You are on page 1of 11

European Journal of Obstetrics & Gynecology and Reproductive Biology 216 (2017) 1–11

Contents lists available at ScienceDirect

European Journal of Obstetrics & Gynecology and


Reproductive Biology
journal homepage: www.elsevier.com/locate/ejogrb

Review article

First-void urine: A potential biomarker source for triage of high-risk


human papillomavirus infected women
Severien Van Keera , Jade Pattyna , Wiebren A.A. Tjalmab , Xaveer Van Ostadec ,
Margareta Ievend, Pierre Van Dammea , Alex Vorstersa,*
a
Centre for the Evaluation of Vaccination (CEV), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of
Antwerp, Belgium
b
Multidisciplinary Breast Clinic, Unit Gynaecologic Oncology, Department of Obstetrics and Gynaecology, Molecular Imaging, Pathology, Radiotherapy,
Oncology (MIPRO), Faculty of Medicine and Health Sciences, Antwerp University Hospital (UZA)—University of Antwerp, Belgium
c
Proteomics, Proteinscience, Proteomics & Epigenetic Signalling (PPES), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp,
Belgium
d
Laboratory of Medical Microbiology (LMM), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of
Antwerp, Belgium

A R T I C L E I N F O A B S T R A C T

Article history:
Received 20 January 2017 Great interest has been directed towards the use of first-void urine as a liquid biopsy for high-risk human
Received in revised form 18 June 2017 papillomavirus DNA testing. Despite the high correlations established between urinary and cervical
Accepted 24 June 2017 infections, human papillomavirus testing is unable to distinguish between productive and transforming
Available online xxx high-risk infections that have the tendency to progress to cervical cancer. Thus far, investigations have
been primarily confined to the identification of biomarkers for triage of high-risk human papillomavirus-
Key words: positive women in cervicovaginal specimens and tissue biopsies. This paper reviews urinary biomarkers
Human papillomavirus for cervical cancer and triage of high-risk human papillomavirus infections and elaborates on the
Cervical cancer
opportunities and challenges that have emerged regarding the use of first-void urine as a liquid biopsy for
Biomarker
the analysis of both morphological- (conventional cytology and novel immunohistochemical techniques)
First-void urine
Self-sampling and molecular-based (HPV16/18 genotyping, host/viral gene methylation, RNA, and proteins)
biomarkers. A literature search was performed in PubMed and Web of Science for studies investigating
the use of urine as a biomarker source for cervical cancer screening. Five studies were identified reporting
on biomarkers that are still in preclinical exploratory or clinical assay development phases and on
assessments of non-invasive (urine) samples. Although large-scale validation studies are still needed, we
conclude that methylation of both host and viral genes in urine has been proven feasible for use as a
molecular cervical cancer triage and screening biomarker in phase two studies. This is especially
promising and underscores our hypothesis that human papillomavirus DNA and candidate human and
viral biomarkers are washed away with the initial, first-void urine, together with exfoliated cells, debris
and impurities that line the urethra opening. Similar to the limitations of self-collected cervicovaginal
samples, first-void urine will likely not fulfil the high-quality cellularity standards required for
morphological biomarkers. Molecular biomarkers will likely overcome this issue to yield high-
throughput, objective, and reproducible results. When using proper sampling, transport, storage,
preanalytical biomarker concentration techniques, and clinically validated assays, first-void urine is
expected to be a valuable source of molecular biomarkers for cervical cancer screening. Furthermore, as
first-void urine can be easily and non-invasively collected, it is a highly preferred technique among
women and offers the ability to test both primary high-risk human papillomavirus and biomarkers in the
same sample. In addition, the use of first-void urine confers opportunities to reduce loss-to follow-up and
non-adherence to screening subjects.
© 2017 The Author(s). Published by Elsevier Ireland Ltd. This is an open access article under the CC BY-NC-
ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

* Corresponding author at: Campus Drie Eiken-Building R2—Universiteitsplein, 1-2610 Wilrijk, Belgium.
E-mail address: alex.vorsters@uantwerp.be (A. Vorsters).

http://dx.doi.org/10.1016/j.ejogrb.2017.06.036
0301-2115/© 2017 The Author(s). Published by Elsevier Ireland Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-
nd/4.0/).
2 S. Van Keer et al. / European Journal of Obstetrics & Gynecology and Reproductive Biology 216 (2017) 1–11

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
State of the art: human papillomavirus and cervical cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Interest in human papillomavirus analysis in first-void urine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
The need for triage of human papillomavirus infected women . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Biomarkers for cervical cancer screening . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Urinary biomarkers for cervical cancer screening reported in the literature . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Opportunities and challenges of first-void urine as a biomarker source . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Microscopy-based markers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Molecular markers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Human papillomavirus DNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
Host and viral gene DNA methylation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
RNA-based biomarkers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Protein biomarkers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9

Introduction Interest in human papillomavirus analysis in first-void urine

State of the art: human papillomavirus and cervical cancer Recently, great interest has been directed towards the use of
urine as a liquid biopsy for HPV DNA testing because of the high
With an estimated 527,624 new cancer cases and 265,672 correlations established between urinary HPV DNA and cervical
deaths in 2012, cervical cancer remains a significant problem infections, the ease of sampling, the lower barrier for women, and
worldwide [1]. Globally, it is the fourth most common cancer in the lower cost of the programme [15–26]. As Papanikolaou
women with an age-standardized incidence rate of 14 cases per previously indicated (1943), superficial cell layers of a cervix
100,000. Strikingly, women living in low- and middle-income carcinoma are exfoliated and subsequently mixed with secretions
countries are most affected by this cancer due to the lack of cervical of the uterus and cervix, which make their way to the vagina and
cancer prevention measures, representing almost 90% of all hence can be recognized in vaginal fluid smears [27]. The theory
cervical cancer deaths [1]. Human papillomavirus, commonly behind identifying biomarkers including HPV DNA for cervical
known as HPV, is the main aetiological factor of cervical cancer [2]. (pre)cancer screening in first-void urine is based on this idea [28],
Although the majority of the population is unaware of the virus, because we are not interested in the urine itself but instead in the
HPV is the most common sexually transmitted infection, affecting mucus and debris from exfoliated cells from the genital tract
nearly all sexually active people during their lifetime [3]. Non- (particularly the cervix) that line the urethra opening and small
sexual, vertical transmission has also been reported [4]. Fortu- labia and are washed away with the initial urine flow. It hence
nately, over 90% of all HPV infections are spontaneously cleared follows that the initial flow of urine (first-void) collects most of
within two years of acquisition [5]. To date, 200 genotypes of this this debris, without interfering with the natural history of an HPV
virus have been identified, from which at least 40 affect the infection by scraping of cervical cells. This finding explains why
anogenital mucosa [6,7]. According to the likelihood of genotypes the first collected portion of a urine void contains more human
to develop cervical cancer, two groups can be distinguished: low- and HPV DNA than the subsequent portion, as was concluded by
risk (LR) and high-risk (HR) HPV genotypes. LR-HPV genotypes the authors of a recent meta-analysis [19]. Our research confirms
mainly cause genital warts, whereas the thirteen genotypes these findings, indicating that first-void urine contains signifi-
classified as (probable) HR-HPV  with HPV16 and 18 causing cantly more HR-HPV (4.8–160 times) and human DNA than the
70% of all cervical cancer cases  can lead to precancerous lesions, subsequent fraction [28].
and if left untreated, cervical cancer [8]. Screening based on HPV testing of self-collected cervicovaginal samples can be
primary HR-HPV DNA detection is currently of great interest, offered as an alternative strategy for women not attending cervical
which is more effective and efficient for preventing cervical cancer cancer screening programmes [29,30] and has been approved for
and the associated mortality compared to cytology [9,10]. This was use in organized, population-based pilot programmes [11].
demonstrated based on the higher sensitivity for detecting high- Recently, it has also been proposed as a primary cervical cancer
grade cervical intraepithelial neoplasia (CIN2+) and the lower screening tool due to its high relative sensitivity and specificity for
cumulative incidence of cervical cancer five years after a negative CIN2+ compared to clinician-collected samples [31]. Population-
HPV test compared to three years after a normal cytology result based approaches, such as sending personal invitation letters and
[9,11,12]. Nonetheless, challenges remain at the sampling level reminders for a scheduled appointment, have proven to be
regarding better up-take in screening programmes and less effective in increasing participation rates [11,32], although barriers
expensive options for follow-up. Additionally, prophylactic HPV for attending cervical cancer screening have persevered. These
vaccinations will result in a lower prevalence of HR-HPV types [13], barriers can be diverse, including practical (e.g., lack of time,
ensuing a drop in the prevalence of true-positive high-grade intending to go but not getting around to it), emotional (e.g., fear of
precancerous lesions (CIN2+). The consequential decrease in the pain, embarrassment, fear of the test results/cancer), and cognitive
‘signal-to-noise’ ratio will thus continue to adversely affect the (e.g., low perceived risk, absence of symptoms) barriers [33,34].
sensitivity, specificity, and positive predictive value (PPV) of Despite the fact that embarrassment was the most mentioned
cytology to a point when it will no longer be suitable for primary barrier, practical barriers such as inconvenience of making an
cervical cancer screening [14]. appointment and not getting around to going for screening, were at
higher odds in a cohort of women overdue for screening, and
S. Van Keer et al. / European Journal of Obstetrics & Gynecology and Reproductive Biology 216 (2017) 1–11 3

therefore, were more predictive of non-attendance to screening aberrations in host cell genes over time. These aberrations can
[33]. Both cervicovaginal and urine self-sampling have been be caused by both genetic (e.g., DNA mutations and chromosomal
proven effective in increasing participation and screening coverage copy number alterations) and epigenetic alterations (e.g., DNA
of target populations [35–38]. However, compared to the currently methylation and histone modifications) [43]. To avoid over-referral
available cervicovaginal self-sampling methods and clinician- and over-treatment of women with clinically irrelevant HR-HPV
collected smears, (first-void) urine collection is highly preferred infections and the associated costs and potential harms caused by
among women [39,40]. First-void urine furthermore allows for the current treatment methods, there is a need for additional triage
easily repetitive sampling if needed. tests that can distinguish productive from transforming HR-HPV
infections. Interesting candidate biomarkers for triage of HR-HPV-
The need for triage of human papillomavirus infected women positive women  hereafter referred to as ‘biomarkers’  has been
a topic of great interest, and many potential biomarkers have been
Despite the high sensitivity of HR-HPV DNA testing, it remains identified to date.
challenging to identify clinically relevant, i.e., transforming, HR-
HPV infections as only approximately 4% of persistent HR-HPV Biomarkers for cervical cancer screening
infections progress to high-grade lesions [41], among which
approximately 30% can progress to cancer if left untreated [42]. A biomarker is “a defined characteristic that is measured as an
Transforming HPV infections differ from productive infections in indicator of normal biological processes, pathogenic processes, or
the way that the normal viral life cycle of HPV is halted, followed by responses to an exposure or intervention, including therapeutic
overexpression of the viral oncogenes E6 and E7, which leads to interventions” [44]. Ideally, a biomarker should be detected at a
chromosome instability and subsequent accumulation of stage early enough to foresee successful treatment and only in the

Fig. 1. Hypothetical one- and two-step triage strategy offering first-void urine self-sampling at home as an alternative to screen non-attendees. Five-yearly primary HR-HPV
testing has been proposed by the European guidelines for quality assurance for cervical cancer screening in organized, population-based cervical cancer screening
programmes [11]. However, it has also been proposed for self-collected cervicovaginal samples for use in organized, population-based pilot programmes for women who do
not attend regular cancer screening programmes despite receiving personal reminders. Nonetheless, due to a lower HPV testing accuracy for use of self-collected versus
clinician-collected samples, one should still be aware that primary HR-HPV testing of self-collected samples is not recommended for national implementation before
successful results have been demonstrated by a pilot study. Currently, a triage of HR-HPV-positive test results is recommended by cytology, followed by repeat testing or
colposcopy [11]. Both microscopy- and molecular-based biomarkers for triage of HR-HPV-positive women were recently reviewed by Ebisch [51] and Luttmer [52]. Discussing
the opportunities and challenges of these biomarkers as urinary biomarker is the main objective of this review. A one-step triage method in which both HR-HPV and
biomarkers can be analysed in the same sample is preferred, as such a method would confer higher compliance to follow-up compared to two-step triage options. HPV:
human papillomavirus.
4 S. Van Keer et al. / European Journal of Obstetrics & Gynecology and Reproductive Biology 216 (2017) 1–11

presence of cervical precancerous lesions that will progress to cytology, and chromosomal aberrations can be classified under
cervical cancer (CIN2+) [45,46]. Notably, CIN2 is an intermediate, morphological biomarkers [50–52]. Examples of promising mo-
more regressive condition including both productive and trans- lecular biomarkers are HPV16/18 genotyping, methylation of host
forming infections  compared to the more histological reproduc- and viral genes, HPV E6 protein, and E6/E7 mRNA-, and miRNA-
ible CIN3 diagnosis [47]. The overall impact of a CIN2+ screening based biomarkers [14,51,52]. Viral load and integration of HPV DNA
test may therefore be overstated, and hence, reporting CIN2+ and into host DNA have been suggested as markers to predict disease
CIN3+ lesions separately may be more efficient [46]. For a outcome [55,56] but further investigations are needed.
biomarker to be acceptable, a PPV and negative predictive value As already mentioned above, none of the biomarkers reported
(NPV) of at least 20% and 98%, respectively, should be obtained in the literature to-date meet the criteria as an ideal cervical cancer
[48,49]. Unfortunately, for cervical cancer screening, none of the screening biomarker for detecting clinically relevant precancerous
biomarkers identified to date offer sufficient prognostic value, and lesions. The only available triage strategy that has been sufficiently
as such, none serve as an ideal triage method [50–52]. It is evaluated for HR-HPV-positive women, without repeat testing, is
furthermore unlikely that a single biomarker will provide HPV16/18 genotyping in combination with cytology [51,52].
sufficient sensitivity to detect all high-grade lesions and cancers Genotyping of HPV16/18 has all the benefits of HPV DNA-based,
(CIN2+) [45,53]. Therefore, a panel of biomarkers is expected to be molecular testing, although high colposcopy referrals will endure.
necessary for accurate testing with good clinical sensitivity and Adding cytology to HPV16/18 genotyping will increase the clinical
specificity. This panel would preferably consist of biomarkers that value of this triage strategy, resulting in an improved NPV,
are involved in cervical carcinogenesis and that complement each although at the cost of the need for a clinician-taken sample for
other concerning, for instance, functionality such that different cytology [51,52]. Their wide evaluation as a triage method makes
aspects of the cancer are targeted for its detection. For example, the them a logical first choice as biomarkers in coming years. However,
biomarkers could be molecules from different cervical cancer with more extensive knowledge on molecular markers from large
pathways (cancer- versus immune-related molecules) and from population-based prospective studies, cervical cancer screening
different biomarker classes (e.g., proteins, (methylated) nucleic may transform to full molecular screening in the future [51].
acids). Thus far, investigations have been primarily confined to the
Recent papers have reviewed the state-of-the-art of triage identification of biomarkers in cervicovaginal specimens and
markers for HR-HPV infections in cervical samples [14,50–52,54]. tissue biopsies. Based on the higher sensitivity of HPV DNA
Briefly, these biomarkers can be divided in two major groups: (i) detection in well-preserved, first-void urine compared to mid-
morphological- or microscopy-based biomarkers and (ii) molecu- stream or random urine samples for the presence of cervical HPV
lar biomarkers. Conventional cytology, novel immunohistochemi- [19,26,40,57], it is expected that biomarkers might also be present
cal techniques such as p16INK4a staining and Ki-67/p16 dual stained in the first part of the urine void. To the best of our knowledge, no

Fig. 2. Flow chart of the literature search: urinary biomarkers for cervical cancer screening. The search yielded seven and ten hits in PubMed imported into Endnote X7 (‘All
Fields Contain’) and Web Of Science within Endnote X7 (‘Title/Keywords/Abstracts’), respectively. The retrieved records were first screened based on the title and abstract,
followed by full text evaluation. Publications were included that reported the performance of biomarkers (other than HPV itself) analysed in human urine for cervical cancer
screening or, more specifically, triage of high-risk HPV-positive women. After screening for eligibility and addition of manually selected publications, 4 papers and 1 abstract
were included, published between 2003 and 2016. The flow chart was adapted from the PRISMA flow diagram [63].
S. Van Keer et al. / European Journal of Obstetrics & Gynecology and Reproductive Biology 216 (2017) 1–11 5

one has reflected on the opportunities and challenges that have HPV-positive women based on microscopy and molecular assays.
emerged regarding the use of first-void urine as a liquid biopsy for Detection of both HR-HPV DNA and biomarkers in the same, self-
triage of HR-HPV-positive women. This paper reviews urinary collected first-void urine sample will be an additional asset to
biomarkers for cervical cancer screening or for triage of HR-HPV- minimize efforts and costs associated with cervical cancer
positive women and reflects emerging challenges with first-void screening. First-void urine self-sampling will furthermore encour-
urine as a liquid biopsy for analysis of triage biomarkers for HR- age women who are currently reluctant to participate in screening

Table 1
Overview of candidate biomarkers in urine for cervical cancer diagnosis reported in the literature.

Candidate biomarker Phase in Study group Colposcopy/ Collection (i) Pre-analytical urine Outcome Refs
(class) biomarker (n = ) biopsy confirmed method; and processing; and (ii) analysis
a
evaluation (n = ) preanalytical of candidate biomarker
storage
conditions
(time span)
Ratio 16a-hydroxy Phase 1 Case-control No, Pap smear and Early (i) 2/3 ml urine (spiked withNo report of sensitivity, Lee SH
estrone (16a-OH E1)/ study (n = 61) cervicogram; morning internal control) applied to a
specificity, PPV, NPV, or AUC [58]
2-hydroxy estrone (2- cervical disease urine in Serdolit XAD-2 resin column 16a-OH E1/2-OH E1 and
OH E1 (estrogen); ratio (n = 18/61), polyethylene (ii) Gas chromatography- THF/5a-THF proposed as a
5b-tetrahydro-cortisol cervical cancer bottles; mass spectrometry-selected dual biomarker panel for
(THF)/5a-THF (n = 18/61), stored at ion-monitoring (GC/MS/ cervical cancer screening due
(androgen/corticoid) controls (n = 25/ 20  C (NA) SIM), normalization to to a significant difference in
(enzyme activity) 61) creatinine concentration concentration between
controls and benign cervical
disease versus cervical
cancers
Collagen I-alpha-I; Phase 1 Case-control Yes, biopsy Spot urine (i) Centrifugation (1,500g, No report of sensitivity, Garbett

collagen XVII-alpha-I study (n = 12) confirmed; CIN2 samples; 15 min, 4 C) ! Amicon specificity, PPV, NPV, or AUC NC [59]
(protein class) (n = 4); cervical stored in Ultra-4 (10,000 NMWCO) of Collagen I-alpha-I increased
cancer (n = 4); aliquots at supernatant ! desalting and in CIN2 > cervical cancer >
controls (n = 4) 80  C (NA) concentration using C18 controls; collagen XVII-
ZipTips alpha-I increased in CIN2 >
(ii) MALDI-TOF(/TOF) MS cervical cancer and is absent
in controls
DAPK1; RARB; TWIST1; Phase 2 Subgroup of Yes, biopsy NA; stored at (i) 5 ml urine HR-HPV detection or Feng QH
and CDH13 women from a confirmed 4 C centrifuged ! cell pellets aberrant methylation of at [61]
(host gene methylation) study (n = 110/129); (processed resuspended in least 1/4 genes: sensitivity of
investigating CIN0 (n = 19), CIN1 within STM ! stored at 84% (ICC) and 64% (CIN2/3),
HPV and CIN (n = 9), CIN2-3/CIS 24 hours) 80  C ! Proteinase K with a specificity of 72% (as
prevalence in (n = 29), ICC digestion (37  C; 1– to CIN0/1) and 81% (as to
West-Africa (n = 72) 2 h) ! DNA isolation of CIN0)
(n = 129) 400 ml urine digest (QIAamp High concordance of HR-HPV
Blood Minicolumn) ! DNA detection or aberrant
concentration to 25 ml methylation of at least 1/4
(Speedvac; RT) genes between paired urine
(ii) Bisulfite conversion of and cervical samples (49–
50–100 ng DNA and 80%) (n = 97/110)
MethyLight (qMSP) analysis
of DAPK1; RARB; TWIST1;
CDH13; U-DNA; M-DNA; and
ACTB
ZNF516; FKBP6; INTS1; Phase 2 Prospective No, paired LBC, NA; flash (i) ccfDNA from 10 ml urine The 4-gene biomarker panel Guerrero-
and HPV16-L1 cohort (n = 40) plasma, and frozen (NA) adsorbed on Q-Sepharose in urinary ccfDNA has Preston R
(host and viral gene serum samples anion-exchange resin, sensitivity of 75%, specificity [62]
methylation) followed by silica-based of 83.3%, PPV of 50%, NPV of
elution with LiCl 93.8%, and AUC of 0.86 for
(ii) Bisulfite conversion and CIN2+ as to NILM/CIN1.
qMSP analysis for ZNF516; Compared to LBC the 4-gene
FKBP6; INTS1; and HPV16-L1. panel has a lower sensitivity,
A target enrichment system better specificity, and similar
was used to sequence the NPV, PPV, and AUC.
HR-HPV epigenome in urine
NA Phase 1 Case-control NA NA Paired cervical scrapes and High accuracy for detecting Rurup R
(host gene methylation) study (n = 53); urine samples were tested cervical cancer (AUC > 0.86 [60]
cervical cancer for HR-HPV DNA and DNA for 4/6 genes)
(n = 29/53), methylation of 6 genes High correlation of DNA
controls (n = 24/ methylation levels between
53) paired urine and cervical
samples (Spearman
correlation = 0.83)
a
: Phase 1: preclinical exploratory studies; Phase 2: clinical assay development for clinical disease and assessment in non-invasive samples; Phase 3: retrospective
longitudinal repository studies; Phase 4: prospective screening studies; Phase 5: prospective intervention study; LBC: liquid-based cytology; NILM: negative for
intraepithelial lesion and malignancy; CIN1-3+: Cervical Intraepithelial Neoplasia grade 1–3+; CIN0: no CIN; CIS: carcinoma in situ; ICC: invasive cervical carcinoma; PPV:
positive predictive value; NPV: negative predictive value; AUC: area under the curve; STM: specimen transport media (Digene); RT: room temperature; ACTB: methylated
b-actin reference gene; U-DNA: unmethylated human sperm DNA control; M-DNA: methylated human sperm DNA control; ccfDNA: circulating cell-free DNA; qMSP:
quantitative methylation-specific PCR; NA: not available.
6 S. Van Keer et al. / European Journal of Obstetrics & Gynecology and Reproductive Biology 216 (2017) 1–11

programmes and may increase their compliance for further follow- clinical trials (www.clinicaltrials.gov) using the same search terms
up if they are HR-HPV positive (Fig. 1) with minimal efforts by the as used for the health science databases yielded 10 and 34 hits,
screened women. respectively. Two of the 10 clinical trials actually investigated
biomarkers for cervical (pre)cancer in urine samples
Urinary biomarkers for cervical cancer screening reported in (NCT02714127 and NCT00290459), but no results were available
the literature so far. The patent search resulted in 2 out of 34 hits describing a
urine detection kit for cervical cancer screening (CN104714030 and
A review of the literature on (bio)markers for cervical cancer CN104198404). Only summaries of the patents were available in
and HPV in urine was performed up to January 3rd 2017 in the two English, providing few details but indicating that the technologies
main databases for health sciences: PubMed (‘All Fields Contain’) are based on changes in urine colour and colorimetry.
and Web Of Science (‘Title/Keywords/Abstracts’) (Fig. 2). In each
database, the following search terms were used: marker* OR Opportunities and challenges of first-void urine as a biomarker
biomarker*; AND HPV*, human papillomavirus*, human papilloma source
virus*, cervical cancer(s), cervical carcinoma*, OR cancer(s) of the
cervix; AND urine. Four papers and one abstract were included In the following sections, evidence is discussed that non-
(Table 1). To promote efficiency and consistency in cancer invasive first-void urine self-sampling for cervical cancer screening
screening biomarker research, a five-step framework was set up is promising and could serve as an alternative method to screen
by the Early Detection Research Network for evaluating the and triage non-attendees in the future.
potential use of a biomarker in cancer screening [45]. Two of the
candidate biomarkers proposed in the articles of our search Microscopy-based markers
reached stage two of the biomarker evaluation, i.e., clinical assay
development [58–60], while the other three only achieved stage Cytological techniques have a 3–4% higher specificity for
one, i.e., the preclinical exploratory phase [45,46,61,62]. Retro- clinically relevant infections compared to primary HR-HPV testing
spective and large-population-based prospective studies are [12], reducing the number of colposcopy referrals and follow-up
required to validate the proposed urinary but also cervicovaginal tests. Novel immunohistochemical techniques (e.g., p16INK4a, Ki-
sample-based biomarkers, as only few candidate biomarkers have 67/p16) have proven to be more sensitive for detecting CIN3+
reached phase three or beyond. None of the studies used first-void lesions for triage of HR-HPV-positive women compared to
urine samples or a urine conservation medium, or they did not conventional cytology, without a substantial increase in colposco-
report such use. For DNA-based biomarkers, use of first-void urine py referrals. Nevertheless, they are not able to distinguish between
and immediate mixing with a urine conservation medium are productive and transforming HR-HPV infections. Chromosomal
essential for high biomarker recovery, as is the use of whole urine aberrations, and more specifically, loss at 3p and 11q and gains at
for analysis instead of pellet or supernatants [19,26,28,57]. In 3q, have been found to be most frequently altered in cervical
contrast, either pellet or cell-free DNA was used in two studies that carcinoma [64]; however, they have not been evaluated in large
investigated DNA methylation biomarkers in urine [61,62]. It must clinical cohorts of HR-HPV-positive women [51,52].
be further investigated whether using whole urine results in In our search for candidate biomarkers in urine, two
higher methylation biomarker yields, as is the case for human and publications reported the use of urine for analysing morphological
HPV DNA. Searches for patents (worldwide.espacenet.com) and markers for urothelial carcinoma (cancer of the bladder, ureters,

Table 2
Summary of the benefits and drawbacks of microscopy-based/morphological and molecular biomarkers with the aim to use self-collected first-void urine samples for triage
of HR-HPV-positive women.

Microscopy-based/morphological biomarkers
Benefits
Specificity, reduction in colposcopy referrals
Widely evaluated as a triage method
Drawbacks
Sensitivity; necessitates repeat testing; can result in loss to follow-up
Subjective (e.g., affected by prior knowledge of HPV status)
Need for trained cytotechnologist/prone to human error
Suboptimal reproducibility
Suboptimal high-throughput capacity
Not reliable in self-collected samplesa due to need of high quality, cell rich material; necessitates extra clinician-collected cervical sample
Two-step triage; can result in drop out

Molecular biomarkers
Benefits
Objective
Less prone to human error
Reproducibility
High-throughput capacity
Multiplexing format enables small sample volumes and less hands-on time
Possible on the same, self-collecteda (first-void urine) sample
Allows one-step triage; decreases drop out and loss to follow-up

Drawbacks
Necessitates large, population-based retro-, and prospective cohort studies (phase 3–5)b in self-collected (first-void urine) samples
a
Self-sampling yields opportunities to increase screening attendance rates, testing of both HR-HPV DNA and molecular biomarkers in the same sample, and hence
minimizing loss of follow-up.
b
Phase 3: retrospective longitudinal repository studies; Phase 4: prospective screening studies; Phase 5: prospective intervention study [45].
S. Van Keer et al. / European Journal of Obstetrics & Gynecology and Reproductive Biology 216 (2017) 1–11 7

urethra, and renal pelvis) [65,66]. Similar to cervical cytology, urinations will positively affect the amount of markers present in
urinary cytology for bladder cancer has a high specificity (ca. 94%) the mucus and cervicovaginal secretions that line the urethra
but is hampered by a low sensitivity (ca. 35%) [67]. Nonetheless, opening and small labia and are subsequently captured in the first
microscopy-based techniques identifying morphological markers urine void. However, recent studies indicate no advantage of
require cell-rich sample material, which cannot be assured in self- testing first-void urine samples taken in the morning versus
collected cervicovaginal samples. The available cervical cells are samples collected later that day using unfractionated urine
often of poor morphological quality and diluted due to the large [40,68,71]. The use of human DNA as a first-void urine sample
amount of vaginal cells present in the sample, resulting in a lower validity control should be interpreted with caution, since we
sensitivity [43,51,52]. In addition to this concentration issue, observed more rapid degradation of HPV than human DNA [57],
reproducibility and high-throughput capacity also limit the possibly resulting in an underreported HPV prevalence. Therefore,
applicability of morphological markers as triage markers for HR- preference is given to the addition of control DNA to the first-void
HPV-positive women due to the subjective nature of the method urine sample immediately upon collection.
and need for a trained cytotechnologist, respectively (Table 2). Care should be taken when interpreting urinary HPV DNA
Molecular-based triage methods, on the other hand, are objective, results for primary screening purposes [21]. The clinical validity of
highly reproducible, suitable for high-throughput testing, and offer HPV testing using urine samples should be investigated under
the opportunity to use self-collected cervicovaginal material optimal conditions. An inferior HPV prevalence was found in the
(Table 2). PaVDaG study where random void urine samples and non-
optimized assay thresholds were used [31]. This is in contrast to
Molecular markers other studies where optimized urine collection, storage, sample
preparation, DNA extraction and amplification methods were used
Human papillomavirus DNA [16,18,40]. These data make evident that, to-date, less attention has
The use of first-void urine as a liquid biopsy for HPV DNA testing been paid to optimal urine sampling, storage, and sample
is promising. In a recent meta-analysis, the sensitivity and preparation methods compared to DNA extraction and amplifica-
specificity for detecting any HPV in urine were 87% (95% CI: 78– tion methods. These optimizations are, however, of utmost
92%) and 94% (82–98%), respectively, when results from cervical importance in order to not underestimate infection rates and to
samples were used as a reference [19]. However, because of a large obtain reproducible results in the context of primary HR-HPV-
heterogeneity in urine collection and testing methods, the pooled based screening but also for triage of HR-HPV-positive women
estimates in this meta-analysis could be incorrectly estimated, using HPV16/18 genotyping.
emphasizing the need for standardized methods for human and
HPV DNA detection in urine samples. The use of random or Host and viral gene DNA methylation
midstream instead of first-void urine has been identified as a An important asset of host cell DNA methylation biomarkers is
source of heterogeneity [22,26,40,57,68], resulting in a 22-fold their ability to distinguish productive from transforming HR-HPV
decrease in accuracy for detecting the presence of cervical HPV infections, especially those with a high short-term risk of
[19]. Ultrafiltration and subsequent DNA extraction of unfractio- progression to cancer. Methylation levels have also been found
nated, DNA preserved, first-void urine has been proven to yield a to rise in parallel with increased severity and duration of disease
greater amount of HPV DNA-positive samples than paired cervical [43,52]. Hereto, host cell DNA methylation has also been proposed
samples, using analytically sensitive HPV DNA assays as a potential biomarker for post-treatment follow-up as it enables
[16,20,57,69,70]. Based on these data and the literature reported differentiation between long-existing and new transforming
to date, we propose that optimized HPV DNA detection in urine lesions [43]. A small set of methylation panels have been evaluated
should include the following: (i) use of first-void urine captured in self-collected lavage and/or brush-based samples, but only one,
with a first-void urine collection device to secure a standardized the multiplex qMSP assay based on the bi-marker panel FAM19A4
urine fraction; (ii) prevention of human/HPV DNA degradation (Family with sequence similarity 19 (chemokine (C-C motif)-like)
during extraction and storage by immediately mixing the first-void member A4)/mir124-2 (microRNA124-2) has been clinically validat-
urine with a conservation medium that is harmless to humans and ed in a large cohort of HPV-positive women. This assay yields
is preferentially prefilled in the vial used for the first-void urine similar sensitivity and specificity for CIN3+ in both lavage and
collection; (iii) processing of a sufficient first-void urine volume; brush-based self-collected samples and clinician-taken cervical
(iv) recovery of cell-free and cell-associated HPV DNA by smears [72].
processing whole first-void urine as opposed to only the pellet In contrast to methylation of human gene promotors resulting
or supernatants [26,28,57]; and (v) the use of HPV assays that fulfil in decreased transcription of tumour suppressor genes due to
the criteria for primary cervical cancer screening [10], with hypermethylation, methylation of viral DNA can result in both
optimized assay thresholds for first-void urine (summarized in increased and decreased viral gene transcription. The exact
Table 3). As we are interested in human and HPV DNA originating mechanism and how viral DNA methylation occurs remains
from the cervix, we expect that a larger time interval between two unclear, but it has been proposed that it is a host defence

Table 3
Key notes for human and HPV DNA detection in urine [10,26,28,57].

Optimized human and HPV DNA detection in urine should include:


- Use of first-void urine; preferentially captured with a first-void urine collection device to secure a standardized urine fraction
- No extensive washing of genitals before urination, as well as collecting the first-void urine sample preceding collection of other cervicovaginal self-samples or clinician-
taken specimens because this could reduce the amount of human/HPV DNA originating from the (exfoliated) cells from the cervix
- Prevention of human/HPV DNA degradation during storage and extraction by immediately mixing the first-void urine with a conservation medium; preferentially
prefilled in the collection vial
- Processing a sufficient amount of first-void urine
- Recovery of both cell-free and cell-associated DNA by processing whole first-void urine rather than only the supernatant or pellet
- Use of validated assays with optimized assay thresholds; for primary HR-HPV screening preferentially HPV assays that fulfil the criteria for primary cervical cancer
screening
8 S. Van Keer et al. / European Journal of Obstetrics & Gynecology and Reproductive Biology 216 (2017) 1–11

mechanism to silence viral replication and transcription [43,73]. validated in large prospective cohorts [43,51]. This is in contrast to
To-date, only specific CpGs in the late genes L1 and L2 of HPV16, 18, E6/E7 mRNA, which has been extensively studied, the over-
31, 33, and 45 have shown consistently higher methylation levels expression of which has been associated with cellular transforma-
with viral persistence and cell immortalization, increasing the tion and increased disease severity [80]. One assay targeting E6/E7
degree of disease, and may be useful for differentiating trans- mRNA from 14 (HR-)HPV types (APTIMA HPV assay, Hologic) has
forming from productive infections after sufficiently validated been fully validated against a standard comparator test according
[50–52,73,74]. to the Meijer protocol, showing a similar pooled sensitivity and
Methylation of host and viral genes as urinary biomarkers superior specificity for CIN2+ in a primary screening setting.
include the opportunity for a fully molecular and easily automated However, these guidelines were developed for HPV DNA-based
assay using both clinician- and self-collected samples (Table 2). assays, and long-term data are not yet available to determine the
However, in contrast to host gene methylation, a unique assay risk of CIN3+ after a negative APTIMA HPV test [10].
needs to be developed and validated for each HR-HPV type due to Liquid biopsy samples have been found to harbour both mRNA
the genetic variation between HPV genotypes. Numerous methyl- and miRNA [81,82]. Specifically, for urine, both mRNA- and miRNA-
ation markers have been proposed as candidate biomarkers, but based biomarkers have been described, such as for bladder and
the greatest need is to validate the existing markers in the near prostate cancer [82–85]. Zika virus RNA is detected at higher levels
future [73]. in urine compared to blood, and a quantitative reverse transcrip-
In theory, DNA methylation analysis is possible for all biological tion-PCR (qRT-PCR) assay has been approved by the FDA for this
sample types containing DNA [73,75,76]. DNA methylation assays use. Similar to HPV, Zika virus is sexually transmitted and has been
for human and viral genes, however, require high (quality) DNA proposed to actively replicate in the genitourinary tract [86].
content. Therefore, the concentrations of both human and HPV Recently, a urinary mRNA biomarker panel for the detection of
DNA in first-void urine are expected to represent an important step bladder cancer confirmed the potential of urine as a liquid biopsy
in the analysis of methylation biomarkers for cervical precancerous source for urothelial cell gene expression signatures [84].
lesions. As such, similar optimization issues as for human and HPV Optimization methods similar to those used for DNA (Table 3)
DNA detection/genotyping purposes are expected to apply here are expected to apply to RNA-based biomarkers as well; for
(Table 3). By multiplexing for a reference gene and different target instance, mRNA has also been shown to degrade due to the
genes in a single PCR reaction, the required amount of sample DNA presence of nucleases in urine samples. On the other hand, miRNA
is further reduced with improved quality control [75]. Multiplex has a high degree of stability in urine [82]. Mixing of the urine with
quantitative methylation-specific PCR (qMSP) is indeed an a buffer containing a chelating agent  inactivating nucleases 
attractive assay that enables the detection of specific DNA might therefore increase the stability of mRNA in urine. The
methylation levels in samples containing a limited amount of question regarding whether mRNA and miRNA should be
methylated DNA in a background of abundant cellular DNA [76,77]. measured in whole urine, the cell pellet or the supernatant should
As this is important for self-collected brush- and lavage-based be further investigated. For the urinary biomarkers reported to-
cervicovaginal samples [75], it is expected to be important for first- date, cell pellets have primarily been investigated for miRNA, and it
void urine. was recommended by van de Vrie and colleagues to use midstream
When considering DNA methylation markers for other diseases urine when investigating biomarkers for kidney pathology due to
such as prostate and bladder cancer, urine is a good source of contamination of cells from the lower urinary tract in the first-void
exfoliated cells from the genitourinary tract [76]. However, [85]. Because we are interested in biomarkers originating from the
exfoliation of tumour cells depends on tumour characteristics cervix, it is expected that first-void urine rather than midstream
(e.g., size, stage, grade) and varies among patients, with non- urine will contain more mRNA and miRNA biomarkers associated
invasive (tumour) lesions shedding less tumour cells compared to with cervical cancer development.
invasive lesions [77,78]. The same phenomenon is seen in cervical An additional path of growing interest is biomarker detection in
(pre)cancerous lesions, with better correlations in HPV DNA exosomes. These are small membrane vesicles (maximal 100 nm
presence between urine and cervical samples in more pronounced diameter) secreted by most cell types into the bloodstream, milk,
cervical lesions [57,79]. The limited amount of DNA in urine indeed and female and male urogenital tract body fluids, including
seems challenging upon detection of methylated DNA. Andersson cervicovaginal specimens and urine. Exosomes have the advantage
and colleagues optimized pre-analytic urine processing, demon- of being highly stable in bodily fluids, containing high concen-
strating that filtration of urine through an 8-mm polycarbonate trations of biological molecules (DNA, proteins/peptides, mRNA,
filter increases the amount of tumour cells and (potentially miRNA, and lipids) protected by their lipid bilayer membrane. In
methylated) DNA retained on the filter and, hence, increases the addition, their ability as protein and RNA biomarker sources has
sensitivity and robustness of the test [78]. For human/HPV DNA, been described for bladder, renal cell, and prostate cancer [87,88].
we have also shown that ultrafiltration of first-void urine increases
the amount of DNA detected in the sample [57]. In contrast, Protein biomarkers
Guerrero-Preston reported a protocol to extract circulating cell- Most of the prominent proteomic studies on cervical cancer
free (trans-renal) DNA from urine for the detection of host and viral have been performed using cell lines, plasma/serum, and cervical
methylated genes for cervical cancer diagnosis [62]. tissue samples. However, a low concordance between studies
exists, and to-date, none of the proposed protein biomarkers have
RNA-based biomarkers been sufficiently validated for clinical use. Elevated levels of E6/E7
In contrast to DNA-based methods, in which the absence or proteins represent an attractive, disease-specific viral biomarker,
presence of the HPV genome is detected, RNA-based assays are as they are the main mediators of HR-HPV-induced malignant
able to detect changes or differences in gene expression profiles transformation and, hence, high-grade CIN. A lateral flow-based
related to cancer development. Messenger RNA (mRNA) plays an assay for detecting E6 protein of HPV16, 18 and 45 has been
important role in protein synthesis, whereas its function is developed (OncoE6, Arbor Vita Corporation) and exhibits high
regulated by small non-coding microRNAs (miRNA) by controlling specificity for CIN3+ using clinician-collected cervical samples
mRNA stability and its translation into proteins. MiR-20a and miR- from HR-HPV-positive women (93.8% (95% CI: 92.1–95.2%)).
21 have been consistently upregulated, and miR-143, miR-03, and However the sensitivity is limited (54.2% (95% CI: 43.7–64.4%))
miR-145 consistently downregulated in studies but have not been [89] possibly because only three HPV genotypes are included. To
S. Van Keer et al. / European Journal of Obstetrics & Gynecology and Reproductive Biology 216 (2017) 1–11 9

the best of our knowledge, the assay has not been validated using infections. In contrast to microscopy-based biomarkers, molecu-
self-collected samples. lar-based biomarkers offer high-throughput capacity, reproducible
Urine is a suitable material for the identification of protein and objective results that are compatible with self-collected
biomarkers due its relatively stable composition of proteins and specimens. Nevertheless, for all biomarkers proposed, both
peptides and its non-invasive, easy method of collection in large retrospective and large population-based prospective studies are
sample volumes [88]. It contains approximately 2000 proteins required for validation in self-collected samples. A limited number
originating from different sites [88], i.e., glomerular ultrafiltration of studies have been reported, especially regarding self-collected
of plasma (systemic), excretion from epithelial cells in the urinary urine, and most available studies are still preclinical exploratory
tract, and shedding of exfoliated epithelial cells from the cervix and studies. However, by using proper sampling, transport, storage,
vagina (local) [17]. However, challenges exist regarding urine preanalytical biomarker concentration techniques, and clinically
processing, including the variability within and between urine validated assays, first-void urine sampling is expected to be a
samples of subjects, making it difficult to distinguish disease valuable method for including non-attending women in cervical
biomarkers from normal bio-physiological variations [90–93]. cancer screening programmes. Indeed, high analytical sensitivities
Furthermore, the typical low protein concentration (factor 1000 have been reported for primary HPV testing using first-void urine
less compared to, e.g., plasma) and the high concentrations of salts but, unfortunately, the clinical performance still needs to be
and other contaminants [88,94] demands additional purification determined. Moreover, the analysis of methylation of both host and
and concentration steps. Thus far, a standardized protocol for first- viral genes in urine has been proven feasible for use as a molecular
void urine collection and processing upon conservation of proteins cervical cancer triage and screening method of urine. This
is lacking. A tentative standard protocol has been described for emphasizes our theory that candidate biomarkers for cervical
urine collection and storage (HKUPP; Human Kidney & Urine cancer are washed away with first-void urine together with
Proteome Project; http://www.hkupp.org/), although it is not exfoliated cells, debris and impurities that line the urethra
based on first-void urine containing cervicovaginal material. opening.
Controversy exists regarding which urine fraction has the least In conclusion, in the near future, the use of self-sampling
variable protein concentrations among single sample collections methods that increase compliance with cervical cancer screening
(e.g., first/second urine sample of the day, random urine) [94,95]. and reduce loss to follow-up will likely increase. We believe that
Nevertheless, preanalytical preparation methods are expected to when appropriately sampled, stored, and processed, first-void
affect peptide abundances more than biological variations urine is a particularly valuable liquid biopsy source for primary
between pools [94]. For cervicovaginal specimen-derived proteins, HPV testing and for analysis of molecular biomarkers via a triage
a ratio of the biomarker of interest over total protein concentration algorithm to distinguish clinically relevant, transforming infec-
has been proposed for normalization [96]. Despite the proven tions from productive infections.
necessity of chelating agents to avoid cell-free DNA degradation by
nucleases, the need for protease inhibitors for the proper storage of
urine specimens remains uncertain. In contrast to blood, where Conflict of interest
proteases are activated upon collection, proteolytic degradation by
proteases in urine may be already completed by the time of voiding P. Van Damme and A. Vorsters are co-founders of a University of
because urine stagnates for hours in the bladder [90]. Studies have Antwerp spin-off company Novosanis (Belgium).
reported that the absence of protease inhibitors in properly
handled urine samples (stored at 80  C without repeated freeze- Acknowledgements
thaws) was not related to differences in the number or identity of
highly abundant proteins for a variable number of years [97,98]. This work was supported by the Industrial Research Fund of the
However, we are not interested in proteins excreted in urine but University of Antwerp (PS ID 32387). S. Van Keer is supported by a
rather proteins excreted in cervicovaginal material that enter the Ph. D. fellowship of the Research Foundation  Flanders (FWO)
first-void urine at the time of voiding. The addition of protease (1120816N). None of the sponsors were involved in the writing of
inhibitors will therefore likely be a requisite for optimal recovery of the manuscript. We would like to thank D. Glorie for the design of
the protein biomarkers of interest, especially when the sample is the illustration.
self-collected at home and transported to the laboratory at room
temperature. Urine preparation methods include, e.g., organic References
solvent precipitation, ultrafiltration, ultracentrifugation, dialysis
[1] GLOBOCAN. Cervical Cancer: Estimated Incidence, Mortality and Prevalence
and lyophilisation, solid phase extraction, and complete evapora- Worldwidein 2012 [Available from: http://globocan.iarc.fr/Pages/fact_-
tion [93,95,97–99], with organic solvent precipitation yielding the sheets_cancer.aspx.]
highest recovery rates. Depletion of high abundance proteins could [2] Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, et al.
Human papillomavirus is a necessary cause of invasive cervical cancer
also increase the concentration of low abundance protein
worldwide. J Pathol 1999;189(1):12–9.
biomarkers [88] but implicates the risk of removing biomarkers [3] Chesson HW, Dunne EF, Hariri S, Markowitz LE. The estimated lifetime
that are associated with the depleted proteins (e.g., albumin). probability of acquiring human papillomavirus in the United States. Sex
Finally, similar to other biomarkers, a panel instead of a single Transm Dis 2014;41(11):660–4.
[4] Burchell AN, Winer RL, de Sanjosé S, Franco EL. Chapter 6: epidemiology and
protein marker will likely increase the accuracy of the assay. Here, transmission dynamics of genital HPV infection. Vaccine 2006;24(Suppl. 3):
the use of multiplex immunoassays would decrease the required S52–61.
sample volume and increase the reproducibility in a less expensive, [5] Plummer M, Schiffman M, Castle PE, Maucort-Boulch D, Wheeler CM. Group
A: a 2-year prospective study of human papillomavirus persistence among
high-throughput format [100]. women with a cytological diagnosis of atypical squamous cells of
undetermined significance or low-grade squamous intraepithelial lesion. J
Conclusion Infect Dis 2007;195(11):1582–9.
[6] de Villiers EM. Cross-roads in the classification of papillomaviruses. Virology
2013;445(1–2):2–10.
There is growing interest in identifying biomarkers from non- [7] Kocjan BJ, Bzhalava D, Forslund O, Dillner J, Poljak M. Molecular methods for
invasive, self-collected samples, which might address both the identification and characterization of novel papillomaviruses. Clin Microbiol
Infec 2015;21(9):808–16.
non-adherence to screening issue and distinguishing clinically [8] IARC. Agents classified by the IARC Monographs. 2016.
relevant, transforming HR-HPV infections from productive
10 S. Van Keer et al. / European Journal of Obstetrics & Gynecology and Reproductive Biology 216 (2017) 1–11

[9] Ronco G, Dillner J, Elfstrom KM, Tunesi S, Snijders PJ, Arbyn M, et al. Efficacy of [36] Racey CS, Withrow DR, Gesink D. Self-collected HPV testing improves
HPV-based screening for prevention of invasive cervical cancer: follow-up of participation in cervical cancer screening: a systematic review and meta-
four European randomised controlled trials. Lancet 2014;383(9916):524–32. analysis. Canad J Publ Health 2013;104(2):e159–66.
[10] Arbyn M, Snijders PJ, Meijer CJ, Berkhof J, Cuschieri K, Kocjan BJ, et al. Which [37] Verdoodt F, Jentschke M, Hillemanns P, Racey CS, Snijders PJ, Arbyn M.
high-risk HPV assays fulfil criteria for use in primary cervical cancer Reaching women who do not participate in the regular cervical cancer
screening? Clin Microbiol Infect 2015;21(9):817–26. screening programme by offering self-sampling kits: a systematic review and
[11] von Karsa L, Arbyn M, De Vuyst H, Dillner J, Dillner L, Franceschi S, et al. meta-analysis of randomised trials. Eur J Cancer 2015;51(16):2375–85.
European guidelines for quality assurance in cervical cancer screening: [38] Ducancelle A, Reiser J, Pivert A, Le Guillou-Guillemette H, Le Duc-Banaszuk
summary of the supplements on HPV screening and vaccination. Papilloma- AS, Lunel-Fabiani F. Home-based urinary HPV DNA testing in women who do
virus Res 2015;1:22–31. not attend cervical cancer screening clinics. J Infect 2015;71(3):377–84.
[12] Arbyn M, Ronco G, Anttila A, Meijer CJ, Poljak M, Ogilvie G, et al. Evidence [39] Sellors JW, Lorincz AT, Mahony JB, Mielzynska I, Lytwyn A, Roth P, et al.
regarding human papillomavirus testing in secondary prevention of cervical Comparison of self-collected vaginal, vulvar and urine samples with
cancer. Vaccine 2012;30(Suppl. 5):F88–99. physician-collected cervical samples for human papillomavirus testing to
[13] Tjalma WA. There are two prophylactic human papillomavirus vaccines detect high-grade squamous intraepithelial lesions. CMAJ 2000;163(5):513–
against cancer, and they are different. J Clin Oncol 2015;33(8):964–5. 8.
[14] Tota JE, Ramana-Kumar AV, El-Khatib Z, Franco EL. The road ahead for cervical [40] Senkomago V, Des Marais AC, Rahangdale L, Vibat CR, Erlander MG, Smith JS.
cancer prevention and control. Curr Oncol 2014;21(2):E255–64. Comparison of urine specimen collection times and testing fractions for the
[15] Burroni E, Bonanni P, Sani C, Lastrucci V, Carozzi F, Hpv ScreeVacc Working G, detection of high-risk human papillomavirus and high-grade cervical
et al. Human papillomavirus prevalence in paired urine and cervical samples precancer. J Clin Virol 2016;74:26–31.
in women invited for cervical cancer screening. J Med Virol 2015;87(3):508– [41] Fey MC, Beal MW. Role of human papilloma virus testing in cervical cancer
15. prevention. J Midwif Women's Health 2004;49(1):4–13.
[16] Combita AL, Gheit T, Gonzalez P, Puerto D, Murillo RH, Montoya L, et al. [42] McCredie MR, Sharples KJ, Paul C, Baranyai J, Medley G, Jones RW, et al.
Comparison between urine and cervical samples for HPV DNA detection and Natural history of cervical neoplasia and risk of invasive cancer in women
typing in young women in Colombia. Cancer Prevent Res (Philadelphia, Pa) with cervical intraepithelial neoplasia 3: a retrospective cohort study. Lancet
2016;9(9):766–71. Oncol 2008;9(5):425–34.
[17] Ducancelle A, Legrand MC, Pivert A, Veillon P, Le Guillou-Guillemette H, De [43] Steenbergen RD, Snijders PJ, Heideman DA, Meijer CJ. Clinical implications of
Brux MA, et al. Interest of human papillomavirus DNA quantification and (epi)genetic changes in HPV-induced cervical precancerous lesions. Nat Rev
genotyping in paired cervical and urine samples to detect cervical lesions. Cancer 2014;14(6):395–405.
Arch Gynecol Obstet 2014;290(2):299–308. [44] FDA. Biomarker Qualification Program 2016 [updated 20/09/2016. Available
[18] Franceschi S, Chantal Umulisa M, Tshomo U, Gheit T, Baussano I, Tenet V, et al. from: http://www.fda.gov/Drugs/DevelopmentApprovalProcess/DrugDeve-
Urine testing to monitor the impact of HPV vaccination in Bhutan and lopmentToolsQualificationProgram/ucm284076.htm.
Rwanda. Int J Cancer 2016;139(3):518–26. [45] Pepe MS, Etzioni R, Feng Z, Potter JD, Thompson ML, Thornquist M, et al.
[19] Pathak N, Dodds J, Zamora J, Khan K. Accuracy of urinary human Phases of biomarker development for early detection of cancer. J Natl Cancer
papillomavirus testing for presence of cervical HPV: systematic review Inst 2001;93(14):1054–61.
and meta-analysis. Bmj 2014;349:g5264. [46] Arbyn M, Ronco G, Cuzick J, Wentzensen N, Castle PE. How to evaluate
[20] Vorsters A, Van Keer S, Biesmans S, Hens A, De Coster I, Goossens H, et al. emerging technologies in cervical cancer screening? Int J Cancer 2009;125
Long-term follow-up of HPV infection using urine and cervical quantitative (11):2489–96.
HPV DNA testing. Int J Mol Sci 201617(5). [47] Carreon JD, Sherman ME, Guillen D, Solomon D, Herrero R, Jeronimo J, et al.
[21] Enerly E, Olofsson C, Nygard M. Monitoring human papillomavirus CIN2 is a much less reproducible and less valid diagnosis than CIN3: results
prevalence in urine samples: a review. Clin Epidemiol 2013;5:67–79. from a histological review of population-based cervical samples. Int J
[22] Smith JS, Smith JB, Faherty BR, Isaacson SC, Des Marais AC, Fakner A, et al., Gynecol Pathol 2007;26(4):441–6.
editors. Effect of urine preservative on HPV testing results among urine [48] Dijkstra MG, van Niekerk D, Rijkaart DC, van Kemenade FJ, Heideman DA,
samples. HPV2017; 2017; Cape-Town, South-Africa. Snijders PJ, et al. Primary hrHPV DNA testing in cervical cancer screening:
[23] Payan C, Ducancelle A, Aboubaker MH, Caer J, Tapia M, Chauvin A, et al. how to manage screen-positive women? A POBASCAM trial substudy. Cancer
Human papillomavirus quantification in urine and cervical samples by using Epidemiol Biomark Prevent 2014;23(1):55–63.
the Mx4000 and LightCycler general real-time PCR systems. J Clin Microbiol [49] Rijkaart DC, Berkhof J, van Kemenade FJ, Coupe VM, Rozendaal L, Heideman
2007;45(3):897–901. DA, et al. HPV DNA testing in population-based cervical screening (VUSA-
[24] Sahasrabuddhe VV, Gravitt PE, Dunn ST, Brown D, Allen RA, Eby YJ, et al. Screen study): results and implications. Br J Cancer 2012;106(5):975–81.
Comparison of human papillomavirus detections in urine, vulvar, and [50] Koeneman MM, Kruitwagen RF, Nijman HW, Slangen BF, Van Gorp T, Kruse AJ.
cervical samples from women attending a colposcopy clinic. J Clin Microbiol Natural history of high-grade cervical intraepithelial neoplasia: a review of
2014;52(1):187–92. prognostic biomarkers. Expert Rev Mol Diagn 2015;15(4):527–46.
[25] Hagihara M, Yamagishi Y, Izumi K, Miyazaki N, Suzuki T, Kato H, et al. [51] Ebisch RM, Siebers AG, Bosgraaf RP, Massuger LF, Bekkers RL, Melchers WJ.
Comparison of initial stream urine samples and cervical samples for Triage of high-risk HPV positive women in cervical cancer screening. Expert
detection of human papillomavirus. J Infect Chemother 2016;22(8):559–62. Rev Anticancer Ther 2016;1–13.
[26] Vorsters A, Micalessi I, Bilcke J, Ieven M, Bogers J, Van Damme P. Detection of [52] Luttmer R, De Strooper LM, Steenbergen RD, Berkhof J, Snijders PJ, Heideman
human papillomavirus DNA in urine: a review of the literature. Eur J Clin DA, et al. Management of high-risk HPV-positive women for detection of
Microbiol Infect Dis 2012;31(5):627–40. cervical (pre)cancer. Expert Rev Mol Diagn 2016;16(9):961–74.
[27] Papanicolaou GN, Marchetti AA. The use of endocervical and endometrial [53] Fuzery AK, Levin J, Chan MM, Chan DW. Translation of proteomic biomarkers
smears in the diagnosis of cancer and of other conditions of the uterus. Am J into FDA approved cancer diagnostics: issues and challenges. Clinical
Obst Gynecol 1943;46(3):421–2. Proteom 2013;10(1):13.
[28] Vorsters A, Van Damme P, Clifford G. Urine testing for HPV: rationale for using [54] Wentzensen N, Silver MI. Biomarkers for cervical cancer prevention
first void. Bmj 2014;349:g6252. programs: the long and winding road from discovery to clinical use. J Low
[29] Arbyn M, Verdoodt F, Snijders PJ, Verhoef VM, Suonio E, Dillner L, et al. Genit Tract Dis 2016;20(3):191–4.
Accuracy of human papillomavirus testing on self-collected versus clinician- [55] Boulet GA, Benoy IH, Depuydt CE, Horvath CA, Aerts M, Hens N, et al. Human
collected samples: a meta-analysis. Lancet Oncol 2014;15(2):172–83. papillomavirus 16 load and E2/E6 ratio in HPV16-positive women:
[30] Landy R, Pesola F, Castanon A, Sasieni P. Impact of cervical screening on biomarkers for cervical intraepithelial neoplasia >or=2 in a liquid-based
cervical cancer mortality: estimation using stage-specific results from a cytology setting? Cancer Epidemiol Biomark Prevent 2009;18(11):2992–9.
nested case-control study. Br J Cancer 2016;115(9):1140–6. [56] Depuydt CE, Jonckheere J, Berth M, Salembier GM, Vereecken AJ, Bogers JJ.
[31] Stanczuk G, Baxter G, Currie H, Lawrence J, Cuschieri K, Wilson A, et al. Serial type-specific human papillomavirus (HPV) load measurement allows
Clinical validation of hrHPV testing on vaginal and urine self-samples in differentiation between regressing cervical lesions and serial virion
primary cervical screening (cross-sectional results from the Papillomavirus productive transient infections. Cancer Med 2015;4(8):1294–302.
Dumfries and Galloway-PaVDaG study). Bmj Open 2016;6(4):e010660. [57] Vorsters A, Van den Bergh J, Micalessi I, Biesmans S, Bogers J, Hens A, et al.
[32] Everett T, Bryant A, Griffin MF, Martin-Hirsch PP, Forbes CA, Jepson RG. Optimization of HPV DNA detection in urine by improving collection, storage,
Interventions targeted at women to encourage the uptake of cervical and extraction. Eur J Clin Microbiol Infect Dis 2014;33(11):2005–14.
screening. Cochrane Database Syst Rev 2016;5:Cd002834. [58] Lee SH, Yang YJ, Kim KM, Chung BC. Altered urinary profiles of polyamines
[33] Waller J, Bartoszek M, Marlow L, Wardle J. Barriers to cervical cancer and endogenous steroids in patients with benign cervical disease and
screening attendance in England: a population-based survey. J Med Screen cervical cancer. Cancer Lett 2003;201(2):121–31.
2009;16(4):199–204. [59] Garbett NC, Merchant ML, Helm CW, Jenson AB, Klein JB, Chaires JB. Detection
[34] Arrossi S, Ramos S, Straw C, Thouyaret L, Orellana L. HPV testing: a mixed- of cervical cancer biomarker patterns in blood plasma and urine by
method approach to understand why women prefer self-collection in a differential scanning calorimetry and mass spectrometry. Plos One 20149(1).
middle-income country. BMC Public Health 2016;16:832. [60] Rurup R, van Trommel N, van Splunter A, Segerink L, Kenter G, Heideman D,
[35] Tamalet C, Le Retraite L, Leandri FX, Heid P, Sancho Garnier H, Piana L. Vaginal et al. Detection of DNA methylation markers in urine of cervical cancer
self-sampling is an adequate means of screening HR-HPV types in women not patients: a feasibility study. Int J Gynecol Cancer 2016;(October):170.
participating in regular cervical cancer screening. Clin Microbiol Infect [61] Feng QH, Hawes SE, Stern JE, Dem A, Sow PS, Dembele B, et al. Promoter
2013;19(1):E44–50. hypermethylation of tumor suppressor genes in urine from patients with
cervical neoplasia. Cancer Epidem Biomar 2007;16(6):1178–84.
S. Van Keer et al. / European Journal of Obstetrics & Gynecology and Reproductive Biology 216 (2017) 1–11 11

[62] Guerrero-Preston R, Valle BL, Jedlicka A, Turaga N, Folawiyo O, Pirini F, et al. [79] Sehgal A, Gupta S, Parashari A, Sodhani P, Singh V. Urine HPV-DNA detection
Molecular triage of premalignant lesions in liquid-based cervical cytology for cervical cancer screening: prospects and prejudices. J Obstet Gynaecol
and circulating cell free DNA from urine, using methylated viral and host 2009;29(7):583–9.
genes. Cancer Prevent Res (Philadelphia Pa) 2016;2016. [80] Cuschieri K, Wentzensen N. Human papillomavirus mRNA and p16 detection
[63] Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for as biomarkers for the improved diagnosis of cervical neoplasia. Cancer
systematic reviews and meta-analyses: the PRISMA statement. PLoS Med Epidemiol Biomark Prevent 2008;17(10):2536–45.
2009;6(7):e1000097. [81] O'Driscoll L. Extracellular nucleic acids and their potential as diagnostic,
[64] Thomas LK, Bermejo JL, Vinokurova S, Jensen K, Bierkens M, Steenbergen R, prognostic and predictive biomarkers. Anticancer Res 2007;27(3a):1257–65.
et al. Chromosomal gains and losses in human papillomavirus-associated [82] Mall C, Rocke DM, Durbin-Johnson B, Weiss RH. Stability of miRNA in human
neoplasia of the lower genital tract  a systematic review and meta-analysis. urine supports its biomarker potential. Biomark Med 2013;7(4):623–31.
Eur J Cancer 2014;50(1):85–98. [83] Hessels D, Schalken JA. The use of PCA3 in the diagnosis of prostate cancer.
[65] Advenier AS, Casalegno JS, Mekki Y, Decaussin-Petrucci M, Mege-Lechevallier Nat Rev Urol 2009;6(5):255–61.
F, Ruffion A, et al. Genotyping of high-risk human papillomaviruses in p16/Ki- [84] Urquidi V, Netherton M, Gomes-Giacoia E, Serie D, Eckel-Passow J, Rosser CJ,
67-positive urothelial carcinoma cells: even a worm will turn. Cytopathology et al. Urinary mRNA biomarker panel for the detection of urothelial
2015;26(2):106–13. carcinoma. Oncotarget 2016;7(25):38731–40.
[66] Piaton E, Casalegno JS, Advenier AS, Decaussin-Petrucci M, Mege-Lechevallier [85] van de Vrie M, Deegens JK, Eikmans M, van der Vlag J, Hilbrands LB. Urinary
F, Ruffion A, et al. p16(INK4a) overexpression is not linked to oncogenic microRNA as biomarker in renal transplantation. Am J Transplant 2016;17(5)
human papillomaviruses in patients with high-Grade urothelial cancer cells. 1160–6 Nov 10.
Cancer Cytopathol 2014;122(10):760–9. [86] Lamb LE, Bartolone SN, Kutluay SB, Robledo D, Porras A, Plata M, et al.
[67] van der Aa MN, Steyerberg EW, Sen EF, Zwarthoff EC, Kirkels WJ, van der Advantage of urine based molecular diagnosis of Zika virus. Int Urol Nephrol
Kwast TH, et al. Patients' perceived burden of cystoscopic and urinary 2016;48(12):1961–6.
surveillance of bladder cancer: a randomized comparison. BJU Int 2008;101 [87] Foster BP, Balassa T, Benen TD, Dominovic M, Elmadjian GK, Florova V, et al.
(9):1106–10. Extracellular vesicles in blood, milk and body fluids of the female and male
[68] Leeman A, Del Pino M, Molijn A, Rodriguez A, Torne A, de Koning M, et al. HPV urogenital tract and with special regard to reproduction. Crit Rev Clin Lab Sci
testing in first-void urine provides sensitivity for CIN2+ detection compara- 2016;53(6):379–95.
ble to a physician-taken smear or brush-based self-sample: cross-sectional [88] Kalantari S, Jafari A, Moradpoor R, Ghasemi E, Khalkhal E. Human urine
data from a triage population. BJOG 2016;2017. proteomics: analytical techniques and clinical applications in renal diseases.
[69] Tshomo U, Franceschi S, Tshokey T, Tobgay T, Baussano I, Tenet V, et al. Int J Proteom 2015;2015:782798.
Evaluation of the performance of Human Papillomavirus testing in paired [89] Qiao YL, Jeronimo J, Zhao FH, Schweizer J, Chen W, Valdez M, et al. Lower cost
urine and clinician-collected cervical samples among women aged over 30 strategies for triage of human papillomavirus DNA-positive women. Int J
years in Bhutan. Virol J 2017;14(1):74. Cancer 2014;134(12):2891–901.
[70] Clifford GM, Vaccarella S, Franceschi S, Tenet V, Umulisa MC, Tshomo U, et al. [90] Decramer S, Gonzalez de Peredo A, Breuil B, Mischak H, Monsarrat B,
Comparison of two widely used human papillomavirus detection and Bascands JL, et al. Urine in clinical proteomics. Mol Cell Proteomics 2008;7
genotyping methods, GP5+/6+-Based PCR followed by reverse line blot (10):1850–62.
hybridization and multiplex type-Specific E7-Based PCR. J Clin Microbiol [91] Zegels G, Van Raemdonck GA, Tjalma WA, Van Ostade XW. Use of
2016;54(8):2031–8. cervicovaginal fluid for the identification of biomarkers for pathologies of
[71] Pattyn J, Dekoeijer M, Van Keer S, Biesmans S, Ieven M, Beyers K, et al. editors. the female genital tract. Proteome Sci 2010;8:63.
HPV DNA detection in urine: effect of a first-void urine collection device and [92] Crosley LK, Duthie SJ, Polley AC, Bouwman FG, Heim C, Mulholland F, et al.
timing of collection. HPV2017. 2017. Cape-Town, South-Africa. Variation in protein levels obtained from human blood cells and biofluids for
[72] De Strooper LM, Verhoef VM, Berkhof J, Hesselink AT, de Bruin HM, van platelet, peripheral blood mononuclear cell, plasma, urine and saliva
Kemenade FJ, et al. Validation of the FAM19A4/mir124-2 DNA methylation proteomics. Genes Nutr 2009;4(2):95–102.
test for both lavage- and brush-based self-samples to detect cervical (pre) [93] Adachi J, Kumar C, Zhang Y, Olsen JV, Mann M. The human urinary proteome
cancer in HPV-positive women. Gynecol Oncol 2016;141(2):341–7. contains more than 1500 proteins, including a large proportion of membrane
[73] Lorincz AT. Cancer diagnostic classifiers based on quantitative DNA proteins. Genome Biol 2006;7(9):R80.
methylation. Expert Rev Mol Diagn 2014;14(3):293–305. [94] Court M, Selevsek N, Matondo M, Allory Y, Garin J, Masselon CD, et al. Toward
[74] Clarke MA, Wentzensen N, Mirabello L, Ghosh A, Wacholder S, Harari A, et al. a standardized urine proteome analysis methodology. Proteomics 2011;11
Human papillomavirus DNA methylation as a potential biomarker for (6):1160–71.
cervical cancer. Cancer Epidem Biomar 2012;21(12):2125–37. [95] Thomas CE, Sexton W, Benson K, Sutphen R, Koomen J. Urine collection and
[75] Snellenberg S, De Strooper LMA, Hesselink AT, Meijer CJLM, Snijders PJF, processing for protein biomarker discovery and quantification. Cancer
Heideman DAM, et al. Development of a multiplex methylation-specific PCR Epidemiol Biomark Prevent 2010;19(4):953–9.
as candidate triage test for women with an HPV-positive cervical scrape. BMC [96] Burgener A, Boutilier J, Wachihi C, Kimani J, Carpenter M, Westmacott G, et al.
Cancer 2012;2012. Identification of differentially expressed proteins in the cervical mucosa of
[76] Renard I, Joniau S, van Cleynenbreugel B, Collette C, Naome C, Vlassenbroeck HIV-1-resistant sex workers. J Proteome Res 2008;7(10):4446–54.
I, et al. Identification and validation of the methylated TWIST1 and NID2 [97] Afkarian M, Bhasin M, Dillon ST, Guerrero MC, Nelson RG, Knowler WC, et al.
genes through real-time methylation-specific polymerase chain reaction Optimizing a proteomics platform for urine biomarker discovery. Mol Cell
assays for the noninvasive detection of primary bladder cancer in urine Proteom 2010;9(10):2195–204.
samples. Eur Urol 2010;58(1):96–104. [98] Voss J, Goo YA, Cain K, Woods N, Jarrett M, Smith L, et al. Searching for the
[77] Roperch JP, Grandchamp B, Desgrandchamps F, Mongiat-Artus P, Ravery V, noninvasive biomarker holy grail: are urine proteomics the answer? Biol Res
Ouzaid I, et al. Promoter hypermethylation of HS3ST2, SEPTIN9 and SLIT2 Nurs 2011;13(3):235–42.
combined with FGFR3 mutations as a sensitive/specific urinary assay for [99] Van Raemdonck GA, Tjalma WA, Coen EP, Depuydt CE, Van Ostade XW.
diagnosis and surveillance in patients with low or high-risk non-muscle- Identification of protein biomarkers for cervical cancer using human
invasive bladder cancer. BMC Cancer 2016;16:704. cervicovaginal fluid. Plos One 2014;9(9):e106488.
[78] Andersson E, Steven K, Guldberg P. Size-based enrichment of exfoliated [100] Khan A. Detection and quantitation of forty eight cytokines, chemokines,
tumor cells in urine increases the sensitivity for DNA-based detection of growth factors and nine acute phase proteins in healthy human plasma,
bladder cancer. Plos One 2014;9(4):e94023. saliva and urine. J Proteom 2012;75(15):4802–19.

You might also like