains. d: both alleles remain, thus normal.

nucleotide within the whole genome, three HPV16 variants were found. The V1 was

the variant with the three common variations and was distributed among most of the

samples. The V2 with all the three variations and in addition a mutation in E2 (nt

3800), was seen in three samples (H2-1, -4 and –5). The V3 also with all the three

variations and in addition a mutation in L1 (nt 6318), was found in another three

samples (H2-9, -15 and –16).

Loss of heterozygosity was analyzed as another assistant clonality marker.

Detection was successful for D3S659 (3p13), D3S1283 (3p24.2-22) and D6S311

(6q22-23) in all samples of this case. The LOH patterns for all samples are shown in

Table 3.

As shown in Table 3 where all data were compiled, five different monoclonal

families were found in the samples from CIN II (H2-13), CIN III and 11/12 invasive

carcinoma. The samples from all normal epithelium, three out of four CIN II samples

and a superficial invasive carcinoma were polyclonal.

5.6.2 Discussion

Inactivation of the X-chromosome occurs at the embryonic stage (130-132), and

the polymorphism of the X-chromosome linked androgen receptor gene is neutral to

the proliferation of the cells. This makes it an ideal clonality marker. However, only

three kinds of result (a, b or ab) can be obtained. The a and b represent monoclonality,

while ab shows polyclonal. If a skewed X-chromosome inactivation or some technical

Page 54: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

53

problem occurs or samples are taken in a biased way, the same inactivation pattern

would probably be presented by cells with different clonal origin in the same cervix

(135). Therefore, assistant clonality markers such as HPV variation or mutation and

LOH are required to resolve this problem.

With the X-chromosome inactivation marker, we identified four types of clonality

in the samples of cervical neoplasia. The a and b represented different monoclonal

samples. The ab and –aa indicated polyclonality of the respective sample. The HPV16

mutation in E2 and the LOH patterns further divided the three b samples of carcinoma

into two groups. The LOH patterns further classified the ten a samples of CIN II, CIN

III and invasive carcinoma into three groups. The X-chromosome inactivation

patterns, the HPV16 mutation in L1 and LOH patterns together divided the four CIN

II samples into four groups: one with monoclonal a pattern and, three different

polyclonal groups. The X-chromosome inactivation and LOH patterns together pull

one CIN II, one CIN III and four invasive carcinoma samples into one monoclonal

group. These findings strongly indicate that this case is of polyclonal origin. Also, in

this case, the neoplasia seems to have developed in two ways: one from normal

epithelium via CIN II, CIN III to invasive carcinoma such as the a family with six

members, and another from normal epithelium possibly directly into invasive

carcinoma such as the two other a clones and the two b clones.

6 General discussion and summary of the conclusion

Cervical carcinoma is one of the most common malignancies in women.

Epidemical, clinical and experimental studies have suggested that HPV may be the

main cause of cervical carcinoma (90, 288). However, only a minority of HPV

infected patients develop any cervical lesions. Specific types of HPV are considered

the major risk factor because several studies associate HPV types with different

morphologies of cervical neoplasia (289). High-risk types of HPV such as HPV16

and18 associate with invasive carcinoma and CIN III. Moderate-risk types of HPV

such as HPV31 and 33 can be seen most frequently in CIN I and CIN II. Low-risk

types of HPV such as HPV6 and 11 present only in benign tumors, i.e., condyloma

and in CIN I. Thus, HPV6 and 11 have been used as a tool to diagnoses condyloma in

some pathology laboratories. Other risk factors associated with the virus may include

persistence of HPV infection, HPV copy number, expression level and patterns of

Page 55: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

54

HPV E6 and E7 genes, HPV physical stage and HPV sequence variations (47, 72,

196, 212, 290).

Since HPV16 is the most commonly seen HPV type in cervical carcinoma and

HPV variations are one of the currently most active subjects in the studies on the

further risk factors, we analyzed HPV types and HPV16 sequence variations or

mutations in cervical neoplasms. In cross sectional case control studies of Swedish

and Russian women, HPV16 E6 variants were distributed nearly even in CIN cases

and invasive carcinoma cases, but when viral oncogene lineages were considered,

additional variations were more frequently seen in invasive carcinoma than in CIN

cases. In a synchronous lesion study, most cases contained an identical type of HPV

or of HPV16 E6 variant from normal epithelium, different grades of CIN lesions to

invasive carcinoma. The results strongly support the opinion that specific infection

with HPV is the cause of cervical carcinoma because identical types of HPV and

HPV16 variants persist from the normal epithelium via CIN lesions to invasive

carcinoma. The results also suggest that a specific HPV16 variant might not be the

further risk factor but that the additional variations of HPV16 oncogene lineages

might be a weak further risk factor.

Recurrent genetic aberrations, such as those observed as LOH, occur frequently in

cervical neoplasms (281), however, no specific oncogene or tumor suppressor gene

has been found to be involed so far. We identified that two common interstitially

deleted regions of chromosome 3p with incidence of LOH higher than 50% of the

informative cases of invasive carcinoma. The six cases with identical LOH patterns

among the synchronous CIN lesions and invasive carcinoma showed a common

deletion in the region between D3S1076 and D3S663 that maps to 3p21.1-3p21.3, and

the region between D3S1260 and D3S1611 that maps to 3p22. The deletions

involving these two regions might contain potential tumor suppressor genes and their

loss may be the early event in the development of the cervical neoplasm. The hMLH1

gene is located in 3p22 and analysis of a microsatellite marker in this gene showed

LOH in 43% of the studied cases, but neither the expression level of protein nor the

genetic sequence of this gene was altered in the cases with LOH in the hMLH1 locus,

suggesting that the hMLH1 gene is not involved in the development of cervical

carcinoma in spite of its frequent involvement in colon carcinoma and other female

tumors.

Cervical invasive carcinoma often co-exists histologically with CINs in the same

patient, and it is assumed that cervical carcinoma is of monoclonal origin and

Page 56: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

55

progresses by multiple steps from normal epithelium via different grades of CINs into

invasive carcinoma. If this assumption is true, all the independent CIN cases and the

invasive carcinoma cases should be monoclonal and synchronous lesions of

carcinoma in a certain patient should share the clonality status. Several studies have

shown that CIN and invasive carcinoma are monoclonal (140) while some studies

supplied the possibility of polyclonality for a fraction of cases of cervical neoplasia

(136, 142). With microsatellite markers for chromosome 3p, 75% (6/8) of informative

cases of cervical carcinoma were found to have identical LOH patterns in these

multiple synchronous lesions, but 25% (2/8) cases had different LOH patterns among

the multiple synchronous lesions. In the “golden case” H2, the clonality status was

investigated extensively. The multiple carcinoma and CIN samples from the lesions

could be divided into several different clonal groups by the X-chromosome

inactivation patterns, HPV16 mutations and LOH patterns. The biggest clonal family

included both CIN II, CIN III and invasive carcinoma samples, while another four

families with a monoclonal pattern of carcinoma did not have associated CIN lesions.

These results indicated a complex clonality status and suggested that cervical

carcinoma can be either monoclonal or polyclonal and develop either directly or via

multiple steps.

Both HPV infection and LOH are known to be common events in cervical

carcinoma (281). Our results and the results of others show that HPV infection occurs

earlier than does LOH because HPV can be seen from normal epithelium and CIN

lesions through invasive carcinoma, while LOH can be found only in the two latter

(223, 291). A suggested molecular mechanism of the carcinogenesis of cervical

neoplasia is that one or more target committed stem cells in the basal layer of the

epithelium are infected by specific types of HPV. The HPV infected stem cells then

go along different paths. One path is that chromosomal deletion occurs in these stem

cells to further equip them with the capacities to give rise to neoplasms. If the event

of deletion has a strong enough biological effect, the stem cells would directly give

rise to carcinoma without CIN lesions, otherwise with CIN lesions. During the

process of carcinoma development, some other risk factors can be added, for instance,

additional variations of HPV16 oncogene lineage, some specific mutations or other

unknown factors, which can increase or reduce the tendency of tumor initiation and

progression. If the HPV infected stem cells do not acquire internal gene deletions,

they will continue to give rise to morphologically normal epithelium. If only one HPV

infected stem cell randomly acquires a strong cellular genetic event, it will

Page 57: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

56

independently cause a cervical neoplasia with monoclonal status, otherwise with

polyclonal status.

Conclusions:

(1) Cervical invasive carcinoma and CINs in the same case may originate either from

a single committed stem cell or from multiple committed stem cells.

(2) Invasive carcinoma may develop directly from the progenitor cell or via different

grades of CINs.

(3) HPV (as a field factor) is the cause of cervical neoplasia.

(4) Additional variations in specific HPV16 oncogene lineages might be one among

other weak further risk factors of cervical carcinoma.

(5) Chromosomal deletions involving 3p22 and 3p21.1-21.3 may be early events in

the development of cervical carcinoma and these areas probably harbor potential

tumor suppressor genes.

(6) The chromosomal deletions, occurring after HPV infection, may be very

important to assist HPV to accomplish the carcinogenesis of cervical neoplasia.

(7) hMLH1, one of the DNA mismatch repair genes, is not involved in the

carcinogenesis of cervical carcinoma.

Page 58: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

57

7 Acknowledgements

During the days and nights working on my Ph.D. project and this dissertation, I

have gotten a lot of help and encouragement from many persons. Here I would like to

express my most sincere thanks to:

Monica Nistér, current supervisor, for kindly helping, encouraging and supporting

me to finish the last half of my Ph.D. project. She scrutinizes the experimental work

patiently and discusses about the interpretation of results repeatedly with me. She

revises the manuscripts and dissertation so seriously that they can become perfect

even without a small punctuation error. From her correction work, I have learnt a lot

of written English which will benefit my future scientific career.

Jan Potén, primary supervisor, for scientific guiding and kindness to me and my

family. I do wish he could read this dissertation and give us comments. For this

dissertation, he already contributed many efforts to supervise me to work on it. Git,

for the hospitality and kindness to us in her family party.

Fredrik Pontén, for being a friendly colleague and a nice referee, for all the help

and encouragement.

Gijs Afink, for being a nice co-author and a nice referee. He speaks and writes

English much better than his native Dutch language. I wish I had that good English

skills like his. He likes to “argue” about scientific stuff in our project with me. I wish

we could have some opportunities to continue the “argument” in the future.

Zhongmin Guo and Feng Wu, for many years of friendship and encouragement.

We share many feeling about life, science, career and society. Guo is also a nice co-

author. Xiang Zou, for good friendship although we have a lot of different ideas about

politics. Xiaoqun Zhang, for the co-operation in the p53 transgenic mouse project.

The Chinese friends who worked or work with me in this department, for their

friendship, Siwei Peng, Zhiping Ren, Qin Li, Weiwei Zhang, Wenru Yu, Siqin Wu

and Guiling Li . The Chinese friends who shared or share a corridor with me, for the

friendship: Xuxia Wu–liang Liu, Ling Ling, Kiu Huang, Xiaoli Liu, Yi Wang, Li Li,

Yuman Zhang, Hao He, Sizhong Bu and Shao Hong.

Former group members: Anna Asplund, Ling Gao, Helena Bäckvall, Cecilia

Wassberg and Natalia Mazurenko, for wonderful co-operation, nice group meetings,

free talks and laughs, delicious food and stimulating alcohol at the group dinner

parties. Anna Asplund is also a nice co-author.

Page 59: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

58

Current group members in Monica´s group for the nice scientific presentations at

the group meeting.

Other co-authors: F Kisseljov, Sonja Andersson and Erik Wilander, for the co-

operation.

All other colleagues who work in the department of Genetics and Pathology, for

the great help.

Finally and particularly, my lovely daughter Xian Hu, my lovely son Ruiwen Hu,

my beloved wife Tianyun Pang and my parents, for their forever love and strong

support. Pang is also a nice co-author.

Page 60: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

59

8 References

1. Reid R. 1983. Genital warts and cervical cancer. II. Is human papillomavirusinfection the trigger to cervical carcinogenesis? Gynecol Oncol 15: 239-252.2. Singer A. 1975. The uterine cervix from adolescence to the menopause. Br JObstet Gynaecol 82: 81-99.3. Abdul-Karim FW, Fu YS, Reagan JW, Wentz WB. 1982. Morphometric studyof intraepithelial neoplasia of the uterine cervix. Obstet Gynecol 60: 210-214.4. Ottaviano M, La Torre P. 1982. Examination of the cervix with the naked eyeusing acetic acid test. Am J Obstet Gynecol 143: 139-142.5. Recher L, Srebnik E. 1981. Histopathologic features of koilocytotic atypia: adetailed description. Acta Cytol 25: 377-382.6. Rubinstein E. 1982. Colposcopic pattern of cervicitis, dysplasia and pre-invasive cancer of the uterine cervix. Acta Obstet Gynecol Scand 61: 253-259.7. Singer A. 1977. Dynamic anatomy of the cervical epithelium. Adv Exp MedBiol 89: 77-89.8. Laverty CR, Russell P, Hills E, Booth N. 1978. The significance ofnoncondylomatous wart virus infection of the cervical transformation zone. A reviewwith discussion of two illustrative cases. Acta Cytol 22: 195-201.9. Ponten J, Guo Z. 1998. Precancer of the human cervix. Cancer Surv 32: 201-229.10. Geirsson G, Woodworth FE, Patten SF, Jr., Bonfiglio TA. 1977. Epithelialrepair and regeneration in the uterine cervix. I. An analysis of the cells. Acta Cytol21: 371-378.11. Czernobilsky B, Zeituni M, Lancet M, Mazor B, Baram A, Deligdish L. 1977.The prevalence of cervicitis, reserve cell hyperplasia, squamous metaplasia, andcervical dysplasia in Jewish women. Obstet Gynecol 49: 587-591.12. Stanek J, Walas-Skolicka E. 1977. Squamous metaplastic cells in cervicalcanal aspiration smears in women with ovarian function disorders. ArchGeschwulstforsch 47: 440-443.13. Purola E, Savia E. 1977. Cytology of gynecologic condyloma acuminatum.Acta Cytol 21: 26-31.14. Fletcher S, Norval M. 1983. On the nature of the deep cellular disturbances inhuman-papilloma- virus infection of the squamous cervical epithelium. Lancet 2: 546-549.15. Matsukura T, Sugase M. 1995. Identification of genital humanpapillomaviruses in cervical biopsy specimens: segregation of specific virus types inspecific clinicopathologic lesions. Int J Cancer 61: 13-22.16. Sherman L, Golan Y, Mitrani-Rosenbaum S, Baram A. 1992. Differentialexpression of HPV types 6 and 11 in condylomas and cervical preneoplastic lesions.Virus Res 25: 23-36.17. Ward BE, Saleh AM, Williams JV, Zitz JC, Crum CP. 1992. Papillaryimmature metaplasia of the cervix: a distinct subset of exophytic cervical condylomaassociated with HPV-6/11 nucleic acids. Mod Pathol 5: 391-395.18. Willett GD, Kurman RJ, Reid R, Greenberg M, Jenson AB, Lorincz AT. 1989.Correlation of the histologic appearance of intraepithelial neoplasia of the cervix withhuman papillomavirus types. Emphasis on low grade lesions including so-called flatcondyloma. Int J Gynecol Pathol 8: 18-25.19. Nathan PM, Moss TR. 1991. Screening colposcopy in genitourinary medicine.Int J STD AIDS 2: 342-345.

Page 61: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

60

20. Fletcher S. 1983. Histopathology of papilloma virus infection of the cervixuteri: the history, taxonomy, nomenclature and reporting of koilocytic dysplasias. JClin Pathol 36: 616-624.21. Syrjanen KJ. 1979. Morphologic survey of the condylomatous lesions indysplastic and neoplastic epithelium of the uterine cervix. Arch Gynecol 227: 153-161.22. Kock KF, Johansen P. 1987. Prevalence of condylomatous atypia and humanpapilloma virus antigen in cervical biopsies in 1972 and 1983. Acta Obstet GynecolScand 66: 111-115.23. Syrjanen KJ. 1986. Human papillomavirus (HPV) infections of the femalegenital tract and their associations with intraepithelial neoplasia and squamous cellcarcinoma. Pathol Annu 21: 53-89.24. Purola EE, Halila H, Vesterinen E. 1983. Condyloma and cervical epithelialatypias in young women. Gynecol Oncol 16: 34-40.25. Syrjanen KJ. 1979. Histological and cytological evidence of a condylomatouslesion in association with an invasive carcinoma of uterine cervix. ArchGeschwulstforsch 49: 436-443.26. Oriel JD. 1981. Genital warts. Sex Transm Dis 8: 326-329.27. Burghardt E. 1973. Early histological diagnosis of cervical cancer. MajorProbl Obstet Gynecol 6: 1-401.28. Quaas J, Budner M, Heinrich J. 2000. Cervical intraepithelial neoplasia--biopsy methods as AIDS for diagnosis and therapy. Ginekol Pol 71: 109-115.29. Gustafsson L, Adami HO. 1989. Natural history of cervical neoplasia:consistent results obtained by an identification technique. Br J Cancer 60: 132-141.30. Ponten J, Adami HO, Bergstrom R, Dillner J, Friberg LG, Gustafsson L,Miller AB, Parkin DM, Sparen P, Trichopoulos D. 1995. Strategies for global controlof cervical cancer [see comments]. Int J Cancer 60: 1-26.31. Bos AB, van Ballegooijen M, van Oortmarssen GJ, van Marle ME, HabbemaJD, Lynge E. 1997. Non-progression of cervical intraepithelial neoplasia estimatedfrom population-screening data. Br J Cancer 75: 124-130.32. Nasiell K, Roger V, Nasiell M. 1986. Behavior of mild cervical dysplasiaduring long-term follow-up. Obstet Gynecol 67: 665-669.33. Nasiell K, Nasiell M, Vaclavinkova V. 1983. Behavior of moderate cervicaldysplasia during long-term follow-up. Obstet Gynecol 61: 609-614.34. Morrow CP, Cozen W. 1995. Perspective on cervical cancer: why prevent? JCell Biochem Suppl 23: 61-70.35. Nelson JH, Jr., Boyce J, Macasaet M, Lu T, Bohorquez JF, Nicastri AD,Fruchter R. 1977. Incidence, significance, and follow-up of para-aortic lymph nodemetastases in late invasive carcinoma of the cervix. Am J Obstet Gynecol 128: 336-340.36. Lerner HM, Jones HW, 3rd, Hill EC. 1980. Radical surgery for the treatmentof early invasive cervical carcinoma (stage IB): review of 15 years' experience. ObstetGynecol 56: 413-418.37. Sparen P, Gustafsson L, Friberg LG, Ponten J, Bergstrom R, Adami HO.1995. Improved control of invasive cervical cancer in Sweden over six decades byearlier clinical detection and better treatment. J Clin Oncol 13: 715-725.38. Brinton LA, Hamman RF, Huggins GR, Lehman HF, Levine RS, Mallin K,Fraumeni JF, Jr. 1987. Sexual and reproductive risk factors for invasive squamouscell cervical cancer. J Natl Cancer Inst 79: 23-30.39. Kjaer SK, de Villiers EM, Haugaard BJ, Christensen RB, Teisen C, MollerKA, Poll P, Jensen H, Vestergaard BF, Lynge E. 1988. Human papillomavirus, herpes

Page 62: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

61

simplex virus and cervical cancer incidence in Greenland and Denmark. Apopulation-based cross-sectional study. Int J Cancer 41: 518-524.40. Villa LL, Franco EL. 1989. Epidemiologic correlates of cervical neoplasia andrisk of human papillomavirus infection in asymptomatic women in Brazil. J NatlCancer Inst 81: 332-340.41. Bauer HM, Ting Y, Greer CE, Chambers JC, Tashiro CJ, Chimera J, ReingoldA, Manos MM. 1991. Genital human papillomavirus infection in female universitystudents as determined by a PCR-based method [see comments]. Jama 265: 472-477.42. Reeves WC, Brinton LA, Garcia M, Brenes MM, Herrero R, Gaitan E,Tenorio F, de BR, Rawls WE. 1989. Human papillomavirus infection and cervicalcancer in Latin America. N Engl J Med 320: 1437-1441.43. Kjaer SK, Engholm G, Teisen C, Haugaard BJ, Lynge E, Christensen RB,Moller KA, Jensen H, Poll P, Vestergaard BF. 1990. Risk factors for cervical humanpapillomavirus and herpes simplex virus infections in Greenland and Denmark: apopulation-based study. Am J Epidemiol 131: 669-682.44. Ley C, Bauer HM, Reingold A, Schiffman MH, Chambers JC, Tashiro CJ,Manos MM. 1991. Determinants of genital human papillomavirus infection in youngwomen. J Natl Cancer Inst 83: 997-1003.45. Campion MJ, McCance DJ, Cuzick J, Singer A. 1986. Progressive potential ofmild cervical atypia: prospective cytological, colposcopic, and virological study.Lancet 2: 237-240.46. Koutsky LA, Holmes KK, Critchlow CW, Stevens CE, Paavonen J,Beckmann AM, DeRouen TA, Galloway DA, Vernon D, Kiviat NB. 1992. A cohortstudy of the risk of cervical intraepithelial neoplasia grade 2 or 3 in relation topapillomavirus infection. N Engl J Med 327: 1272-1278.47. Londesborough P, Ho L, Terry G, Cuzick J, Wheeler C, Singer A. 1996.Human papillomavirus genotype as a predictor of persistence and development ofhigh-grade lesions in women with minor cervical abnormalities. Int J Cancer 69: 364-368.48. Goldberg GL, Vermund SH, Schiffman MH, Ritter DB, Spitzer C, Burk RD.1989. Comparison of Cytobrush and cervicovaginal lavage sampling methods for thedetection of genital human papillomavirus. Am J Obstet Gynecol 161: 1669-1672.49. Vermund SH, Schiffman MH, Goldberg GL, Ritter DB, Weltman A, BurkRD. 1989. Molecular diagnosis of genital human papillomavirus infection:comparison of two methods used to collect exfoliated cervical cells. Am J ObstetGynecol 160: 304-308.50. Kiviat NB, Koutsky LA, Critchlow CW, Galloway DA, Vernon DA, PetersonML, McElhose PE, Pendras SJ, Stevens CE, Holmes KK. 1990. Comparison ofSouthern transfer hybridization and dot filter hybridization for detection of cervicalhuman papillomavirus infection with types 6, 11, 16, 18, 31, 33, and 35. Am J ClinPathol 94: 561-565.51. Schiffman MH, Bauer HM, Lorincz AT, Manos MM, Byrne JC, Glass AG,Cadell DM, Howley PM. 1991. Comparison of Southern blot hybridization andpolymerase chain reaction methods for the detection of human papillomavirus DNA. JClin Microbiol 29: 573-577.52. Finch JT, Klug A. 1965. The structure of viruses of the papilloma-polyomatype 3. Structure of rabbit papilloma virus, with an appendix on the topography ofcontrast in negative-staining for electron-microscopy. J Mol Biol 13: 1-12.53. Yabe Y, Sadakane H, Isono H. 1979. Connection between capsomeres inhuman papilloma virus. Virology 96: 547-552.54. Gissmann L, Pfister H, Zur Hausen H. 1977. Human papilloma viruses(HPV): characterization of four different isolates. Virology 76: 569-580.

Page 63: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

62

55. Orth G, Favre M, Croissant O. 1977. Characterization of a new type of humanpapillomavirus that causes skin warts. J Virol 24: 108-120.56. Lancaster WD, Olson C. 1978. Demonstration of two distinct classes ofbovine papilloma virus. Virology 89: 372-379.57. Chen EY, Howley PM, Levinson AD, Seeburg PH. 1982. The primarystructure and genetic organization of the bovine papillomavirus type 1 genome.Nature 299: 529-534.58. Danos O, Katinka M, Yaniv M. 1982. Human papillomavirus 1a completeDNA sequence: a novel type of genome organization among papovaviridae. Embo J1: 231-236.59. Schwarz E, Durst M, Demankowski C, Lattermann O, Zech R, Wolfsperger E,Suhai S, zur Hausen H. 1983. DNA sequence and genome organization of genitalhuman papillomavirus type 6b. Embo J 2: 2341-2348.60. Kremsdorf D, Favre M, Jablonska S, Obalek S, Rueda LA, Lutzner MA,Blanchet-Bardon C, Van Voorst Vader PC, Orth G. 1984. Molecular cloning andcharacterization of the genomes of nine newly recognized human papillomavirustypes associated with epidermodysplasia verruciformis. J Virol 52: 1013-1018.61. Seedorf K, Krammer G, Durst M, Suhai S, Rowekamp WG. 1985. Humanpapillomavirus type 16 DNA sequence. Virology 145: 181-185.62. Danos O, Engel LW, Chen EY, Yaniv M, Howley PM. 1983. Comparativeanalysis of the human type 1a and bovine type 1 papillomavirus genomes. J Virol 46:557-566.63. Syrjanen KJ, Mantyjarvi R, Vayrynen M, Yliskoski M, Syrjanen SM,Saarikoski S, Nurmi T, Parkkinen S, Castren O. 1987. Cervical smears in assessmentof the natural history of human papillomavirus infections in prospectively followedwomen. Acta Cytol 31: 855-865.64. Kataja V, Syrjanen K, Syrjanen S, Mantyjarvi R, Yliskoski M, Saarikoski S,Salonen JT. 1990. Prospective follow-up of genital HPV infections: survival analysisof the HPV typing data. Eur J Epidemiol 6: 9-14.65. Cunningham M. 1983. Spot blot: a hybridization assay for specific DNAsequences in multiple samples. Anal Biochem 128: 415-421.66. Wagner D, Ikenberg H, Boehm N, Gissmann L. 1984. Identification of humanpapillomavirus in cervical swabs by deoxyribonucleic acid in situ hybridization.Obstet Gynecol 64: 767-772.67. Grussendorf EI, zur Hausen H. 1979. Localization of viral DNA-replication insections of human warts by nucleic acid hybridization with complementary RNA ofhuman papilloma virus Type 1. Arch Dermatol Res 264: 55-63.68. Gravitt P, Hakenewerth A, Stoerker J. 1991. A direct comparison of methodsproposed for use in widespread screening of human papillomavirus infections. MolCell Probes 5: 65-72.69. Brandsma J, Burk RD, Lancaster WD, Pfister H, Schiffman MH. 1989. Inter-laboratory variation as an explanation for varying prevalence estimates of humanpapillomavirus infection. Int J Cancer 43: 260-262.70. Bartholoma NY, Adelson MD, Forbes BA. 1991. Evaluation of twocommercially available nucleic acid hybridization assays for the detection and typingof human papillomavirus in clinical specimens. Am J Clin Pathol 95: 21-29.71. Xi LF, Koutsky LA, Galloway DA, Kuypers J, Hughes JP, Wheeler CM,Holmes KK, Kiviat NB. 1997. Genomic variation of human papillomavirus type 16and risk for high grade cervical intraepithelial neoplasia [see comments]. J NatlCancer Inst 89: 796-802.72. zur Hausen H. 2000. Papillomaviruses causing cancer: evasion from host-cellcontrol in early events in carcinogenesis. J Natl Cancer Inst 92: 690-698.

Page 64: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

63

73. Gross G, Ellinger K, Roussaki A, Fuchs PG, Peter HH, Pfister H. 1988.Epidermodysplasia verruciformis in a patient with Hodgkin's disease: characterizationof a new papillomavirus type and interferon treatment. J Invest Dermatol 91: 43-48.74. Corley E, Pueyo S, Goc B, Diaz A, Zorzopulos J. 1988. Papillomaviruses inhuman skin warts and their incidence in an Argentine population. Diagn MicrobiolInfect Dis 10: 93-101.75. Bergeron C, Barrasso R, Beaudenon S, Flamant P, Croissant O, Orth G. 1992.Human papillomaviruses associated with cervical intraepithelial neoplasia. Greatdiversity and distinct distribution in low- and high-grade lesions. Am J Surg Pathol16: 641-649.76. de Villiers EM, Neumann C, Le JY, Weidauer H, zur Hausen H. 1986.Infection of the oral mucosa with defined types of human papillomaviruses. MedMicrobiol Immunol 174: 287-294.77. Lindeberg H, Krogdahl A. 1999. Laryngeal cancer and human papillomavirus:HPV is absent in the majority of laryngeal carcinomas. Cancer Lett 146: 9-13.78. Azzimonti B, Hertel L, Aluffi P, Pia F, Monga G, Zocchi M, Landolfo S,Gariglio M. 1999. Demonstration of multiple HPV types in laryngeal premalignantlesions using polymerase chain reaction and immunohistochemistry. J Med Virol 59:110-116.79. Tsuhako K, Nakazato I, Hirayasu T, Sunakawa H, Iwamasa T. 1998. Humanpapillomavirus DNA in adenosquamous carcinoma of the lung. J Clin Pathol 51: 741-749.80. West AB, Soloway GN, Lizarraga G, Tyrrell L, Longley JB. 1996. Type 73human papillomavirus in esophageal squamous cell carcinoma: a novel association.Cancer 77: 2440-2444.81. He D, Zhang DK, Lam KY, Ma L, Ngan HY, Liu SS, Tsao SW. 1997.Prevalence of HPV infection in esophageal squamous cell carcinoma in Chinesepatients and its relationship to the p53 gene mutation. Int J Cancer 72: 959-964.82. Lavergne D, de Villiers EM. 1999. Papillomavirus in esophageal papillomasand carcinomas. Int J Cancer 80: 681-684.83. Van Den Brule AJ, Walboomers JM, Du Maine M, Kenemans P, Meijer CJ.1991. Difference in prevalence of human papillomavirus genotypes incytomorphologically normal cervical smears is associated with a history of cervicalintraepithelial neoplasia. Int J Cancer 48: 404-408.84. Young LS, Bevan IS, Johnson MA, Blomfield PI, Bromidge T, Maitland NJ,Woodman CB. 1989. The polymerase chain reaction: a new epidemiological tool forinvestigating cervical human papillomavirus infection. Bmj 298: 14-18.85. Ishi K, Suzuki F, Saito A, Kubota T. 2000. Prevalence of humanpapillomavirus infection and its correlation with cervical lesions in commercial-sexworkers in Japan. J Obstet Gynaecol Res 26: 253-257.86. Liaw KL, Hsing AW, Chen CJ, Schiffman MH, Zhang TY, Hsieh CY, GreerCE, You SL, Huang TW, Wu TC. 1995. Human papillomavirus and cervicalneoplasia: a case-control study in Taiwan. Int J Cancer 62: 565-571.87. Becker TM, Wheeler CM, McGough NS, Parmenter CA, Jordan SW, StidleyCA, McPherson RS, Dorin MH. 1994. Sexually transmitted diseases and other riskfactors for cervical dysplasia among southwestern Hispanic and non-Hispanic whitewomen. Jama 271: 1181-1188.88. Coker AL, Jenkins GR, Busnardo MS, Chambers JC, Levine LZ, Pirisi L.1993. Human papillomaviruses and cervical neoplasia in South Carolina. CancerEpidemiol Biomarkers Prev 2: 207-212.89. Das BC, Sharma JK, Gopalkrishna V, Das DK, Singh V, Gissmann L, zurHausen H, Luthra UK. 1992. A high frequency of human papillomavirus DNA

Page 65: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

64

sequences in cervical carcinomas of Indian women as revealed by Southern blothybridization and polymerase chain reaction. J Med Virol 36: 239-245.90. Schiffman MH, Bauer HM, Hoover RN, Glass AG, Cadell DM, Rush BB,Scott DR, Sherman ME, Kurman RJ, Wacholder S. 1993. Epidemiologic evidenceshowing that human papillomavirus infection causes most cervical intraepithelialneoplasia. J Natl Cancer Inst 85: 958-964.91. Lorincz AT, Reid R, Jenson AB, Greenberg MD, Lancaster W, Kurman RJ.1992. Human papillomavirus infection of the cervix: relative risk associations of 15common anogenital types. Obstet Gynecol 79: 328-337.92. Crum CP, Mitao M, Levine RU, Silverstein S. 1985. Cervicalpapillomaviruses segregate within morphologically distinct precancerous lesions. JVirol 54: 675-681.93. Beaudenon S, Kremsdorf D, Croissant O, Jablonska S, Wain-Hobson S, OrthG. 1986. A novel type of human papillomavirus associated with genital neoplasias.Nature 321: 246-249.94. Reid R, Greenberg M, Jenson AB, Husain M, Willett J, Daoud Y, Temple G,Stanhope CR, Sherman AI, Phibbs GD. 1987. Sexually transmitted papillomaviralinfections. I. The anatomic distribution and pathologic grade of neoplastic lesionsassociated with different viral types. Am J Obstet Gynecol 156: 212-222.95. Lorincz AT, Lancaster WD, Temple GF. 1986. Cloning and characterizationof the DNA of a new human papillomavirus from a woman with dysplasia of theuterine cervix. J Virol 58: 225-229.96. Lorincz AT, Quinn AP, Lancaster WD, Temple GF. 1987. A new type ofpapillomavirus associated with cancer of the uterine cervix. Virology 159: 187-190.97. Kiviat NB, Koutsky LA. 1993. Specific human papillomavirus types as thecausal agents of most cervical intraepithelial neoplasia: implications for current viewsand treatment. J Natl Cancer Inst 85: 934-935.98. Bosch FX, Manos MM, Munoz N, Sherman M, Jansen AM, Peto J, SchiffmanMH, Moreno V, Kurman R, Shah KV. 1995. Prevalence of human papillomavirus incervical cancer: a worldwide perspective. International biological study on cervicalcancer (IBSCC) Study Group [see comments]. J Natl Cancer Inst 87: 796-802.99. Burger RA, Monk BJ, Kurosaki T, Anton CH, Vasilev SA, Berman ML,Wilczynski SP. 1996. Human papillomavirus type 18: association with poor prognosisin early stage cervical cancer [see comments]. J Natl Cancer Inst 88: 1361-1368.100. Xi LF, Demers GW, Koutsky LA, Kiviat NB, Kuypers J, Watts DH, HolmesKK, Galloway DA. 1995. Analysis of human papillomavirus type 16 variantsindicates establishment of persistent infection. J Infect Dis 172: 747-755.101. Dyson N, Howley PM, Munger K, Harlow E. 1989. The human papillomavirus-16 E7 oncoprotein is able to bind to the retinoblastoma gene product. Science243: 934-937.102. Phelps WC, Munger K, Yee CL, Barnes JA, Howley PM. 1992. Structure-function analysis of the human papillomavirus type 16 E7 oncoprotein. J Virol 66:2418-2427.103. von KDM, Rittmuller C, Aengeneyndt F, Jansen DP, Spitkovsky D. 1994.Reversible repression of papillomavirus oncogene expression in cervical carcinomacells: consequences for the phenotype and E6-p53 and E7-pRB interactions. J Virol68: 2811-2821.104. Werness BA, Levine AJ, Howley PM. 1990. Association of humanpapillomavirus types 16 and 18 E6 proteins with p53. Science 248: 76-79.105. Burger RA, Monk BJ, Kurosaki T, Anton-Culver H, Vasilev SA, Berman ML,Wilczynski SP. 1996. Human papillomavirus type 18: association with poor prognosisin early stage cervical cancer. J Natl Cancer Inst 88: 1361-1368.

Page 66: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

65

106. Zaninetti P, Franceschi S, Baccolo M, Bonazzi B, Gottardi G, Serraino D.1986. Characteristics of women under 20 with cervical intraepithelial neoplasia. Int JEpidemiol 15: 477-482.107. Kanetsky PA, Gammon MD, Mandelblatt J, Zhang ZF, Ramsey E, Wright TC,Jr., Thomas L, Matseoane S, Lazaro N, Felton HT, Sachdev RK, Richart RM, CurtinJP. 1998. Cigarette smoking and cervical dysplasia among non-Hispanic blackwomen. Cancer Detect Prev 22: 109-119.108. Brinton LA, Huggins GR, Lehman HF, Mallin K, Savitz DA, Trapido E,Rosenthal J, Hoover R. 1986. Long-term use of oral contraceptives and risk ofinvasive cervical cancer. Int J Cancer 38: 399-344.109. Layde PM, Broste SK. 1989. Carcinoma of the cervix and smoking. BiomedPharmacother 43: 161-165.110. Reeves WC, Caussy D, Brinton LA, Brenes MM, Montalvan P, Gomez B, deBritton RC, Morice E, Gaitan E, de Lao SL. 1987. Case-control study of humanpapillomaviruses and cervical cancer in Latin America. Int J Cancer 40: 450-454.111. Ylitalo N, Sorensen P, Josefsson A, Frisch M, Sparen P, Ponten J, GyllenstenU, Melbye M, Adami HO. 1999. Smoking and oral contraceptives as risk factors forcervical carcinoma in situ. Int J Cancer 81: 357-365.112. Herrero R, Brinton LA, Reeves WC, Brenes MM, Tenorio F, de Britton RC,Gaitan E, Garcia M, Rawls WE. 1989. Invasive cervical cancer and smoking in LatinAmerica. J Natl Cancer Inst 81: 205-211.113. Hildesheim A, Herrero R, Castle PE, Wacholder S, Bratti MC, Sherman ME,Lorincz AT, Burk RD, Morales J, Rodriguez AC, Helgesen K, Alfaro M, HutchinsonM, Balmaceda I, Greenberg M, Schiffman M. 2001. HPV co-factors related to thedevelopment of cervical cancer: results from a population-based study in Costa Rica.Br J Cancer 84: 1219-1226.114. Brinton LA. 1991. Oral contraceptives and cervical neoplasia. Contraception43: 581-595.115. Reeves WC, Brinton LA, Brenes MM, Quiroz E, Rawls WE, De Britton RC.1985. Case control study of cervical cancer in Herrera Province, Republic of Panama.Int J Cancer 36: 55-60.116. Vessey M, Grice D. 1989. Carcinoma of the cervix and oral contraceptives:epidemiological studies. Biomed Pharmacother 43: 157-160.117. Chang AR. 1989. Hormonal contraceptives, human papillomaviruses andcervical cancer; some observations from a colposcopy clinic. Aust N Z J ObstetGynaecol 29: 329-331.118. Celentano DD, Klassen AC, Weisman CS, Rosenshein NB. 1987. The role ofcontraceptive use in cervical cancer: the Maryland Cervical Cancer Case-ControlStudy. Am J Epidemiol 126: 592-604.119. Pike MC, Spicer DV. 2000. Hormonal contraception and chemoprevention offemale cancers. Endocr Relat Cancer 7: 73-83.120. Magnusson PK, Sparen P, Gyllensten UB. 1999. Genetic link to cervicaltumours. Nature 400: 29-30.121. Magnusson PK, Lichtenstein P, Gyllensten UB. 2000. Heritability of cervicaltumours. Int J Cancer 88: 698-701.122. Maciag PC, Schlecht NF, Souza PS, Franco EL, Villa LL, Petzl-Erler ML.2000. Major histocompatibility complex class II polymorphisms and risk of cervicalcancer and human papillomavirus infection in Brazilian women. Cancer EpidemiolBiomarkers Prev 9: 1183-1191.123. Hemminki K, Dong C, Vaittinen P. 1999. Familial risks in cervical cancer: isthere a hereditary component? Int J Cancer 82: 775-781.

Page 67: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

66

124. Hemminki K, Li X, Mutanen P. 2001. Familial risks in invasive and in situcervical cancer by histological type. Eur J Cancer Prev 10: 83-89.125. Griffiths DF, Davies SJ, Williams D, Williams GT, Williams ED. 1988.Demonstration of somatic mutation and colonic crypt clonality by X- linked enzymehistochemistry. Nature 333: 461-463.126. Noguchi S, Motomura K, Inaji H, Imaoka S, Koyama H. 1992. Clonal analysisof human breast cancer by means of the polymerase chain reaction. Cancer Res 52:6594-6597.127. Sidransky D, Frost P, Von Eschenbach A, Oyasu R, Preisinger AC, VogelsteinB. 1992. Clonal origin bladder cancer. N Engl J Med 326: 737-740.128. Fearon ER, Hamilton SR, Vogelstein B. 1987. Clonal analysis of humancolorectal tumors. Science 238: 193-197.129. Paradis V, Laurent A, Flejou JF, Vidaud M, Bedossa P. 1997. Evidence forthe polyclonal nature of focal nodular hyperplasia of the liver by the study of X-chromosome inactivation. Hepatology 26: 891-895.130. Lyon MF. 1989. X-chromosome inactivation as a system of gene dosagecompensation to regulate gene expression. Prog Nucleic Acid Res Mol Biol 36: 119-130.131. Li E, Beard C, Jaenisch R. 1993. Role for DNA methylation in genomicimprinting. Nature 366: 362-365.132. Riggs AD, Pfeifer GP. 1992. X-chromosome inactivation and cell memory.Trends Genet 8: 169-174.133. Allen RC, Zoghbi HY, Moseley AB, Rosenblatt HM, Belmont JW. 1992.Methylation of HpaII and HhaI sites near the polymorphic CAG repeat in the humanandrogen-receptor gene correlates with X chromosome inactivation. Am J Hum Genet51: 1229-1239.134. Edwards A, Hammond HA, Jin L, Caskey CT, Chakraborty R. 1992. Geneticvariation at five trimeric and tetrameric tandem repeat loci in four human populationgroups. Genomics 12: 241-253.135. Diaz-Cano SJ. 2000. Designing a molecular analysis of clonality in tumours. JPathol 191: 343-344.136. Park TW, Richart RM, Sun XW, Wright TC, Jr. 1996. Association betweenhuman papillomavirus type and clonal status of cervical squamous intraepitheliallesions. J Natl Cancer Inst 88: 355-358.137. Enomoto T, Haba T, Fujita M, Hamada T, Yoshino K, Nakashima R, Wada H,Kurachi H, Wakasa K, Sakurai M, Murata Y, Shroyer KR. 1997. Clonal analysis ofhigh-grade squamous intra-epithelial lesions of the uterine cervix. Int J Cancer 73:339-344.138. Enomoto T, Fujita M, Inoue M, Tanizawa O, Nomura T, Shroyer KR. 1994.Analysis of clonality by amplification of short tandem repeats. Carcinomas of thefemale reproductive tract. Diagn Mol Pathol 3: 292-297.139. Guo Z, Thunberg U, Sallstrom J, Wilander E, Ponten J. 1998. Clonalityanalysis of cervical cancer on microdissected archival materials by PCR-based X-chromosome inactivation approach. Int J Oncol 12: 1327-1332.140. Guo Z, Ponten F, Wilander E, Ponten J. 2000. Clonality of precursors ofcervical cancer and their genetical links to invasive cancer. Mod Pathol 13: 606-613.141. Guo Z, Wu F, Asplund A, Hu X, Mazurenko N, Kisseljov F, Ponten J,Wilander E. 2001. Analysis of intratumoral heterogeneity of chromosome 3pdeletions and genetic evidence of polyclonal origin of cervical squamous carcinoma.Mod Pathol 14: 54-61.142. Ko HM, Choi C, Park CS, Juhng SW. 1997. Analysis of clonality by Xchromosome inactivation in uterine cervix cancer. J Korean Med Sci 12: 322-326.

Page 68: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

67

143. Chuaqui R, Silva M, Emmert-Buck M. 2001. Allelic deletion mapping onchromosome 6q and X chromosome inactivation clonality patterns in cervicalintraepithelial neoplasia and invasive carcinoma. Gynecol Oncol 80: 364-371.144. Sawada M, Azuma C, Hashimoto K, Noguchi S, Ozaki M, Saji F, TanizawaO. 1994. Clonal analysis of human gynecologic cancers by means of the polymerasechain reaction. Int J Cancer 58: 492-496.145. Stoehr R, Hartmann A, Hiendlmeyer E, Murle K, Wieland W, Knuechel R.2000. Oligoclonality of early lesions of the urothelium as determined bymicrodissection-supported genetic analysis. Pathobiology 68: 165-172.146. Kersemaekers AM, van de Vijver MJ, Fleuren GJ. 2000. Comparison of thegenetic alterations in two epithelial collision tumors of the uterine cervix. A report oftwo cases. Int J Gynecol Pathol 19: 225-230.147. Koyama N, Nishihira J, Nakabayashi H, Nishi S, Hoshi N, Fujimoto S,Shimizu M, Yoshiki T, Sugihara T. 2000. Aneuploidy of sex chromosomes in basalcell carcinoma: its clonality and involvement in the development of carcinogenesis.Int J Oncol 16: 15-23.148. Medeiros LJ, Carr J. 1999. Overview of the role of molecular methods in thediagnosis of malignant lymphomas. Arch Pathol Lab Med 123: 1189-1207.149. Rinaldi F, Mairs RJ, Wheldon TE, Katz F, Chessells JM, Gibson BE. 1999.Clonality analysis suggests that early-onset acute lymphoblastic leukaemia is ofsingle-cell origin and implies no major role for germ cell mutations in parents. Br JCancer 80: 909-913.150. Niho S, Yokose T, Suzuki K, Kodama T, Nishiwaki Y, Mukai K. 1999.Monoclonality of atypical adenomatous hyperplasia of the lung. Am J Pathol 154:249-254.151. Siu IM, Robinson DR, Schwartz S, Kung HJ, Pretlow TG, Petersen RB,Pretlow TP. 1999. The identification of monoclonality in human aberrant crypt foci.Cancer Res 59: 63-66.152. Bevilacqua RA, Nunes DN, Stroun M, Anker P. 1998. The use of geneticinstability as a clinical tool for cancer diagnosis. Semin Cancer Biol 8: 447-453.153. Tanaka T, Chang L, Umesaki N, Ogita S. 1997. Clonal analysis of cancer cellsthat survived anticancer drug treatment. Osaka City Med J 43: 259-265.154. van Oijen MG, Leppers Vd Straat FG, Tilanus MG, Slootweg PJ. 2000. Theorigins of multiple squamous cell carcinomas in the aerodigestive tract. Cancer 88:884-893.155. Shibata A, Tsai YC, Press MF, Henderson BE, Jones PA, Ross RK. 1996.Clonal analysis of bilateral breast cancer. Clin Cancer Res 2: 743-748.156. Yamamoto T, Kajino K, Kudo M, Sasaki Y, Arakawa Y, Hino O. 1999.Determination of the clonal origin of multiple human hepatocellular carcinomas bycloning and polymerase chain reaction of the integrated hepatitis B virus DNA.Hepatology 29: 1446-1452.157. Dijkhuizen SM, Entius MM, Clement MJ, Polak MM, Van den Berg FM,Craanen ME, Slebos RJ, Offerhaus GJ. 1997. Multiple hyperplastic polyps in thestomach: evidence for clonality and neoplastic potential. Gastroenterology 112: 561-566.158. Gill PS, Tsai YC, Rao AP, Spruck CH, 3rd, Zheng T, Harrington WA, Jr.,Cheung T, Nathwani B, Jones PA. 1998. Evidence for multiclonality in multicentricKaposi's sarcoma. Proc Natl Acad Sci U S A 95: 8257-8261.159. Magda JL, Minger BA, Rimm DL. 1998. Polymerase chain reaction-baseddetection of clonality as a non- morphologic diagnostic tool for fine-needle aspirationof the breast. Cancer 84: 262-267.

Page 69: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

68

160. Malamou-Mitsi VD, Krikoni OC, Agnantis NJ. 1996. Clonal analysis by PCRand RFLP in breast cancer and precancerous lesions. Preliminary data. AnticancerRes 16: 3943-3948.161. Hartmann A, Rosner U, Schlake G, Dietmaier W, Zaak D, Hofstaedter F,Knuechel R. 2000. Clonality and genetic divergence in multifocal low-gradesuperficial urothelial carcinoma as determined by chromosome 9 and p53 deletionanalysis. Lab Invest 80: 709-718.162. Garcia SB, Novelli M, Wright NA. 2000. The clonal origin and clonalevolution of epithelial tumours. Int J Exp Pathol 81: 89-116.163. Palefsky JM, Holly EA. 1995. Molecular virology and epidemiology ofhuman papillomavirus and cervical cancer. Cancer Epidemiol Biomarkers Prev 4:415-428.164. Hawley-Nelson P, Vousden KH, Hubbert NL, Lowy DR, Schiller JT. 1989.HPV16 E6 and E7 proteins cooperate to immortalize human foreskin keratinocytes.Embo J 8: 3905-3910.165. Sedman SA, Barbosa MS, Vass WC, Hubbert NL, Haas JA, Lowy DR,Schiller JT. 1991. The full-length E6 protein of human papillomavirus type 16 hastransforming and trans-activating activities and cooperates with E7 to immortalizekeratinocytes in culture. J Virol 65: 4860-4866.166. White AE, Livanos EM, Tlsty TD. 1994. Differential disruption of genomicintegrity and cell cycle regulation in normal human fibroblasts by the HPVoncoproteins. Genes Dev 8: 666-677.167. Havre PA, Yuan J, Hedrick L, Cho KR, Glazer PM. 1995. p53 inactivation byHPV16 E6 results in increased mutagenesis in human cells. Cancer Res 55: 4420-4424.168. Klingelhutz AJ, Foster SA, McDougall JK. 1996. Telomerase activation bythe E6 gene product of human papillomavirus type 16. Nature 380: 79-82.169. Smotkin D, Wettstein FO. 1987. The major human papillomavirus protein incervical cancers is a cytoplasmic phosphoprotein. J Virol 61: 1686-1689.170. Halbert CL, Demers GW, Galloway DA. 1991. The E7 gene of humanpapillomavirus type 16 is sufficient for immortalization of human epithelial cells. JVirol 65: 473-478.171. Dyson N, Guida P, Munger K, Harlow E. 1992. Homologous sequences inadenovirus E1A and human papillomavirus E7 proteins mediate interaction with thesame set of cellular proteins. J Virol 66: 6893-6902.172. Jones DL, Alani RM, Munger K. 1997. The human papillomavirus E7oncoprotein can uncouple cellular differentiation and proliferation in humankeratinocytes by abrogating p21Cip1-mediated inhibition of cdk2. Genes Dev 11:2101-2111.173. Arroyo M, Bagchi S, Raychaudhuri P. 1993. Association of the humanpapillomavirus type 16 E7 protein with the S- phase-specific E2F-cyclin A complex.Mol Cell Biol 13: 6537-6546.174. Kessis TD, Connolly DC, Hedrick L, Cho KR. 1996. Expression of HPV16E6 or E7 increases integration of foreign DNA. Oncogene 13: 427-431.175. Hwang ES, Nottoli T, Dimaio D. 1995. The HPV16 E5 protein: expression,detection, and stable complex formation with transmembrane proteins in COS cells.Virology 211: 227-233.176. Schlegel R, Wade-Glass M, Rabson MS, Yang YC. 1986. The E5transforming gene of bovine papillomavirus encodes a small, hydrophobicpolypeptide. Science 233: 464-467.

Page 70: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

69

177. Conrad M, Bubb VJ, Schlegel R. 1993. The human papillomavirus type 6 and16 E5 proteins are membrane- associated proteins which associate with the 16-kilodalton pore-forming protein. J Virol 67: 6170-6178.178. Gu Z, Matlashewski G. 1995. Effect of human papillomavirus type 16oncogenes on MAP kinase activity. J Virol 69: 8051-8056.179. Crusius K, Auvinen E, Alonso A. 1997. Enhancement of EGF- and PMA-mediated MAP kinase activation in cells expressing the human papillomavirus type16 E5 protein. Oncogene 15: 1437-1444.180. Tsao YP, Li LY, Tsai TC, Chen SL. 1996. Human papillomavirus type 11 and16 E5 represses p21(WafI/SdiI/CipI) gene expression in fibroblasts and keratinocytes.J Virol 70: 7535-7539.181. Venuti A, Salani D, Poggiali F, Manni V, Bagnato A. 1998. The E5oncoprotein of human papillomavirus type 16 enhances endothelin-1-inducedkeratinocyte growth. Virology 248: 1-5.182. Bible JM, Mant C, Best JM, Kell B, Starkey WG, Shanti Raju K, Seed P,Biswas C, Muir P, Banatvala JE, Cason J. 2000. Cervical lesions are associated withhuman papillomavirus type 16 intratypic variants that have high transcriptionalactivity and increased usage of common mammalian codons. J Gen Virol 81 Pt 6:1517-1527.183. Biswas C, Kell B, Mant C, Jewers RJ, Cason J, Muir P, Raju KS, Best JM.1997. Detection of human papillomavirus type 16 early-gene transcription by reversetranscription-PCR is associated with abnormal cervical cytology. J Clin Microbiol 35:1560-1564.184. Dowhanick JJ, McBride AA, Howley PM. 1995. Suppression of cellularproliferation by the papillomavirus E2 protein. J Virol 69: 7791-7799.185. Yasugi T, Benson JD, Sakai H, Vidal M, Howley PM. 1997. Mapping andcharacterization of the interaction domains of human papillomavirus type 16 E1 andE2 proteins. J Virol 71: 891-899.186. Del Vecchio AM, Romanczuk H, Howley PM, Baker CC. 1992. Transientreplication of human papillomavirus DNAs. J Virol 66: 5949-5958.187. Romanczuk H, Howley PM. 1992. Disruption of either the E1 or the E2regulatory gene of human papillomavirus type 16 increases viral immortalizationcapacity. Proc Natl Acad Sci U S A 89: 3159-3163.188. Crum CP, Barber S, Symbula M, Snyder K, Saleh AM, Roche JK. 1990.Coexpression of the human papillomavirus type 16 E4 and L1 open reading frames inearly cervical neoplasia. Virology 178: 238-246.189. Doorbar J, Ely S, Sterling J, McLean C, Crawford L. 1991. Specificinteraction between HPV-16 E1-E4 and cytokeratins results in collapse of theepithelial cell intermediate filament network. Nature 352: 824-827.190. Komly CA, Breitburd F, Croissant O, Streeck RE. 1986. The L2 open readingframe of human papillomavirus type 1a encodes a minor structural protein carryingtype-specific antigens. J Virol 60: 813-816.191. Romanczuk H, Villa LL, Schlegel R, Howley PM. 1991. The viraltranscriptional regulatory region upstream of the E6 and E7 genes is a majordeterminant of the differential immortalization activities of human papillomavirustypes 16 and 18. J Virol 65: 2739-2744.192. Yamada T, Manos MM, Peto J, Greer CE, Munoz N, Bosch FX, Wheeler CM.1997. Human papillomavirus type 16 sequence variation in cervical cancers: aworldwide perspective. J Virol 71: 2463-2472.193. Alvarez-Salas L, Wilczynski S, Burger R, Monk B, Dipaolo J. 1995.Polymorphism of the HPV-16 E6 gene of cervical carcinoma. International Journal ofOncology 7: 261-266.

Page 71: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

70

194. Yamada T, Wheeler CM, Halpern AL, Stewart AC, Hildesheim A, JenisonSA. 1995. Human papillomavirus type 16 variant lineages in United Statespopulations characterized by nucleotide sequence analysis of the E6, L2, and L1coding segments. J Virol 69: 7743-7753.195. Zehbe I, Voglino G, Delius H, Wilander E, Tommasino M. 1998. Risk ofcervical cancer and geographical variations of human papillomavirus 16 E6polymorphisms [letter]. Lancet 352: 1441-1442.196. Zehbe I, Wilander E, Delius H, Tommasino M. 1998. Human papillomavirus16 E6 variants are more prevalent in invasive cervical carcinoma than the prototype.Cancer Res 58: 829-833.197. Stoppler MC, Ching K, Stoppler H, Clancy K, Schlegel R, Icenogle J. 1996.Natural variants of the human papillomavirus type 16 E6 protein differ in theirabilities to alter keratinocyte differentiation and to induce p53 degradation. J Virol 70:6987-6993.198. Matsumoto K, Yoshikawa H, Nakagawa S, Tang X, Yasugi T, Kawana K,Sekiya S, Hirai Y, Kukimoto I, Kanda T, Taketani Y. 2000. Enhanced oncogenicityof human papillomavirus type 16 (HPV16) variants in Japanese population. CancerLett 156: 159-165.199. Icenogle JP, Sathya P, Miller DL, Tucker RA, Rawls WE. 1991. Nucleotideand amino acid sequence variation in the L1 and E7 open reading frames of humanpapillomavirus type 6 and type 16. Virology 184: 101-107.200. Luxton J, Mant C, Greenwood B, Derias N, Nath R, Shepherd P, Cason J.2000. HPV16 E6 oncogene variants in women with cervical intraepithelial neoplasia.J Med Virol 60: 337-341.201. Fujinaga Y, Okazawa K, Nishikawa A, Yamakawa Y, Fukushima M, Kato I,Fujinaga K. 1994. Sequence variation of human papillomavirus type 16 E7 inpreinvasive and invasive cervical neoplasias. Virus Genes 9: 85-92.202. Terry G, Ho L, Cuzick J. 1997. Analysis of E2 amino acid variants of humanpapillomavirus types 16 and 18 and their associations with lesion grade and HLADR/DQ type. Int J Cancer 73: 651-655.203. Bontkes HJ, van DM, de GT, Duggan KM, Walboomers JM, Stukart MJ,Verheijen RH, Helmerhorst TJ, Meijer CJ, Scheper RJ, Stevens FR, Dyer PA, SinnottP, Stern PL. 1998. HPV 16 infection and progression of cervical intra-epithelialneoplasia: analysis of HLA polymorphism and HPV 16 E6 sequence variants. Int JCancer 78: 166-171.204. Jeon S, Allen HB, Lambert PF. 1995. Integration of human papillomavirustype 16 into the human genome correlates with a selective growth advantage of cells.J Virol 69: 2989-2997.205. Cullen AP, Reid R, Campion M, Lorincz AT. 1991. Analysis of the physicalstate of different human papillomavirus DNAs in intraepithelial and invasive cervicalneoplasm. J Virol 65: 606-612.206. Kalantari M, Blennow E, Hagmar B, Johansson B. 2001. Physical state ofHPV16 and chromosomal mapping of the integrated form in cervical carcinomas.Diagn Mol Pathol 10: 46-54.207. Samoylova EV, Shaikhaiev GO, Petrov SV, Kisseljova NP, Kisseljov FL.1995. HPV infection in cervical-cancer cases in Russia. Int J Cancer 61: 337-341.208. Kristiansen E, Jenkins A, Holm R. 1994. Coexistence of episomal andintegrated HPV16 DNA in squamous cell carcinoma of the cervix. J Clin Pathol 47:253-256.209. Choo KB, Pan CC, Liu MS, Ng HT, Chen CP, Lee YN, Chao CF, Meng CL,Yeh MY, Han SH. 1987. Presence of episomal and integrated human papillomavirusDNA sequences in cervical carcinoma. J Med Virol 21: 101-107.

Page 72: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

71

210. Hall WS, Goto-Mandeville R, Shih HA, Shank PR, Braun L. 1997. Molecularanalysis of episomal human papillomavirus type 16 DNA in a cervical carcinoma cellline. Virus Res 51: 183-195.211. Pirami L, Giache V, Becciolini A. 1997. Analysis of HPV16, 18, 31, and 35DNA in pre-invasive and invasive lesions of the uterine cervix. J Clin Pathol 50: 600-604.212. Lehn H, Villa LL, Marziona F, Hilgarth M, Hillemans HG, Sauer G. 1988.Physical state and biological activity of human papillomavirus genomes inprecancerous lesions of the female genital tract. J Gen Virol 69: 187-196.213. el Awady MK, Kaplan JB, O'Brien SJ, Burk RD. 1987. Molecular analysis ofintegrated human papillomavirus 16 sequences in the cervical cancer cell line SiHa.Virology 159: 389-398.214. Lazo PA. 1988. Rearrangement of both alleles of human chromosome 8 inHeLa cells, one of them as a result of papillomavirus DNA integration. J Biol Chem263: 360-367.215. Choo KB, Lee HH, Liew LN, Chong KY, Chou HF. 1990. Analysis of theunoccupied site of an integrated human papillomavirus 16 sequence in a cervicalcarcinoma. Virology 178: 621-625.216. Gallego MI, Schoenmakers EF, Van de Ven WJ, Lazo PA. 1997. Complexgenomic rearrangement within the 12q15 multiple aberration region induced byintegrated human papillomavirus 18 in a cervical carcinoma cell line. Mol Carcinog19: 114-121.217. Jones MH, Nakamura Y. 1992. Deletion mapping of chromosome 3p infemale genital tract malignancies using microsatellite polymorphisms. Oncogene 7:1631-1634.218. Kohno T, Takayama H, Hamaguchi M, Takano H, Yamaguchi N, Tsuda H,Hirohashi S, Vissing H, Shimizu M, Oshimura M, et al. 1993. Deletion mapping ofchromosome 3p in human uterine cervical cancer. Oncogene 8: 1825-1832.219. Karlsen F, Rabbitts PH, Sundresan V, Hagmar B. 1994. PCR-RFLP studies onchromosome 3p in formaldehyde-fixed, paraffin- embedded cervical cancer tissues.Int J Cancer 58: 787-792.220. Larson AA, Kern S, Curtiss S, Gordon R, Cavenee WK, Hampton GM. 1997.High resolution analysis of chromosome 3p alterations in cervical carcinoma. CancerRes 57: 4082-4090.221. Chu TY, Shen CY, Chiou YS, Lu JJ, Perng CL, Yu MS, Liu HS. 1998. HPV-associated cervical cancers show frequent allelic loss at 3p14 but no apparentaberration of FHIT mRNA. Int J Cancer 75: 199-204.222. Guo Z, Wilander E, Sallstrom J, Ponten J. 1998. Deletion of chromosome 3pis an early event in malignant progression of cervical cancer. Anticancer Res 18: 707-712.223. Guo Z, Hu X, Afink G, Ponten F, Wilander E, Ponten J. 2000. Comparison ofchromosome 3p deletions between cervical precancers synchronous with and withoutinvasive cancer. Int J Cancer 86: 518-523.224. Rader JS, Gerhard DS, O'Sullivan MJ, Li Y, Li L, Liapis H, Huettner PC.1998. Cervical intraepithelial neoplasia III shows frequent allelic loss in 3p and 6p.Genes Chromosomes Cancer 22: 57-65.225. Sozzi G, Veronese ML, Negrini M, Baffa R, Cotticelli MG, Inoue H, TornielliS, Pilotti S, De Gregorio L, Pastorino U, Pierotti MA, Ohta M, Huebner K, CroceCM. 1996. The FHIT gene 3p14.2 is abnormal in lung cancer. Cell 85: 17-26.226. Hayashi S, Tanimoto K, Hajiro-Nakanishi K, Tsuchiya E, Kurosumi M,Higashi Y, Imai K, Suga K, Nakachi K. 1997. Abnormal FHIT transcripts in human

Page 73: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

72

breast carcinomas: a clinicopathological and epidemiological analysis of 61 Japanesecases. Cancer Res 57: 1981-1985.227. Zou TT, Lei J, Shi YQ, Yin J, Wang S, Souza RF, Kong D, Shimada Y,Smolinski KN, Greenwald BD, Abraham JM, Harpaz N, Meltzer SJ. 1997. FHIT genealterations in esophageal cancer and ulcerative colitis (UC). Oncogene 15: 101-105.228. Wilke CM, Hall BK, Hoge A, Paradee W, Smith DI, Glover TW. 1996.FRA3B extends over a broad region and contains a spontaneous HPV16 integrationsite: direct evidence for the coincidence of viral integration sites and fragile sites.Hum Mol Genet 5: 187-195.229. Muller CY, O'Boyle JD, Fong KM, Wistuba, II, Biesterveld E, Ahmadian M,Miller DS, Gazdar AF, Minna JD. 1998. Abnormalities of fragile histidine triadgenomic and complementary DNAs in cervical cancer: association with humanpapillomavirus type. J Natl Cancer Inst 90: 433-439.230. Strand M, Prolla TA, Liskay RM, Petes TD. 1993. Destabilization of tracts ofsimple repetitive DNA in yeast by mutations affecting DNA mismatch repair. Nature365: 274-276.231. Alani E, Reenan RA, Kolodner RD. 1994. Interaction between mismatchrepair and genetic recombination in Saccharomyces cerevisiae. Genetics 137: 19-39.232. Li GM, Modrich P. 1995. Restoration of mismatch repair to nuclear extractsof H6 colorectal tumor cells by a heterodimer of human MutL homologs. Proc NatlAcad Sci U S A 92: 1950-1954.233. Thibodeau SN, French AJ, Roche PC, Cunningham JM, Tester DJ, LindorNM, Moslein G, Baker SM, Liskay RM, Burgart LJ, Honchel R, Halling KC. 1996.Altered expression of hMSH2 and hMLH1 in tumors with microsatellite instabilityand genetic alterations in mismatch repair genes. Cancer Res 56: 4836-4840.234. Benachenhou N, Guiral S, Gorska-Flipot I, Labuda D, Sinnett D. 1999.Frequent loss of heterozygosity at the DNA mismatch-repair loci hMLH1 andhMSH3 in sporadic breast cancer. Br J Cancer 79: 1012-1017.235. Magnusson PK, Wilander E, Gyllensten U. 1996. Analysis of loss ofheterozygosity in microdissected tumor cells from cervical carcinoma usingfluorescent dUTP labeling of PCR products. Biotechniques 21: 844-847.236. Helland A, Borresen-Dale AL, Peltomaki P, Hektoen M, Kristensen GB,Nesland JM, de la Chapelle A, Lothe RA. 1997. Microsatellite instability in cervicaland endometrial carcinomas. Int J Cancer 70: 499-501.237. Mitra AB, Murty VV, Li RG, Pratap M, Luthra UK, Chaganti RS. 1994.Allelotype analysis of cervical carcinoma. Cancer Res 54: 4481-4487.238. Mullokandov MR, Kholodilov NG, Atkin NB, Burk RD, Johnson AB, KlingerHP. 1996. Genomic alterations in cervical carcinoma: losses of chromosomeheterozygosity and human papilloma virus tumor status. Cancer Res 56: 197-205.239. Huettner PC, Gerhard DS, Li L, Gersell DJ, Dunnigan K, Kamarasova T,Rader JS. 1998. Loss of heterozygosity in clinical stage IB cervical carcinoma:relationship with clinical and histopathologic features. Hum Pathol 29: 364-370.240. Kersemaekers AM, Kenter GG, Hermans J, Fleuren GJ, van de Vijver MJ.1998. Allelic loss and prognosis in carcinoma of the uterine cervix. Int J Cancer 79:411-417.241. Theile M, Seitz S, Arnold W, Jandrig B, Frege R, Schlag PM, Haensch W,Guski H, Winzer KJ, Barrett JC, Scherneck S. 1996. A defined chromosome 6qfragment (at D6S310) harbors a putative tumor suppressor gene for breast cancer.Oncogene 13: 677-685.242. Sandhu AK, Kaur GP, Reddy DE, Rane NS, Athwal RS. 1996. A gene on 6q14-21 restores senescence to immortal ovarian tumor cells. Oncogene 12: 247-252.

Page 74: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

73

243. Ning Y, Weber JL, Killary AM, Ledbetter DH, Smith JR, Pereira-Smith OM.1991. Genetic analysis of indefinite division in human cells: evidence for a cellsenescence-related gene(s) on human chromosome 4. Proc Natl Acad Sci U S A 88:5635-5639.244. Hampton GM, Larson AA, Baergen RN, Sommers RL, Kern S, Cavenee WK.1996. Simultaneous assessment of loss of heterozygosity at multiple microsatelliteloci using semi-automated fluorescence-based detection: subregional mapping ofchromosome 4 in cervical carcinoma. Proc Natl Acad Sci U S A 93: 6704-6709.245. Park SY, Kang YS, Kim BG, Lee SH, Lee ED, Lee KH, Park KB, Lee JH.1995. Loss of heterozygosity on the short arm of chromosome 17 in uterine cervicalcarcinomas. Cancer Genet Cytogenet 79: 74-78.246. Fujita M, Inoue M, Tanizawa O, Iwamoto S, Enomoto T. 1992. Alterations ofthe p53 gene in human primary cervical carcinoma with and without humanpapillomavirus infection. Cancer Res 52: 5323-5328.247. Helland A, Karlsen F, Due EU, Holm R, Kristensen G, Borresen-Dale A.1998. Mutations in the TP53 gene and protein expression of p53, MDM 2 andp21/WAF-1 in primary cervical carcinomas with no or low human papillomavirusload. Br J Cancer 78: 69-72.248. Storey A, Thomas M, Kalita A, Harwood C, Gardiol D, Mantovani F, BreuerJ, Leigh IM, Matlashewski G, Banks L. 1998. Role of a p53 polymorphism in thedevelopment of human papillomavirus- associated cancer. Nature 393: 229-234.249. Thomas M, Kalita A, Labrecque S, Pim D, Banks L, Matlashewski G. 1999.Two polymorphic variants of wild-type p53 differ biochemically and biologically.Mol Cell Biol 19: 1092-1100.250. Summersgill KF, Smith EM, Kirchner HL, Haugen TH, Turek LP. 2000. p53polymorphism, human papillomavirus infection in the oral cavity, and oral cancer.Oral Surg Oral Med Oral Pathol Oral Radiol Endod 90: 334-339.251. Szarka K, Veress G, Juhasz A, Konya J, Sapy T, Soos G, Hernadi Z, GergelyL. 2000. Integration status of virus DNA and p53 codon 72 polymorphism in humanpapillomavirus type 16 positive cervical cancers. Anticancer Res 20: 2161-2167.252. Zehbe I, Voglino G, Wilander E, Genta F, Tommasino M. 1999. Codon 72polymorphism of p53 and its association with cervical cancer. Lancet 354: 218-219.253. Tachezy R, Mikyskova I, Salakova M, Van Ranst M. 1999. Correlationbetween human papillomavirus-associated cervical cancer and p53 codon 72arginine/proline polymorphism. Hum Genet 105: 564-566.254. Kim JW, Lee CG, Park YG, Kim KS, Kim IK, Sohn YW, Min HK, Lee JM,Namkoong SE. 2000. Combined analysis of germline polymorphisms of p53,GSTM1, GSTT1, CYP1A1, and CYP2E1: relation to the incidence rate of cervicalcarcinoma. Cancer 88: 2082-2091.255. Kawaguchi H, Ohno S, Araki K, Miyazaki M, Saeki H, Watanabe M, TanakaS, Sugimachi K. 2000. p53 polymorphism in human papillomavirus-associatedesophageal cancer. Cancer Res 60: 2753-2755.256. Makni H, Franco EL, Kaiano J, Villa LL, Labrecque S, Dudley R, Storey A,Matlashewski G. 2000. P53 polymorphism in codon 72 and risk of humanpapillomavirus-induced cervical cancer: effect of inter-laboratory variation. Int JCancer 87: 528-533.257. Dokianakis DN, Spandidos DA. 2000. P53 codon 72 polymorphism as a riskfactor in the development of HPV- associated cervical cancer. Mol Cell Biol ResCommun 3: 111-114.258. Agorastos T, Lambropoulos AF, Constantinidis TC, Kotsis A, Bontis JN.2000. p53 codon 72 polymorphism and risk of intra-epithelial and invasive cervicalneoplasia in Greek women. Eur J Cancer Prev 9: 113-118.

Page 75: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

74

259. Baek WK, Cho JW, Suh SI, Suh MH, Shin DH, Cho CH, Lee TS, Cha SD.2000. p53 codon 72 polymorphism and risk of cervical carcinoma in Korean women.J Korean Med Sci 15: 65-67.260. Brady CS, Duggan-Keen MF, Davidson JA, Varley JM, Stern PL. 1999.Human papillomavirus type 16 E6 variants in cervical carcinoma: relationship to hostgenetic factors and clinical parameters. J Gen Virol 80: 3233-3240.261. Giannoudis A, Graham DA, Southern SA, Herrington CS. 1999. p53 codon 72ARG/PRO polymorphism is not related to HPV type or lesion grade in low- and high-grade squamous intra-epithelial lesions and invasive squamous carcinoma of thecervix. Int J Cancer 83: 66-69.262. Hamel N, Black MJ, Ghadirian P, Foulkes WD. 2000. No association betweenP53 codon 72 polymorphism and risk of squamous cell carcinoma of the head andneck. Br J Cancer 82: 757-759.263. Hildesheim A, Schiffman M, Brinton LA, Fraumeni JF, Jr., Herrero R, BrattiMC, Schwartz P, Mortel R, Barnes W, Greenberg M, McGowan L, Scott DR, MartinM, Herrera JE, Carrington M. 1998. p53 polymorphism and risk of cervical cancer.Nature 396: 531-532.264. Madeleine MM, Shera K, Schwartz SM, Daling JR, Galloway DA, Wipf GC,Carter JJ, McKnight B, McDougall JK. 2000. The p53 Arg72Pro polymorphism,human papillomavirus, and invasive squamous cell cervical cancer. Cancer EpidemiolBiomarkers Prev 9: 225-227.265. Minaguchi T, Kanamori Y, Matsushima M, Yoshikawa H, Taketani Y,Nakamura Y. 1998. No evidence of correlation between polymorphism at codon 72 ofp53 and risk of cervical cancer in Japanese patients with human papillomavirus 16/18infection. Cancer Res 58: 4585-4586.266. Ngan HY, Liu VW, Liu SS. 1999. Risk of cervical cancer is not increased inChinese carrying homozygous arginine at codon 72 of p53. Br J Cancer 80: 1828-1829.267. Tenti P, Vesentini N, Rondo Spaudo M, Zappatore R, Migliora P, Carnevali L,Ranzani GN. 2000. p53 codon 72 polymorphism does not affect the risk of cervicalcancer in patients from northern Italy. Cancer Epidemiol Biomarkers Prev 9: 435-438.268. Wang NM, Tsai CH, Yeh KT, Chen SJ, Chang JG. 1999. P53 codon 72Argpolymorphism is not a risk factor for carcinogenesis in the chinese. Int J Mol Med 4:249-252.269. Wong YF, Chung TK, Cheung TH, Nobori T, Hampton GM, Wang VW, LiYF, Chang AM. 2000. p53 polymorphism and human papillomavirus infection inHong Kong women with cervical cancer. Gynecol Obstet Invest 50: 60-63.270. Wank R, Thomssen C. 1991. High risk of squamous cell carcinoma of thecervix for women with HLA- DQw3. Nature 352: 723-725.271. Ferrera A, Olivo A, Alaez C, Melchers WJ, Gorodezky C. 1999. HLA DOA1and DOB1 loci in Honduran women with cervical dysplasia and invasive cervicalcarcinoma and their relationship to human papillomavirus infection. Hum Biol 71:367-379.272. Odunsi K, Terry G, Ho L, Bell J, Cuzick J, Ganesan TS. 1995. Associationbetween HLA DQB1 * 03 and cervical intra-epithelial neoplasia. Mol Med 1: 161-171.273. Tabrizi SN, Fairley CK, Chen S, Borg AJ, Baghurst P, Quinn MA, GarlandSM. 1999. Epidemiological characteristics of women with high grade CIN who doand do not have human papillomavirus. Br J Obstet Gynaecol 106: 252-257.274. Cuzick J, Terry G, Ho L, Monaghan J, Lopes A, Clarkson P, Duncan I. 2000.Association between high-risk HPV types, HLA DRB1* and DQB1* alleles andcervical cancer in British women. Br J Cancer 82: 1348-1352.

Page 76: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

75

275. Allen M, Kalantari M, Ylitalo N, Pettersson B, Hagmar B, Scheibenpflug L,Johansson B, Petterson U, Gyllensten U. 1996. HLA DQ-DR haplotype andsusceptibility to cervical carcinoma: indications of increased risk for development ofcervical carcinoma in individuals infected with HPV 18. Tissue Antigens 48: 32-37.276. Glew SS, Duggan-Keen M, Cabrera T, Stern PL. 1992. HLA class II antigenexpression in human papillomavirus-associated cervical cancer. Cancer Res 52: 4009-4016.277. Glew SS, Duggan-Keen M, Ghosh AK, Ivinson A, Sinnott P, Davidson J,Dyer PA, Stern PL. 1993. Lack of association of HLA polymorphisms with humanpapillomavirus- related cervical cancer. Hum Immunol 37: 157-164.278. Chen S, Tabrizi SN, O'Sullivan H, Fairley CK, Quinn MA, Garland SM. 1999.Lack of association between HLA DQB1 alleles with HPV infection and histologyfindings in cervical cancer in Australian women. Int J Gynecol Cancer 9: 220-224.279. Walboomers JM, Meijer CJ, Steenbergen RD, van Duin M, Helmerhorst TJ,Snijders PJ. 2000. [Human papillomavirus and the development of cervical cancer:concept of carcinogenesis]. Ned Tijdschr Geneeskd 144: 1671-1674.280. Zehbe I, Sallstrom JF, Evander M, Edlund K, Rylander E, Wadell G, WilanderE. 1996. Nonradioisotopic detection and typing of human papillomaviruses by use ofpolymerase chain reaction and single-strand conformation polymorphism. Diagn MolPathol 5: 206-213.281. Lazo PA. 1999. The molecular genetics of cervical carcinoma. Br J Cancer 80:2008-2018.282. Kolodner RD, Hall NR, Lipford J, Kane MF, Morrison PT, Finan PJ, Burn J,Chapman P, Earabino C, Merchant E. 1995. Structure of the human MLH1 locus andanalysis of a large hereditary nonpolyposis colorectal carcinoma kindred for mlh1mutations. Cancer Res 55: 242-248.283. Nystrom-Lahti M, Parsons R, Sistonen P, Pylkkanen L, Aaltonen LA, LeachFS, Hamilton SR, Watson P, Bronson E, Fusaro R. 1994. Mismatch repair genes onchromosomes 2p and 3p account for a major share of hereditary nonpolyposiscolorectal cancer families evaluable by linkage. Am J Hum Genet 55: 659-665.284. Bronner CE, Baker SM, Morrison PT, Warren G, Smith LG, Lescoe MK,Kane M, Earabino C, Lipford J, Lindblom A. 1994. Mutation in the DNA mismatchrepair gene homologue hMLH1 is associated with hereditary non-polyposis coloncancer. Nature 368: 258-261.285. Han HJ, Maruyama M, Baba S, Park JG, Nakamura Y. 1995. Genomicstructure of human mismatch repair gene, hMLH1, and its mutation analysis inpatients with hereditary non-polyposis colorectal cancer (HNPCC). Hum Mol Genet4: 237-242.286. Bergthorsson JT, Egilsson V, Gudmundsson J, Arason A, Ingvarsson S. 1995.Identification of a breast tumor with microsatellite instability in a potential carrier ofthe hereditary non-polyposis colon cancer trait. Clin Genet 47: 305-310.287. Boyer JC, Umar A, Risinger JI, Lipford JR, Kane M, Yin S, Barrett JC,Kolodner RD, Kunkel TA. 1995. Microsatellite instability, mismatch repairdeficiency, and genetic defects in human cancer cell lines. Cancer Res 55: 6063-6070.288. Song S, Pitot HC, Lambert PF. 1999. The human papillomavirus type 16 E6gene alone is sufficient to induce carcinomas in transgenic animals. J Virol 73: 5887-5893.289. Matsukura T, Sugase M. 1995. Identification of genital humanpapillomaviruses in cervical biopsy specimens: segregation of specific virus types inspecific clinicopathologic lesions. Int J Cancer 61: 13-22.290. Ylitalo N, Sorensen P, Josefsson AM, Magnusson PK, Andersen PK, PontenJ, Adami HO, Gyllensten UB, Melbye M. 2000. Consistent high viral load of human

Page 77: Molecular Pathogenesis of Cervical Carcinoma160985/FULLTEXT01.pdfPathology presented at Uppsala University in 2001 Abstract Hu, X. 2001. Molecular Pathogenesis of Cervical Carcinoma:

76

papillomavirus 16 and risk of cervical carcinoma in situ: a nested case-control study.Lancet 355: 2194-2198.291. Chu TY, Shen CY, Lee HS, Liu HS. 1999. Monoclonality and surface lesion-specific microsatellite alterations in premalignant and malignant neoplasia of uterinecervix: a local field effect of genomic instability and clonal evolution. GenesChromosomes Cancer 24: 127-134.