You are on page 1of 14

doi:10.1111/jog.14168 J. Obstet. Gynaecol. Res. Vol. 46, No.

2: 201–214, February 2020

Screening and management of preinvasive lesions of the


cervix: Good clinical practice recommendations from the
Federation of Obstetrics and Gynaecologic Societies of India
(FOGSI)

1 1 2 3 4
Neerja Bhatla , Seema Singhal , Usha Saraiya , Shikha Srivastava , Sarita Bhalerao , Saritha
5 6 7 8
Shamsunder , Niranjan Chavan , Partha Basu , and CN Purandare , (on behalf of FOGSI Expert
group)
1
All India Institute of Medical Sciences, 5Vardhmaan Mahaveer Medical College & Safdarjung Hospital, New Delhi, 2Breach
Candy, Saifee, Elizabeth & Cumballa Hill Hospitals, 4Reliance HNH, Saifee and Bhatia Hospitals, 6LTMMC & LTMGH, Sion,
8
Saifee Hospital, Mumbai, 3Population Services International (PSI), Delhi, India and 7Early Detection and Prevention Section
(EDP)/Screening Group (SCR) International Agency for Research on Cancer, World Health Organization, Lyon, France

Abstract
In India, there are marked variations in resources for cervical cancer screening. For the first time, resource-stratified
screening guidelines have been developed that will be suitable for low middle-income countries with similar diversities.
The current article describes the process and outcomes of these resource stratified guidelines for screening and treatment
of preinvasive lesions of cervix. Evidence from literature was collated and various guidelines were reviewed by an
expert panel. Based on the level of evidence, guidelines were developed for screening by human papillomavirus (HPV)
testing, cytology and visual inspection after appli-cation of acetic acid (VIA), and management of screen positive
lesions in different resource settings. Expert opinion was used for certain country-specific situations. The healthcare
system was stratified into two resource settings – good or limited. The mode of screening and treatment for each was
described. HPV test-ing is the preferred method for cervical cancer screening. VIA by trained providers is especially
suitable for low resource settings until an affordable HPV test becomes available. Healthcare providers can choose the
most appropriate screening and treatment modality. A single visit approach is encouraged and treatment may be offered
based on colposcopy diagnosis (‘see and treat’) or even on the basis of HPV test or VIA results (‘screen and treat’), if
compliance cannot be ensured. The Federation of Obsterician and Gyn-aecologists of India Good Clinical Practice
Recommendations (FOGSI) GCPR are appropriately designed for countries with varied resource situations to ensure an
acceptable cervical cancer prevention strategy.

Key words: cervical cancer screening, good clinical practice, prevention, recommendations, resource-based, single
visit approach.

Received: June 14 2019.


Accepted: November 13 2019.
Correspondence: Dr Neerja Bhatla, Department of Obstetrics & Gynaecology, Room no 3081 Teaching Block, All India Institute of
Medical Sciences, New Delhi, India. Email: neerja.bhatla07@gmail.com
Members of FOGSI Expert Group: Agarwal P, Arora M, Bhatla N, Basu P, Bhalerao S, Batra S, Banerjee D, Chavan N, Dhorepatil B,
Gupta B, Gandhi G, Gupta K, Ganesh P, Joshi S, Kriplani A, Khanna R, Kumar S, Maheshwari A, Meena J, Nayak B, Natarajan J,
Pai R, Pathak R, Prabhu RB, Peedicayil A, Rajaram S, Sekhon R, Shamsunder S, Srivastava S, Singh U, Singhal S, Saraiya U, Zutshi V.

© 2019 Japan Society of Obstetrics and Gynecology 201


N. Bhatla et al.
Introduction
Cervical cancer is a major public health problem, being the
1
second most common cancer among Indian women. India
contributes to one quarter of the global burden with 96 922
1
incident cases and 60 078 deaths in 2018. The age-
standardized incidence and mortal-ity rates in India (ASR) distributed among the experts who reviewed the liter-ature
are 14.7/100 000 and 9.2/100 000 women, respectively.
1 with reference to study designs and participants,
intervention, comparisons, outcomes (PICOS). Studies
There are regional variations in ASR, from 24.3/100 000 in
2 were evaluated for quality, and strength was assigned to
Aizawl to 5.6/100 000 in Dibrugarh district. The 5-year 7
relative survival rate for cancer cervix in India continues to each practice recommendation. Previously publi-shed
be low, approximately 46% (range 34–60%), predomi- guidelines by major international organizations as well as
3 guidelines appropriate for resource-constrained situations
nantly because of late detection.
by the World Health Organiza-tion 2014, Government of
In developed countries, establishment of national India 2016 and the American
screening programs with population-based invitation of Society of Clinical Oncology (ASCO) 2016 were ana-lyzed
6,8,9
eligible women and a recall system for screen posi-tives for their applicability in the Indian context. A
4 synopsis was prepared and circulated in the group.
resulted in prevention of almost 70% of cervical cancer.
India is a land of diversity with enormous variations in Thereafter, in a face-to-face meeting, the document with
sociocultural practices and healthcare infrastructure.
5 various algorithms was discussed and subse-quent
Shortage of trained manpower and infrastructure has comments were circulated until unanimous agreement was
limited the establishment of effec-tive, standardized, reached. The draft recommendations were posted online on
cytology-based screening program, which is currently used the Federation of Obsterician
only for opportunistic screen-ing. Ministry of Health and and Gynaecologists of India (FOGSI) website for pub-lic
10,11
Family Welfare (MoHFW), Govt. of India has formulated comments. The final consensus document was
operational framework for screening of cervical cancer presented and was then approved at the FOGSI Man-aging
based on visual inspection of cervix after application of Committee Meeting in January 2018.
6
acetic acid (VIA) in 2016. However, in urban settings the
option of high-risk human papillomavirus (HPV) test-ing is
now available in many cities. Results and Discussion
Resource stratification
In the absence of national guidelines, healthcare
providers have been following different international Initially the expert panel planned to stratify the healthcare
recommendations, which were limited in their appli- system into four groups in accordance with ASCO resource
9
cability to the Indian setting. Therefore, a need was felt for stratified clinical practice guidelines, but later for easy
an expert group to develop evidence-based understanding and usability, resource stratification was
recommendations that are suitable for diverse resource simplified into two strata as good and limited resource
situations. The current article describes the process and settings. Settings with-out lack of facilities including
outcomes of resource stratified guidelines for screening and trained doctors, labora-tories and equipment are considered
treatment of preinvasive lesions of cervix. Though good resource settings. Settings that lack any of these are
developed in India they are also suit-able for low and termed limited resource settings.
middle income countries (LMICs) with similar diversities.
The suggested modalities for screening and triage in each
setting are shown in Table 1. Considering the var-iations in
cervical cancer screening practices, expert group agreed to
suggest preferred and acceptable modalities for follow-up
Methods and treatment. Single visit approach (SVA), in which
A panel of gynecologists, gynecologic oncologists and screening and treatment can be completed at the same
public health experts with experience in cervical cancer sitting, was identified as the preferred modality for all
prevention from all regions of the country was consti-tuted. resource settings, especially when patient is likely to be lost
Relevant questions were formulated and to follow-up.

Choosing an appropriate screening modality


The three main modalities of screening in use are HPV
testing, cytology and visual inspection with

202 © 2019 Japan Society of Obstetrics and Gynecology


Cervical Cancer Screening & Management of CIN

Table 1 Resource-based strategies for cervical cancer screening and management of CIN
Good Resource settings Limited Resource settings
Screening modality HPV testing VIA (visual inspection of cervix with acetic
•Primary HPV testing acid)
•Co-testing (HPV and cytology) Affordable HPV test (if available),
Cytology including self-sampling, can be used
VIA
Triage tool For ASCUS cytology: Colposcopy if available

High-risk HPV test Biopsy
For HPV test:

Cytology newer modalities
Or HPV Genotyping for types 16/18

Or colposcopy and biopsy

Or VIA and biopsy
Management options All CIN2 and CIN3 can be treated by LEEP. All grades of CIN fulfilling the criteria for
§
for histopathology CKC can be performed for AIS or if early ablation should be treated by cryotherapy
proved CIN invasive cancer is suspected. or thermal ablation.
CIN1 can be followed up and be treated All grades of CIN not fulfilling the criteria
if it progresses or persists after should be treated by LEEP.
2 years. CKC can be performed for AIS or if early
Cryotherapy or thermal ablation can be invasive cancer is suspected.
performed for all grades of CIN as an
alternative to LEEP, provided the
§
criteria for ablation are fulfilled.
Approaches to reduce the ‘See-and-Treat’ strategy is preferred to treat ‘See-and-Treat’ strategy should be followed,
number of visits women with colposcopically suspected if colposcopy facilities are available.
high-grade lesions. Treatment with If colposcopy is not available, VIA or HPV
LLETZ/LEEP or ablation depending on positive women may be offered imme-
suitability of lesion for ablative treatment. diate treatment (‘Screen-and-Treat’). All
Histopathology report should be reviewed cases should initially be assessed for
at follow-up. suitability of ablative treatment
(cryotherapy or thermal ablation).
Eligibility of HPV positive women to be
assessed after applying 5% acetic acid
(VIA). A punch biopsy should be
obtained prior to ablative treatment and
the report reviewed at follow-up.
LLETZ/LEEP, large loop excision of transformation zone/loop electrosurgical excision procedure; CKC, cold knife colonization; CIN, cer-vical intra
epithelial lesion.; †Newer modalities (p16, Ki 67 testing, mRNA testing, E6, E7 protein testing).; ‡Biopsy from any abnormal area(s). and §Criteria for
ablation: (i) Lesion should be entirely visible and not occupy more than two quadrants of cervix, (ii) The entire lesion should be located on ecto cervix
without any vaginal or endo-cervical extension, (iii) Lesion should be entirely covered by largest cryotherapy probe available, (iv) No suspicion of
invasive disease, (v) Contraindicated in cases with post coital or postmenopausal bleed-ing, obvious cervical growth, irregular surface or bleeds on
touch.

acetic acid (VIA). Figure 1 describes the algorithm to is expensive and not widely available. It is rec-ommended
choose the most appropriate screening modality. Vali-dated for women aged over 30 years using vali-dated tests.
primary HPV screening, the most effective method, was Validated tests are those that have demonstrated efficacy in
recommended for screening in good resource settings. randomized controlled trials or have shown test accuracy
However, the centers with an established cytology program comparable to a vali-dated test. The results of such tests are
with good quality con-trol may continue with cytology. In generally reported as detected/not detected for a pool of 13
limited resource settings one may screen with VIA. or 14 high-risk HPV genotypes; some also report indi-
vidual genotypes for the most oncogenic types (HPV
There is ample evidence that HPV testing is the most 16/18). Example of validated tests are Hybrid Cap-ture 2,
sensitive modality, suitable for screening in all resource careHPV, Cobas, Xpert, Cervista, APTIMA etc. HPV tests
12,13
settings. In an Indian study, even a single round of based on routine polymerase chain reaction (PCR)
HPV testing was shown to significantly reduce the detecting only a few genotypes should not be
14
incidence of cervical cancer. However, it

© 2019 Japan Society of Obstetrics and Gynecology 203


N. Bhatla et al.

Figure 1 : How to choose the most appropriate screening modality?

advised as they are of high analytical sensitivity and a 16


and high false positivity. Currently it is the only test that
positive result has little clinical relevance. Affording can be used in low resource settings with considerable
centers/individuals, can consider co-testing with both accuracy and safety but in future, the availability of
cytology and HPV test or switch over to primary HPV affordable HPV testing may change this paradigm.
15
testing. The incremental benefit of co-testing over HPV
test alone is, however, small. Whenever possible, colposcopy should be used to obtain
Cytology facilities are available in most cities and a guided biopsy; if not available, biopsy can be guided by
tertiary hospitals, but cytology has an inherent fal- VIA too. It is advisable to link screening with treatment by
lacy of low sensitivity (60–70%), which therefore minimizing the number of visits to the clinics, especially
13,14,16 when there is a risk of loss to follow up. When screening
necessitates frequent rounds of testing. Its
major advantage lies in its high specificity (93.5%). Liquid and treatment can be com-pleted in the same sitting, the
based cytology (LBC) does not improve the sensitivity of strategy is known as the single visit approach (SVA).
conventional cytology, though it does decrease the rates of Biopsy can be reviewed post-hoc for adequacy of treatment
unsatisfactory smears and also allows the same sample to and to rule out invasive cancer. Women with high-grade
17
be used for HPV test-ing. Centers with established abnormalities on cytology (ASC-H or HSIL) and an
cytology programs should assess quality indicators abnormal colposcopy can be treated without waiting for
18
including diagnostic accuracy, training, coverage, etc. If histopathological verification. Such an approach is known
these are found adequate they may continue with the same. as ‘See-and-Treat’. It is also applicable to women who are
If not, they should consider switching to primary HPV HPV or VIA positive and are referred for colposcopy. In
screening or even substitute or supplement with VIA. VIA low resource settings where colposcopy facilities are not
has sensitivity comparable to cytology but poorer available, treat-ment by cryotherapy or thermal ablation
16
specificity. It can be used in all resource settings by all based on abnormal HPV test or VIA is also admissible and
levels of healthcare providers. How-ever, a larger number is known as a ‘Screen-and-Treat’ strategy. This will ensure
of women will require further triage by colposcopy because that women with cervical intraepithelial
of its low specificity

204 © 2019 Japan Society of Obstetrics and Gynecology


Cervical Cancer Screening & Management of CIN

neoplasia (CIN) lesions that fulfill the criteria do not remain feasible, it should be targeted at women between the age of
14
untreated. With this there is an acceptable risk of over- 35–40 years.
treatment and the referral is minimized to only those with The upper age limit is decided based on life expec-tancy.
large, suspicious or endocervical lesions. If screening was continued till 90 years, the estimated
chance of preventing cervical cancer was only 1.6 cases per
20
1000 women with substantial bur-den of colposcopies.
Age of screening Therefore, women over 65 years with previously adequate
To be most cost-effective, the age of starting and stop-ping negative screening and no history of CIN2+ within the last
20,21
screening is determined by the age-specific can-cer burden. 20 years need not be screened further. Adequate
In India, the burden of cervical cancer under the age of 30 negative prior screening is defined as three consecutive
years is very low and it is negligi-ble under the age of 25 negative cytology or two consecutive negative co-tests
19
years. According to Globocan 2018, the age standardized within
incidence rate below 29 years is 0.46/100 000 women and 10 years, with the last test having taken place within the
20,21
below 24 years, is as low as 0.06 per 100 000 women.
1 last 5 years. In women who have undergone
There-fore, screening should not be initiated before 25 hysterectomy with a report of CIN2+ lesions, screen-ing
years in asymptomatic women, regardless of the age of ini- should be continued for 20 years from the age of surgery.
tiation of sexual activity/resources. In limited resource Women who underwent hysterectomy for any other benign
settings, screening is recommended after the age of 30 disease need not continue screening.
years. If only one round of screening is The screening age and periodicity recommenda-tions for
each resource setting are depicted in Table 2.

Table 2 Resource-based cervical cancer screening recommendations


Good resource settings Limited resource settings
Target age group (years) 25–65 years 30–65 years (NB: In post-menopausal
women, screening with VIA may not
be as effective)
Age to start (years) Cytology at 25 years
Primary HPV testing/co-testing VIA at 30 years
at 30 years
Frequency Primary HPV testing or co-testing – Every 5 years (at least 1–3 times in a
every 5 years lifetime)
Cytology alone – every 3 years
Age to stop (years) 65 years, with consistent negative results in last 15 years
Women with no prior screening should undergo tests once at 65 years and, if
negative, they should exit screening
Follow up after treatment (method HPV testing (preferred) or cytology or VIA, 12 months
and interval) colposcopy, 12 months
Screening following abnormal
reports ≥ CIN 2+, irrespective of 20 years
method of treatment
Screening in hysterectomized
women Following hysterectomy in which cervix was removed for benign causes: no need for
screening, unless there is history of previous cervical intraepithelial neoplasia.

Absence of cervix must be confirmed by clinical records or examination. If


indications for hysterectomy unclear, screening may be performed at
Follow up in women with CIN in clinician’s discretion.
hysterectomy HPE report Need to be screened with HPV at 6 months and 18 months.
Screening of immunocompromised
women Start within 1 year of initiation of Start within 1 year of initiation of
sexual activity or as soon as HIV is sexual activity or as soon as HIV is
detected in sexually active women detected in sexually active women
HPV testing/co-testing/cytology/ VIA/affordable HPV testing
VIA (if available)
Every 2–3 years Every 3 years (at least twice as often
as general population)

© 2019 Japan Society of Obstetrics and Gynecology 205


N. Bhatla et al.
VIA every 5 years, starting from 30 upto 65 years is the
recommended screening modality by Government of
6
India. The accuracy of VIA in postmenopausal women
24,25
decreases as the transformation zone (TZ) recedes into the countries including Australia and some parts of Europe.
endocervical canal. Other resource-based guidelines have In an RCT the detection rates of high-
recommended screen-ing at least one to three times till the grade precancer/cancer (CIN2+) lesions in the initial rounds
8,9 were higher in the HPV arm than cytology but were either
age of 50 years. In a randomized trial in India, a single
round of VIA based screening led to 30% reduction in same or lower in the subsequent rounds of screening with
cervical cancer incidence and a 42% reduction in cervical greater sensitivity (ratio = 1.29, 95% CI, 1.18–1.39) and
26
cancer mortality. The maximum reduction in the incidence lower specificity (ratio = 0.94, 95% CI 0.92–0.96).
of cervical cancer was seen among women aged 30–39 Another study observed the cumulative incidence rates of
21
years. Another study demon-strated 25–31% reduction in CIN3 and cancer in HPV negative women aged >25 years
the lifetime risk of invasive cervical cancer with VIA, 30– as low as 0.34% (95% CI, 0.10–0.65), in cytology-negative
36% reduction with HPV DNA testing, carried out once at women 0.78%(95% CI, 0.53–1.09) and least in co-test
the age of 35 years. Two rounds of screening done at the 27
negative women 0.30% (95% CI, 0.05–0.62). How-ever,
22
age of 35 and 40 years reduced lifetime risk by 40%. this small decrease in cancer risk associated with co-testing
may not be affordable in developing coun-tries with
important competing health priorities. HPV test also
provides better reassurance that high-grade lesion is
Screening paradigms currently absent and thus provides an oppor-tunity to safely
The efficacy of the cervical cancer screening modali-ties; extend the screening intervals to 5 years or even 10 years.
co- testing, primary HPV testing, cytology and VIA are Compared to Pap test, HPV test misses fewer
28
investigated in large clinical trials and based on the adenocarcinomas or adenocarci-nomas in situ.
generated evidence, clinical practice points are formulated
as shown in Table 3.
The specificity and positive predictive value of HPV test
Co-testing with HPV testing and cytology are low. To reduce referral to colposcopy, HPV positive
The combination of a highly sensitive validated HPV test women may be triaged with another test (cytology or VIA
with highly specific cytology every 5 years is the most or HPV 16/18 genotyping), and triage positive cases may
effective and preferred screening modality in India. Co- 29
then be referred to colpos-copy. The risk of CIN3+ in
testing can detect 51% more cases of CIN2/3 or invasive HPV 16/18 positive cases is 21.1% while it is 5.4% with
23 27
cancer than cytology alone. Schiffman et al. investigated other non-16/18 strains. Hence HPV 16,18 positive
the relative performance of each component and found that women may be immediately referred to colposcopy and
the first co-test could detect 67.9% cases likely to progress those with other strains may have cytology or VIA. In
12
in next 10 years. Only a small fraction, i.e., 3.5% cases of good resource settings, women should be screened every 5
preinvasive and 5.9% cases of invasive disease were years with an approved HPV test or co-test. FOGSI Good
detected in women who were HPV−/cytology+ results and Clinical Practice Recommendations (GCPR) rec-ommends
15
were actually benefited by adding cytol-ogy to HPV test. VIA as a feasible option to triage HPV posi-tive cases
Although this number may be small (five cases/million where genotyping or good quality cytology are not
women/year) and the com-bination is costly but considering available. Direct colposcopy is acceptable for noncompliant
that in India major-ity of the women will be possibly cases (Fig. 2).
having infrequent screening or even once in a life-time
screening, it is prudent to use co-testing. Management of
women with abnormal co-testing based on specific Cytology as the primary screening modality
abnormal-ity and resource availability is depicted in Figure Cytology-based programs have reduced the burden of
2. cervical cancer in developed countries. The sensi-tivity of a
single Pap test for detection of CIN2/3 reportedly ranges
30
from 51% to 53% but the specificity is as high as 96.3%.
Primary HPV testing Therefore, currently cytology is best used as a triage tool
Primary HPV testing alone has replaced cytology in the for HPV positive cases to reduce unnecessary
12
cervical cancer screening programs in various colposcopy. The GCPR provide algorithms for the
management of women with both
squamous and glandular abnormalities in different age
10,11
groups.

206 © 2019 Japan Society of Obstetrics and Gynecology


Cervical Cancer Screening & Management of CIN


Table 3 Resource-based good clinical practice recommendations for cervical cancer screening and treatment of preinvasive lesions

FOGSI good clinical practice recommendation


1. In limited resource settings, women should be screened with VIA, at least one to three times in a lifetime, every 5 years
(Level A).
In good resource settings, women should be screened every 5 years with a validated HPV test or co-test (HPV +
cytology) (Level A).
2. HPV positive cases should be triaged with either HPV genotyping or cytology. Referral to colposcopy if HPV 16/18
positive or cytology result is abnormal (ASCUS or worse). Others should have a repeat HPV test after 1 year.
Colposcopy referral if the HPV test is persistently positive (Level A).
In limited resource settings, VIA can be used to triage HPV positive women. Direct colposcopy is an
acceptable option where compliance is an issue (Level B).
3. Women with co-testing (HPV + cytology)
Women with both HPV positive and cytology abnormal report should be advised colposcopy and directed biopsy
(Level A).
Women with HPV positive and cytology negative report should be advised HPV genotyping or repeat co-testing
after 1 year, depending on the availability of resources (Level B).
Women with HPV negative and cytology abnormal report should be advised further follow-up depending on the type of
cytological abnormality. All cases with ASC-H, HSIL, atypical glandular cells or suspected cancer on cytology
should be referred to colposcopy irrespective of HPV test result (Level A).
4. Women with ASCUS cytology:
Women aged 30–64 years with ASCUS cytology should be triaged preferably with HPV test, or if not
available with repeat cytology at 1 year (Level A).
Younger women should be followed up with annual cytology for 2 years (Level A).
If compliance is a concern, or HPV test is not available, colposcopy/VIA and directed biopsy is an acceptable option for
all age groups (Level C).
Women with LSIL cytology:
Women with LSIL cytology should be managed preferably with colposcopy (Level A).
If compliance is an issue, ‘See-and-Treat’ approach is an acceptable option (Level C).
Colposcopy in younger women with LSIL cytology should be done only when cytology results are persistent or severe
(Level B).
In postmenopausal women, LSIL cytology should be triaged with either colposcopy or reflex HPV or repeat cytology
(Level B).
Women with ASC-H or HSIL cytology:
Colposcopy should be done with special emphasis on visualization of the transformation zone (TZ) and
endocervical evaluation for managing women with ASC-H or HSIL (Level A).
Women with atypical glandular cells with any of the sub-categories should be evaluated with colposcopy and
directed biopsy along with endocervical sampling. Cold knife colonization or LEEP is required if the cytology is
repeatedly showing glandular abnormalities (Level A).
If colposcopy normal endometrial and adnexal pathology should be ruled out (Level B).
5. VIA based screening
VIA based screening is the most feasible modality in low resource settings and also in good resource settings with high
volume of patients (Level B).
VIA positive cases, depending on their compliance and need, can be either managed with colposcopy and biopsy or
can be treated in the same sitting with ablation if eligibility criteria for ablative treatment are fulfilled (Level C).

6. HIV positive women can be screened with any available modality (Level A).
Screen positive HIV positive women should be managed in the same way as general population (Level A).
7. In women with CIN1 preceded by ASCUS or LSIL cytology treatment in form of excision or ablation is
warranted if it persists for 2 years (Level A).
In women with CIN1 preceded by ASC-H or HSIL cytology should be followed up either with yearly co-testing or
with 6-monthly cytology for 2 years. If the abnormality persists, a diagnostic excisional procedure should be advised.
Alternatively, if the patient cannot follow up, a diagnostic excisional procedure can be recommended (Level B).

Patients with CIN2/3 who have type I transformation zone on colposcopy should be managed by either excision
(LEEP) or ablation of the TZ (cryotherapy or thermal ablation). Conversely, if TZ is type II or III on colposcopy or
endocervical sampling shows CIN2/3 or in recurrent CIN, excisional procedure (either cold knife conization or LEEP)
should be performed (Level B).
†Level A: Recommendations are based on good and consistent scientific evidence; Level B: Recommendations are based on limited or
inconsistent scientific evidence; Level C: Recommendations are based primarily on consensus and expert opinion.

© 2019 Japan Society of Obstetrics and Gynecology 207


N. Bhatla et al.

Figure 2 (a) Management of abnormal primary HPV testing. HR HPV, high-risk HPV; VIA, visual inspection after appli-cation of
acetic acid; NILM, negative for intra epithelial lesion or malignancy; CIN, cervical intra epithelial neoplasia.
(b) Management of abnormal co-testing. ASCUS, atypical squamous cells of undetermined significance; HSIL, high-grade
squamous intra epithelial lesion.

Management of cytology showing atypical squamous cells reflex HPV or repeat cytology at 6 months or immedi-ate
with undetermined significance Atypical squamous cells colposcopy. The detection of cumulative cases of CIN3+
with undetermined significance (ASCUS) cytol-ogy is seen was highest in HPV triage group (72.3%), followed by
in 2.8% of women between 30 and conservative management (55%), and least(54.6%) in
64 years and 23–74% of these women are HPV posi- immediate colposcopy group. HPV tri-age could detect
31,32
tive. The ASCUS/LSIL Triage Study (ALTS) 92.4% of the CIN3 cases. Repeat cytology needed two
investigated triage of ASCUS cytology either with visits to demonstrate similar

208 © 2019 Japan Society of Obstetrics and Gynecology


sensitivity (95.4%) and would have to refer a large number Cervical Cancer Screening & Management of CIN
of cases (67.1%) for colposcopy and thus was not cost-
33
effective compared to HPV testing. There-fore, HPV
triage is equally sensitive as immediate col-poscopy and the accuracy of cytology is uncertain, HPV testing may be
32 used for triage. If compliance is an issue, ‘See-and-Treat’
also reduces unnecessary colposcopy referrals. Women
approach is acceptable (Fig. 3). Colposcopy and treatment
aged 30–64 years with ASCUS should be triaged
may do more harms than benefits in women aged <30 years
preferably with HPV test or if not available with repeat
because of higher regression (70% with CIN2) and lower
cytology at 1 year and women ≤30 years with ASCUS or 35
low grade squamous intra-epithelial lesion (LSIL) should progression (0.5% with CIN3) rates. Therefore,
be followed with annual cytology for 2 years. However, colposcopy should be done with severe/persistent cytology
where compli-ance is a concern or HPV testing is not results. Immediate colposcopy is also preferred in post-
available, col-poscopy/VIA and directed biopsy is menopausal women. However, due to likelihood of
acceptable for all age groups (Fig. 3). regression of transformation zone (TZ), use of endocervical
specu-lum for clear delineation is highly desirable.

Management of LSIL cytology In the ALTS trial, 1572


women with LSIL were followed either with colpos-copy Management of women with ASC-H (atypical squamous
or HPV or with repeat cytology at 6 months. More than cells: Cannot exclude high-grade squamous intraepithelial
80% were HPV positive thus obviating the need for HPV lesion) or high-grade squamous intraepithelial lesion
triage in women with LSIL.
34
Thus col-poscopy is cytology HPV triage is not advisable because 65.8% of
preferred for managing women with LSIL cytology. If ASC-H and 89–97% of high-grade squamous intra-
colposcopy is not readily available or if epithelial lesion (HSIL) cytology are HPV positive and are
managed with immediate colposcopy with endo-cervical
35
evaluation. If TZ is not visualized, diagnostic excisional
procedure is preferred except during

Figure 3 Management of abnormal cytology test. (a) Management of ASCUS. ASCUS, atypical squamous cells of un determined
significance; LSIL, low grade squamous intra epithelial lesion; VIA, visual inspection after application of acetic acid; HSIL, high-
grade squamous intra epithelial lesion. (b) Management of LSIL. (c) Management of ASC-H/ HSIL. ASC-H, atypical squamous
cells cannot exclude HSIL. (d) Management of HSIL.

© 2019 Japan Society of Obstetrics and Gynecology 209


N. Bhatla et al.

Figure 4 Management of abnormal VIA test. VIA, visual inspection of cervix after application of acetic acid; CIN, cervical intra
epithelial neoplasia.

pregnancy. In noncompliant cases, SVA with cervical 19


from 16% to 82.6%, and 82.1% to 96.8%, respec-tively.
ablation is acceptable if criteria is fulfilled, but without a VIA when compared with cytology has higher sensitivity
diagnostic specimen (Fig. 3). In younger women with (90% vs 50%) but lower specificity
normal colposcopy and Type-I TZ after a high-grade (37% vs 93.5%) thus raising the burden of false posi-tive
19,40,41
smear, regular follow-up is emphasized and a diagnostic cases. VIA permits an SVA for screening
excisional procedure is recommended when an abnormal in LMICs because of instant results and linkage with
screening test persists for 2 years. 7,8,16
treatment. VIA-positive cases can be either
managed with colposcopy and biopsy or can be treated in
Management of women with glandular cell abnormalities the same sitting with ablation, if criteria are fulfilled (Fig.
About 30% women aged >40 years with 4).
atypical glandular cytology will have preinvasive or
36,37
invasive disease. Majority of these lesions are squa-
mous lesions but the probability of finding endometrial Screening in HIV Infected Women
malignancy is 2–3%, cervical adenocarcinoma in situ is 3–
4%, invasive adenocarcinoma cervix is 2% and cervi-cal Infection with HIV increases a woman’s lifetime risk of
38
squamous cell carvcinoma is 1%. Hence, a thor-ough developing cervical cancer and approxi-mately 10% HIV
evaluation to rule out endometrial, ovarian or cervical positive women develop CIN2+ each year compared to 1–
38 42
malignancy is essential. Women with atypi-cal glandular 2% HIV negative women. The prevalence of invasive
cells with any of the sub-categories should be evaluated cervical can-
with colposcopy and directed biopsy along with cer was 4% and CIN2+ lesions was 8.5% among HIV
endocervical sampling (Fig. 3). positive women.
42,43
Women who are HIV pos-
itive should begin screening with any of the screen-ing
VIA as the primary screening modality modalities as soon as they are diagnosed with HIV. The
VIA is a promising tool for screening in low- expert group has endorsed the recommen-dations by WHO
resource settings because it is economical, with minimal and ASCO that the screening frequency in HIV+ women
39,40 7,9
loss to follow up. The pooled sensitiv- should be twice as often as the general population.
ity and specificity of VIA to detect CIN2+ ranged ART(antiretroviral

210 © 2019 Japan Society of Obstetrics and Gynecology


Cervical Cancer Screening & Management of CIN

Figure 5 Management of women with CIN on histology. CIN, cervical intraepithelial neoplasia; ASC-H, atypical squa-mous cells
cannot exclude HSIL; HSIL, high-grade squamous intra epithelial lesion. (a) Management of women with CIN-1. (b) Management
of women with CIN2/3

therapy) services should be integrated with cervical Management of CIN


44
cancer screening program. Screen positive cases
should be managed in the same way as general The aim of CIN management is to prevent progression
population (Tables 2 and 3). of disease while avoiding over-treatment of lesions that

© 2019 Japan Society of Obstetrics and Gynecology 211


N. Bhatla et al.
45
might otherwise regress. CIN1 preceded by ASCUS or
LSIL cytology is likely to progress to CIN2/3 in 4–13%
cases in next 2 years. Thus treatment by ablation or exci-
sion is warranted if the abnormality persists for 2 years.
46 Contributions of PSI (Population Services Interna-tional
The risk of progression of CIN1 when pre-ceded by ASC-H India) are appreciated for arranging the expert group
or HSIL is more than 15%, thus imme-diate colposcopy meeting.
46
examination is advised. About 40–58% of CIN2 will
regress and only 22% will progress to CIN3; the
cumulative incidence of invasive disease in untreated CIN3 Disclosure
47
is 31% after 10 years. Patients with CIN2/3 with type-I or
None declared.
type-II TZ on colposcopy may be treated by excision or
ablation. Risk of invasive cancer is as high as 7% in
women with type-III TZ on colposcopy or in recurrent References
CIN, therefore excisional procedures with either
LLETZ/LEEP (large loop excision of TZ) or cold knife 1. Cancer Today-Global cancer observatory. [Cited 12 Oct 2018].
colonization are performed and ablation should be avoided Available from URL: https://gco.iarc.fr/today/home.
47 2. Sreedevi A, Javed R, Dinesh A. Epidemiology of cervical cancer
in such situation. with special focus on India. Int J Women’s Health 2015; 7: 405–
In women ≤30 years the probability of regression is 414.
higher and 70% of CIN2 lesions would regress. There-fore, 3. Sankaranarayanan R, Swaminathan R, Lucas E. Cancer Sur-vival
conservative management is preferred and treat-ment is in Africa, Asia, Caribbean and Central America: Survcan. Lyon:
45,47 IARC scientific publication International Agency for Research on
advisable when CIN2 persists for over 24 months. The
Cancer, 2010.
management of CIN depending on the severity and age 4. Labeit A, Peinemann F, Kedir A. Cervical cancer screening
group is depicted in Figure 5 and summarized in Table 3. service utilization in UK. Sci Rep 2013; 3: 2362.
5. Bhatla N, Dar L, Patro AR et al. Can human papilloma virus DNA
The FOGSI recommendations have been drafted to testing of self collected vaginal samples compare with physician
empower healthcare professionals with evidence-based collected cervical samples and cytology for cervi-cal cancer
screening in developing countries? Cancer Epidemiol 2009; 33:
simplified algorithms applicable in daily practice regard- 10.
less of available resources. However, dissemination to 6. Ministry of Health and Family Welfare, Government of India.
remote areas still remains a challenge. Expanding access Operational framework. Management of common cancers. 2016.
through newer technologies such as mHealth is being [Cited 1 Feb 2019.] Available from URL:
considered. http://cancerindia.org.in/cp/images/PDF/Operational Framework
Management of Common Cancers.pdf/.
7. Harris RP, Helfand M, Woolf SH et al. Current methods of the US
preventive services task force: A review of the pro-cess. Am J
Conclusion and Perspective Prev Med 2001; 20: 21–35.
8. WHO. Guidelines for Screening and Treatment of Precancerous
Implementation of universal screening for cervical can-cer Lesions for Cervical Cancer Prevention. Geneva: World Health
irrespective of resources is the need of the hour. The Organization, 2013.
9. Jeronimo J, Castle PE, Temin S et al. Secondary prevention of
screening effort should be linked with optimum treatment cervical cancer: ASCO resource-stratified clinical practice
and follow-up. The FOGSI recommenda-tions not only guideline. J Global Oncol 2016; 3: 635–657.
empower clinicians with the option of using any available 10. FOGSI GCPR on Screening and Treatment of Preinvasive lesions
modality, but also provide a com-prehensive overview of of the cervix and HPV vaccination. [30 Jan 2019.] Available from
follow-up and treatment. Thus they are suitable not only for URL https://www.fogsi.org/fogsi-gcpr-on-screening-mangement-
India but also for many LMICs with similar diverse of-cervical-precancerous-lesions/
practices and inequality. 11. FOGSI GCPR on Screening and Treatment of Preinvasive lesions
of the cervix and HPV vaccination – Summary flow-charts. [1 Feb
2019.] Available from URL: http://www.fogsi.
org/wp=content/uploads/2018/03/FOGSI-GCPR-Final-March-
Acknowledgments 2018.pdf.
12. Tota JE, Bentley J, Blake J et al. Introduction of molecular HPV
The contributions of Sankaranarayanan R, Ahuja M, Joshi testing as the primary technology in cervical cancer screening:
Acting on evidence to change the current para-digm. Prev Med
R, Purandare N and Natarajan J to development of the
2017; 98: 5–14.
GCPR are deeply appreciated. 13. Isidean SD, Mayrand MH, Ramanakumar AV et al. Human
papillomavirus testing versus cytology in primary cervical cancer
screening: End-of-study and extended follow-up results from the
Canadian cervical cancer screening trial. Int J Cancer 2016; 139:
2456–2466.

212 © 2019 Japan Society of Obstetrics and Gynecology


Cervical Cancer Screening & Management of CIN
14. Sankaranarayanan R, Nene BM, Shastri SS et al. HPV screen-ing
for cervical cancer in rural India. NEJM 2009; 360: 1385–1394.
primary cervical cancer screening. Int J Cancer 2006; 119:
15. Schiffman M, Kinney WK, Cheung LC et al. Relative perfor- 1095–1101.
mance of HPV and cytology components of co-testing in cer-vical 31. Katki HA, Schiffman M, Castle PE et al. Benchmarking Cin 3 +
screening. J Natl Cancer Inst 2017; 110: 501–508. risk as the basis for incorporating HPV and pap cotesting into
16. Arbyn M, Sankaranarayanan R, Muwonge R et al. Pooled analysis cervical screening and management guidelines. J Low Genit Tract
of the accuracy of five cervical cancer screening tests assessed in Dis 2013; s28: 17.
eleven studies in Africa and India. Int J Can-cer 2008; 123: 153– 32. Katki HA, Schiffman M, Castle PE et al. Five year risks of Cin 2+
160. and Cin 3+ among women with HPV positive and HPV negative
17. Arbyn M, Bergeron C, Klinkhamer P, Martin-Hirsch P, Siebers LSIL pap results. J Low Genit Tract Dis 2013; s43: 17.
AG, Bulten J. Liquid compared with conventional cervical
cytology: A systematic review and meta-analysis. Obstet Gynecol 33. ASCUS LSIL Triage Study (ALTS) Group. Results of a ran-
2008; 111: 167–177. domized trial on the management of cytology interpreta-tions of
18. Guducu N, Sidar G, Bassullu N, Turkmen I, Dunder I. Three-step atypical squamous cells of undetermined significance. Am J
approach versus see-and-treat approach in patients with Obstet Gynecol 2003; 188: 1383.
cytological abnormalities. Int J Clin Exp Med 2013; 6: 372–376. 34. ASCUS LSIL Triage Study (ALTS) group. A randomized trial on
the management of low-grade squamous intra-epithelial lesion
19. Bobdey S, Sathwara J, Jain A, Balasubramaniam G. Burden of cytology interpretations. Am J Obstet Gynecol 2003; 188: 1393–
cervical cancer and role of screening in India. Indian J Med 1400.
Paediatr Oncol 2016; 37: 278–285. 35. Castle PE, Fetterman B, Cox JT et al. The age-specific rela-
20. Kulasingam SL, Havrilesky L, Ghebre R, Myers ER. Screen-ing tionships of abnormal cytology and human papillomavirus DNA
for Cervical Cancer: A Decision Analysis for the US Preven-tive results to the risk of cervical precancer and cancer. Obstet
Services Task Force. Rockville, MD: Agency for Healthcare Gynecol 2010; 116: 76–84.
Research and Quality, 2011. 36. Marques JP, Costa LB, Pinto AP et al. Atypical glandular cells
21. Sankaranarayanan R, Esmy PO, Rajkumar R et al. Effect of visual and cervical cancer: Systematic review. Rev Assoc Med Bras
screening on cervical cancer incidence and mortality in Tamil 2011; 57: 234–238.
Nadu, India: A cluster-randomised trial. Lancet 2007; 370: 398– 37. Zhao C, Florea A, Onisko A, Austin RM. Histologic follow-up
406. results in 662 patients with pap test findings of atypical glandular
22. Goldie SJ, Gaffikin L, Goldhaber-Fiebert JD et al. Cost- cells: Results from a large academic womens hos-pital laboratory
effectiveness of cervical-cancer screening in five developing employing sensitive screening methods. Gynecol Oncol 2009;
countries. NEJM 2005; 353: 2158–2168. 114: 383–389.
23. Naucler P, Ryd W, Törnberg S et al. Human papillomavirus and 38. Schnatz PF, Guile M, O’sullivan DM, Sorosky JI. Clinical sig-
papanicolaou tests to screen for cervical cancer. New Engl J Med nificance of atypical glandular cells on cervical cytology. Obstet
2007; 357: 1589–1597. Gynecol 2006; 107: 701–708.
24. Australian Government Department of Health. Overview of the 39. Shastri SS, Mittra I, Mishra GA et al. Effect of VIA screen-ing by
Renewal. 2017. [Cited 14 Sept 2018.] Available from URL: primary health workers: Randomized controlled study in Mumbai,
http://www.cancerscreening.gov.au/internet/screening/ India. J Natl Cancer Inst 2014; 106: dju009.
publishing.nsf/Content/overview-of-the-renewal.
40. Mustafa RA, Santesso N, Khatib R et al. Systematic reviews and
25. Ponti A, Anttila A, Ronco G, Senore C. Cancer Screening in the
meta-analyses of the accuracy of HPV tests, visual inspection with
European Union (2017). Report on the implementation of the
acetic acid, cytology, and colposcopy. Int J Gynecol Obstet 2016;
Council Recommendation on cancer screening. [Cited 14 Sept
132: 259–265.
2018.] Available from URL: https://ec.europa.eu/
41. Poli UR, Bidinger PD, Gowrishankar S. Visual inspection with
health/sites/health/files/major_chronic_diseases/docs/2017_
acetic acid (VIA) screening program: Seven years experience in
cancerscreening_2ndreportimplementation_en.pdf
early detection of cervical cancer and pre-cancers in rural South
26. Richardson L, Tota J, Franco E. Optimizing technology for
India. Indian J Community Med 2015; 40: 203–207.
cervical cancer screening. Expert Rev Obstet Gynecol 2011; 6:
343–353.
42. Joshi S, Sankaranarayanan R, Muwonge R, Kulkarni V,
27. Wright TC Jr, Stoler MH, Behrens CM, Apple R, Derion T,
Somanathan T, Divate U. Screening of cervical neoplasia in HIV-
Wright TL. The ATHENA human papillomavirus study: Design,
infected women in India. AIDS 2013; 27: 607–615.
methods, and baseline results. Am J Obstet Gynecol 2012; 206:
46–e1.
43. Clifford GM, Tully S, Franceschi S. Carcinogenicity of human
28. Gage JC, Schiffman M, Katki HA et al. Reassurance against
papillomavirus (HPV) types in HIV-positive women: A meta-
future risk of precancer and cancer conferred by a negative human
analysis from HPV infection to cervical cancer. Clin Infect Dis
papillomavirus test. J Natl Cancer Inst 2014; 106: 1–4. 2017; 64: 1228–1235.
44. Sahasrabuddhe VV, Bhosale RA, Kavatkar AN et al. Com-parison
29. Bhatla N, Puri K, Kriplani A et al. Adjunctive testing for cer-vical
of visual inspection with acetic acid and cervical cytology to
cancer screening in low resource settings. Aust N Z J Obstet
detect high-grade cervical neoplasia among HIV-infected women
Gynaecol 2012; 52: 133–139.
in India. Int J Cancer 2012; 130: 234–240.
30. Cuzick J, Clavel C, Petry KU et al. Overview of the European and
north American studies on HPV testing in
45. Elit L, Levine MN, Julian JA et al. Expectant management versus
immediate treatment for low-grade cervical

© 2019 Japan Society of Obstetrics and Gynecology 213


N. Bhatla et al.
intraepithelial neoplasia : A randomized trial in Canada and
Brazil. Cancer 2011; 117: 1438–1445.
46. Katki HA, Gage JC, Schiffman M et al. Follow-up testing post-
colposcopy: Five-year risk of CIN2+ after a colposcopic diag-
nosis of CIN1 or less. J Lower Genit Tract Dis 2013; 17: S69.
47. McCredie MR, Sharples KJ, Paul C et al. Natural history of
cervical neoplasia and risk of invasive cancer in women with
cervical intraepithelial neoplasia
3: A retrospective cohort study. Lancet Oncol 2008; 9:
425–434.

214 © 2019 Japan Society of Obstetrics and Gynecology

You might also like