Vous êtes sur la page 1sur 9

Annals of Oncology Advance Access published January 19, 2014

review

Annals of Oncology 00: 19 , 2014


doi:10.1093/annonc/mdt538

Cervical cancer screening: on the way to a shift from


cytology to full molecular screening
M. G. Dijkstra1,2, P. J. F. Snijders1, M. Arbyn3, D. C. Rijkaart1, J. Berkhof4 & C. J. L. M. Meijer1*
1
Department of Pathology, VU University Medical Center, Amsterdam; 2Department of Obstetrics and Gynaecology, St. Lukas Andreas Ziekenhuis, Amsterdam, the
Netherlands; 3Unit of Cancer Epidemiology, Scientic Institute of Public Health, Brussels, Belgium; 4Department of Clinical Epidemiology and Biostatistics, VU University
Medical Center, Amsterdam, the Netherlands

Received 10 June 2013; revised 10 September 2013; accepted 29 October 2013

introduction
Currently, cervical cancer is the fourth leading cause of cancer
death in women worldwide, causing more than 275 000 deaths
annually. The disease has a very uneven global distribution; over
85% of cases are found in low-resource countries, with incidence
and death rates being the highest in sub-Saharan Africa, Central
America, South-Central Asia, and Melanesia [1, 2]. This imbalance in disease burden can be explained by differences in background risk [exposure to human papillomavirus (hrHPV)
infection], and the fact that cervical cancer is preventable by an
effective screening and intervention system. Therefore, the
lowest incidence and mortality rates are recorded in countries
where screening is available to women. The impact of population-based screening is reected in a substantial reduction in the
incidence of cervical cancer over the past 50 years in countries
with established cytology-based screening programs [36].
Especially, quality-assured population-based programs have
shown to be very effective [6]. However, cytology-based cervical
screening also has some limitations. The major problem is the
low sensitivity of a single smear to detect high-grade precursor
lesions (50%70%), which require frequent testing [7]. In addition, cytology has low reproducibility, leading to variable
*Correspondence to: Prof. Chris J. L. M. Meijer, Department of Pathology, VU University
Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands. Tel: +31-20-4444070; Fax: +31-20-444-2964; E-mail: cjlm.meijer@vumc.nl

accuracy [8, 9]. Moreover, by repeating cytology, the number of


false positives increases substantially over time [10]. Finally, the
decrease in the incidence of cervical cancer induced by
cytology-based screening is mainly restricted to squamous cell
carcinoma, whereas no change is observed in the incidence of
cervical adenocarcinoma [11, 12], suggesting that cytology fails
to detect adenocarcinomas and its precursors. Consequently,
there is a need for a better primary screening test, and thus a
new screening algorithm.

human papillomavirus testing in the


prevention of cervical cancer
cross-sectional sensitivity
Infection with hrHPV is a necessary event in the multistep
process of cervical carcinogenesis [13]. Thirteen hrHPV types
have been identied [14], of which HPV16 and HPV18 are the
most important types, causing 70% of squamous cell carcinomas, and >90% of adenocarcinomas [15, 16]. Recently, two
prophylactic vaccines against HPV16 and HPV18 (Cervarix
GSK and Gardasil Merck) have shown good protection against
vaccine type-related cervical intraepithelial neoplasia grade 2 or
worse (CIN2+) precursor lesions. However, cervical screening is
still required, because the current vaccines do not protect
against all carcinogenic HPV types. In addition, despite the high
vaccine uptake among women in countries with a school-based

The Author 2014. Published by Oxford University Press on behalf of the European Society for Medical Oncology.
All rights reserved. For permissions, please email: journals.permissions@oup.com.

review

Downloaded from http://annonc.oxfordjournals.org/ by guest on February 4, 2014

Cytology-based nation-wide cervical screening has led to a substantial reduction of the incidence of cervical cancer in
western countries. However, the sensitivity of cytology for the detection of high-grade precursor lesions or cervical cancer
is limited; therefore, repeated testing is necessary to achieve program effectiveness. Additionally, adenocarcinomas and
its precursors are often missed by cytology. Consequently, there is a need for a better screening test. The insight that infection with high-risk human papillomavirus (hrHPV) is the causal agent of cervical cancer and its precursors has led to
the development of molecular tests for the detection of hrHPV. Strong evidence now supports the use of hrHPV testing in
the prevention of cervical cancer. In this review, we will discuss the arguments in favor of, and concerns on aspects of implementation of hrHPV testing in primary cervical cancer screening, such as the age to start hrHPV-based screening,
ways to increase screening attendance, requirements for candidate hrHPV tests to be used, and triage algorithms for
screen-positive women.
Key words: hrHPV DNA, cervical cancer screening, triage, self-sampling

review

Annals of Oncology

the rst round, has a lower specicity for CIN3+ than usually
expected [31]. Possible reasons for these discrepancies could,
rst, be attributed to overdiagnosis of lesions, as LBC was just
introduced at that time and secondly, to incomplete follow-up
of hrHPV-positive women with normal cytology, resulting in
lower detection rates of high-grade CIN in the hrHPV arm at
baseline [31, 32].

long-term protection
At present, four randomized trials conducted in Europe have
published longitudinal data on CIN3+ diagnosed at subsequent screening rounds, which took place in 35 years
(Figure 2) [2426, 28, 33]. All trials reported an 50% lower
CIN3+ detection rate in the second screening round among
women who were hrHPV negative at baseline than those who
had normal cytology. The pooled detection rate ratio of CIN3
+, of hrHPV testing versus cytology, was 0.42 [95% condence
interval (CI): 0.320.55]. The consistency of results was
reected by the absence of interstudy heterogeneity (P = 0.68).
Thus, a negative hrHPV test provides a better protection

Table 1. Trials comparing cytology and hrHPV testing in cervical cancer screening
Study

Description

Interval

Reference

POBASCAM

HPV (GP5+/6+-PCR) and cytology versus cytology


alone
HPV (HC2) combined with cytology (LBC) versus
cytology (LBC) alone
HPV (GP5+/6+-PCR) and cytology versus cytology
alone
HPV (HC2) alone versus HPV (HC2) and cytology
(LBC) versus cytology alone
HPV (HC2) and cytology versus cytology and HPV
(HC2) (randomized order of collection)
HPV (HC2) and cytology triage versus cytology
alone
HPV (HC2) versus cytology versus visual inspection
with acetic acid (VIA) versus no screening

5 years
3 years

Bulkmans et al. [24, 109]


Rijkaart et al. [33]
Kitchener et al. [25, 110]

35 years (by age)

Naucler et al. [26, 27]

3 years

Ronco et al. [28, 111, 112]

1 year

Mayrand et al. [29]

5 years

Leinonen et al. [113]

Shankaranarayanan et al. [41]

ARTISTIC
SwedeScreen
NTCC
CCCaST
Finnish screening trial
India screening trial

Study or Subgroup

Risk Ratio
M-H, Fixed, 95% Cl Year

Ronco 2006
Naucler 2007
Ronco 2008
Leinonen 2009
Kitchener 2009
Rijkaart 2012

1.24 [0.78, 2.00]


1.28 [0.90, 1.83]
2.06 [1.16, 3.68]
1.49 [0.88, 2.55]
0.94 [0.73, 1.21]
1.17 [0.93, 1.46]

Total (95% Cl)

1.18 [1.03, 1.35]

Risk Ratio
M-H, Fixed, 95% Cl

2006
2007
2008
2009
2009
2012

Total events
Heterogeneity: Chi2 = 7.69, df = 5 (P = 0.17); I 2 = 35%
0.1
1
0.01
Test for overall effect: Z = 2.36 (P = 0.02)
Favours experimental

10
100
Favours control

* Ronco et al. is restricted to women of 35 years or older


Figure 1. Detection rate ratio in screening with hrHPV versus cytology in the rst screening round of randomized trials for outcome CIN3+.

| Dijkstra et al.

Downloaded from http://annonc.oxfordjournals.org/ by guest on February 4, 2014

program [1719], the uptake remains suboptimal in, e.g. the


Netherlands, France, Germany, and the USA [2023].
The causal relationship between infection with hrHPV and
cervical cancer has stimulated the application of hrHPV DNA
testing, which has been proposed, either alone or in combination with cytology, as a means to improve existing cervical
screening programs. In the past 15 years, large randomized
trials designed to evaluate the performance of hrHPV testing
have provided important arguments for the implementation of
this assay as a primary screening tool (Table 1). First, ve of
these trials showed, in cross-sectional studies, that hrHPV
testing is 30% more sensitive in detecting CIN2+, and four of
these studies showed that it is also 20% more sensitive in
detecting CIN3+ (Figure 1). Most of these lesions are HPV16associated. The higher cross-sectional sensitivity was conrmed by two more studies analyzing the baseline data of
screening populations [29, 30]. The study published by
Kitchener et al. [25] showed somewhat different results; rst, it
demonstrated similar cross-sectional sensitivity for hrHPV
testing and liquid-based cytology (LBC) in primary cervical
screening, and secondly, the data suggest that LBC, as used in

review

Annals of Oncology

Study or Subgroup
Naucler 2007
Kitchener 2009
Ronco 2010
Rijkaart 2012

Risk Ratio
M-H, Fixed, 95% CI Year
0.53 [0.29, 0.98]
0.52 [0.28, 0.97]
0.34 [0.15, 0.75]
0.39 [0.27, 0.56]

Risk Ratio
M-H, Fixed, 95% CI

2007
2009
2010
2012

Total (95% CI)


0.42 [0.32, 0.55]
Total events
Heterogeneity: Chi2 = 1.51, df = 3 (P = 0.68); I2 = 0%
0.01
0.1
1
Test for overall effect: Z = 6.37 (P < 0.00001)
Favours experimental

10

100

Favours control

Figure 2. Detection rate ratio of screening with hrHPV versus cytology in the second screening round, for outcome CIN3+, among women who were hrHPV
negative versus cytology negative in the rst screening round.

round by hrHPV testing represent non-regressing, clinically


relevant lesions [33].
Finally, an Indian cluster-randomized trial [42] found that, in
a low-resource setting, a single round of hrHPV screening was
associated with a signicant decline in the rate of advanced cervical cancers (FIGO stage II+), when compared with an unscreened control group [hazard ratio 0.47 (95% CI 0.320.69)].
No signicant reduction was observed in the study arms with
cytology or visual inspection with acetic acid screening.
Collectively, the available evidence indicates that sole hrHPV
testing should replace cytology as a primary screening tool in
cervical screening.

hrHPV detection methods


hrHPV detection methods include both HPV DNA assays and
E6/E7 mRNA assays. The drawback of hrHPV testing is that it
has an (apparently) unavoidable tradeoff between sensitivity and
specicity. Overall, hrHPV testing has a 3%4% lower specicity
than cytology [at cutoff atypical squamous cells of undetermined signicance or worse (ASC-US+)] [40] due to its inability to distinguish between persistent hrHPV infections
associated with ( precursor lesions of ) cervical cancer and transient hrHPV infections. The specicity could even be lowered
>25% when an ultra-sensitive hrHPV test was applied [43].
From a clinical point of view, testing for hrHPV is only useful
when a positive hrHPV test result is informative about the presence or absence of CIN2+ (clinical sensitivity and specicity).
Thus, in order to prevent excessive follow-up procedures for
women with transient hrHPV infections or hrHPV-positive
women without cervical lesions, candidate hrHPV tests to be
used for cervical screening should be clinically validated. Two
hrHPV DNA tests, i.e. HC2 and GP5+/6+, have shown, in large
clinical trials, to perform better in reducing the incidence of
CIN3+ and are thus considered as clinically validated prototype
assays. Guidelines for hrHPV test requirements and clinical validation were developed, based on the available data from large
prospective screening studies [44]. These guidelines can be used
to assess the clinical performance of a candidate HPV test, relative to one of two prototype hrHPV tests with proven good clinical performance (i.e. HC2 or GP5+6+-PCR) by a crosssectional clinical equivalence analysis in a screening setting [45].
In short, the candidate test should have a clinical sensitivity for

doi:10.1093/annonc/mdt538 |

Downloaded from http://annonc.oxfordjournals.org/ by guest on February 4, 2014

against CIN3+, than a negative Pap smear. Several cohort


studies have also shown a consistently lower long-term cumulative incidence rate of CIN2+ among women negative for
hrHPV than those with normal cytology [30, 34, 35]. In addition, in two trials, the detection rate of cervical cancer at the
second screening round was signicantly lower among women,
who were hrHPV negative at baseline [odds ratio (OR): 0.19
(95% CI 0.070.53)] [28, 33]. Most importantly, in a pooled
analysis of four European trials following women for at least
two screening rounds, it was recently conrmed that hrHPVbased screening provides better protection against cervical
cancer than cytology [36]. Consequently, screening intervals
might be extended when primary hrHPV testing has been
introduced. Berkhof et al. [37] have shown that an extension to
6 or 8 years is possible without increasing the lifetime cancer
risk, using a simulation model. However, it makes sense to
extend the screening interval for hrHPV negatives only, as
hrHPV-positive women, even those with normal cytology,
have a non-negligible CIN3+ risk (5.2%) [33, 35]. The risk is
too high to delay follow-up to the next screening round (35
years) [38, 39], but too low to refer these women for immediate
colposcopy [40]. Therefore, hrHPV-positive women with normal cytology at baseline require further triage testing and/or
follow-up, which will be discussed below.
Another important and consistent nding in the ve trials
with combined hrHPV and cytology co-testing was that cotesting has virtually no additional value compared with single
hrHPV screening [40]. Although slightly more sensitive, no
signicant differences for CIN2+ or CIN3+ detection were
found [sensitivity ratio cytology and hrHPV versus hrHPV
alone: 1.06 (95% CI 0.961.18) for CIN2+, and 1.03 (95% CI
0.891.20) for CIN3+] [31]. A recent study from the USA also
showed that hrHPV and cytology co-testing have no advantage
over sole cytology screening [35]. Collectively, these data show
that sole hrHPV testing is sufcient for cervical screening, and
argue against the use of combined hrHPV and cytology cotesting in women aged 3060 years, as recently recommended
by the American Cancer Society (ASC) [41]. Moreover, the data
from the POBASCAM trial, in which hrHPV and cytology cotesting were used in both study arms in the second screening
round, show that the total number of CIN3+ detected over two
rounds is equal in both arms. These results indicate that at least
a part of the surplus of CIN2/3+ lesions detected in the rst

review

management of hrHPV-positive women


cytology and hrHPV genotyping
Epidemiological studies have estimated that HPV16 and HPV18
cause 70% of cervical cancers worldwide, and that the cumulative 10-year risk of CIN3+ in HPV16/18-positive women ranges
from 10% to 20% [60, 61]. These facts have led to several studies
evaluating the value of hrHPV genotyping, with or without cytology, to triage hrHPV-positive women.
In the VUSA-screen study, evaluating 14 triage strategies
[59], cytology triage at baseline and repeat cytology testing at 12
months emerged as the optimal strategy; it showed the highest
positive predictive value [PPV 37.5% (95% CI 32.642.6)] in
combination with a high negative predictive value (NPV) for

| Dijkstra et al.

CIN3+ [99.3% (95% CI 89.199.8)]. A CIN3+ risk of <2% (corresponding with a NPV for CIN3+ of >98%) was considered to
be acceptable for dismissal from further follow-up. This threshold was based on the 5-year CIN3+ risk of women with borderline or mild dyskaryosis (BMD) cytology at baseline, and
normal cytology at 6- and 18-month follow-up (1.2%), which is
presently accepted in the Netherlands [38].
In addition, we have recently carried out a post hoc analysis
of data from the POBASCAM trial [39] to evaluate useful triage
strategies, including HPV16/18 genotyping and cytology. Three
triage strategies met the criteria for both the NPV (98%) and
PPV (20%) that were set to minimize the risk of over investigation and excessively aggressive management; i.e. the NPVs
ranged between 98.1% and 99.6%, while the PPVs ranged
between 25.6% and 34.0%. The eligible triage strategies were:
(i) cytology and HPV16/18 genotyping at baseline without
repeat testing, (ii) cytology at baseline with repeat cytology
testing after 6 months, and (iii) cytology and HPV16/18 genotyping at baseline followed by repeat cytology examination at 6
months [39]. Furthermore, a nested evaluation of the Swedish
SWEDESCREEN study suggested to follow-up hrHPV-positive
women with normal cytology at baseline, by one repeat hrHPV
test [27]. This strategy showed comparable results in terms of
sensitivity for CIN3+ (96.0%), however, with somewhat lower
PPV (22.0%) than obtained with the preferred strategies in the
POBASCAM sub-study. Moreover, implementation of this strategy leads to a substantial increase in the colposcopy referral rate,
and thus possible overtreatment [39, 59].
Finally, Castle et al. [62] evaluated the performance of Cobas
HPV testing (Roche Molecular Systems, USA) as a primary
screening test among women aged 25 years and older. The preferred strategy in this sub-analysis of the ATHENA study was
cytology triage (threshold low-grade squamous intraepithelial
lesion or worse) in combination with the detection of HPV16,
HPV18, or both types, resulting in a sensitivity for CIN3+ of
72.2% (95% CI 66.477.4) and a PPV of 13.9% (95% CI 12.8
15.0). Although the authors recommended re-testing of screen
negatives after 1 year, because the sensitivity for CIN3+ was
<80% and the NPV <98%.
Thus, several triage strategies seem to be feasible, because the
results of the different studies also indicate that it does not
appear that the exact triage protocol has any effect on the
outcome. However, as preferences and the quality of cytology
will vary between countries, policymakers will have to weigh the
pros and cons of the different triage strategies when making a
choice. Especially, the balance between the safety of a triage
strategy (NPV) and the burden of screening on patients and
clinicians (PPV and referral rate) is important [63].

HPV mRNA, p16INK4a IHC, and methylation markers


In future cervical screening, it might be expected that the role of
cytology, as a triage test, will become more limited, as the
hrHPV test result may inuence the subjective reading of cytology. Promising biomarkers involved in cervical carcinogenesis have already emerged. Especially, biomarkers that indicate a
shift from the productive phase of hrHPV infection to the transforming phase are valuable. hrHPV E6/E7 oncogenes are highly
expressed in ( para)basal cells of high-grade CIN and interact

Downloaded from http://annonc.oxfordjournals.org/ by guest on February 4, 2014

CIN2+ not less than 90%, and a clinical specicity not less than
98% of that of the reference assays.
Thus far, three additional hrHPV DNA tests, i.e. Cobas 4800
Roche, Real-Time (RT) PCR Abbott Molecular, and
Papillocheck Bio-Greiner (when only 14 hrHPV types are considered), have fullled the criteria provided in these guidelines,
with sensitivities ranging between 100% and 95.8%, and specicities from 96.7% to 92.3% [4652]. Thus, these assays can be
considered as clinically validated for primary hrHPV-based cervical cancer screening.
In contrast to the HPV DNA tests, the APTIMA HPV Assay
(GenProbe) relies on aggregate detection of mRNA of 14
hrHPV types [53]. Two studies [54, 55] have evaluated the performance of this assay for primary screening compared with the
HC2 test, and the results were summarized in a recent review
[40]; the relative sensitivity ratio (outcome CIN2+) of APTIMA
versus HC2 was 1.02 (95% CI 0.861.20), and the relative specicity ratio was 1.07 (95% CI 1.051.08). Thus, the APTIMA
assay seems to perform with sensitivity closely to that of the
DNA test and with a possible slightly higher specicity. A recent
study comparing the APTIMA HPV Assay with GP5+/6+ PCR
showed similar results, and indicated that also the APTIMA
HPV Assay is clinically non-inferior to GP5+/6+ PCR with
respect to sensitivity and specicity for CIN2+ [56]. Another approach in trying to validate new hrHPV tests was used by
Cuzick et al. [57] in the predictors study: the sensitivity was primarily analyzed in a cytology-based referral population [58],
and specicity in a cytology-based screening population [57].
This study evaluated six HPV tests and established that four
tests (Roche Cobas, GenProbe APTIMA, Abott Real-Time PCR,
and (Becton, Dickinson and Company) HPV assay) achieved
the required sensitivity and specicity compared with HC2.
This would also qualify the BD test as clinically validated.
However, the predictors screening study only included cytologydriven CIN2+ lesion, which made the sensitivity criterion too
soft to consider it in line with the guideline described earlier
[44]. In addition, no details of any previous screening history of
participants were available. The use of validated HPV test assays
alone, however, is not sufcient to reduce the number of falsepositive tests among healthy women; triage testing of hrHPVpositive women is necessary to keep the number of invasive
follow-up examinations, and thus the costs within acceptable
limits [59].

Annals of Oncology

Annals of Oncology

expression. In many cancers, tumor suppressor genes were


found to be inactivated by hypermethylation of their promoter
region. Therefore, detection of hypermethylation of tumor suppressor genes involved in cervical cancer genesis may provide
powerful biomarkers for cancer detection [85, 86], especially as
methylation has been detected already at precancerous stages
[87, 88]. In fact, a recent study of Bierkens et al. showed that
methylation levels of two genes (i.e. CADM1 and MAL)
increased with the grade of underlying CIN and were highest in
carcinomas. Moreover, cervical scrapes of women with CIN 2/3
lesions with long-lasting hrHPV infections (5 years) had
higher methylation levels than those with a shorter duration of
preceding hrHPV infection (<5 years) [89]. These ndings indicate that at least lesions with a longer duration of existence are
detected by such methylation markers, further strengthening
their value for triage testing of hrHPV-positive cervical scrapes.
Recently, a study on scrapes from women participating in
screening was carried out to evaluate the value of an objective
real-time PCR assays that assess the methylation status of the
promoter regions of CADM1 and MAL to triage hrHPV-positive women for CIN3+ [90]. Because of the design of the study,
the results were compared with CIN3+ sensitivity and specicity
of cytology only, and of cytology combined with HPV16/18
genotyping. An optimal threshold resulted in a sensitivity of
84.2%, and a corresponding specicity of 52.5% for the methylation assay; for cytology, these were 65.8% and 78.8%, and for cytology with HPV16/18 genotyping, these were 84.2% and 54.0%,
respectively. Consequently, the authors concluded that, in
hrHPV positive women, this methylation marker panel was at
least equally discriminatory for high-grade CIN as cytology, or
as cytology with the detection of HPV16/18. These results indicate that complete cervical screening by objective, non-morphological molecular methods seems to be feasible. However,
further validation is needed before any of these tests can be considered for screening. Currently, a large randomized, controlled
trial to validate methylation markers to triage hrHPV-positive
women is being carried out in the Netherlands.

primary hrHPV testing: at what age


should we start?
Another important issue is the age at which hrHPV testing
should be offered for primary screening. Most experts agree
that, in women younger than 25 years of age, hrHPV testing is
not recommended, because the prevalence is very high, and at
this age, hrHPV infections are commonly transient.
Ronco et al. [28] reported that hrHPV testing in women aged
2534 years could lead to substantial overdiagnosis of regressive
CIN2+ lesions, particularly when hrHPV positives in this age
group are directly referred for colposcopy, without further triage
testing. The results from the POBASCAM trial [33] showed that
hrHPV screening does not have to be postponed until age 36
years or older, but can be started at age 30 years. The long-term
follow-up results of this study showed that the cumulative detection of CIN2+ and CIN3+ lesions over two screening rounds
did not differ between women aged 2933 years, and women of
34 years of age and older [33]. Thus, hrHPV screening does not
result in excessive diagnosis of lesions destined to regress, even

doi:10.1093/annonc/mdt538 |

Downloaded from http://annonc.oxfordjournals.org/ by guest on February 4, 2014

with p53 and pRB, respectively; in this way, they interfere with
cell cycle control [64]. As a consequence, uncontrolled proliferation and chromosomal instability occur, resulting in additional
(epi) genetic changes [65, 66]. Therefore, detection of elevated
E6/E7 mRNA levels in cervical smears has been suggested to be
an attractive biomarker [67].
Two commercial hrHPV mRNA assays can be used for triage
testing, i.e. the PreTect HPV-Proofer (NorChip) and the
NucliSENS Easy Q HPV (bioMrieux). Both are based on the
same technology and are marketed under different brand names
in different countries. These assays detect HPV E6/E7 mRNA
from the ve most prevalent hrHPV types in cervical cancer
(HPV16, 18, 31, 33, and 45) [68]. A recent study [69] showed
that the HPV-Proofer assay is particularly of value to triage
hrHPV DNA-positive women with normal cytology, given their
markedly increased risk of CIN2+ in case of a positive mRNA
result [55% (95% CI 34%76%]. However, this study also
revealed that hrHPV-positive cytological normal women who
are mRNA test negative would still need follow-up, as their
CIN2+ risk was 20% (95% CI 7%33%). Thus, as a primary
screening test, the clinical applicability of these HPV mRNA
tests is insufcient
An alternative is the detection of cellular host genes that are
specically up-regulated and overexpressed, or silenced in cells
that have undergone the shift into the transforming phase of
hrHPV infection [70]. Cyclin-dependent kinase inhibitor
p16INK4a is up-regulated as a consequence of hrHPV E7 expression in proliferating cells [71, 72], and several studies have
shown that p16INK4a-based cytology, either alone or in combination with hrHPV testing, can detect underlying CIN2+ with
high sensitivity [7378]. However, the results for p16INK4a were
less favorable in the predictors studies [58, 79], comparing the
sensitivity and specicity of several tests for the detection of
high-grade CIN in a cytology-based referral population. In addition, even after p16INK4a immunostaining, still morphological
interpretation necessary to differentiate hrHPV-transformed
cells from endometrial cells with non-hrHPV induced p16INK4a
expression [80]. To overcome this limitation, a double staining
kit for p16INK4a and Ki-67 has been developed that allows simultaneous detection of p16INK4a and nuclear Ki-67 expression in
dividing cervical cells (CINtec Plus, MTM laboratories). The
potential of this double staining kit to identify women at risk for
underlying high-grade disease was shown in women with abnormal cytology [81, 82], as well as in hrHPV positives with normal
cytology, when used as a reex test [83]. At present, these good
results will need to be conrmed in prospective studies.
Furthermore, a recently published retrospective study evaluating
different triage strategies in a cytology-based referral population
suggested that further testing for p16INK4a and HPV16 genotyping may also be an important strategy to determine which
women are in need of referral. They advocate this strategy, especially for hrHPV screen positives with normal cytology [84].
Currently, these strategies should be evaluated in prospective
studies.
Although, a hrHPV infection can induce immortalization
and trigger chromosomal instability, additional changes in
oncogene expression and loss of function of tumor suppressor
genes are necessary, to obtain a full blown invasive cancer cell.
Methylation of CpG islands is an epigenetic modier of gene

review

review

Annals of Oncology

in women in the 3034 years of age category. These ndings indicate that primary hrHPV screening should be recommended
for women aged 3060 years. Additional follow-up, after the age
of 60, is indicated for women with a positive hrHPV test at the
last screening round. The residual risk of CIN3+ is too high to
dismiss them from follow-up, even when triage testing in
hrHPV-positive women is negative [91]. These women can only
be discharged from further hrHPV testing after they have
cleared the virus. Future studies should investigate whether
methylation markers might be used as a more specic triage test
for hrHPV-positive women in the younger age range (below 30
years) to detect clinically relevant lesions, as high methylation
levels seem to be related to the degree and duration of underlying CIN and highest levels are found in carcinomas [89].

increasing the screening coverage

Table 2. Systematic reviews and meta-analyses of the literature


comparing self-collected and physician-taken samples for the
detection of hrHPV DNA
Reference

No. of studies
included

Kappa value for agreement


(range)

Ogilvie et al. [103]


Petignat et al. [95]
Schmeink et al. [114]

12 studies
18 studies
19 studies

0.451.00
0.500.82
0.450.81

| Dijkstra et al.

conclusion
All the evidence collected so far suggest that the time has come
for the implementation of hrHPV testing as a primary screening
test, as it provides a superior protection against cervical ( pre)cancerous lesions compared with cytology. hrHPV testing
detects 30% more CIN2+, and 20% more CIN3+ lesions in
women over 30 years of age. Although any hrHPV test used
should be clinically validated, hrHPV-based primary screening
should be implemented preferably within a population-based
screening program with a call and recall system. The somewhat
lower specicity, which may cause excess false-positive tests
among healthy women, can be overcome by triage of hrHPVpositive women. At this time, triaging these women by cytology
at baseline and repeat cytology testing after 6 months, possibly
in combination with baseline HPV16/18 genotyping, seems to
be very suitable for this purpose. Although, as preferences and
the quality of cytology will vary between countries, policymakers will have to weigh the pros and cons of the different
triage strategies when making a choice. In future screening,
however, it is likely that the role of cytology becomes more
limited and validated (molecular) biomarkers gain attention;
among these, p16INK4a/Ki-67 double staining and host genome
or viral DNA methylation markers appear to be promising.

Downloaded from http://annonc.oxfordjournals.org/ by guest on February 4, 2014

One other problem concerning the effectiveness of current cervical screening programs remains nonattendance [92, 93].
Especially nonparticipating women are at increased risk of
cancer [3, 94], as half of the cervical carcinomas are found in
non-attending women. Therefore, targeting non-attendees is
important in achieving optimal protection from screening programs.
Self-sampling is a less costly and less invasive collection
method [95], and several studies have shown that non-attendees
actually do take part in self-sampling studies [92, 9699]. Thus,
there is a basis for self-sampling in cervical cancer screening. In
addition, self-collection makes cervical screening accessible to
women in medium- and low-income countries [100102]. That
is why, in recent years, several studies have focused on the use of
self-collected samples for hrHPV testing.
In most studies, a moderate-to-good agreement between
hrHPV test positivity in self-collected and physician-taken
samples was found. The data have been summarized in (systematic) reviews and meta-analyses [95, 103], in which study parameters included concordance in hrHPV detection rates between
self- and physician-collected samples through kappa values,
which varied between 0.45 and 1.00 (Table 2). However, the
hrHPV test positivity rates varied across the studies. Most likely
due to differences in study populations (age range and country
of origin), the use of different collection devices for self- and
physician-sampling, as well as differences in hrHPV tests and
protocols. For example, in studies using the HC2 method, a
higher hrHPV detection rate in self- compared with physiciancollected samples was observed [97, 100, 104, 105]. The HC2
assay, however, is known to show some cross-reactivity to

low-risk HPV types [106], and these types tend to more commonly affect vaginal than cervical mucosa. In other studies, a
higher hrHPV detection rate was found in the physiciancollected cervical samples [107, 108]. Yet, overall the data show
that self-sampling is concordant with physician-sampling in
detecting hrHPV DNA.
However, before its use in cervical cancer screening, it is most
important to know how self-sampling performs with regard to
relevant disease outcomes. In other words, hrHPV self-sampling
procedures should be clinically validated in terms of sensitivity
and specicity to detect CIN2+ lesions. Various cross-sectional
studies have been carried out to compare the value of hrHPV
testing on self- with physician-collected samples to detect CIN2+,
and the results have recently been summarized by Snijders et al.
[99]. This review showed that hrHPV testing on self-sampled
specimen is as sensitive for CIN2+ in several studies as hrHPV
detection on physician-taken cervical samples, although sometimes less specic. However, the authors also reported that both
the type of self-sampling device and the type of hrHPV test
seem to inuence the clinical performance of hrHPV testing on
self-samples. Thus, for reaching clinical equivalence, in terms of
detecting high-grade CIN, between self- and physician-sampling, a certied combination of self-sampling device and validated hrHPV test is important. Overall, the data show that selfsampling for primary hrHPV testing offers possibilities to increase screening coverage by reaching non-responders, and in
future, might even be offered as a safe alternative screening
method to regular attendees.
Additionally, hrHPV-oriented cervical screening programs
will increase public awareness of the link between hrHPV and
cervical cancer, which may, in turn, lead to a higher uptake of
the prophylactic HPV vaccines and consequently a further reduction in cervical cancer incidence.

Annals of Oncology

As for the age to commence screening, we advocate the introduction of primary hrHPV testing in women from the age of 30,
to ensure that mostly clinically relevant, non-regressing highgrade lesions are detected. Furthermore, since a negative hrHPV
test offers a better protection (50%) against CIN3+ than normal
cytology, hrHPV testing can be implemented together with an
extension of the screening interval for hrHPV screen negative
women. In addition, to achieve optimal protection and screening coverage, self-sampling might be offered to non-attendees,
and in future possibly as an alternative method to regular attendees, under the condition that a validated combination of selfsampling device and hrHPV assay is used.
Collectively, these data show that the time has come to implement primary hrHPV testing in population-based cervical
screening, thereby offering women maximum protection against
high-grade CIN with less uncertainty and fewer screening
rounds.

PJFS declares that he has received speakers fee from Roche,


Abbott, GenProbe, and Qiagen. He is also shareholder of Selfscreen B.V. JB declares that he has received speakers fee from
Qiagen and a consultancy fee from GSK. CJLMM declares that
he has received speakers fee from Qiagen, Roche, and Merck.
He has received consultancy fees from Qiagen until 2010, and
he is minority shareholder of Self-screen B.V. All remaining
authors have declared no conicts of interest.

references
1. IARC. WHO/ICO information centre on HPV and cervical cancer (HPV Information
Centre). Human papillomavirus and related cancers in World. 2010. Ref Type:
Report.
2. Arbyn M, Castellsague X, de SS et al. Worldwide burden of cervical cancer in
2008. Ann Oncol 2011; 22: 26752686.
3. Peto J, Gilham C, Fletcher O et al. The cervical cancer epidemic that screening
has prevented in the UK. Lancet 2004; 364: 249256.
4. de KI, van der Aa MA, van BM et al. Trends in cervical cancer in the Netherlands
until 2007: has the bottom been reached? Int J Cancer 2011; 128:
21742181.
5. Gustafsson L, Ponten J, Zack M et al. International incidence rates of invasive
cervical cancer after introduction of cytological screening. Cancer Causes Control
1997; 8: 755763.
6. Arbyn M, Raifu AO, Weiderpass E et al. Trends of cervical cancer mortality in the
member states of the European Union. Eur J Cancer 2009; 45: 26402648.
7. Cuzick J, Clavel C, Petry KU et al. Overview of the European and North American
studies on HPV testing in primary cervical cancer screening. Int J Cancer 2006;
119: 10951101.
8. Nanda K, McCrory DC, Myers ER et al. Accuracy of the Papanicolaou test in
screening for and follow-up of cervical cytologic abnormalities: a systematic
review. Ann Intern Med 2000; 132: 810819.
9. Fahey MT, Irwig L, Macaskill P. Meta-analysis of Pap test accuracy. Am J
Epidemiol 1995; 141: 680689.
10. Katki HA, Wentzensen N. How might HPV testing be integrated into cervical
screening? Lancet Oncol 2012; 13: 810.
11. Bulk S, Visser O, Rozendaal L et al. Cervical cancer in the Netherlands 1989
1998: decrease of squamous cell carcinoma in older women, increase of
adenocarcinoma in younger women. Int J Cancer 2005; 113: 10051009.
12. Anttila A, Pukkala E, Soderman B et al. Effect of organised screening on cervical
cancer incidence and mortality in Finland, 19631995: recent increase in
cervical cancer incidence. Int J Cancer 1999; 83: 5965.

13. Walboomers JM, Jacobs MV, Manos MM et al. Human papillomavirus is a


necessary cause of invasive cervical cancer worldwide. J Pathol 1999; 189:
1219.
14. Bouvard V, Baan R, Straif K et al. A review of human carcinogenspart B:
biological agents. Lancet Oncol 2009; 10: 321322.
15. Munoz N, Bosch FX, Castellsague X et al. Against which human papillomavirus
types shall we vaccinate and screen? The international perspective. Int J Cancer
2004; 111: 278285.
16. Castellsague X, Diaz M, de Sanjose S et al. Worldwide human papillomavirus
etiology of cervical adenocarcinoma and its cofactors: implications for screening
and prevention. J Natl Cancer Inst 2006; 98: 303315.
17. Franceschi S. Monitoring HPV16/18 immunisation in England and elsewhere. Br
J Cancer 2010; 103: 157158.
18. Garland SM, Skinner SR, Brotherton JM. Adolescent and young adult HPV
vaccination in Australia: achievements and challenges. Prev Med 2011; 53(Suppl
1): S29S35.
19. Shearer BD. HPV vaccination: understanding the impact on HPV disease. Purple
Papers 2011; 34: 118.
20. van Lier EA. Immunization coverage National Immunization programme in The
Netherlands. RIVM report 210021014/2011. 1-1-2011. Ref Type: Report.
21. Bartlett JA, Peterson JA. The uptake of human papillomavirus (HPV) vaccine
among adolescent females in the United States: a review of the literature. J Sch
Nurs 2011; 27: 434446.
22. Arbyn M, de SS, Saraiya M et al. EUROGIN 2011 roadmap on prevention and
treatment of HPV-related disease. Int J Cancer 2012; 131: 19691982.
23. Etter DJ, Zimet GD, Rickert VI. Human papillomavirus vaccine in adolescent
women: a 2012 update. Curr Opin Obstet Gynecol 2012; 24: 305310.
24. Bulkmans NW, Berkhof J, Rozendaal L et al. Human papillomavirus DNA testing
for the detection of cervical intraepithelial neoplasia grade 3 and cancer: 5-year
follow-up of a randomised controlled implementation trial. Lancet 2007; 370:
17641772.
25. Kitchener HC, Almonte M, Thomson C et al. HPV testing in combination with
liquid-based cytology in primary cervical screening (ARTISTIC): a randomised
controlled trial. Lancet Oncol 2009; 10: 672682.
26. Naucler P, Ryd W, Tornberg S et al. Human papillomavirus and Papanicolaou
tests to screen for cervical cancer. N Engl J Med 2007; 357: 15891597.
27. Naucler P, Ryd W, Tornberg S et al. Efcacy of HPV DNA testing with cytology
triage and/or repeat HPV DNA testing in primary cervical cancer screening. J Natl
Cancer Inst 2009; 101: 8899.
28. Ronco G, Giorgi-Rossi P, Carozzi F et al. Efcacy of human papillomavirus testing
for the detection of invasive cervical cancers and cervical intraepithelial
neoplasia: a randomised controlled trial. Lancet Oncol 2010; 11: 249257.
29. Mayrand MH, Duarte-Franco E, Rodrigues I et al. Human papillomavirus DNA
versus Papanicolaou screening tests for cervical cancer. N Engl J Med 2007;
357: 15791588.
30. Cuzick J, Szarewski A, Mesher D et al. Long-term follow-up of cervical
abnormalities among women screened by HPV testing and cytologyresults
from the Hammersmith study. Int J Cancer 2008; 122: 22942300.
31. Arbyn M, Ronco G, Meijer CJ et al. Trials comparing cytology with human
papillomavirus screening. Lancet Oncol 2009; 10: 935936.
32. Sasieni P, Castle PE, Cuzick J. Further analysis of the ARTISTIC trial. Lancet
Oncol 2009; 10: 841842.
33. Rijkaart DC, Berkhof J, Rozendaal L et al. Human papillomavirus testing for the
detection of high-grade cervical intraepithelial neoplasia and cancer: nal results
of the POBASCAM randomised controlled trial. Lancet Oncol 2012; 13: 7888.
34. Dillner J, Rebolj M, Birembaut P et al. Long term predictive values of cytology and
human papillomavirus testing in cervical cancer screening: joint European cohort
study. BMJ 2008; 337: a1754.
35. Katki HA, Kinney WK, Fetterman B et al. Cervical cancer risk for women
undergoing concurrent testing for human papillomavirus and cervical cytology: a
population-based study in routine clinical practice. Lancet Oncol 2011; 12:
663672.
36. Ronco G, Dillner J, Elfstrm KM et al. Efcacy of HPV-based screening for
prevention of invasive cervical cancer: follow-up of four European randomised
controlled trials. The International HPV screening working group. Lancet. 2013

doi:10.1093/annonc/mdt538 |

Downloaded from http://annonc.oxfordjournals.org/ by guest on February 4, 2014

disclosure

review

review
37.
38.

39.

40.

41.

42.

44.

45.
46.

47.
48.

49.

50.

51.
52.

53.

54.

55.

56.

57.

| Dijkstra et al.

58. Szarewski A, Mesher D, Cadman L et al. Comparison of seven tests for highgrade cervical intraepithelial neoplasia in women with abnormal smears: the
Predictors 2 study. J Clin Microbiol 2012; 50: 18671873.
59. Rijkaart DC, Berkhof J, van Kemenade FJ et al. Evaluation of 14 triage strategies
for HPV DNA-positive women in population-based cervical screening. Int J
Cancer 2012; 130: 602610.
60. Clifford GM, Smith JS, Plummer M et al. Human papillomavirus types in invasive
cervical cancer worldwide: a meta-analysis. Br J Cancer 2003; 88: 6373.
61. Khan MJ, Castle PE, Lorincz AT et al. The elevated 10-year risk of cervical
precancer and cancer in women with human papillomavirus (HPV) type 16 or 18
and the possible utility of type-specic HPV testing in clinical practice. J Natl
Cancer Inst 2005; 97: 10721079.
62. Castle PE, Stoler MH, Wright TC, Jr et al. Performance of carcinogenic human
papillomavirus (HPV) testing and HPV16 or HPV18 genotyping for cervical cancer
screening of women aged 25 years and older: a subanalysis of the ATHENA
study. Lancet Oncol 2011; 12: 880890.
63. Meijer CJ, Berkhof J. Screening: cervical cancershould we abandon cytology
for screening? Nat Rev Clin Oncol 2012; 9: 558559.
64. von Knebel DM. New markers for cervical dysplasia to visualise the genomic
chaos created by aberrant oncogenic papillomavirus infections. Eur J Cancer
2002; 38: 22292242.
65. Duensing S, Duensing A, Flores ER et al. Centrosome abnormalities and genomic
instability by episomal expression of human papillomavirus type 16 in raft
cultures of human keratinocytes. J Virol 2001; 75: 77127716.
66. Duensing S, Munger K. The human papillomavirus type 16 E6 and E7
oncoproteins independently induce numerical and structural chromosome
instability. Cancer Res 2002; 62: 70757082.
67. Sotlar K, Stubner A, Diemer D et al. Detection of high-risk human papillomavirus
E6 and E7 oncogene transcripts in cervical scrapes by nested RT-polymerase
chain reaction. J Med Virol 2004; 74: 107116.
68. Molden T, Kraus I, Skomedal H et al. PreTect HPV-Proofer: real-time detection
and typing of E6/E7 mRNA from carcinogenic human papillomaviruses. J Virol
Methods 2007; 142: 204212.
69. Rijkaart DC, Heideman DA, Coupe VM et al. High-risk human papillomavirus
(hrHPV) E6/E7 mRNA testing by PreTect HPV-Proofer for detection of
cervical high-grade intraepithelial neoplasia and cancer among hrHPV DNApositive women with normal cytology. J Clin Microbiol 2012; 50:
23902396.
70. Klaes R, Friedrich T, Spitkovsky D et al. Overexpression of p16(INK4A) as a
specic marker for dysplastic and neoplastic epithelial cells of the cervix uteri. Int
J Cancer 2001; 92: 276284.
71. Khleif SN, DeGregori J, Yee CL et al. Inhibition of cyclin D-CDK4/CDK6 activity is
associated with an E2F-mediated induction of cyclin kinase inhibitor activity. Proc
Natl Acad Sci USA 1996; 93: 43504354.
72. McLaughlin-Drubin ME, Crum CP, Munger K. Human papillomavirus E7
oncoprotein induces KDM6A and KDM6B histone demethylase expression and
causes epigenetic reprogramming. Proc Natl Acad Sci USA 2011; 108:
21302135.
73. Guo M, Hu L, Baliga M et al. The predictive value of p16(INK4a) and hybrid
capture 2 human papillomavirus testing for high-grade cervical intraepithelial
neoplasia. Am J Clin Pathol 2004; 122: 894901.
74. Nieh S, Chen SF, Chu TY et al. Is p16(INK4A) expression more useful than human
papillomavirus test to determine the outcome of atypical squamous cells of
undetermined signicance-categorized Pap smear? A comparative analysis using
abnormal cervical smears with follow-up biopsies. Gynecol Oncol 2005; 97:
3540.
75. Yoshida T, Fukuda T, Sano T et al. Usefulness of liquid-based cytology specimens
for the immunocytochemical study of p16 expression and human papillomavirus
testing: a comparative study using simultaneously sampled histology materials.
Cancer 2004; 102: 100108.
76. Carozzi F, Confortini M, Dalla PP et al. Use of p16-INK4A overexpression to
increase the specicity of human papillomavirus testing: a nested substudy of the
NTCC randomised controlled trial. Lancet Oncol 2008; 9: 937945.
77. Denton KJ, Bergeron C, Klement P et al. The sensitivity and specicity of p16
(INK4a) cytology vs HPV testing for detecting high-grade cervical disease in the

Downloaded from http://annonc.oxfordjournals.org/ by guest on February 4, 2014

43.

November 1. doi:pii: S0140-6736(13)62218-7. 10.1016/S0140-6736(13)


62218-7. [epub ahead of print]
Berkhof J, Coupe VM, Bogaards JA et al. The health and economic effects of
HPV DNA screening in The Netherlands. Int J Cancer 2010; 127: 21472158.
Rijkaart DC, Berkhof J, van Kemenade FJ et al. HPV DNA testing in populationbased cervical screening (VUSA-Screen study): results and implications. Br J
Cancer 2012;106: 975981.
Dijkstra M, van ND, Rijkaart D et al. Primary hrHPV DNA testing in cervical cancer
screening: how to manage screen positive women? A POBASCAM trial substudy.
Cancer Epidemiol Biomarkers Prev 2013 June 3 [epub ahead of print], doi:
10.1158/1055-9965.EPI-13-0173.
Arbyn M, Ronco G, Anttila A et al. Evidence regarding human papillomavirus
testing in secondary prevention of cervical cancer. Vaccine 2012; 30(Suppl 5):
F88F99.
Saslow D, Solomon D, Lawson HW et al. American cancer society, American
society for colposcopy and cervical pathology, and American society for clinical
pathology screening guidelines for the prevention and early detection of cervical
cancer. Am J Clin Pathol 2012; 137: 516542.
Sankaranarayanan R, Nene BM, Shastri SS et al. HPV screening for cervical
cancer in rural India. N Engl J Med 2009; 360: 13851394.
Hesselink AT, Van Ham MA, Heideman DA et al. Comparison of GP5+/6+-PCR
and SPF10-line blot assays for detection of high-risk human papillomavirus in
samples from women with normal cytology results who develop grade 3 cervical
intraepithelial neoplasia. J Clin Microbiol 2008; 46: 32153221.
Meijer CJ, Berkhof J, Castle PE et al. Guidelines for human papillomavirus DNA
test requirements for primary cervical cancer screening in women 30 years and
older. Int J Cancer 2009; 124: 516520.
Meijer CJ, Berkhof H, Heideman DA et al. Validation of high-risk HPV tests for
primary cervical screening. J Clin Virol 2009; 46(Suppl 3): S1S4.
Castle PE, Sadorra M, Lau T et al. Evaluation of a prototype real-time PCR assay for
carcinogenic human papillomavirus (HPV) detection and simultaneous HPV genotype
16 (HPV16) and HPV18 genotyping. J Clin Microbiol 2009; 47: 33443347.
Snijders PJ, Heideman DA, Meijer CJ. Methods for HPV detection in exfoliated
cell and tissue specimens. APMIS 2010; 118: 520528.
Hesselink AT, Heideman DA, Berkhof J et al. Comparison of the clinical
performance of PapilloCheck human papillomavirus detection with that of the
GP5+/6+-PCR-enzyme immunoassay in population-based cervical screening. J
Clin Microbiol 2010; 48: 797801.
Heideman DA, Hesselink AT, Berkhof J et al. Clinical validation of the cobas
4800 HPV test for cervical screening purposes. J Clin Microbiol 2011; 49:
39833985.
Carozzi FM, Burroni E, Bisanzi S et al. Comparison of clinical performance of
Abbott RealTime High Risk HPV test with that of hybrid capture 2 assay in a
screening setting. J Clin Microbiol 2011; 49: 14461451.
Poljak M, Cuzick J, Kocjan BJ et al. Nucleic acid tests for the detection of alpha
human papillomaviruses. Vaccine 2012; 30(Suppl 5): F100F106.
Hesselink AT, Meijer CJ, Poljak M et al. Clinical validation of the Abbott RealTime
High Risk (HR) HPV assay according to the guidelines for HPV DNA test
requirements for cervical screening. J Clin Microbiol 2013; 51: 24092410.
Getman D, Aiyer A, Dockter J et al. Efciency of the APTIMA HPV Assay for
detection of HPV RNA and DNA targets. J Clin Virol 2009; 45(Suppl 1):
S49S54.
Wu R, Belinson SE, Du H et al. Human papillomavirus messenger RNA assay for
cervical cancer screening: the Shenzhen Cervical Cancer Screening Trial I. Int J
Gynecol Cancer 2010; 20: 14111414.
Monsonego J, Hudgens MG, Zerat L et al. Evaluation of oncogenic human
papillomavirus RNA and DNA tests with liquid-based cytology in primary cervical
cancer screening: the FASE study. Int J Cancer 2011; 129: 691701.
Heideman DA, Hesselink AT, van Kemenade FJ et al.. The Aptima HPV assay
fullls the cross-sectional clinical and reproducibility criteria of international
guidelines for human papillomavirus test requirements for cervical screening. J
Clin Microbiol 2013; 51: 36533657.
Cuzick J, Cadman L, Mesher D et al. Comparing the performance of six human
papillomavirus tests in a screening population. Br J Cancer 2013; 108:
908913.

Annals of Oncology

Annals of Oncology

78.

79.

80.

81.

82.

83.

85.
86.

87.
88.
89.

90.

91.
92.

93.

94.

95.

96.

97. Gok M, Heideman DA, van Kemenade FJ et al. HPV testing on self collected
cervicovaginal lavage specimens as screening method for women who do not
attend cervical screening: cohort study. BMJ 2010; 340: c1040.
98. Virtanen A, Nieminen P, Luostarinen T et al. Self-sample HPV tests as an
intervention for nonattendees of cervical cancer screening in Finland: a
randomized trial. Cancer Epidemiol Biomarkers Prev 2011; 20: 19601969.
99. Snijders PJ, Verhoef VM, Arbyn M et al. High-risk HPV testing on self-sampled
versus clinician-collected specimens: a review on the clinical accuracy and
impact on population attendance in cervical cancer screening. Int J Cancer
2012; 32: 22232236.
100. Holanda F, Jr, Castelo A, Veras TM et al. Primary screening for cervical cancer
through self sampling. Int J Gynaecol Obstet 2006; 95: 179184.
101. Qiao YL, Sellors JW, Eder PS et al. A new HPV-DNA test for cervical-cancer
screening in developing regions: a cross-sectional study of clinical accuracy in
rural China. Lancet Oncol 2008; 9: 929936.
102. Lazcano-Ponce E, Lorincz AT, Cruz-Valdez A et al. Self-collection of vaginal
specimens for human papillomavirus testing in cervical cancer prevention
(MARCH): a community-based randomised controlled trial. Lancet 2011; 378:
18681873.
103. Ogilvie GS, Patrick DM, Schulzer M et al. Diagnostic accuracy of self collected
vaginal specimens for human papillomavirus compared to clinician collected
human papillomavirus specimens: a meta-analysis. Sex Transm Infect 2005; 81:
207212.
104. Dannecker C, Siebert U, Thaler CJ et al. Primary cervical cancer screening by
self-sampling of human papillomavirus DNA in internal medicine outpatient
clinics. Ann Oncol 2004; 15: 863869.
105. Khanna N, Mishra SI, Tian G et al. Human papillomavirus detection in selfcollected vaginal specimens and matched clinician-collected cervical specimens.
Int J Gynecol Cancer 2007; 17: 615622.
106. Castle PE, Schiffman M, Burk RD et al. Restricted cross-reactivity of hybrid
capture 2 with nononcogenic human papillomavirus types. Cancer Epidemiol
Biomarkers Prev 2002; 11: 13941399.
107. Baldwin S, Santos C, Mendez BE et al. Comparison of type-specic human
papillomavirus data from self and clinician directed sampling. Gynecol Oncol
2005; 97: 612617.
108. Nobbenhuis MA, Helmerhorst TJ, van den Brule AJ et al. Primary screening for
high risk HPV by home obtained cervicovaginal lavage is an alternative screening
tool for unscreened women. J Clin Pathol 2002; 55: 435439.
109. Bulkmans NW, Rozendaal L, Snijders PJ et al. POBASCAM, a population-based
randomized controlled trial for implementation of high-risk HPV testing in cervical
screening: design, methods and baseline data of 44,102 women. Int J Cancer
2004; 110: 94101.
110. Kitchener HC, Almonte M, Wheeler P et al. HPV testing in routine cervical
screening: cross sectional data from the ARTISTIC trial. Br J Cancer 2006; 95:
5661.
111. Ronco G, Giorgi-Rossi P, Carozzi F et al. Human papillomavirus testing and
liquid-based cytology in primary screening of women younger than 35 years:
results at recruitment for a randomised controlled trial. Lancet Oncol. 2006; 7:
54755.
112. Ronco G, Giorgi-Rossi P, Carozzi F et al. Results at recruitment from a
randomized controlled trial comparing human papillomavirus testing alone with
conventional cytology as the primary cervical cancer screening test. J Natl Cancer
Inst 2008; 100: 492501.
113. Leinonen M, Nieminen P, Kotaniemi-Talonen L et al. Age-specic evaluation of
primary human papillomavirus screening vs conventional cytology in a
randomized setting. J Natl Cancer Inst 2009; 101: 16121623.
114. Schmeink CE, Bekkers RL, Massuger LF et al. The potential role of self-sampling
for high-risk human papillomavirus detection in cervical cancer screening. Rev
Med Virol 2011; 21: 13953.

doi:10.1093/annonc/mdt538 |

Downloaded from http://annonc.oxfordjournals.org/ by guest on February 4, 2014

84.

triage of ASC-US and LSIL pap cytology results. Am J Clin Pathol 2010; 134:
1221.
Carozzi F, Gillio-Tos A, Confortini M et al. Risk of high-grade cervical
intraepithelial neoplasia during follow-up in HPV-positive women according to
baseline p16-INK4A results: a prospective analysis of a nested substudy of the
NTCC randomised controlled trial. Lancet Oncol 2013; 14: 168176.
Szarewski A, Ambroisine L, Cadman L et al. Comparison of predictors for highgrade cervical intraepithelial neoplasia in women with abnormal smears. Cancer
Epidemiol Biomarkers Prev 2008; 17: 30333042.
Wentzensen N, Bergeron C, Cas F et al. Evaluation of a nuclear score for p16
(INK4a)-stained cervical squamous cells in liquid-based cytology samples.
Cancer 2005; 105: 461467.
Schmidt D, Bergeron C, Denton KJ et al. p16/ki-67 dual-stain cytology in the
triage of ASCUS and LSIL papanicolaou cytology: results from the European
equivocal or mildly abnormal Papanicolaou cytology study. Cancer Cytopathol
2011; 119: 158166.
Wentzensen N, Schwartz L, Zuna RE et al. Performance of p16/Ki-67
immunostaining to detect cervical cancer precursors in a colposcopy referral
population. Clin Cancer Res 2012; 18: 41544162.
Petry KU, Schmidt D, Scherbring S et al. Triaging Pap cytology negative, HPV
positive cervical cancer screening results with p16/Ki-67 Dual-stained cytology.
Gynecol Oncol 2011; 121: 505509.
Mesher D, Szarewski A, Cadman L et al. Comparison of human papillomavirus
testing strategies for triage of women referred with low-grade cytological
abnormalities. Eur J Cancer 2013 February 26 [epub ahead of print].
Herman JG, Baylin SB. Gene silencing in cancer in association with promoter
hypermethylation. N Engl J Med 2003; 349: 20422054.
Henken FE, Wilting SM, Overmeer RM et al. Sequential gene promoter
methylation during HPV-induced cervical carcinogenesis. Br J Cancer 2007; 97:
14571464.
Steenbergen RD, Kramer D, Braakhuis BJ et al. TSLC1 gene silencing in cervical
cancer cell lines and cervical neoplasia. J Natl Cancer Inst 2004; 96: 294305.
Esteller M. Epigenetics in cancer. N Engl J Med 2008; 358: 11481159.
Bierkens M, Hesselink AT, Meijer CJ et al. CADM1 and MAL promoter
methylation levels in hrHPV-positive cervical scrapes increase proportional to
degree and duration of underlying cervical disease. Int J Cancer 2013; 133:
12931299.
Hesselink AT, Heideman DA, Steenbergen RD et al. Combined promoter
methylation analysis of CADM1 and MAL: an objective triage tool for high-risk
human papillomavirus DNA-positive women. Clin Cancer Res 2011; 17:
24592465.
Dijkstra et al. Long-term CIN3+ risks following primary hrHPV DNA and triage
testing in Cervical Cancer screening. submitted.
Bais AG, van Kemenade FJ, Berkhof J et al. Human papillomavirus testing on selfsampled cervicovaginal brushes: an effective alternative to protect nonresponders
in cervical screening programs. Int J Cancer 2007; 120: 15051510.
Raab SS, Grzybicki DM, Zarbo RJ et al. Frequency and outcome of cervical
cancer prevention failures in the United States. Am J Clin Pathol 2007; 128:
817824.
Sasieni P, Adams J, Cuzick J. Benet of cervical screening at different ages:
evidence from the UK audit of screening histories. Br J Cancer 2003; 89:
8893.
Petignat P, Faltin DL, Bruchim I et al. Are self-collected samples comparable to
physician-collected cervical specimens for human papillomavirus DNA testing? A
systematic review and meta-analysis. Gynecol Oncol 2007; 105: 530535.
Brink AA, Meijer CJ, Wiegerinck MA et al. High concordance of results of testing
for human papillomavirus in cervicovaginal samples collected by two methods,
with comparison of a novel self-sampling device to a conventional endocervical
brush. J Clin Microbiol 2006; 44: 25182523.

review

Vous aimerez peut-être aussi