Download as pdf or txt
Download as pdf or txt
You are on page 1of 78

CHAPTER 13 

Cervical Squamous Neoplasia


Christopher P. Crum, Emily E. Meserve, and William A. Peters III

Chapter Outline
INTRODUCTION Cervical Cancer Prevention and Management of the Abnormal Papanicolaou Test

Definition Management of ASCUS and Squamous Intraepithelial Lesions

Identifying Patients at Risk for Cervical Neoplasia Histopathologic Diagnosis of Preinvasive Disease

The Squamocolumnar Junction Diagnostic Criteria for Squamous Intraepithelial Lesions

Human Papillomaviruses Low-Grade Squamous Intraepithelial Lesions

Non-Viral Factors Endocervical Curettage

Human Papillomavirus Testing Resolving Discordant Papanicolaou Test and Biopsy Results

Current Screening Algorithms Management of Squamous Intraepithelial Lesions

CYTOLOGY DIAGNOSIS AND MANAGEMENT OF INVASIVE SQUAMOUS


CELL CARCINOMA
NONDIAGNOSTIC SQUAMOUS ATYPIA (ATYPICAL SQUAMOUS
CELLS OF UNDETERMINED SIGNIFICANCE) Introduction

Introduction Diagnosis of Invasion

Cytologic Alterations That Are Best Left Out of the ASCUS Category Immunohistochemistry

Cytologic Criteria for the Diagnosis of ASCUS Defining Superficially Invasive (Microinvasive) Squamous Cell Carcinoma

Cytologic Criteria for Squamous Intraepithelial Lesions Management of Superficially Invasive Squamous Cell Carcinoma (Stage IA)

Low-Grade Squamous Intraepithelial Lesions Pathology of Invasive Squamous Carcinoma

High-Grade Squamous Intraepithelial Lesions Treatment and Outcome of Squamous Carcinoma

Differential Diagnosis of Low-Grade and High-Grade Squamous Intraepithelial Lesion Management of Squamous Cell Carcinoma

Introduction a prior abnormal Pap test. Risk factors classically associated


with cervical cancer—early age of first intercourse, multiple
Definition sexual partners, and increasing parity—are sufficiently
common that a high percentage of women would qualify
Cervical squamous neoplasia is defined as all squamous cell for Pap smear screening by these criteria. Moreover, timing
alterations that occur in or near the cervical transformation of sexual activity may be as or more important than
zone and are causally related to human papillomavirus absolute number of sexual partners in conferring risk of
(HPV) infections. The terms cervical intraepithelial neoplasia current HPV infection.2 The following is a series of risk
(CIN), dysplasia, and squamous intraepithelial lesion (SIL) factors associated with cervical neoplasia.
apply to this group of lesions as well. In this chapter, the
terms CIN1, flat condyloma, and exophytic condyloma are
used interchangeably with low-grade squamous intraepithelial
The Squamocolumnar Junction
lesion (LSIL), and the terms CIN2 and CIN3/carcinoma in It is understood that the cervix has a tenfold to twentyfold
situ are synonymous with high-grade squamous intraepithelial greater risk of malignancy than other anogenital sites,
lesion (HSIL). The recent conclusions of the Lower Ano- including vulva, vagina, and anus. It has long been known
genital Squamous Terminology (LAST) project, held in that the great majority of carcinomas arise near the entrance
2012, are in line with this.1 The choice of diagnostic ter- to the cervix at the squamocolumnar junction (SCJ). This
minology (dysplasia, CIN, SIL) is critical only in terms of is due to a unique target cell population at the SCJ that
the decision point for recommending excision versus is absent in these other sites, including the anus (Fig.
observation. The impact of this terminology on manage- 13.1). The specific properties of this junction are discussed
ment is discussed later in this chapter. later in the chapter.3 Suffice to say that this chapter would
be much shorter if unique SCJ cells did not exist!
Identifying Patients at Risk for Cervical Neoplasia
In clinical practice, there is no algorithm that will decide
Human Papillomaviruses
whether a given reproductive-age woman should or should HPV infection is the etiologic agent for the great majority
not have a Papanicolaou (Pap) test, short of clinical evi- of cervical epithelial neoplasms. The powerful association
dence of HPV infection, lower genital tract symptoms, or between HPV infection and cervical neoplasia has been
298
Cervical Squamous Neoplasia 13

Abstract ning of this concept is the absence of any biomarker that


can be depended upon to segregate low-grade squamous
This chapter has been extensively revised to address several intraepithelial lesion (LSIL) from high-grade squamous
important issues in the field of cervical neoplasia. These intraepithelial lesion (HSIL) in light of the widespread
include new information about the origin of cervical variations in interpretation between observers and the lack
squamous neoplasia and its impact on our perceptions of compelling information to support p16 as either a
of lesion development. This leads in to a discussion of marker of HSIL or a predictor thereof. Other important
the conundrum of lesion grading, specifically the laboratory topics include new approaches to superficially invasive
management of lesions that fall between cervical intraepi- squamous carcinoma, new management schemes, and the
thelial neoplasia grade 1 (CIN1) and grade 3 (CIN3). promise of vaccination programs. Finally, the concept of
Strategies for managing this problem are offered, and the prophylactic ablation of the squamocolumnar junction
concept of squamous intraepithelial lesion (SIL) of inter- (SCJ) is addressed as another possible approach to cervical
mediate (or indeterminate) grade is unveiled. The underpin- cancer risk reduction in vulnerable populations.

Keywords
cervical intraepithelial neoplasia (CIN)
human papillomavirus (HPV)
p16
squamous cell carcinoma
squamous intraepithelial lesion (SIL)

298.e1
Cervical Squamous Neoplasia 13

A B
Fig. 13.1.  The squamocolumnar junction (SCJ) and cervical neoplasia. A, Tissue section of the junction
of the ectocervical (or mature metaplastic) epithelium with the endocervical mucosa. The arrow highlights
the SCJ. B, Immunofluorescence for cytokeratin 7 (CK7) of a comparable region in another cervix
demonstrates intense staining of a cuboidal cell population at the junction of the ectocervix and endocervix.
This population is postulated to be the origin of cervical squamous and columnar neoplasia.

Prior exposure Squamo-columnar


(antibodies) junction cell Lower-risk types
Condoms Impaired local immunity Immune response

ACIS

Columnar
differentiation
HPV Cell SIL Persistence HSIL Cancer

Viral HPV HLA


load variants HPV Viral 3q25–27
interact integration amplification

High
risk
Centrosome abnormalities
Cell cycle disruption
Genomic instability
Promotor methylation

4 to 24 months

4 months to 5 years

1 to 25 years

Fig. 13.2.   Schematic of human papillomavirus (HPV) infection. ACIS, Adenocarcinoma in situ; SIL,

squamous intraepithelial lesion; HSIL, high-grade squamous intraepithelial lesion; HLA, human leukocyte
antigen.

established, and the accumulated experimental, molecular, early reproductive years, (3) infections are transient, often
and clinical evidence has left no doubt that HPV directly appearing and disappearing without cytologic abnormality,
influences the pathogenesis of cervical neoplasia. Table but (4) persistent infection by the same HPV type is strongly
13.1 summarizes the varied incidences of cervical cancer associated with risk of a current or subsequent cervical
worldwide. Fig. 13.2 illustrates what takes place locally neoplasm.4,5 The conclusion to this chapter reiterates the
in the cervix and the factors involving infection in the most compelling link between HPV and cervical neoplasia—
SCJ. Over the past several years, the realization has emerged that immunization with vaccines derived from high-risk
that (1) HPV infection is ubiquitous in the young sexually HPVs will ultimately have a major impact on the prevalence
active population, (2) frequency of infection peaks in the rates of significant HPV-associated cervical neoplasms.6 It
299
Diagnostic Gynecologic and Obstetric Pathology

Table 13.1  Incidence of Cervical Carcinoma Worldwidea Table 13.2  Classification of Human Papillomavirus Types

Adjusted Adjusted Human


Locale Incidenceb Mortalityb Papillomavirus Cancers (%) Controls (%) Odds Ratio

Worldwide 16.1 8.0 16 50.5 3.3 435

More developed 11.4 4.1 18 13.1 1.3 248

Less developed 18.7 9.8 45 5.5 0.7 198

East Africa 44.3 24.2 31 2.7 0.6 124

Mid Africa 25.1 14.2 52 2.7 0.3 200

North Africa 16.8 9.1 33 1.0 0.1 373

South Africa 30.3 16.5 58 2.3 0.5 115

West Africa 20.3 10.9 35 1.1 0.5 74

Caribbean 35.8 16.8 59 1.3 0.1 419

Central America 40.3 17.0 51 1.0 0.3 67

South America 30.9 12.0 56 0.7 0.4 45

North America 8.3 3.2 39 0.6 0.0 —

East Asia 6.4 3.2 73 0.4 0.1 106

Southeast Asia 18.3 9.7 68 0.2 0.1 54


High-risk: Types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 68, 73, 82.
South Central Asia 26.5 15.0 Possibly high-risk: Types 26, 53, 66.
Low-risk: Types 6, 11, 40, 42, 43, 44, 54, 61, 72, 81, cp6108.
Western Asia 4.8 2.5 From Munoz N, Bosch FX, de Sanjose S, et al: Epidemiologic classification of human
papillomavirus types associated with cervical cancer. N Engl J Med 348(6):518-527, 2003.
Eastern Europe 16.8 6.2

Northern Europe 9.8 4.0


studies, it is possible to ascertain the relative strength of
Southern Europe 10.2 3.3 association between certain HPV groups and not only
cancer but also preinvasive atypias. Kahn et al. followed
Western Europe 10.4 3.7
women with mild cytologic atypia and correlated the
Australia/New Zealand 7.7 2.7 outcome of CIN3 at 10 years with HPV type.7 The relatively
strong association between HPVs 16 and 18 and CIN3
Melanesia 43.8 23.8 (15% to 18%) versus all other high-risk HPV types (3%)
Micronesia 12.3 6.2
and low-risk HPV types (<1%) is a compelling endorsement
of HPV-16 as the prototypic high-risk type. A more recent
Polynesia 32.9 17.4 study by Castle et al. showed that two high-risk or onco-
a
Data from http://globocan.iarc.fr/old/FactSheets/cancers/cervix-new.asp. genic HPV tests within approximately 1 year conferred a
b
Per 100,000 per year. risk of CIN2+ biopsy at 3 years of 17%, which increased
to 41% for HPV-16 (Table 13.3).8 However, as noted earlier,
infection by HPV-16 is not a guarantee that HSIL will
ensue, and other factors, such as the degree of cytologic
also addresses the potential value of pre-emptive removal abnormality and colposcopic findings, have a major influ-
or destruction of the SCJ. ence on whether HSIL will be found on biopsy.9
Cancer-associated (“high-risk”) HPV types impose a broad The relationships between HPVs and cervical neoplasia vary
gradient of risk, with HPV-16 conferring the greatest risk. both for lesion grade and cell type. HPVs 6 and 11 are not
Low-risk HPV types may confer risk as surrogate markers associated with cervical carcinomas or HSILs. HPV-16 is
of “at-risk behavior.” Nearly 100 HPVs have been character- detected in nearly 50% of HSIL and squamous carcinomas
ized, and more than 40 have been identified in the genital of the cervix and is the “prototypic” cancer-causing virus.
tract. Genital HPVs have traditionally been divided into In contrast, HPV-56 is associated with fewer than 1 in 50
those with low, intermediate, and high association with cancers. HPV-18 is associated with less than 15% of
cervical carcinoma.7 Currently, those HPVs with any squamous carcinomas, predominating in adenocarcinomas
association are termed “high-risk” HPV types. Table 13.2 in situ (ACISs) and invasive adenocarcinomas. Small cell
reviews the types of HPV and the evidence linking them neuroendocrine carcinomas are almost exclusively associ-
to cervical carcinoma. From this information and prior ated with HPV-18. A fourth group, consisting of newly
300
Cervical Squamous Neoplasia 13

20
Table 13.3  Follow-Up as a Function of Short-Term % HPV+
Persistence of High-Risk Human Papillomavirusa SCC incidence

Cervical Intraepithelial 15
High-Risk Human Papillomavirus Neoplasia Grade 2 + Biopsy
Status (Two Tests) (3 Years)

Negative/negative 0.5%
10
Negative/positive 3.4%

Positive/negative 1.2%
5
Positive/positive 17%

HPV-16+/HPV-16+ 40.8%
a
Defined as two positive tests within an average of 1 year. 0
From Castle PE, Rodríguez AC, Burk RD, et al: Proyecto Epidemiológico Guanacaste
(PEG) Group. Short term persistence of human papillomavirus and risk of cervical 24 29 34 39 44 49 54 59 64
precancer and cancer: population based cohort study. BMJ 339:b2569, 2009.
Fig. 13.3.  Distribution of human papillomavirus (HPV) frequency
in the population and cervical cancer as a function of age. A progressive
decline in HPV index coincides with an increase in cancer risk. SCC,
Squamous cell carcinoma.
discovered HPV types and having uncertain or controversial
association with cancer, exists and presumably confers a
low risk of cancer.10
Is there regional variation in HPV type? Prevalence rates This risk drops significantly with increasing age, which
of high-risk HPVs vary somewhat among different regions is associated with increasing risk of cancer (Fig. 13.3).
of the world and different countries. Nevertheless, HPVs Risk drops further with the approach of menopause,
16 and 18 are the dominant HPVs detected, ranging from paralleling the drop in HPV prevalence with age. The
64% to 77% across different continents.11 progressive drop in risk with age has been attributed
Does the amount of virus present influence risk? There is to an effective immune response to the virus that follows
general agreement that higher levels of viral DNA (viral the onset of sexual activity and exposure to HPVs.
load) correlate with risk of SIL. However, viral load will Protection is long lasting, given the low rates of HPV
not discriminate LSIL from HSIL, because viral production positivity in middle-age women. There is conflicting
is often higher in low-grade lesions.12 For this reason, viral evidence regarding HPV rates in older women, but some
load is not used as a predictor for HSIL. studies support a small but distinct increase in positive
Persistent infection by the same HPV type is strongly associated rate after menopause.17
with risk of cervical neoplasia. The majority of HPV infections • Immune status: Defects in cell-mediated immunity
are transient. Only a few cases score positive for the same imposed by transplant therapy significantly increase
HPV type on successive tests. Prospective studies have the risk of cervical cancer. Human immunodeficiency
shown that persistent infections by the same type correlate virus (HIV)-infected individuals are prone to HPV
with increased risk of lesion detection, particularly after infection, persistent HPV infection, and a higher risk
cone biopsy, and retrospective studies of patients with of precursor lesions. The impact of HIV infection on
cervical cancer have also documented prior infection and cervical cancer risk is controversial, but prognosis appears
an association between persistent HPV positivity and worse in patients with severe immunodeficiency. HIV
neoplasia.13,14 Studies have shown that more than 90% of is clearly associated with increased risk of anal cancer
patients who developed smear abnormalities had persistent in men.18
high-risk HPV infection.15 Women post-transplant have higher than normal rates
of HPV positivity.19 Cancer rates are higher in this
population, and in general anogenital cancers occur
Non-Viral Factors at a younger age. One study reported a fivefold
A number of non-viral factors have been implicated in increase in risk of HSIL (50% vs. 10%) in transplant
risk, most of which are not usually taken into account in recipients.20 In one study, lower genital cytopathology
clinical management. They include the following: was evaluated in 105 immunosuppressed renal
• Human leukocyte antigen (HLA) type: The evidence transplant recipients. Evidence of HPV infection was
suggests that specific HLA class II haplotypes may found in 17.5% and of lower genital neoplasia in
influence HPV antigen presentation and the immune 9.5%. The rate of the virus infection in the immu-
response to HPV infection, in turn influencing the risk nosuppressed was nine times greater than in a general
of developing invasive cervical carcinoma. However, population and 17 times greater than in a matched
the precise mechanisms underlying these associations immunocompetent population.21 In one-third of
remain to be clarified.16 patients with HPV lesions and half of patients with
• Age: Young, sexually active women are at greatest risk neoplastic lesions, multiple lower genital sites were
for HPV infection and preinvasive cervical neoplasia. also involved.
301
Diagnostic Gynecologic and Obstetric Pathology

HIV infection has been the most intensively studied other women did not increase the risk independently,
and permits the breakdown of risk according to whereas a history of genital warts and absence of
several outcome measures: condom use did.39
• The risk of HPV positivity is increased in women • Condoms protect against HPV infections but incom-
who are HIV positive. Approximately 60% of pletely. Winer et al. reported an adjusted hazard
HIV-infected women versus 36% of uninfected ratio of 0.3 for HPV infection in young women
women score positive for HPV, based on testing whose partners always used condoms versus those
of cervical samples. Frequencies of HPV-16, who used condoms less than 5% of the time. Thus,
HPV-18, and multiple infections are also signifi- condoms significantly reduce the risk of cervical HPV
cantly more common in the HIV-infected group.22,23 infection.40
• The risk of persistent HPV infection is significantly • Circumcision reduces the risk of infection in the
increased in HIV-infected women. Estimates place male partner and his female contacts, producing an
the risk of high-risk HPV infection at least twofold RR of 0.65 in the male. Other infections, such as
of non–HPV-infected controls, and the risk of syphilis and HIV, are also significantly reduced in
persistent infection is nearly six times higher. circumcised men.41
Furthermore, in one study, persistence was 1.9 • Oral contraceptives: Oral contraceptive pill (OCP) use
(95% confidence interval [CI], 1.5–2.3) times modestly increases the risk of cervical neoplasia. A large
greater than normal if the subject had a CD-4 cell meta-analysis postulated an RR of 1.90 with use of
count greater than 200 cells/µL (vs. less than 500 greater than 5 years and a decline in risk with cessation
cells/µL).24 of use.42
• The risk of a subsequent SIL is significantly • Smoking: Smoking increases the risk of cervical neo-
higher in HIV-infected women and viral load plasia. The theoretic basis is presumably the presence
and low CD-4 counts correlate with cytologic of DNA adducts in the cervical mucus, exposing the
abnormalities.25-27 transformation zone mucosa to carcinogens.43 A retro-
• The proportion of HSILs appears to be not sub- spective study in a Nordic population showed an odds
stantially higher in HIV-infected women. Ellerbrock ratio of 2.7 for women with nicotine in their serum
et al. also showed that 91% and 75% of SILs in after controlling for the presence of HPV-16/18 antibod-
the two groups, respectively, were LSILs.25 ies.44 An RR of 1.58 was computed in another study
• The risk of persistent SIL appears higher in women examining environmental exposure.45
with HIV, being 76% versus 18% in the study by • Chlamydia infection: The association between chlamydia
La Ruche et al.28 infection and cervical neoplasia is controversial. Several
• The incidence of invasive carcinoma is variably studies have shown a relationship between genital infec-
influenced by HIV infection. Three studies reported tions and HPV or HSIL.46 Others, however, have not
up to fifteenfold greater risk in HIV-infected shown a relationship between antibodies to chlamydia
women.29-31 However, Chokunonga et al. and La and cervical neoplasia.47 A recent study showed an odds
Ruche et al. did not consider HIV a major factor ratio for cytologic abnormalities of 10.7 when coinfec-
in cervical cancer incidence in Central Africa.32,33 tion with both chlamydia and HPV was documented.48
Another study from South Africa found no sig- • Women who have sex with women: Risk of cervical
nificant difference between stage of presentation neoplasia is increased in women who have sex with
and HIV status, although some correlation was other women. Studies have shown high rates of sexually
observed for patients with CD-4 counts of less transmitted infections in women who have sex pre-
than 200 cells/µL.34 dominantly with other women, although the rates of
• The male partners: The role of the male partner can be genital condylomata were low. However, the risk of
summarized as follows: developing HSIL was not determined. Two reports have
• Sexual history of the male sexual partner influences shown that CIN2 may occur in lesbians.49,50
risk of cervical neoplasia, both in number of lifetime In summary, a multitude of factors, viral and host related,
sexual partners and sexual practices (unprotected sex influences risk of cervical neoplasia before, during, and
with prostitutes).35,36 One group showed that monoga- following exposure and lesion progression. The complexity
mous wives of husbands who had sexual relationships of this relationship underscores the importance of addi-
both before and during the marriage had a substantial tional risk factors that result in only a minority of women
risk of cervical cancer (relative risk [RR] = 6.9 [CI, developing cervical cancer following exposure to HPV.
2.3–20.7]).37 However, the degree of risk conferred
by extramarital sexual experiences has varied from
study to study.
Human Papillomavirus Testing
• There is little evidence that reinfection of the woman As a screening tool, HPV testing has been approved for
with the same HPV type influences recurrence. Krebs cervical cancer prevention, keeping the following in mind:
and Helmkamp showed that treating the male partner • HPV positivity increases the risk of cervical neoplasia
did not influence the risk of recurrent genital warts.38 fortyfold.
The implication of this study is that partners are • An abnormal smear plus high-risk HPV confers a 20%
not reinfected by the same virus, consistent with a risk of HSIL.
functioning immune system. Kjaer et al. found that • Consecutive positive high-risk HPV confers an up to
repeated sexual contacts of the male partner with 33% risk of HSIL outcome.
302
Cervical Squamous Neoplasia 13

• A single positive Hybrid Capture II test, particularly of HPV-16/18 testing.56 The reader can envision that as
in young women, has less value. However, the high time passes, the proportion of women scoring positive
negative predictive value of this test, combined with a for HPV-16/18 will diminish and the algorithm might
normal Pap smear, virtually ensures the patient is free evolve, or other biomarkers will be proposed if they have
of a cancer precursor. Testing is superior in sensitivity a stronger predictive value than HPV testing. At present,
to the Pap smears and has emerged as a cost-effective other adjuncts to the Pap smear have not been approved
alternative.51 for screening.
• Combined negative HPV testing and normal smear may
increase the duration of the “protective effect.” Because
HPV is detected prior to the development of a cytologic Cytology
abnormality, it is likely that patients negative by both
HPV testing and smear may be followed at longer A more comprehensive treatise on the cytopathologic
intervals than by smear alone. diagnosis of cervical neoplasia can be found in any of
• Women under age 20 frequently score positive for HPV, several texts addressing this field. The purpose of this
but many of these infections will resolve; hence the discussion is to update and integrate several facets of
recommendation that this group not be tested for HPV.4 cytologic screening that have been evolving, are directly
• The index of HPV positivity drops markedly over age relevant to patient care, and are thus of interest to anyone
30, and this group has been selected for screening. involved in the laboratory or clinical management of
The degree to which an HPV test is more specific for cervical neoplasia. These include the following:
neoplasia risk in this group varies according to the • Realistic expectations from cytologic screening: The
study. However, the percentage of women over 30 years majority of squamous carcinomas of the cervix are
of age who score positive is sufficiently low that HPV preceded by preinvasive SILs. Studies of cancer inci-
testing is considered a cost-effective adjunct to cervical dence rates have shown an approximately two-thirds
cytology.52 reduction since the late 1940s and the introduction of
• A significant fraction of menopausal women may score the Pap test. Predictably, the most striking reduction
positive for HPV, with the significance of this finding in rates occurs immediately following introduction of
still unclear. A recent study using a sensitive assay for screening, where a precipitous reduction (from 28 to
detecting HPV found two peaks of HPV DNA preva- four cases per 100,000 in the studies by Bryans et al.)
lence, including a first peak of 16.7% in women occurs simply by removing those with occult cancer.57
younger than 25 years old. HPV DNA prevalence Once this is accomplished, the incidence drops to a
declined to 3.7% in the age group 35 to 44 years old, steady state of approximately four cases per 100,000
then increased progressively to 23% among women and is favorably affected by screening of women at a
65 years old and older.53 Another study found the young age. According to Miller, the maximum achievable
rates to be lower than the above figure postmenopause reduction in cancer by conscientious Pap test screening
(6.2%), with the frequency of HPV-16 in CIN2+ (29%) is approximately 90%.58
less than half that seen in similar lesions in younger Moreover, since the mid-1980s, there have been progres-
women.54 sive declines in the prevalence of and death rate for
• A number of studies have emphasized the importance cervical cancer, a trend that should continue with
of HPV-16 and HPV-18, the two most prevalent HPVs the use of HPV testing in screening algorithms and
in cervical cancer, relative to other high-risk HPVs, the widespread use of papillomavirus vaccines.59
particularly the fact that of all the high-risk HPVs, these • The Bethesda System for cytologic diagnosis is the
two are much more likely to be associated with follow-up standard for evaluation of cervical cytology since 1988.60
cervical abnormality.55 In 2001, the system was refined with the following:
• Eliminating the adequacy category “satisfactory but
limited”
Current Screening Algorithms • Combining the category previously termed “negative
Screening algorithms have undergone a two-step evolution. and benign cellular changes” into a single “negative”
The first algorithm proposed a high-risk HPV test (Hybrid category
Capture II) in conjunction with cervical cytology for women • Renaming atypical squamous cells of undetermined
over age 30. Women negative with both tests are followed significance (ASCUS) atypical squamous cells (ASC)
at 3-year intervals. More recently, with the adoption of and contracting its subcategories to ASCUS and atypi-
type specific (HPV-16/18) testing as an adjunct and with cal squamous cells, cannot rule out HSIL (ASC-H)
the availability of this probe set as a U.S. Food and Drug • Renaming atypical glandular cells of undetermined
Administration (FDA)–approved test, the algorithm has significance (AGCUS) as atypical glandular cells and
been improved to permit exclusion of a significant number contracting its subcategories to atypical glandular
of HPV-positive women from immediate follow-up. In cells, not otherwise specified (AGC NOS) and atypical
essence, HPV-positive women who score negative for glandular cells, favor neoplasia (AGC favor neopla-
HPV-16 or HPV-18 can be followed at a longer interval sia).61 This has been revised recently in 2014 with
with HPV testing and cytology. If both are negative, the the following minor changes:
patient can return to a 3-year screening interval. If either • Benign endometrial cells (which conceivably
is positive, the patient is evaluated further. This approach indicate endometrial pathology) are reported
takes advantage of the stronger positive predictive value starting at age 45 rather than age 40.
303
Diagnostic Gynecologic and Obstetric Pathology

• Cytologic preparations with LSIL and a “few abnormal squamous cytologic changes.62 (See Table 14.2 for
cells suggesting HSIL” are now reported with two the classification and reporting of glandular abnormalities.)
diagnoses, LSIL and ASCUS favor HSIL (ASCH). Glandular lesions are discussed in greater detail in
This was recommended in order to maintain Chapter 14, but three issues relating to diagnostic errors
the two-tier system and avoid an “intermedi- in Pap test interpretation should be mentioned:
ate” category. However, in the opinion of these • Detection errors are more common with adenocarci-
authors, the two-tier system will always be an nomas than squamous carcinomas.
oversimplification. The reader is referred to the • Adenocarcinomas represent a greater proportion of
most recent text, which updates both figures and cancers now detected in reproductive-age women.
recommendations.62,63 • Adenocarcinomas also constitute the bulk of the
Box 13.1 summarizes the most recent Bethesda classifica- “surprises” that are being encountered in the 20- to
tion system for the interpretation of benign findings and 30-year-old group. The latter fact has important

BOX 13.1  The Bethesda System of Cytologic Classification (2014)


Specimen Type Other
Indicate conventional smear (Pap smear) versus liquid-based preparation • Endometrial cells (in a woman ≥45 years old)
versus other (Specify if “negative for squamous intraepithelial lesion”)

Specimen Adequacy Epithelial Cell Abnormalities


• Satisfactory for evaluation (describe presence or absence of Squamous Cell
endocervical/transformation zone component and any other quality • Atypical squamous cells (ASCs)
indicators, e.g., partially obscuring blood, inflammation, etc.) • Of undetermined significance (ASC-US)
• Unsatisfactory for evaluation (specify reason) • Cannot exclude HSIL (ASC-H)
• Specimen rejected/not processed (specify reason) • Low-grade squamous intraepithelial lesion (LSIL)
• Specimen processed and examined, but unsatisfactory for evaluation (Encompassing: Human papillomavirus (HPV)/mild dysplasia/CIN1)
of epithelial abnormality because of (specify reason) • High-grade squamous intraepithelial lesion (HSIL)
(Encompassing: Moderate and severe dysplasia, CIS; CIN2 and CIN3)
General Categorization (Optional) • With features suspicious for invasion (if invasion is suspected)
• Negative for intraepithelial lesion or malignancy • Squamous cell carcinoma
• Other: See Interpretation/Result (e.g., endometrial cells in a woman
≥45 years old) Glandular Cell
• Epithelial cell abnormality: See Interpretation/Result (specify • Atypical
“squamous” or “glandular” as appropriate) • Endocervical cells (NOS or specify in comments)
• Endometrial cells (NOS or specify in comments)
Interpretation/Result • Glandular cells (NOS or specify in comments)
Negative for Intraepithelial Lesion or Malignancy • Atypical
(When there is no cellular evidence of neoplasia, state this in the General • Endocervical cells, favor neoplastic
Categorization above and/or in the Interpretation/Result section of the • Glandular cells, favor neoplastic
report—whether or not there are organisms or other non-neoplastic • Endocervical adenocarcinoma in situ
findings.) • Adenocarcinoma
• Endocervical
• Endometrial
Non-Neoplastic Findings (Optional to Report)
• Extrauterine
• Non-neoplastic cellular variations • NOS
• Squamous metaplasia
• Keratotic changes Other Malignant Neoplasms (Specify)
• Tubal metaplasia
• Atrophy Adjunctive Testing
• Pregnancy-associated changes Provide a brief description of the test method(s) and report the result so
• Reactive cellular changes associated with: that it is easily understood by the clinician.
• Inflammation (includes typical repair)
• Lymphocytic (follicular cervicitis) Computer-Assisted Interpretation of Cervical Cytology
• Radiation If case examined by an automated device, specify device and result.
• Intrauterine contraceptive device (IUD)
• Glandular cells status posthysterectomy Educational Notes and Comments Appended to Cytology
Reports (Optional)
Organisms
Suggestions should be concise and consistent with clinical follow-up guidelines
• Trichomonas vaginalis published by professional organizations (references to relevant publications
• Fungal organisms morphologically consistent with Candida spp. may be included).
• Shift in flora suggestive of bacterial vaginosis
• Bacteria morphologically consistent with Actinomyces spp.
• Cellular changes consistent with herpes simplex virus (HSV)
• Cellular changes consistent with cytomegalovirus
ACIS, Adenocarcinoma in situ; CIN1, cervical intraepithelial neoplasia grade 1; CIN2, cervical intraepithelial neoplasia grade 2; CIN3, cervical intraepithelial neoplasia grade 3; NOS, not
otherwise specified.
From Nayar R, Wilbur DC (Eds.): The Bethesda System for reporting cervical cytology: definitions, criteria, and explanatory notes, ed 3, New York, 2015, Springer.
304
Cervical Squamous Neoplasia 13

implications (and a mandate) for cancer detection Cytologic Alterations That Are Best Left Out
strategies (HPV testing) that augment the Pap test in of the ASCUS Category
this group of reproductive-age women and the important
role of vaccines, which target the principal HPVs associ- These include the following:
ated with this group of tumors.64 1. Mild superficial cell karyomegaly without hyperchro-
masia: This applies particularly to the changes seen in
Nondiagnostic Squamous Atypia association with menopause or women older than 40
(Atypical Squamous Cells of years old (Fig. 13.4A and B).67
Undetermined Significance) 2. Parakeratosis and anucleated squames: Superficial sheets
of parakeratotic cells may contain mild degrees of
Introduction anisokaryosis (see Fig. 13.4C).68
3. Focal nuclear enlargement in atrophic smears (see
ASCUS connotes squamous abnormalities that cannot be Fig. 13.4D): In the absence of hyperchromasia, pleo-
confidently classified as negative or SIL. ASC interpretations morphic forms, or binucleation, these are best interpreted
carry, in aggregate, an approximate 10% risk of a coexisting as benign cellular changes.
HSIL. Historically, this category received considerable 4. Immature metaplasia with uniform nuclei (see
scrutiny with efforts to subclassify the type of ASCUS.65 Fig. 13.4E)
However, with the advent of highly sensitive HPV testing, 5. Reactive squamous epithelial cells with mild karyomegaly
there is less emphasis on cytologic precision and more should not be interpreted as ASCUS. However, the
on the HPV results. distinction from ASCUS is subjective in such instances
The outcome of studies using HPV testing has demon- (see Fig. 13.4F).
strated two important findings, as shown in Table 13.4:
• The disparity in risk between HPV-positive and HPV-
negative ASCUS is approximately twentyfold (20% vs.
Cytologic Criteria for the Diagnosis of ASCUS
1.1%), in contrast to an ASCUS subclassification system, The term “atypia” as defined by the Bethesda System was
in which the disparity between “favor HSIL” and “favor reserved for cases in which the cellular changes are of
reactive” is only tenfold.66 uncertain significance but suggestive of SIL.69 Inevitably,
• Subclassification of ASCUS does not add significant this category includes cases that ultimately prove to be
value to what is already gained by HPV testing. The inflammatory, reparative, or atrophic changes, as well as
role of HPV testing in managing ASCUS is discussed cases that prove to be SIL. If this terminology is to be
here.61 used, it is critical that alterations such as “benign atypia,”
“inflammatory atypia,” or “reactive atypia” and other
recognizably benign alterations be excluded. Whether
alterations “suggestive but not diagnostic of koilocytotic
Table 13.4  Risk of High-Risk Human Papillomavirus, atypia” should be included depends on whether the
High-Grade Squamous Intraepithelial Lesion, or Cancer as a diagnosis of ASCUS is based on cytoplasmic halos alone.
Function of Cytologic Diagnosis In essence, ASCUS is best applied to those alterations that
raise the possibility of a cancer precursor. It is unlikely
Parameter Reference Outcomea Percentage that mild superficial karyomegaly, cytoplasmic halos, and
normal non-atypical parakeratotic cells qualify as significant
ASCUS 115 HPV(+) 30 to 60 cancer risk factors.
ASCUS 115 HSIL 10
The cytologic criteria for ASCUS are designed to identify
abnormalities that are intermediate in severity between
186 Cancer 0.1 those of clear-cut reactive or reparative processes and SILs.
Nuclear enlargement exceeds that typically associated with
ASCUS (HPV+) 115 HSIL 15 to 27b
known benign proliferations but falls short of SIL; hence,
311 Cancer 0.5 the compromise on 2.5-fold differences in size. Nuclear
hyperchromasia may be greater than normal, but the
ASCUS (HPV−) 115 HSIL 1.1 diagnosis of SIL may not be made, because the nuclei do
not contain coarse chromatin or nuclear membrane
Follow-up smear (−) 115 HSIL 3.7
irregularity, are associated with a low nuclear/cytoplasmic
LSIL smear 311 HSIL 27.6c (N/C) ratio, appear degenerated, or are arranged in regularly
spaced cells more characteristic of a benign process.
LSIL cyt/Colpo (−) 311 HSIL 11.3 Alternatively, the cells may be obscured by inflammation,
311 HSIL 11.7
blood, or drying artifact, forcing a qualification of the
LSIL cyt/Biopsy (−)
diagnosis. Finally, the number of cells may be so few that
LSIL cyt/LSIL biopsy 311 HSIL 13 the cytopathologist is uncomfortable with an absolute
a
Outcomes with follow-up of up to 2 years.
diagnosis.68
b
Varies with length of follow-up and cut-off threshold for HPV positive. ASCUS can be subdivided conceptually into several
c
2-year follow-up. morphologic patterns illustrated in Fig. 13.5. It should be
ASCUS, Atypical squamous cells of undetermined significance; colpo, colposcopy; cyt,
cytology; HPV, human papillomavirus; HSIL, high-grade squamous intraepithelial lesion; stressed that these patterns overlap and may not be
LSIL, low-grade squamous intraepithelial lesion. reproducible, but the reader should be aware of them to
305
Diagnostic Gynecologic and Obstetric Pathology

understand the range of nondiagnostic squamous atypias halos and minor degrees of nuclear atypia (see Fig. 13.5,
and how they may be interpreted. For example, a category A-D). It may be argued that this type of ASCUS is the least
(such as, cells suggesting koilocytosis) may be interpreted supportable, inasmuch as cytoplasmic halos without
by one cytopathologist as a mimic of koilocytosis and diagnostic nuclei may warrant a diagnosis of reactive
another as diagnostic. Moreover, superficial cells with mild changes only. The spectrum of such changes begins with
cytologic atypia inherently generate less concern than reactive changes (such as is seen with trichomoniasis) and
immature metaplastic cells with atypia, the latter overlap- ends with koilocytotic atypia.70,71 Cells containing the classic
ping with HSILs that have a metaplastic appearance. irregular densely bordered halos with slight nuclear atypia
Atypical superficial cells resembling koilocytes comprise fall into the category of ASCUS. Cells without any nuclear
a pattern of cellular changes, which include cytoplasmic atypia, irrespective of the appearance of the cytoplasmic

A B

C D
Fig. 13.4.   Changes that should not be called atypical squamous cells of undetermined significance

(ASCUS) include: A and B, mild superficial cell karyomegaly in perimenopausal women; C, parakeratosis
without atypia; D, focal nuclear enlargement in atrophic smears; E, immature metaplasia with uniform
nuclei and discrete cell borders; and F, reactive squamous cells.
306
Cervical Squamous Neoplasia 13

E F
Fig. 13.4, cont’d.

A B
Fig. 13.5.   Cytologic patterns often classified as atypical squamous cells of undetermined significance

(ASCUS) include: A, cells bordering on low-grade squamous intraepithelial lesion (LSIL); B and C, mildly
atypical maturing metaplastic cells; D, atypical parakeratotic cells; E, atypical metaplasia; F, atypical
immature cells with a metaplastic phenotype; G, hyperchromatic clusters of cells with a high nuclear/
cytoplasmic (N/C) ratio; and, H, degenerated atypical cells (less common with liquid-based preparations).
A and D cannot be reproducibly distinguished from LSIL; B, C, and E are least impressive and might
also be classified as reactive changes; F and G were associated with biopsy-proven high-grade squamous
intraepithelial lesion (HSIL) and cervical carcinoma, respectively. Continued

307
Diagnostic Gynecologic and Obstetric Pathology

C D E

F G H
Fig. 13.5, cont’d.

halo, would best be classified as reactive, inasmuch as Fig. 13.5E and F). If the nuclear changes are interpreted
their contribution to cancer risk is negligible. to be those of an SIL, the cytology will be that of a high-
Non-koilocytotic nuclear atypia in mature squamous grade lesion, almost by definition. For most clinicians,
cells, including squamous metaplasia, is another form of this will result in more aggressive management.
ASCUS, which typically presents with two- to threefold Key features in AISM include nuclear sizes of 35 to
nuclear enlargement with mild increases in nuclear chro- 100 µm, with cyanophilic or granular staining cytoplasm,
masia and no alterations in nuclear contour (see Fig. 13.5). N/C ratios generally over 60%, and irregular nuclear
The distinction between this form of ASCUS and SIL is contours. Significant increases in nuclear chromasia are
based on the degree of chromatin coarsening or distur- less likely, particularly in liquid-based preparations. The
bances in cytoplasmic maturation. cells tend to occur in strings and small groups on con-
Atypical immature squamous metaplasia (AISM) poses ventional smears but are frequently isolated on liquid-based
a diagnostic problem as a consequence of the small cell cytology (LBC) preparations. Because of their small size,
size and hence, higher N/C ratio of metaplastic cells (see they can be easily overlooked or mistaken for benign
308
Cervical Squamous Neoplasia 13

histiocytes, small (reserve) squamous cells, or endometrial keeping in mind that some women will continue with
cells. Because of the potential that AISM may accompany, persistent squamous atypia, despite estrogen treatment.
progress to, or represent an HSIL, Pap smears displaying Cohesive sheets or clumps of immature cells, so-called
this abnormality should be carefully scrutinized. This hyperchromatic crowded groups (HCGs), may describe a
pattern is most often reported as ASC-H. variety of conditions, ranging from benign endocervical,
Atypical repair reactions can demonstrate cellular endometrial, or lower uterine segment epithelial cells to
crowding and overlap (as contrasted with typical repair, HSIL, ACIS, and invasive carcinomas. They are discussed
which is in flat sheets) marked variation in nuclear size, here and in Chapter 14.75
prominent and irregular nucleoli, and irregular chromatin Degenerated atypical cells may produce problems in
distribution. Such cases may be difficult to distinguish interpretation, primarily because the degree of nuclear
from invasive carcinoma and SIL (see Fig. 13.5G). Car- irregularity or hyperchromasia may be marked (see Fig.
cinomas often have a tumor diathesis and many isolated 13.5H). In cases where degenerative changes are clearly
atypical cells, features that are usually absent in repair present, in particular when they occur in small numbers
reactions. of cells, follow-up smears should be considered. If degenera-
Regarding squamous atypia in the postmenopausal tion appears to be a secondary phenomenon and the
patient, it is important to preface the remarks below with numbers of cells in question are plentiful, colposcopy
two comments. First, screening is not recommended for should be performed.
those women with no history of a cervical cytologic
abnormality. Second, if an abnormality is detected, HPV Cytologic Criteria for Squamous
testing will likely be part of the workup, and the patient
will be managed accordingly. That being said, the patholo-
Intraepithelial Lesions
gist should keep the following in mind: Cytologic criteria for squamous intraepithelial lesions
• Smears obtained from postmenopausal women not follow the histologic spectrum for early squamous neoplasia
receiving hormone replacement often are air-dried with as classically defined. Fig. 13.6 depicts the traditional
apparent nuclear enlargement, a significant inflammatory histologic categories of CIN1 to CIN3 with some modern
component, and clusters of small dark cells, most likely adjustments, the most substantive of which is the com-
small “parabasal” cells. A common finding is the appear- bining of CIN1, flat or exophytic condyloma, into the
ance of “blue blobs”—degenerated nuclear material, category of LSIL. Exophytic condylomas are excluded
as well as scattered squamous atypia, may be found. from CIN1 by many, but this distinction cannot be made
• Some atypias may resolve with estrogen therapy. Others cytologically except when the high-risk HPV test is nega-
may not, yet still not be corroborated with an abnormal tive, as will occur for HPV-6 or HPV-11 associated lesions.
tissue biopsy. However, for simplicity and ease of management, these
• Squamous atypia in general in older women is less three categories are combined and, by definition, exhibit
likely to be followed by a tissue diagnosis of SIL. principally surface atypia. Progressive loss of epithelial
Symmans et al. correlated abnormal cervical cytology maturation accompanied by greater nuclear atypia heralds
and biopsies with HPV DNA detection using in situ the transition to CIN2 or CIN3, the latter termed high-grade
hybridization.72 The rate of HPV detection in pre- and squamous intraepithelial lesions (HSILs). The following is a
postmenopausal patients with abnormalities was 50% guideline for translating this concept into cytologic criteria
and 10%, respectively, with similar disparities in biopsy- (Figs. 13.7 through 13.9).
proven disease (46% vs. 17%) indicating a low frequency
of HPV DNA in postmenopausal atypias.
• Kaminski et al. found that a single smear with squamous
Low-Grade Squamous Intraepithelial Lesions
atypia in patients older than 50 years old was associated On cytologic smears, the changes associated with LSIL can
with CIN or HPV (biopsy diagnoses) in fewer than 5% be divided into three types:
of cases.73 Moreover, only 9% with repeat cytologic • Intermediate or superficial cells with nuclear enlargement
abnormalities had a biopsy proven SIL. Thus, non- (usually threefold) and hyperchromasia, which sharply
diagnostic squamous atypia in the postmenopausal distinguishes these cells from the adjacent normal cells
woman, even when persistent, has a low likelihood of (see Fig. 13.7A and B): The nuclei exhibit relatively smooth
SIL outcome.74 perimeters and small irregularities in nuclear shape and
In summary, squamous atypia in the postmenopausal contour. Hyperchromasia is present and may take the
period is associated with significant cervical pathology in form of a finely granular chromatin or uniformly increased
a far smaller proportion of cases than in a younger age nuclear density with opaque or smudged appearance.
group. This seems to hold true even in cases with repeated Nucleoli are inconspicuous in both LSILs and HSILs,
atypias; and the “estrogen test,” although helpful in revers- excepting those associated with inflammatory change.
ing atrophy atypia for a significant number of women, • The classic koilocyte: These cells should have enlarged
still resulted in repeat squamous atypia without biopsy- (twofold to threefold) nuclei, and they are usually
proven disease in 14%. hyperchromatic (see Fig. 13.7A). One defining feature
On a practical level, postmenopausal women with is a sharply etched perinuclear halo with an irregular
squamous atypia in a setting of atrophy should be offered perimeter of dense cytoplasm. Another feature support-
an estrogen test and repeat smear to check for reversal of ing the diagnosis of LSIL is nuclear enlargement
the atypia with epithelial maturation. Colposcopy or biopsy and hyperchromasia. Perinuclear halos without any
should be performed in those cases where atypias persist, nuclear enlargement, binucleation, or hyperchromasia
309
Diagnostic Gynecologic and Obstetric Pathology

A B C D E

F G H I J

Normal CIN1 CIN2 CIN3/CIS


(LSIL) (HSIL) (HSIL)
Fig. 13.6.  A classic schematic of cervical intraepithelial neoplasia (CIN; lower) defines the cytopathologic
(A-E) and histopathologic (F-J) transitions from normal to low-grade squamous intraepithelial lesion
(LSIL) (cervical intraepithelial neoplasia grade 1 [CIN1]) to high-grade squamous intraepithelial lesion
(HSIL) (cervical intraepithelial neoplasia grade 2 [CIN2]/cervical intraepithelial neoplasia grade 3 [CIN3]).
This composite addresses the common aspects of histologic-cytologic correlation but does not take into
account nuances of transformation zone differentiation that may further influence morphology (see Figs.
13.23 to 13.27). CIS, Carcinoma in situ.

are not sufficient for a diagnosis of an SIL, inasmuch • The abnormal cells are characteristically less mature
as perinuclear halos alone have not been shown to and exhibit a higher N/C ratio than those of LSIL.
correlate independently with the presence of HPV. • Nuclear enlargement is generally in the same range as
Binucleation is common in LSIL and is found in in LSILs, but because the N/C ratio is increased, the
intermediate or superficial cells. Just as with the peri- cells appear smaller.
nuclear halo, binucleation is more specific for the • Hyperchromasia, coarse chromatin, and membrane
diagnosis of LSIL if it is accompanied by some of the contour irregularity are all more conspicuous than
other features, such as nuclear enlargement or in LSIL.
hyperchromasia. • Architecturally, the cells of HSILs are arranged in two
• The most subtle manifestation of LSIL is a change main patterns: (1) as cohesive groups of cells with
confined predominantly in the parabasal-type cells. The indistinct cell borders (syncytial-like groupings) or
degree of parabasal cell atypia may vary, and in smears (2) as individual cells arranged in rows or streams.
containing LSIL, conspicuous atypia is not present. In Small, immature squamous cells and mature keratin-
immature condylomas (discussed later), cells with a izing cells with marked nuclear atypia are classified
parabasal/intermediate appearance are most prominent as HSIL.
and show slight nuclear enlargement, slightly more The reader can surmise from the previous discussion that
granular chromatin, and binucleation. We have termed it takes more than just an assessment of cell maturity to
a subset of such cells “abortive koilocytes” inasmuch distinguish low-grade from high-grade precursors. The
as they exhibit some features of these cells (nuclear nuclear abnormalities associated with high-grade precursors
enlargement, indistinct perinuclear halos) but lack the can be appreciated in not only the basal/parabasal cells but
more striking nuclear hyperchromasia and perinuclear also in intermediate and superficial-type cells. It should be
clearing. emphasized that a portion of HSILs will be associated with
low-grade morphology. As many as three-quarters of SIL
lesions of all grades have been reported to contain coexisting
High-Grade Squamous Intraepithelial Lesions koilocytes.76 From 10% to 20% of LSILs on a Pap test are
On cytologic preparations, the cells of HSIL (see Fig. 13.8) confirmed as HSIL on biopsy.77 Moreover, high-risk HPV
display the following: types are also associated with LSIL.3 At the practical level,
310
Cervical Squamous Neoplasia 13

A B

C
Fig. 13.7.  Cytology of low-grade squamous intraepithelial lesions (LSILs), including koilocytes (A),
and superficial cell karyomegaly and binucleation (B, C). The principal features are variation (and
increase) in nuclear size and chromatin density.

the diagnostic pathologist must always consider the possibil- Differential Diagnosis of Low-Grade and
ity that an LSIL, even one that closely resembles a condyloma, High-Grade Squamous Intraepithelial Lesion
may comprise a portion of (or coexist with) a larger lesion
that contains high-grade morphology. Occasionally, a classic The following are the most common pitfalls in the diagnosis
LSIL will merge with a CIN3. These facts must also be of either LSIL or HSIL on cytologic preparations. The reader
appreciated when interpreting cytology. should bear in mind that many of the “pitfalls” of cytologic
One particular presentation that should be noted is HSIL interpretation also fall under the category of nondiagnostic
composed of small, metaplastic-type cells. Some HSILs squamous atypias:
are composed of exfoliated small, metaplastic-type cells • Mimics of LSIL include the following: (1) trichomonas
with mild to moderate atypias, a correlate of a subset of infections with mild perinuclear halos, (2) postmeno-
similar-appearing lesions on histology.78,79 The extremely pausal nuclear karyomegaly (see Fig. 13.9A), and (3)
coarse chromatin and bizarre shapes typifying conventional nonspecific reactive changes leading to mild superficial
HSIL are not present. Because of the relatively mild atypia, nuclear atypia.
the diagnosis of such lesions is poorly reproducible, and • Mimics of HSIL include (1) atrophic changes, which
they are often diagnosed as ASCUS or ASC-H, if not include a high N/C ratio but greater regularity in
overlooked completely (see Fig. 13.8C and D). The nuclear contour and absence of coarse chromatin (see
important features distinguishing this subset of HSIL from Fig. 13.9B and C); (2) sampling of lower uterine
benign metaplasia are the small size of the cells, increased segment, in which syncytial groups of lower uterine
N/C ratio, and irregularly shaped nuclei. The latter changes segments may be confused with HSIL; and (3) adeno-
may be subtle. carcinoma in situ, which may also be difficult to
311
Diagnostic Gynecologic and Obstetric Pathology

B C
Fig. 13.8.   Cytology of high-grade squamous intraepithelial lesions (HSILs), including: A, abnormal

cells with moderate cytoplasm, anisokaryosis, and coarse-appearing chromatin; B and C, similar nuclear
features; and a higher nuclear/cytoplasmic (N/C) ratio.

separate from HSIL (see Chapter 14). Occasionally the diagnosis of HSIL is justified, and it may not be
immature condylomas may be difficult to distinguish possible to exclude the potential of invasive carcinoma.
from HSIL on cytology (see Fig. 13.9D); however, this If the changes are of a lesser degree and the distinction
dilemma is uncommon. between LSIL and HSIL is not clear, the diagnosis “SIL,
• Is it LSIL or HSIL? Pitfalls in the diagnosis include difficult to grade” is appropriate.
keratinizing lesions, including dyskeratosis or atypical
parakeratosis (Fig. 13.10A-C). Mild surface binucleation
in the absence of hyperchromasia correlates poorly with Cervical Cancer Prevention and Management of
LSIL (see Fig. 13.10A). In general, keratinizing cells the Abnormal Papanicolaou Test
consisting of small round to oval hyperchromatic nuclei Current Guidelines for Screening and Human
with uniform chromatin distribution and binucleation Papillomavirus Testing
and plaquelike clusters correlate with LSIL (see Fig. Presently the following are recommended for the most
13.10B and C). Greater disturbances in cellular dif- cost-effective cervical cancer prevention:
ferentiation, characterized by densely hyperchromatic • Pap tests are not performed until age 21.
nuclei, variable keratinization, and increased N/C ratio, • Pap tests are performed between age 21 and age 30.
are characteristic of HSIL (see Fig. 13.10D-F). When • After age 30, women can be followed by HPV testing
the pathologist encounters changes of this magnitude, alone.
312
Cervical Squamous Neoplasia 13

A B

C D
Fig. 13.9.   Cytologic mimics of squamous intraepithelial lesion (SIL). A, Menopausal karyomegaly

with cytoplasmic halos mimics a low-grade squamous intraepithelial lesion (LSIL). Mimics of high-grade
squamous intraepithelial lesion (HSIL) include, B and C, atrophic changes and, rarely; D, immature
condyloma.

Co-testing with smear and HPV testing versus HPV testing field at the specter of a missed or undiagnosed cancer or
alone is still being debated. precursor that could develop in such an accessible site
and eventually kill a woman. This chapter does not list
Management of ASCUS and Squamous all of these algorithms, but readers are referred to the
ASCCP website (www.asccp.org). The principles behind
Intraepithelial Lesions these algorithms include the following:
Numerous algorithms, some rather ponderous, have been • High risk HPV-positive ASCUS is typically referred to
established for the management of women with cytologic colposcopy with two caveats:
abnormalities. The complexity of these algorithms contrasts • Management of women younger than 21 to 25 years
with the very low risk overall of developing cervical cancer, old with any abnormal Pap test is more conservative
but they reflect the continued fear by all members of the to avoid precipitating a path to unnecessary cone
313
Diagnostic Gynecologic and Obstetric Pathology

(loop electrosurgical excision procedure [LEEP]) referred to colposcopy, but as mentioned earlier, a
biopsy. follow-up interval is permitted in women age 21 to 25.
• More refined testing for HPV-16/18 permits a more • Atypical glandular cells are always pursued with care,
conservative approach to high-risk HPV-positive although negative HPV testing provides a follow-up
ASCUS, allowing continued follow-up. option.
• HPV testing can be taken into account for not only • Follow-up intervals are lengthened to 3 years following
ASCUS but ASC-H and LSIL. ASC-H and HSIL are always negative HPV and cytologic findings.

A B

C D
Fig. 13.10.   Abnormal parakeratosis and abnormal keratinizing cells. A and B, Binucleation and more

subtle differences in nuclear size and staining intensity in mature cells are generally more common in
low-grade squamous intraepithelial lesion (LSIL). C and D, More pronounced nuclear disturbances with
coarse chromatin raise the possibility of high-grade squamous intraepithelial lesion (HSIL).
314
Cervical Squamous Neoplasia 13

E F
Fig. 13.10, cont’d. E and F, Striking abnormalities in nuclear contour and chromasia coupled with
dense cytoplasmic keratinization characterize HSIL and may be associated with invasive carcinoma.

an abnormality in the cervical transformation zone that


Histopathologic Diagnosis of Preinvasive Disease requires ablation or surgical (LEEP) removal.
Expectations From the Clinical Setting • A woman younger than 25 years old should be managed
When the clinician sends a biopsy to the pathologist for with care, particularly if there is a question of whether
a diagnosis, it is usually based on an abnormal Pap smear. her cervical abnormality qualifies as an HSIL or is a
Although the practitioner may expect the biopsy to explain questionable CIN2. The diagnosis of CIN2 imposes a
the abnormality on the smear, approximately 70%, 45%, LEEP excision on fewer patients due to the more recent
and 20% of cytologic interpretations of ASCUS, LSIL, and guidelines but is subject to a high level of interobserver
HSIL, respectively, will not be verified on cervical biopsy. discordance. When relegating a patient to LEEP excision,
The colposcopist should expect this. Explanations for such review of such cases by more than one observer may
discrepancies include clinical sampling error and diagnostic be helpful in coming to a therapeutic decision.
imprecision, particularly in the interpretation of ASCUS.
The role of colposcopy is to exclude cancer and
Practical Considerations Before Examining the Biopsy
assess the transformation zone (Fig. 13.11A-D). In many
cases, the colposcopist will not identify a lesion and will Examination of the cervical specimen requires an under-
not perform a biopsy. The pathologist may receive an standing of four components germane to interpreting
endocervical curettage (ECC) only and, again, should cervical squamous neoplasia:
approach this specimen with realistic expectations. The • Excluding non-neoplastic infections, such as herpes or
pathologist must avoid overcalling minor histologic changes chlamydia
in an effort to achieve consistency between the cytologic • Understanding the transformation zone and the plethora
and histologic findings. In turn, the colposcopist should of benign alterations in epithelial differentiation that
be wary of cytologic/histologic correlation rates that are characterize this region
high (over 80%) or diagnoses of SIL that follow negative • Applying the criteria for determining whether a lesion
or equivocal colposcopic findings. is LSIL, HSIL, or of indeterminate grade
The pathologist who is interpreting the biopsy should • Understanding the influence of the various differentia-
be aware of the previously described odds of detecting an tion patterns in the region of the SCJ and their influence
SIL and realize several important realities that will be on morphology of SILs
repeated later in this section:
• That an HPV-positive ASCUS or LSIL diagnosis on
Excluding Infections Other Than
cytology that is confirmed to be LSIL or less on biopsy
Human Papillomavirus
carries a 5% to 13% risk at 2 years of HSIL on
follow-up.80,81 Ostensibly, cervical sampling permits the identification and
• The most important practical issue, following exclusion qualification of HPV-related cervical neoplasia. However,
of malignancy, is to determine whether the patient has the issue of infection by other microbial agents must be
315
Diagnostic Gynecologic and Obstetric Pathology

A B

C D
Fig. 13.11.  A, Colposcopy photographs of prominent transformation zone in a diethylstilbestrol-exposed
patient. B and C, Low-grade squamous intraepithelial lesions (LSILs). D, Following cryotherapy, the
transformation zone is ablated and reepithelialized with mature squamous mucosa. (Photographs courtesy
of Richard U. Levine and Alex Ferenczy.)

considered when evaluating any cervix. Infections that can smudged (“ground glass”) chromatin and are typically at
be identified or suspected by histologic examination of the the periphery of the ulcer, in the spongiotic epithelium, or
cervix include herpes simplex viruses (HSVs) (Fig. 13.12A as single desegregated cells. Special stains may be helpful if
and B), cytomegalovirus (see Fig. 13.12C and D), Chlamydia the diagnosis is uncertain on morphologic grounds alone.
trachomatis (see Fig. 13.12E-G), and, rarely, adenovirus.82,83 C. trachomatis infection does not elicit specific epithelial
HSV should be suspected and excluded in any ulcer, par- changes but has been associated with follicular cervicitis
ticularly if it is accompanied by epithelial cell necrosis and in one report.84 This may not be sufficient to confirm the
acute inflammatory exudate. The diagnostic cells contain association, but we do mention follicular cervicitis and
one or multiple nuclei with either discrete inclusions or the possible association when it is encountered.
316
Cervical Squamous Neoplasia 13

A B C

D E F

G H
Fig. 13.12.   Infections of the cervix that can be identified morphologically include: A, herpes simplex,

presenting as an ulcer with epithelial necrosis and, B, intranuclear inclusions; C, cytomegalovirus, and,
D, chlamydia, which may be associated with follicular cervicitis. E, Inclusions are best seen by electron 317
microscopy (arrows). F, They contain primary and secondary particles (small and large arrowheads). By light
microscopy, inclusions are extremely subtle (G), but may be identified by immunohistochemistry (H).
Diagnostic Gynecologic and Obstetric Pathology

The fact that most significant precancerous lesions arise


The Squamocolumnar Junction and the
in the SCJ implies that infection of those cells or their
Transformation Zone
immediate precursors, either on the surface or in subjacent
The cell of origin for this spectrum of squamous and pos- crypts, is paramount to the genesis of cervical cancer.
sibly columnar differentiation resides in or near the SCJ. Inasmuch as the progeny of the SCJ cells can evolve to
Recent work has highlighted a population of cells at the SCJ either mature squamous or columnar mucosa, it is reason-
that may be important progenitors to the entire spectrum able to assume that as these cells progressively mature,
of cellular differentiation (see Fig. 13.1). Moreover, one the risk of significant neoplasia diminishes. Fig. 13.14A
can envision that the spectrum of at-risk epithelium is illustrates a schematic of the SCJ and the transformation
derived from this population. Furthermore, it is possible zone, depicting the regions less vulnerable to carcinogenic
that the risk of a given epithelium evolving to produce an HPV infection (ectocervix) and the region most at risk
HSIL following HPV-16 infection is linked to its location (SCJ). The evidence for this gradient of vulnerability is
(i.e., within the SCJ vs. more mature squamous elements largely indirect but based on the following:
in the transformation zone) (Fig. 13.13). • Reserve cells, which are also produced during embryonic
Based on the work by Herfs et al., the SCJ develops development and are common more cephalad to the
during fetal and early postnatal life as a result of three SCJ junction, as well as in endocervical polyps and
phases: microglandular hyperplasia: None of these sites are
• A population of p63-positive basal cells emerges beneath particularly vulnerable to HSIL, suggesting that once reserve
cytokeratin 7 (CK7)-positive embryonic müllerian cells are established, their risk of HPV-associated neo-
epithelium. plastic transformation is reduced.
• The CK7-positive müllerian epithelium exfoliates as • Immature squamous metaplasia, like reserve cells, has
the squamous differentiation progresses. This process a similar distribution yet a very low risk of HSIL away
is virtually identical to what can be seen in microglan- from the SCJ, suggesting that once squamous metaplasia
dular hyperplasia in adults, wherein immature columnar ensues, the risk of neoplastic transformation more closely
cells give rise to reserve cells that in turn mature to approximates that of the ectocervical or vaginal mucosa.
form squamous metaplasia. • Mature squamous epithelium a few millimeters away
• The result is a SCJ where mature squamous epithelium on the ectocervix is particularly resistant to HSIL.
joins a mature tall endocervical columnar population. • The risk of HSIL once the SCJ is removed is low. More-
However, on close examination, a small monolayer over, most of the recurrences develop on the ectocervix,
of cuboidal cells remains at this junction, with a few implying that the vulnerable SCJ cells do not regenerate.
cells extending over the leading edge of the squamous In particular, once persistent disease is excluded by an
mucosa. These cells have been designated residual interval without an abnormal smear or the presence of
embryonic cells. They are illustrated in red in the HPV, the risk of HSIL is exceedingly low.86,87
schematic in Fig. 13.14A and in the CK7 and p16 The aforementioned model supports the notion that
stained section of an early lesion in Fig. 13.14B. The there is a population of cells at the SCJ that could be
implication is that these cells, like their embryonic destined for both squamous and columnar cell differentia-
predecessors, still maintain the ability to undergo tion (metaplasias) that develop in response to inflammatory
squamous and possibly columnar differentiation (see stimuli and alterations in pH that occur following the
Figs. 13.1 and 13.13).3,85 onset of menarche. The squamous component emerges

Endocervix J Transformation Ectocervix


(columnar) u zone (squamous)
n
c (squamous)
t
i
o
n

n
ctio
Jun

Fig. 13.13.   Cytokeratin 7 (CK7)-stained section of the transformation zone and squamocolumnar

junction (SCJ). Note there are four different epithelial “targets” for human papillomavirus (HPV) infection,
including the ectocervix, metaplastic epithelium between the ectocervix and SCJ (transformation zone),
the SCJ, and the endocervical epithelium. (From Herfs M, Yamamoto Y, Laury A, et al: A discrete population
of squamocolumnar junction cells implicated in the pathogenesis of cervical cancer. Proc Natl Acad Sci
USA 109:10516–10521, 2012.)
318
HPV Conventional bottom-up
HPV infection/differentiation

SCJ-negative LSIL

Krt7

Novel top-down HPV Expansion of


Infection/differentiation
the SCJ cells
Postinfection

SCJ-positive HSIL p16

A B

C
Fig. 13.14.   A, Schematic of the lesion development in the squamocolumnar junction (SCJ) and transformation zone. This depicts different

scenarios of infection. The traditional mode of infection was via the virus accessing basal cells through defects in the epithelium, typically mature
metaplastic or ectocervical epithelium. In contrast, direct infection of the SCJ leads to outgrowth of infected cells which undergo squamous differentia-
tion. B, Early high-risk infection of the SCJ with partial expansion of cytokeratin 7 (CK7) and p16-positive cells. C, Two high-grade squamous
intraepithelial lesions (HSILs), in two columns. Upper images depict CK7 positivity in the superficial cell layers emblematic of the SCJ cells (arrows).
Middle images depict strong p16 staining in all layers including the surface (arrows). Bottom images illustrate human papillomavirus (HPV) nucleic
acids in the surface cells too. The impression is that the residual SCJ cells are being undermined by outward “top-down” expansion of the HSIL.
(C, From Herfs M, Vargas SO, Yamamoto Y, et al: A novel blueprint for ‘top down’ differentiation defines the cervical squamocolumnar junction 319
during development, reproductive life, and neoplasia. J Pathol 229:460-468, 2013.)
Diagnostic Gynecologic and Obstetric Pathology

in the reserve cells that are highlighted by p63, which is (shown in red in Fig. 13.15A) HSILs develop by “top
recognized in basal cells of skin appendages, myoepithelial down” differentiation (i.e., they develop via neoplastic
cells in the breast and salivary gland, and sub-columnar squamous metaplasia by induction of p63 within the
cells in the prostate.88 This gene is critical to the mainte- SCJ population). Consequently, these SCJ cells transition
nance of stem cell characteristics in squamous epithelium to a neoplastic population and are both HPV and p16
but in the cervix marks the transition from columnar to positive (see Fig. 13.15C). In essence, the process is
squamous lineage as a reserve cell marker.89,90 This transition identical to what happens during embryogenesis with
and the squamous epithelium that eventuates from the the exception that it is driven by an oncogenic HPV.
expansion of these cells define the transformation zone • In contrast, LSILs are more heterogeneous with respect
(see Fig. 13.13). The transformation zone is delineated to CK7 staining. CK7-negative LSILs tend to be cytologi-
by the squamous epithelium extending over recognizable cally bland and appear to arise in mature transformation
crypt epithelia signifying the region where the columnar zone epithelium or ectocervix. Moreover, these lesions
cells became replaced by squamous metaplasia. The reader have a lower risk of HSIL outcome, presumably because
can imagine two zones of susceptibility, that of the trans- they are not the consequence of direct infection of the
formation zone and that of the SCJ, the latter more vulner- SCJ cells. They also have a lower rate of p16ink4+ and
able to HSIL following infection by oncogenic HPVs because HPV-16+.91,92
the cells have yet to differentiate (see Fig. 13.14). • Unlike their counterparts in the ectocervix or mature
SCJ cells stain intensely with CK7, a property unique transformation zone, CK7-positive LSILs appear to arise
relative to the mature transformation zone epithelium in the SCJ or in immediate metaplastic progeny. These
and to a lesser degree, the mature endocervical epithelium. lesions may have a higher likelihood of HSIL outcome,
CK7 staining revealed the following in the cervix, sum- a higher rate of p16ink4+ and HPV-16+. Moreover, we
marized and illustrated in Fig. 13.15: found that these “LSILs” might generate greater disagree-
• We discovered that the majority of HSILs in the cervix ment between observers.92,93
stained positive for CK7, suggesting an origin within • Thus, we concluded that most HSILs will stain for SCJ
the SCJ.3,85 Fig. 13.15C illustrates how many HSILs markers and that LSILs arising in the SCJ may be more
contain strong CK7 staining, and this marker highlights problematic diagnostically. However, despite this, lesions
cells on the surface reminiscent of SCJ cells. The impres- classified as LSIL by multiple observers still had a very low
sion is that under conditions that favor neoplastic rate of HSIL outcome. These three groups of lesions are
expansion of the SCJ cells following HPV infection illustrated in Fig. 13.15.91

LSIL InSIL HSIL

A C E

B D F

SCJ marker Negative Positive Positive


HR HPV+ 50% 100% 100%
HPV-16+ 10% 60% 60%
p16ink4+ 27% 74% 90%
Agreement High Low High
Fig. 13.15.  Lesions emerging from the squamocolumnar junction (SCJ) and transformation zone may
exhibit a range of histologies, and the origin of the lesion will influence histology and consequently,
interpretation. A diagnosis of low-grade squamous intraepithelial lesion (LSIL) in the ectocervix (SCJ-)
(A and B) can be made with good agreement. Similarly a diagnosis of high-grade squamous intraepithelial
lesion (HSIL) (CIN2-3) in the SCJ (E and F, SCJ+) will have a high reproducibility. However, a subset of
lesions in the SCJ will be more problematic (C and D), generating less agreement. We interpret these as
lesions of uncertain or intermediate grade (CIN1-2). Associations with high-risk human papillomavirus
(HR HPV), HPV-16 and p16 staining patterns are enumerated. What is clear is that any lesion arising in
the SCJ is highly likely to contain high-risk HPV and be strongly positive for p16, yet may defy classification
as either LSIL or HSIL. InSIL, Intermediate-grade squamous intraepithelial lesion.
320
Cervical Squamous Neoplasia 13

additional patterns not accounted for in traditional clas-


Milestones in the Classification of Squamous
sifications. Lesion categories are summarized in Table 13.5.
Intraepithelial Lesions
The classification and management of SILs has evolved
SILs are defined as squamous alterations in the cervical since the late 1960s.154 This is summarized in Fig. 13.16.
transformation zone that are induced by HPV infection. The key points in this evolution are as follows:
Differences in the differentiation level or differentiation • CIN was originally defined as a proliferation containing
pathway of the underlying mucosa may influence the atypias in all layers of the epithelium.94 The reader can
morphologic presentation of cervical lesions, leading to imagine that lesions classified as CIN in the 1960s were

Table 13.5  Descriptive Categories of Low-Grade Squamous Intraepithelial Lesion and High-Grade Squamous
Intraepithelial Lesion

Category Descriptors Human Papillomavirus p16 Immunostaining

LSIL CIN1, flat condyloma, mild dysplasia HR (70%) Usually diffuse

Exophytic condyloma LR Negative or patchy

Immature condyloma (papillary immature metaplasia) LR Negative or patchy

Immature flat metaplastic LSIL HR Usually diffuse

HSIL CIN2 or moderate dysplasia HR (45% type 16) Diffuse

CIN3 or severe dysplasia/carcinoma in situ HR (60% type 16) Diffuse

Keratinizing SIL HR Diffuse

HSIL with immature metaplastic phenotype HR Diffuse

Papillary carcinoma in situ HR Diffuse

Adenosquamous carcinoma in situ HR Diffuse


CIN, Cervical intraepithelial lesion; HR, high risk; HSIL, high-grade squamous intraepithelial lesion; LR, low risk; LSIL, low-grade squamous intraepithelial lesion; SIL, squamous
intraepithelial lesion.

1960s Mild Moderate Severe CIS

Follow Cone/hysterectomy

1970–80s Condyloma/CIN1 CIN2 CIN3

Cryotherapy Laser/cone

1990s LSIL HSIL (CIN2) HSIL (CIN3)

Follow LEEP

Future? LSIL HSIL (CIN2)* HSIL (CIN3)

Follow LEEP
Fig. 13.16.  Management of cervical lesions placed in historical perspective. Note that the distinction
of moderate from severe dysplasia became less important when the therapeutic options included cryotherapy
and laser ablation (1970s through 1980s). In contrast, when the therapeutic options included loop
electrosurgical excision procedure (LEEP) versus follow-up, the distinction of cervical intraepithelial
neoplasia grade 1 (CIN1) (low-grade squamous intraepithelial lesion [LSIL]) from cervical intraepithelial
neoplasia grade 2 (CIN2) (high-grade squamous intraepithelial lesion [HSIL]) became more critical and
small differences in interpretation had significant impact on management. However, in the vaccine era
and with the application of biomarkers or new algorithms, CIN2* conceivably may be viewed more
critically to allow for conservative management of a subset of women with this diagnosis. CIN3, Cervical
intraepithelial neoplasia grade 3; CIS, Carcinoma in situ.
321
Diagnostic Gynecologic and Obstetric Pathology

unlikely to be confused with condylomas of any type. • The range of epithelial changes associated with HPV
In fact, the classic treatise by Burkhardt contains no has expanded further as markers for high-risk HPV (p16)
examples of flat or exophytic condylomas.95 Koss implied have been employed.
in particular in 1955 that lesions containing koilocytosis • An excessive reliance on p16 as a marker to discriminate
were not technically dysplasias because the basal epi- not only lesion from no lesion but also LSIL from HSIL
thelial cell layers were not atypical.96 (CIN2).
• Meisels and Purola and Savia in 1976 popularized the • The realization that the interobserver reproducibility
cervical flat condyloma, which logically became more for the diagnosis of CIN2 (vs. CIN1) is sufficiently poor
prevalent with earlier workup of abnormal cervical to call into question the significance of a diagnosis of
cytology. Meisels assigned the term “atypical condyloma” CIN2 when it is rendered by a single observer.92
to those lesions harboring both koilocytosis and basal • The fact that even an adjudicated CIN2 (one that
atypias.97-99 multiple observers agree on) carries a 40% likelihood
• This created for the time being a four-class system, flat of resolving in 6 months and an over 60% likelihood
condyloma, CIN1, CIN2, and CIN3, one favored by of resolving within 2 years without treatment.102,105
the British group.100 Less common exophytic condylomas • The accumulative data now show that LEEP excision
were segregated from this classification or lumped with carries an increasing albeit variable risk of premature
flat condyloma. We would emphasize that this approach rupture of membranes and premature labor in subse-
stays true to the original notion that CIN should not quent pregnancies.103 This management tool is too drastic
contain so-called “pure” HPV infections that do not for a condyloma or CIN1, which can be managed
display atypia in the more basal epithelial layers. The by observation alone because of the low risk of HSIL
only caveat is the difficulty in consistently separating outcome.81
three grades of CIN once flat and exophytic condylomas • LEEP is probably overused, given the distinction of CIN1
have been removed. This is one reason why a three-grade from CIN2 is made with variable interobserver reproduc-
system now classifies the flat condyloma and related ibility. This is the basis for recommendations to employ
lesions as CIN1. a period of follow-up when managing women younger
• The shift in philosophy for the CIN classification was than 25 years old.
facilitated by the Bethesda System for cytologic classifica-
tion, which equated condyloma and CIN1 on cytology. Interobserver Agreement for the Diagnosis of CIN2 Is
CIN1, once considered an important precursor, was Fair at Best
“decriminalized” to be combined with condyloma. Thus, Based on published reports, the interobserver agreement
traditional CIN lesions were reduced to two groups, for the diagnosis of CIN2 is highly variable. Overall, in
CIN2 and CIN3, and this concept spread to histologic one study employing experienced pathologists a unanimous
interpretation. verdict (based on independent review) of CIN2 was reached
• Cryotherapy, used extensively in the 1980s to treat CIN1 in only 19.4% of cases reviewed, and in 31.6% and 47.5%
and CIN2, was usually replaced by LEEP excision, with of CIN1 and CIN3, respectively.104 In the ASCUS/LSIL triage
CIN1 lesions relegated to follow-up. This resulted in study (ALTS), quality control (QC) reviewers downgraded
CIN2 being the diagnosis that would result in LEEP.101 29% of CIN2 to CIN1 or less.105 In another study, the
• Reproducibility of CIN2 has been shown to be poor, diagnosis of CIN2 by initial reviewers was proven highly
and follow-up studies have shown that up to two-thirds unreliable, with two study pathologists agreeing on only
of CIN2s in young women will regress in 2 years, 40% 13% and 32%.106 A summary of four studies comparing
in 6 months.102 interobserver reproducibility for the diagnosis of CIN2
• Recently the LAST project redefined CIN1 as LSIL and noted kappa values of 0.22 (poor), 0.41 and 0.44 (fair),
CIN2-3 as HSIL, with the proviso that CIN2 be managed and 0.57 (good). The authors noted a high rate of interob-
conservatively in young women by follow-up when server agreement for scoring lesions as p16 positive (κ =
possible.1 0.87–0.91) and that by combining p16 and H&E interpreta-
• In addition, as closer attention has been paid to the tion, the kappas ranged from 0.62 to 0.75. The implication
SCJ and to immature proliferations using biomarkers was that the presence of p16 positivity facilitated the
such as p16, it has become clear that the range diagnosis of CIN2.92
of proliferations containing HPV nucleic acids has In our practice, relatively inexperienced pathologists had
expanded. Many of these are immature “metaplastic” good agreement on CIN1 or CIN3 with kappa values of
SILs that must be addressed within the classification approximately 0.67. However, agreement on CIN2 was
system.78 fair with values of 0.22.

Association Between p16 Immunostaining


Classification Systems and Approaches to Squamous
and SIL Grade
Intraepithelial Lesion Diagnosis
There is a consistent association in the literature between
In the past edition, the discussion of SILs proceeded with “strong” p16 immunoreactivity and either HSIL or the
classification (LSIL vs. HSIL), criteria, and a discussion of presence of a high-risk HPV type. Sano et al. noted that
biomarkers (p16ink4, MIB1, etc.) in diagnosis. Since then, over 98% of high-risk HPV infections exhibited diffuse
classification has become impacted by several variables: p16 staining, usually defined as linear strong staining.107
• Attention to the SCJ has uncovered early forms of SIL Subsequent reports noted positive staining from 64% to
previously underappreciated. 100% of HSILs, the lesser estimate possibly an artifact
322
Cervical Squamous Neoplasia 13

created by the lack of a clear biologic distinction between sia.20 There are three valid reasons for questioning the use
strong and diffuse and strong but focal staining.86,109 Stain- of this diagnostic term:
ing frequencies of LSILs have varied as a function of the • There is much greater reproducibility in separating CIN3
frequency of associated low-risk versus high-risk HPVs. from CIN1.107
Klaes et al. and Sano et al. noted no diffuse staining in • There is a high rate of short-term resolution for lesions
low-risk HPV infections or associated LSILs.108 Klaes et al. classified as CIN2.
observed that 87% of LSILs associated with high-risk HPV • There is the recent widespread adoption of the terms
exhibited diffuse staining. Herfs et al. noted lower rates LSIL and HSIL to connote lesions with different risks
of p16 positivity in CK7-negative LSILs versus CK7-positive of cancer outcome.2
lesions, but this appears to reflect differences in association These facts leave the practitioner with several options
with high-risk HPVs between the two groups.91 Overall, for managing cervical biopsies with suspected SIL:
approximately a low estimate of one-third to a high estimate • Classify the lesions as LSIL or HSIL and rely on a second
of two-thirds of LSILs stain strongly for p16.3 observer to confirm a suspected lesion at the low end
of HSIL (CIN2).
Predicting Cervical Intraepithelial Neoplasia • Rely on p16 staining to adjudicate the diagnosis of
Grade 2–Positive Outcome of p16 and/or borderline lesions, classifying p16 positive cases as CIN2.
Cytokeratin 7–Positive Low-Grade Squamous • Apply strict criteria for the diagnosis of LSIL and HSIL,
Intraepithelial Lesion reserving a “gray zone” for lesions that cannot be
A number of reports tracking the histologic outcome of diagnosed as HSIL with confidence. Lesions in the gray
CIN1 cases stained with p16 noted a distinctly higher zone would be classified as SIL of intermediate (or
frequency of CIN2-3 on follow-up.109,110 In these studies, indeterminate) grade (INSIL). This term would connote
from 10.3% to 35.4% of p16-positive cases had an HSIL lesions in the CIN1 to CIN2 category. The reader might
outcome. In contrast, a much narrower range of p16-negative take issue with this, inasmuch as the accepted diagnostic
LSILs (0%–4.4%) had an HSIL outcome. Several studies term is CIN2. However, if this entity cannot be diagnosed
showed the association between p16 and HSIL outcome reproducibly and if the biomarker used (p16) does not
to be either of arguable significance or not significant. predict outcome with any degree of precision, a term
One found no differences in risk, but the HSIL outcome reflecting this uncertainty (CIN1 or CIN2) has merit.
for both groups was inordinately high. Another found The discussion later of diagnostic criteria for SIL will
p16 only predictive of an HSIL outcome in the first year be made with an appreciation of this conundrum.
of follow-up. Another found a significant association
between p16 and HSIL outcome but it was not corroborated Diagnostic Criteria for Squamous
in multivariate analysis.111 One study found no significant
association between p16 positivity and HSIL outcome. In
Intraepithelial Lesions
that study, the HSIL outcome for p16 positive and negative The diagnostic approach to SILs involves not only address-
LSILs was 15.0% and 9.4%, respectively. In our experience, ing the distribution and quality of the atypias but also
it was 10.3% and 5.7%, respectively.92 the maturation of the epithelium. This discussion addresses
Two notable observations from the literature pertain to first the more conventional patterns followed by the
interstudy predictions of HSIL outcome from p16 staining “immature metaplastic” patterns. In both groups, we will
and observer agreement. Sagasta et al. summarized sensitiv- segregate them into obvious LSIL and HSIL and illustrate
ity, specificity, and positive and negative predictive values a group that is more problematic, so-called “intermediate
for p16 staining as predictors of HSIL outcome based on grade” SILs.
prior reports.113 Respective ranges for these parameters were
59 to 100, 37 to 72, 10 to 35, and 90 to 100. The most
consistent finding across all of these studies was the negative
Low-Grade Squamous Intraepithelial Lesions
predictive value for HSIL outcome of a p16-negative LSIL The diagnosis of LSIL presumes the presence of high- or
(90–100). low-risk HPV infection that produces a degree of cytologic
If the reader surveys most of the studies that address atypia that falls beneath a threshold for worrisome changes
“progression,” he or she will note that very few illustrate that might herald an HSIL or higher. The diagnosis requires
entry and exit (outcome) histology. Given the lack of first excluding benign mimics of LSIL, which is discussed
illustrations in most studies and the lack of reproducibility later. The second step is to confirm the presence of the
across multiple observers for the diagnosis of CIN2, it is characteristic cytologic atypia.
easy to speculate that making claims of progression from The features of LSIL in mature squamous epithelium
CIN1 to CIN2 is a hazardous exercise when evaluating include the following (Fig. 13.17):
the value of any biomarker, such as p16 or CK7. Moreover, • Conspicuous superficial cell atypia with binucleation,
the magnitude of the association may be insufficient to twofold nuclear enlargement, and variable nuclear
validate the biomarkers as predictors of outcome. This chromasia
seems to be the case in the more recent studies evaluating • Generally low N/C ratio in maturing epithelial cells
both p16 and CK7.93,108,112,113 with preserved cytoplasmic differentiation
The lack of clarity regarding the diagnostic criteria • A subtle expansion of the lower third of the epithelium,
for—and significance of—CIN2 begs the question as to signifying a mild delay in epithelial cell maturation
whether this diagnosis has any real value in the current • A range of nuclear features in the lower third of the
management of women with preinvasive cervical neopla- epithelium that includes either euchromasia or uniform
323
Diagnostic Gynecologic and Obstetric Pathology

Ectocx/Mature TZ SCJ/Immature TZ

Normal

Low

High
A B

C D

E F
Fig. 13.17.   Flat mature low-grade squamous intraepithelial lesions (LSILs) (cervical intraepithelial

neoplasia grade 1 [CIN1] or flat condyloma). A, Schematic. B and C, Variable parabasal cellularity with
orderly maturation and superficial cell atypia (koilocytosis). D, Atypia in the lower third can occur but
is within maturing keratinocytes. E and F, Basal cells are uniform with nucleoli and uniformly distributed
chromatin. SCJ, Squamocolumnar junction; TZ, transformation zone.

hyperchromasia, with minimal variation in nuclear size • Exophytic lesions associated with HPV-6/11 will exhibit
and shape: Enlarged nuclei in the lower layers usually patchy or negative p16 staining
have minimal chromatin complexity and reside within • Most flat lesions will exhibit diffuse p16 staining in
cells undergoing cytoplasmic maturation. the lower epithelial layers
• Generally well-preserved polarity with uniform transi- As outlined, the category of LSILs comprises those lesions
tions to mature epithelium in which the stigmata of “classic” CIN—including high
324
Cervical Squamous Neoplasia 13

mitotic index, parabasal atypia, loss of cell polarity, abnor- third of the epithelium. In contrast to exophytic/papillary
mal mitoses, and marked disturbances of maturation—are lesions, flat condylomata are frequently associated with
not present or less pronounced. Presumably, a low- or intermediate- and high-risk HPV types. For this reason,
high-risk HPV may be present but does not produce criti- they will usually exhibit diffuse immunostaining with
cal biologic changes in the replicating (lower) epithelial p16.
layers. This group of lesions can be subdivided into four • Mature exophytic LSIL (exophytic condyloma, condy-
overlapping categories. A sixth category—lesions falling loma acuminatum): Exophytic condylomata are less
between LSIL and HSIL—will also be discussed, inasmuch common in the cervical transformation zone and, when
as these lesions are inevitably encountered in practice. present, are associated strongly with HPV types 6 and
LSIL patterns include the following: 11 (Fig. 13.18). As defined earlier, these lesions exhibit
• Flat mature LSIL (flat condyloma or CIN1): The first acanthosis, verruciform growth pattern, and viral
category of LSIL consists of the flat condyloma (CIN1), cytopathic effect (koilocytotic atypia). The verrucous
which exhibits features similar to condyloma acumi- pattern is typified by blunt papillae, in contrast to the
natum but lacks the exophytic or papillary growth slender filiform papillae of immature condyloma
pattern (see Fig. 13.17). The degree of epithelial matura- (discussed later). The superficial atypia is characterized
tion and koilocytosis may vary, but there is minimal by karyomegaly, nuclear enlargement with binucleation,
nuclear hyperchromasia and pleomorphism in the lower irregularities in nuclear membrane, and hyperchromasia.

Ectocx/Mature TZ SCJ/Immature TZ

Normal

Low

High
B
A

C D
Fig. 13.18.  Exophytic mature low-grade squamous intraepithelial lesion (LSIL) (condyloma).
A, Schematic. B to D, Exophytic growth with orderly maturation and koilocytosis. SCJ, Squamocolumnar
junction; TZ, transformation zone.
325
Diagnostic Gynecologic and Obstetric Pathology

These lesions closely resemble vulvar condylomata but or metastasized (illustrated later in Fig. 13.53). These
with more conspicuous cytopathic effect. Because of are extremely rare, however.
their association with low-risk HPVs, exophytic LSILs
will usually stain weakly or negative with p16. High-Grade Squamous Intraepithelial Lesions
• Extensive exophytic LSIL (“giant condyloma”): An Histologic Criteria
uncommon but important variant of exophytic con- HSILs (Figs. 13.20 and 13.21) exhibit atypia in all layers
dyloma is extensive or “giant” condyloma of the cervix. of the epithelium similar to those classically defined as
These lesions present clinically as a large tumor of CIN. In essence, the extent and degree of nuclear atypia
the cervix and distal vagina. This entity can be mistak- exceed the limits of that described in flat or exophytic
enly classified as a malignancy (verrucous carcinoma), condylomas (LSIL). These features include the following:
because it may exhibit a minimum of koilocytosis (Fig. • In general, less maturation, higher nuclear density, and
13.19). The reader is cautioned that any extensive a less orderly transition from the immature to mature
well-differentiated verruciform lesion in the cervix in epithelial layers
a young woman should be approached with this pos- • Abnormal cell differentiation
sibility in mind. However, we have also encountered • Greater differences in nuclear size and staining as well
extreme examples of large condylomatous lesions of the as contours and chromasia in the lower epithelial layers:
cervix that contained low-risk HPVs yet had invaded In essence, these features reflect the effects of oncogenic

A B

C
Fig. 13.19.  A, Extensive (giant) cervical condyloma showing a bulky exophytic architecture. At higher
magnification the exophytic papillae are devoid of atypia (B) and the growth is nondestructive, with
extension into endocervical crypts (C).
326
Cervical Squamous Neoplasia 13

Ectocx/Mature TZ SCJ/Immature TZ

Normal

Low

High
B
A

C D
Fig. 13.20.   High-grade squamous intraepithelial lesions (HSILs) with maturation. A, Schematic. Types

of HSIL include, B and C, well-differentiated or koilocytotic lesions with prominent basal atypia (cervical
intraepithelial neoplasia grade 2 [CIN2]), and, D, keratinizing lesions. SCJ, Squamocolumnar junction;
TZ, transformation zone.

papillomaviruses on the biology of the replicating (lower that closely mimic immature metaplastic epithelium,
epithelial) cell layers. with more subtle degrees of karyomegaly or anisokaryo-
• In addition to a broader distribution of nuclear atypia sis. Such cases may be difficult to recognize on both
and loss of cell polarity, these lesions are distinguished cytologic and histologic examination, and other strate-
to some degree from LSIL by increased mitotic index gies (such as, the use of biomarkers) may be required
and abnormal mitotic figures. CIN3 lesions (including to distinguish them from inflammatory or reparative
carcinomas in situ) contain full-thickness nuclear atypia. metaplastic changes.
However, the degree of variation in nuclear size, contour, HSILs can display a range of maturation, but a constant
and staining may vary, giving rise to variants of CIN3 feature is full or near full-thickness atypia.
327
Diagnostic Gynecologic and Obstetric Pathology

Ectocx/Mature TZ SCJ/Immature TZ

Normal

Low

High
B
A
Fig. 13.21.   Schematic (A) and a lesion (B) fulfilling the criteria for cervical intraepithelial neoplasia

grade 3 (CIN3) (or carcinoma in situ). SCJ, Squamocolumnar junction; TZ, transformation zone.

Fig. 13.23A-C shows additional examples of lesions that


Squamous Intraepithelial Lesions of Intermediate
might be difficult to classify as either LSIL or HSIL. A
(Indeterminate) Grade (CIN1-CIN2)
common theme with these cases is greater cellularity than
If there is one fact that most pathologists agree on, it typically seen in LSIL and less maturation, yet preservation
is that deciding what falls between CIN1 and CIN3 is of polarity, inconspicuous mitotic activity, and uniform
difficult to determine. In our experience, among relatively nuclear morphology with uniform chromatin distribution
inexperienced pathologists, the degree of agreement for and relatively little nuclear overlap.
CIN2 is fair at best. This problem comes up when
attempting to identify a threshold for CIN2. The transi-
Squamous Intraepithelial Lesions With a Predominate
tion to HSIL is based primarily on the presence of
Metaplastic Phenotype
sufficient atypia in the latter that does not fit within
the spectrum of a flat or exophytic condyloma. One The aforementioned LSILs, intermediate SILs, and HSILs
approach is to focus on the lower epithelial layers, where comprise a spectrum that is most often illustrated in the
the progenitor cell population is derived and contains major texts. In those lesions, maturation usually parallels
the cells that are most likely to confer a diagnosis of grade (i.e., loss of maturation usually but not always heralds
HSIL in a cytologic preparation. Fig. 13.22 compares the presence of HSIL). In contrast, lesions arising within
the lower half of the epithelium in six lesions. The first or presenting as an immature metaplastic epithelium can
(see Fig. 13.22A) is a typical flat condyloma (LSIL). The be more problematic, inasmuch as the degree of cytologic
next (see Fig. 13.22B) is another LSIL with some expan- atypia may be mild and at odds with the lack of cellular
sion of the basal layer but without any change in nuclear maturation. Like the more traditional SILs, this group may
features. The third (see Fig. 13.22C) depicts lower layers appear low or high grade once the degree of atypia is
with somewhat more hyperchromasia but with preserved taken into account; they may also be problematic and
polarity, minimal nuclear variation, and without nuclear many may fall into an indeterminate group. Virtually all
overlap. This fits best with LSIL although some might of these lesions will contain high-risk HPV and will be
consider it indeterminate (CIN1-CIN2). In Fig. 13.22D, strongly positive for p16 with the exception of the so-called
there is now nuclear overlap, hyperchromasia, and greater “immature condyloma.” This entity is discussed first, after
variations in nuclear size. This would likely be interpreted which we address the spectrum of flat immature metaplastic
as either indeterminate or CIN2. Fig. 13.22E and F depict lesions.
irregular nuclear spacing, nuclear overlap, large nuclei,
loss of polarity, and so on, features most would classify
Immature Exophytic Low-Grade Squamous
as CIN2 (HSIL). From these photomicrographs, the
Intraepithelial Lesion (Immature Condyloma,
reader can appreciate the futility of trying to draw a
Squamous Papilloma, Papillary Immature Metaplasia)
sharp line between LSIL and CIN2 (HSIL). It is the most
cogent argument for having a three-grade system, LSIL Immature condylomas fall into the third category of LSIL
(see Fig. 13.22A and B), HSIL (see Fig. 13.22E and F), variants (Fig. 13.24). Conceptually, immature condylomas
and SIL of intermediate or indeterminate grade (CIN1- may be viewed as an infection of transformation zone epi-
CIN2; see Fig. 13.22C and D). thelium by low-risk HPV types (6 or 11) that fails to mature
328
Cervical Squamous Neoplasia 13

A B

C
D

E F
Fig. 13.22.  A spectrum of changes in the lower layers of squamous intraepithelial lesions (SILs).
A and B (low-grade squamous intraepithelial lesion [LSIL]) and E and F (high-grade squamous intraepithelial
lesion [HSIL]) would likely generate reasonable agreement among pathologists. C and D are more
problematic.

and maintains a phenotype closely resembling immature small filiform papillae, which is pathognomonic; and (3)
metaplasia. Because the atypia in mature condylomas is a frequent preservation of overlying columnar cell layers, a
function of viral cytopathic effect and because viral cyto- pattern that is characteristic of a metaplastic process. Some
pathic effect depends on maturation, immature condylomas of these features fall within the description of “papillary
will manifest with the degree of atypia usually seen in immature metaplasia.” Like exophytic condylomas, imma-
the immature cell layers of a condyloma, which is mild. ture condylomas contain HPV types 6 and 11, meriting
Immature condylomas as described are virtually identical definition as immature variants of these lesions. They will
to those variously described as squamous papilloma and stain weakly or negative for p16 and typically display a
papillary immature metaplasia. Conceivably, some of these low to moderate MIB1 index with sparing of the upper
lesions may also be termed transitional papillomas. These epithelial layers. However, the reader must be aware that
lesions present as a spectrum and are illustrated in Fig. occasional lesions associated with high-risk HPV infections
13.24A-D. Patterns of immature condyloma include the might mimic immature condylomas; an example is shown
following: (1) a close similarity to squamous metaplasia in Fig. 13.24E. Moreover, it is important to distinguish
with (a) mild increases in N/C ratio, (b) preservation immature condyloma from papillary carcinomas in situ
of nucleoli, (c) slight nuclear crowding, and (d) a near and papillary carcinomas. The latter exhibit a high mitotic
absence of keratinocyte maturation; (2) the formation of index, as well as greater nuclear atypia (see Fig. 13.24F).
329
Diagnostic Gynecologic and Obstetric Pathology

A B

C
Fig. 13.23.  A to C, Squamous intraepithelial lesions (SILs) difficult to grade are typically more hypercel-
lular and demonstrate less maturation than typical low-grade squamous intraepithelial lesions (LSILs)
but exhibit less nuclear overlap, anisokaryosis, and variations in chromatin staining and complexity than
high-grade squamous intraepithelial lesions (HSILs). These are truly lesions of uncertain or “intermediate”
grade.

Immature Flat Metaplastic Squamous that strong p16 positivity can sometimes be encountered in
Intraepithelial Lesions metaplastic epithelium with minimal atypia (Fig. 13.25A
and B). In such cases, a descriptive diagnosis or one of
Because most lesions arising in the cervix do so near the “LSIL in immature metaplasia” is appropriate.
SCJ, they are by definition “metaplastic” in their origin. • Nuclei should be evenly spaced without overlap, and
However, depending on the degree of atypia and matura- the nuclear chromatin should be similar to uninfected
tion, many have been overlooked prior to the age of p16 cells. Small nucleoli, similar to those seen in immature
immunostaining. They are also less well understood being condyloma, are usually conspicuous.
largely near the SCJ and often containing high-risk HPV • In some cases, columnar cell differentiation will be
types. Thus their diagnosis should be made with this appreciated in the upper epithelial layers (see Fig.
in mind. 13.25C). However, the level of atypias is distinct from
that of a high-grade lesion with columnar differentiation
Immature Metaplastic Low-Grade Squamous (Fig. 13.26E and F).
Intraepithelial Lesions • The lesions will stain strongly for p16, but the MIB1
These lesions contain features similar to immature con- index is usually modest (refer to Fig. 13.28, later). The
dylomas described in the prior paragraphs: columnar cells are often p16 positive as well, in keeping
• They contain a higher nuclear density throughout the epi- with their contribution to the lesion. In essence, the
thelium. This usually distinguishes them from uninfected lesion may exhibit both squamous and columnar
metaplastic epithelium, but the reader must be aware differentiation.
330
Cervical Squamous Neoplasia 13

Ectocx/Mature TZ SCJ/Immature TZ

Normal

Low

High
B
A

C D

E F
Fig. 13.24.   Immature exophytic low-grade squamous intraepithelial lesion (LSIL) (immature condyloma).

A, Schematic. B and C, Papillary architecture at low magnification. D, At higher magnification the epithelium
is composed of uniform spaced nuclei with small nucleoli and infrequent mitoses. The p16 staining
(inset) will be patchy or negative. E, A squamous intraepithelial lesion (SIL) variant resembles immature
condyloma but has strong p16 staining (right inset) and greater nuclear atypia (left inset). F, High magnifica-
tion of papillary squamous carcinoma. Note the conspicuous nuclear atypia and high mitotic activity.
SCJ, Squamocolumnar junction; TZ, transformation zone.

331
Diagnostic Gynecologic and Obstetric Pathology

Ectocx/Mature TZ SCJ/Immature TZ

Normal

Low

High
B
A

C D
Fig. 13.25.   Flat immature low-grade squamous intraepithelial lesion (LSIL). A, Schematic. B and

C, Metaplasia with minimal or mild atypia that might be classified as reactive by some. D, Another
example with some columnar differentiation. Insets depict strong p16 staining, consistent with high-risk
human papillomavirus (HPV) infections in the region of the squamocolumnar junction (SCJ). We
would classify these lesions as LSIL within immature metaplasia. TZ, transformation zone.

Immature Metaplastic High-Grade Squamous One variant, which is discussed in Chapter 14, is a stratified
Intraepithelial Lesions mucin-producing intraepithelial lesion (SMILE). These
These lesions resemble their low-grade counterparts, with lesions contain a uniform distribution of vacuoles, giving
a high nuclear density but with the following features: a honeycomb appearance. Although they resemble HSIL
• There is greater variation in nuclear size and staining by their growth pattern, we interpret them as variants of
with less conspicuous nucleoli and more opaque or adenocarcinoma in situ particularly because of their strong
granular chromatin (see Fig. 13.26C and D). association with coexisting conventional ACIS. Because
• Nuclear density is higher, and there is some nuclear these stratified mucin-producing intraepithelial lesions are
overlap. typically seen in association with both HSIL and ACIS,
• p16 will be strongly positive; MIB1 index will be they suggest a “transition” between the two. We classify
elevated usually with greater than 50% of nuclei staining them as ACIS, stratified variant. In our experience, the
positive. frequency of coexisting invasion appears increased in the
presence of stratified ACIS, although the absolute risk is
Metaplastic High-Grade Squamous Intraepithelial unknown.113
Lesion With Columnar Differentiation
It is not uncommon to encounter columnar differentiation Metaplastic Squamous Intraepithelial Lesions of
in SILs, often as more superficially arranged vacuoles. These Intermediate Grade (CIN1-CIN2)
cells may signify displaced non-neoplastic columnar cells, Given that the distinction between a low- and high-grade
but in some instances superficial columnar differentiation SIL with a metaplastic growth pattern may be difficult
accompanies a lesion that is otherwise classified as SIL. enough to make, it goes without saying that determining
332
Cervical Squamous Neoplasia 13

Ectocx/Mature TZ SCJ/Immature TZ

Normal

Low

High

B
A

C D

E F
Fig. 13.26.   Metaplastic high-grade squamous intraepithelial lesion (HSIL). A, Schematic. B, HSIL

occurring in immature metaplastic epithelium. C, The proliferative index (MIB1) is high. D, Another
immature metaplastic HSIL. E and F, Stratified mucin-producing intraepithelial lesions. These are stratified
variants of adenocarcinoma in situ that overlap histologically with metaplastic variants of HSIL. The
mucicarmine stain of the lesion in F highlights the diffuse distribution of mucin in the epithelium. SCJ,
Squamocolumnar junction; TZ, transformation zone.
333
Diagnostic Gynecologic and Obstetric Pathology

A B

C D
Fig. 13.27.   Squamous intraepithelial lesions (SILs) in metaplastic epithelium that may be difficult to

classify (cervical intraepithelial neoplasia grade 1 [CIN1] to cervical intraepithelial neoplasia grade 2
[CIN2]). A, An expanded population of uniform-appearing immature metaplastic cells. This case had a
low proliferative index. B, Another immature metaplastic proliferation falling between CIN1 and CIN2.
C, This proliferation is more in keeping with CIN2 but is still quite uniform in appearance. D, A metaplastic
SIL populates a prior region of microglandular change.

the “middle ground” between these two entities would Fig. 13.25 and 13.28A) and the other that exhibits greater
seem impossible. Suffice it to say that this spectrum is nuclear density and atypia that may be more difficult to
challenging, but the images in Fig. 13.27 illustrate prolifera- pigeonhole into an LSIL or HSIL. As shown in the figures,
tive lesions that show an increase in cellularity but are this decision can be difficult.
difficult to pigeonhole as to grade. Another form of problematic lesion is that which occurs
This category encompasses some of the entities previously in the setting of small microglandular expansions of
popularized by Ma et al., who termed them eosinophilic metaplastic epithelium (see Fig. 13.27D). The pattern
dysplasia.114 They suggested classifying these lesions as HSIL, closely resembles early squamous metaplasia in areas of
but depending on the nuances of cytology, this type of microglandular change, with a lobular or nesting arrange-
lesion may be more suited for a diagnosis of “indetermi- ment of immature squamous cells, preservation of cyto-
nate” grade. In essence, these lesions are characterized by plasmic maturation, and mucin droplets. The mitotic index
uniform populations of immature metaplastic epithelial is low, and the nuclei are evenly spaced with minimal
cells that maintain a mildly increased nuclear density crowding but exhibit prominent karyomegaly with mul-
throughout the epithelium and a mild degree of aniso- tinucleation. The characteristic features include a lack of
karyosis. The more subtle lesions in this category could reduction in nuclear density in the upper layers, the
easily be misclassified as reactive squamous metaplasia. appearance of a syncytium of nuclei in the superficial
This is a problematic diagnostic area because the pathologist epithelium, and nuclear hyperchromasia. Occasionally
must distinguish two types of “metaplastic dysplasia,” one they may undermine intact columnar mucosa or otherwise
that exhibits mild atypia and can be followed (LSIL; see resemble reserve cells, recapitulating reserve cells.
334
Cervical Squamous Neoplasia 13

A B

C D
Fig. 13.28.  A, A problematic proliferation looks like a squamous intraepithelial lesion (SIL) but is
difficult to characterize. B, The p16 stain is strong. C, However, the MIB1 index is low, arguing against
high-grade squamous intraepithelial lesion (HSIL). D, This inflamed epithelium contains considerable
atypia. However, it is p16 negative (inset) in keeping with either low-grade squamous intraepithelial
lesion (LSIL) or a reactive atypia.

Immunohistochemistry (Table 13.6) • p16ink4, a cyclin-dependent kinase inhibitor, is the


most reliable marker, being expressed strongly in lesions
Because we rely primarily on histologic criteria and because associated with intermediate- and high-risk HPV types,
p16 is not sufficiently specific for HSIL or HSIL outcome in contrast to low-risk HPV infection. Although this
to warrant its routine use in management, we use biomark- marker will not discriminate LSIL from HSIL, it has a
ers sparingly. This discussion is limited to MIB1 and p16, high negative predictive value (i.e., if negative the lesion
because they are the ones most likely to be used in a is highly unlikely to be HSIL). The following are gener-
practical sense. ally true about p16 immunostaining:
• Generic cell cycle proliferative marker (Ki-67) is typically • Intense continuous p16 staining characterizes SIL
confined to the suprabasal cells of the lower third of and usually signifies an HPV type in intermediate
normal mucosal epithelium. The presence of Ki-67–posi- or high-risk group.
tive cells in the upper epithelial layers is characteristic • Patchy or weak p16 staining is less specific, particu-
of HPV-related SIL, which induces cell cycle activity in larly if present in mildly atypical epithelium.
these cells.115 Thus, Ki-67 staining may be helpful in • Low-risk HPV infections exhibit a distinctly different
distinguishing reactive epithelial changes from LSIL or pattern, with concentration of staining in superficial
HSIL, or atrophic mucosa from HSIL. However, denuded cell and koilocytic nuclei, implying a link to viral
inflamed epithelium often has a high proliferative index. replication.
Moreover, the distinction of LSIL from non–HPV-infected • In the vulva and skin, p16 has been associated with
mucosa is of marginal significance given the nearly non–HPV-related proliferations, a phenomenon that
identical follow-up algorithm. may reflect the relationship between p16 and cell
335
Diagnostic Gynecologic and Obstetric Pathology

As mentioned earlier, staining of reactive changes (such


Table 13.6  Differential Diagnosis of Squamous
as, microglandular change) will stain focally positive
Intraepithelial Lesions
(Fig. 13.30).
Category Mimic Distinguishing Features
Differential Diagnosis of Squamous
Intraepithelial Lesions
LSIL Mucosal polyp (vaginal) Minimal acanthosis, no
koilocytosis
Mimics of Low-Grade Squamous
Intraepithelial Lesion
Reactive epithelial Mild superficial karyomegaly, The reader is reminded that the distinction between LSIL
changes occasional binucleated and non-lesional epithelium has little significance, because
intermediate cells the risk of HSIL outcome in 2 years is approximately the
Postmenopausal Superficial cell karyomegaly, same (5% to 13%). We do not employ special stains to
changes cytoplasmic halos resolve these minor conundrums but keep in mind the
potential mimics of LSIL. A variety of mature epithelial
HSIL Immature reactive/ Basal hyperchromasia, uniform alterations may mimic koilocytosis and are illustrated in
repair nuclear spacing and nuclear
contour, nucleoli
Figs. 13.31 and 13.32 and summarized in Table 13.7. They
include the following:
Immature metaplasia Uniform maturation, minimal • Filiform mucosal excrescences of the vagina (see Fig.
surface hyperchromasia 13.31A): These are discussed in Chapter 11 but are
revisited because of their frequent inclusion with cervical
Atrophy No mitoses, uniform
chromasia, nuclear density biopsies. These are essentially plicaform mucosal polyps
and are no more likely to harbor HPV nucleic acids
Atypical atrophy Enlarged nuclei, uncommon than normal squamous epithelium.117
• Reactive epithelial changes (nonspecific): Occasionally,
Implantation site Uniform and wide nuclear
spacing, bizarre nuclei reparative or reactive epithelial changes will undergo
incomplete maturation, in which case, nuclear hyper-
Endometrial histiocytes Small indented nuclei, granular chromasia will be present in association with the
cytoplasm, lack of polarity cytoplasmic halos. The most important discriminating
HSIL, High-grade squamous intraepithelial lesion; LSIL, low-grade squamous feature is absence of variation, in either cell size or
intraepithelial lesion. staining intensity, in the cell population (see Fig.
13.31B-D). Binucleated cells may or may not be present
but will usually be inconspicuous.
• Postmenopausal squamous atypia (see Fig. 13.32A
cycle arrest. This is an issue with severely inflamed and B): Prior reports have identified cells resembling
epithelia or epithelia undergoing repair irrespective koilocytes in postmenopausal women.118 This spectrum
of site (see Chapter 6). Staining is typically patchy. includes maturation disturbances with “pseudokoilo-
• Endocervical columnar epithelium, including micro- cytosis,” transitional metaplasia, and classic atrophic
glandular change, often stains focally and intensely for changes. In some instances, all three may be present
p16, a phenomenon that must be kept in mind when in the same biopsy. Pseudokoilocytosis is character-
interpreting stains. The reader is encouraged to always ized by epithelial maturation, cytoplasmic halos, and
correlate morphology with p16 immunostaining.116 variable (usually mild) alterations in nuclear size or
• Disparities in staining, in both immature metaplastic staining. Occasionally, binucleation is present, and
epithelium and atrophy, where p16 is positive and the process may closely mimic koilocytosis. In some
the Ki-67 index was very low or no HPV is detected, cases, peripheral condensation of pale cytoplasm
can occur. In such cases, it is probably best to issue an around the uniform halos will give a target-like or “fried
explanatory note and make recommendations based egg” appearance to the cells. This pattern is similar
on the level of morphologic atypia and coexisting to that produced by navicular cells in pregnancy and
clinical and cytologic findings. Mild atypias that stain postpartum.
modestly for p16 are best given a descriptive diagnosis
and managed by follow-up cervical cytology. Differential Diagnosis of Squamous Intraepithelial
As discussed previously in this chapter, we do not recom- Lesion of High or Indeterminate Grade
mend the regular use of p16 or MIB1 to either classify SIL With the advent of biomarkers such as p16, most of these
or predict SIL outcome. We use these markers sparingly conundrums can be resolved with an immunostain when
and under the following scenarios: necessary. The entities that might be encountered include
• Resolving the status of atypical immature metaplastic the following:
proliferations regarding HPV origin (see Fig. 13.28A-C) • Reactive/reparative changes can be difficult when there is
• Addressing the neoplastic potential of some inflamma- hypercellularity and little epithelial maturation present;
tory atypias (see Fig. 13.28D) however, if one or two layers of normal superficial
• Distinguishing atypical atrophy from SIL within atrophy cells can be ascertained, the process is usually benign.
(Fig. 13.29A-F) Prominent nucleoli, regular nuclear spacing, and absence
• Determining if a squamous neoplasm is more or less of marked anisokaryosis or surface cell multinucleation
likely to be derived from the cervix characterize reactive changes (Fig. 13.33A).
336
Cervical Squamous Neoplasia 13

A B

C D

E F
Fig. 13.29.   Atypia in the atrophic setting. A, This cervical biopsy in a postmenopausal woman contains

atrophy with mild squamous atypias. B, The p16 stain is negative (patchy). C, The MIB1 index is low,
in keeping with atrophic changes. D, Another atypia in the setting of atrophy. E, It is strongly p16 positive.
F, The MIB1 index is slightly increased. This would be classified as a squamous intraepithelial lesion
(SIL) in of uncertain grade.

337
Diagnostic Gynecologic and Obstetric Pathology

• Immature squamous metaplasia is usually not difficult • Atrophic changes include the following, apart from the
to distinguish from HSIL, but excluding SIL may be pseudokoilocytic changes noted earlier:
difficult when there is an increased nuclear density, in • Conventional atrophy with immature but cytologically
which case an SIL is likely (see Fig. 13.33B and C). This bland morphology (Fig. 13.34A and B)
can be resolved with an immunostain for p16 (see Figs. • Atrophy with partial maturation and focal nuclear
13.28 and 13.29). enlargement or conspicuous nuclear hyperchromasia
• Artifacts that induce nuclear enlargement and hyper- (see Fig. 13.34C)
chromasia as a function of the thermal injury are • Highly cellular lesions with mitotic activity that signify
common, particularly in LEEP specimens. Importantly, a coexisting HSIL (see Fig. 13.34D). Features common
the enlargement is typically uniform and not accom- to atrophic processes include:
panied by anisonucleosis or nuclear crowding (see • Hyperchromatic but generally uniform nuclei
Fig. 13.33D and E). • Frequent elongated and sometimes grooved nuclei
• Absence of conspicuous atypia in the upper
epithelial layers
• Absence of mitotic figures
• Atrophic epithelia with partial maturation and
enlarged atypical nuclei comprise one of the more
difficult categories, and distinction from SIL is based
on even spacing of nuclei with conspicuous intercel-
lular bridges and an absence of mitotic activity.
• Radiation effect can involve both the endocervical and
squamous epithelial cells. The prominent changes
include nuclear enlargement and hyperchromasia
associated with abundant cytoplasm, uniform nuclear
spacing with minimal crowding, a low mitotic index,
and evidence of cytoplasmic degeneration with vacuoles
(see Chapter 11). The nuclear chromatin is often
indistinct or smudged, in contrast to the coarse chro-
masia associated with neoplasia. The preservation of a
N/C ratio is important.
• Alterations due to nonsquamous epithelium include
placental implantation site (see Fig. 13.34E). These are
much less common than seen in the endometrium but
Fig. 13.30.   Focal nonspecific staining for p16 in microglandular change. are sufficiently distinctive that misdiagnosis is unlikely

Table 13.7  Biomarkers in Laboratory Management of Biopsies

Problem Biomarker Notes

Atrophy versus SIL MIB1 Staining of >30% nuclei or upper layer staining in keeping with SIL

Reactive versus SIL p16ink4 p16 more reliable. MIB1 will stain most of the epithelium but will diminish in the superficial cell layers of reactive
changes.

R/0 SIL p16ink4 Diffuse and often intense staining in immature layers of SILs associated with high-risk HPVs but will also stain at
least 50% of LSILs and many subtle metaplastic atypias
Lesions associated with low-risk HPVs stain variably.
Will stain endocervical cells variably and strongly, including polyps and microglandular change.
More recent studies do not support p16 as either a predictor of LSIL progression or for separating LSIL from HSIL.

CK7 Stains squamocolumnar junction (SCJ).


CK7-positive LSILs have a slightly higher rate of HSIL outcome but the stain is not useful for predicting.
One study suggested that CK7-negative LSILs might have a very low HSIL outcome.

Squamous p63 Intense staining of squamous carcinomas; negative or weak differentiation


Weak in poorly differentiated adenocarcinomas and versus other small cell neuroendocrine carcinomas

Implantation site HLA-G, Intense staining of trophoblast, inhibin variable but specific
versus CIN/carcinoma inhibin
CIN, Cervical intraepithelial neoplasia; CK7, cytokeratin 7; HPV, human papillomavirus; HSIL, high-grade squamous intraepithelial lesion; LSIL, low-grade squamous intraepithelial lesion;
SIL, squamous intraepithelial lesion.
338
Cervical Squamous Neoplasia 13

A B

C D
Fig. 13.31.   Differential diagnosis of low-grade squamous intraepithelial lesion (LSIL), including mucosal

polyps (vaginal) (A), mild disturbances in maturation (B), mild nuclear irregularity with cytoplasmic
halos associated with inflammation (C, D).

once the pathologist has some experience with this will be positive and distinguish this from squamous
lesion. The implantation site usually presents as an epithelium.119 Endometrial macrophages (see Fig. 13.34F)
eosinophilic (by virtue of the abundant keratin-positive are another phenomenon that may mimic squamous
matrix), irregularly contoured process in the superficial lesions. The distinction from an epithelial neoplasm is
cervical stroma that is punctuated by scattered hyper- based on the small size of the cells, the small slightly
chromatic nuclei. On closer examination, the irregular vesicular nuclei, granular cytoplasm without distinct
margins of the lesion are composed of a series of small boundaries, and a feathering of the peripheral line of
rounded borders, and the atypical nuclei are separated demarcation without a sharp basal lamina. Mitoses may
by abundant pale to eosinophilic matrix. Occasionally, be present, and eosinophils are common. Rarely, a
these lesions may present as plaquelike lesions resem- marked macrophage response to prior injury (cautery)
bling intraepithelial lesions. An immunostain for inhibin can mimic a squamous neoplasm (see Fig. 13.34G).
339
Diagnostic Gynecologic and Obstetric Pathology

A B
Fig. 13.32.   A and B, Menopausal-related changes can be associated with cytoplasmic halos (fried-egg

cells) mimicking koilocytosis.

Endocervical Curettage epithelium consistent with HSIL or HSIL detached frag-


ments (see Fig. 13.35B). Large papillary, irregular or poorly
ECC is typically used on three occasions: oriented fragments of neoplastic epithelium should be
• During the initial colposcopic evaluation classified as strips of neoplastic squamous epithelium,
• At the time of LEEP excision with the disclaimer that squamous cell carcinoma cannot
• During follow-up after excisional therapy be excluded (see Fig. 13.35C) or, if severe, simply classified
Because a diagnosis of HSIL now requires a LEEP or as fragments of squamous carcinoma (see Fig. 13.35D).
cone biopsy, the precise role of the ECC in managing
women with HSIL is less well defined than previously Resolving Discordant Papanicolaou Test and
when the ECC would dictate whether the patient received
cryotherapy or a cone biopsy. In the reproductive-age
Biopsy Results
woman with a satisfactory colposcopic examination, the As discussed previously, lesion heterogeneity or sampling
ECC is rarely unexpectedly positive. However, the results issues may result in discordance between the Pap test and
of the ECC appear to have some prognostic significance. biopsy (Fig. 13.36A-F). Resolving these discrepancies is
Kobak et al. found that no patients with negative ECC important prior to proceeding to LEEP or cone biopsy.
on initial colposcopy had invasive disease on the final Common scenarios are as follows:
conization specimen, whereas all of those with invasion • The Pap is abnormal and the biopsy shows no lesion.
had positive ECC.120 Other studies have corroborated the This occurs in approximately 15% and 40% of HSIL
value of ECC in ruling out disease in the endocervical and LSIL Pap tests, respectively. If review of the Pap
canal so that conservative ablation therapy can be con- confirms the discrepancy, follow-up is necessary. A
sidered.121 Another option is the endobrush, although this significant minority of these cases will be proven to
may be associated with a higher false positive rate.122 have a lesion on a subsequent evaluation. HPV testing
Currently, a common scenario is the negative ECC that is is of limited value unless the cytologic diagnosis comes
unaccompanied by a biopsy, usually following a negative into question.
colposcopic examination for an HPV-positive ASCUS smear. • The Pap shows LSIL, but the biopsy is HSIL. This occurs
In this setting, the ECC is principally for reassurance. in 10% to 20% of LSIL smears. The cause is usually
Our approach to the ECC is to classify them as (1) sampling artifact, which can be “vertical” (sampling
fragments of benign endocervix, if deemed adequate, (2) only the surface cells) or “horizontal” (sampling only
scant fragments of endocervix if only a few small fragments a portion of the lesion) (see Fig. 13.36A-C). Management
are seen in a few fields, and (3) no endocervix or essentially is dictated by the severity of the histologic lesion. In
no endocervix identified if endocervix is absent or confined some instances, the cytologic discrepancy is the product
to rare epithelial cells. of coexisting low- and high-grade lesions. In most
Fragments of LSIL can be classified as “LSIL, detached instances, when such lesions are present in continuity,
fragments” (Fig. 13.35A). Polarized strips of higher-grade they are derived from the same HPV infection. In a
neoplastic epithelium can be classified as strips of neoplastic minority of cases, two distinct HPV infections may
340
Cervical Squamous Neoplasia 13

A B

C D

E
Fig. 13.33.   A to C, Illustrate immature squamous proliferations that raise the possibility of a squamous

intraepithelial lesion (HSIL). A, Reactive changes exhibit a reduction in nuclear density with minimal
nuclear atypia on the surface or slightly more hypercellular features with readily apparent cytoplasmic
maturation and distinct cell borders. B, High surface nuclear density with less apparent cell borders or,
C, surface hyperchromasia is consistent with SIL in this context. D, Benign cauterized epithelium can be
problematic. E, Lesional epithelium can usually be distinguished by greater surface nuclear density and
anisokaryosis in the latter.

341
A B

C D

E G
Fig. 13.34.   Differential diagnosis of high-grade squamous intraepithelial lesion (HSIL). Spectrum of atrophic atypia, including conventional

atrophy with variable maturation (A, B), atrophy with mild atypias. C, Atypical atrophy. D, A squamous intraepithelial lesion (SIL) in the setting
342 of atrophy. E, Implantation site trophoblastic tumor, mimicking SIL. F, Sheets of histiocytes can be confused with an epithelial lesion. G, Reactive
changes and histiocytes at a biopsy site.
Cervical Squamous Neoplasia 13

A B

C D
Fig. 13.35.  A, Strips of detached LSIL in detached LSIL in the endocervical/endometrial sampling.
B, Thin polarized strips of neoplastic epithelium from an HSIL; C, fragments of papillary neoplastic
epithelium cannot be guaranteed to come from a noninvasive squamous neoplasm; D, fragments of
squamous carcinoma are shown for comparison.

produce two lesions. Fig. 13.36D-F illustrates two


Management of Low-Grade Squamous
instances where two HPV infections—one high- and
Intraepithelial Lesion
one low-risk—coexist in the same area.
• The Pap shows HSIL but the biopsy shows LSIL. This LSIL is managed by observation only. Follow-up may be
is an uncommon discrepancy but one that requires by either HPV DNA testing every 12 months or with cervical
careful review of the cytology and is associated with a cytology obtained every 6 to 12 months. If LSIL persists
substantial risk of subsequent HSIL. If the cytology is for more than 2 years, then ablative treatment is an option,
confirmed as HSIL, the cervical lesion should be carefully as is continued observation. Therapeutic intervention is
reviewed. Again, a small number of these discrepancies particularly discouraged in adolescence. In a recent study,
can be attributed to coexisting low- and high-risk HPV Cox et al. found that the 2-year outcomes of HSIL following
infections. It is particularly important that when faced (1) a biopsy diagnosis of LSIL, (2) an LSIL-positive smear
with a cytologic diagnosis of HSIL the pathologist with a negative biopsy, and (3) an HPV-positive ASCUS
carefully avoid the overdiagnosis of LSIL on histology. that was colposcopically negative were from 11% to 13%.123
Such a diagnosis can be particularly confusing, because In our experience, the follow-up HSIL rate (confirmed by
it may mislead the clinician into assuming LSIL is present review) is less than 10%.81
when in fact the HSIL has eluded sampling. Lesions associated with HPV-6 and HPV-11 confer
essentially no direct risk of cancer and relatively little but
Management of Squamous not negligible surrogate risk. Low-grade intraepithelial
lesions associated with intermediate- and high-risk HPV
Intraepithelial Lesions types have a high rate of spontaneous regression and
The current guidelines for managing biopsy proven SIL empirically low risk of progressing to invasive carcinoma.
are available on the ASCCP website and are summarized Recent studies are consistent with the fact that many LSILs
in the following sections. spontaneously regress.
343
Diagnostic Gynecologic and Obstetric Pathology

A B

E F

Fig. 13.36.   Papanicolaou (Pap) smear/biopsy discrepancies may be produced by vertical sampling

error defined as cytologic sampling (A) of the surface of well-differentiated high-grade squamous
intraepithelial lesion (HSIL) (B), and horizontal sampling error in which multiple grades are present in
the horizontal plane (C). Immature condyloma (low-grade squamous intraepithelial lesion [LSIL]; D)
merges with HSIL (E, right). F, Note the differences in Ki-67 staining (insets). Coexisting LSIL (HPV-II)
and adenocarcinoma in situ (right, HPV-16).

344
Cervical Squamous Neoplasia 13

Invasive cancers are rarely associated with a histologic vaporization is uniquely applicable for patients with HSIL
diagnosis of LSIL. We have seen three cases of LSIL cytology extending into the upper vagina. All of these techniques
followed by cancer. One was a keratinizing lesion that have comparable success rates, and success is influenced
was underdiagnosed, the second was a case of koilocytosis by appropriate patient selection.
accompanied by some cells diagnostic of moderate dys-
plasia, and the last was a CIN1 concurrent with invasive
Management of Young Women (Age 21 to 25)
carcinoma. Thus, only one of the three was a “bona fide”
LSIL. However, as mentioned earlier, classic LSILs, including Three issues emphasize the importance of managing
condylomata, may be associated with HSIL as a conse- younger women with greater care. First is the importance
quence of either progression or two coexisting HPV of ensuring that the diagnosis of CIN2 or greater is correct,
infections. Recognizing this, the practitioner should use given the potential for interobserver disagreement. The
caution when following LSILs that have been preceded by second is the potential impact of treatment on fertility
a cytologic diagnosis of HSIL. and pregnancy outcome. A meta-analysis showed an
Invasive carcinomas rarely follow LSIL and are almost association of cold knife cone with prematurity, low birth
always preceded by HSIL. Studies investigating the first weight, and increased cesarean section rate.124 LEEP was
abnormal smear preceding cervical cancer have found that shown to increase the rate of prematurity and of premature
more than 80% contain CIN3 (severe dyskaryosis), even rupture of the membranes. Laser ablation was not associated
as long as 20 years before the diagnosis of invasive carci- with an increased risk of adverse pregnancy outcomes.
noma. As mentioned earlier, koilocytosis uncommonly is There was insufficient data to evaluate cryotherapy in this
present on smears immediately predating cancer and, when meta-analysis, but it seems unlikely that cryotherapy would
present, is associated with younger age, which calls into have a greater effect on pregnancy than would CO2 laser
question its etiologic significance in such cases. ablation. All methods of treatment have the potential for
In essence, conservative management of women with causing infertility related to cervical stenosis or inadequate
LSIL diagnosis on biopsy requires that (1) the biopsy cervical mucus production, but there are no reliable
correlates with the Pap smear, (2) the biopsy findings comparative studies. The third issue is the absolute risk
correlate with the colposcopic impression, and (3) follow- of a cancer outcome in women who are followed after a
up can be ensured. Ablation must be considered if a diagnosis of CIN2. In our experience and that of others,
subsequent smear displays a persistent LSIL (more than at least 40% of consensus (agreement by at least two of
1 year) or higher-grade lesion that is verified by colposcopic three observers) CIN2 or CIN3 in women from 14 to 25
examination. years old will regress in 6 months, with up to 71% of
CIN2 regressing in adolescents alone.102 In one study, we
encountered one case in 150 HSILs reviewed. In another,
Management of High-Grade Squamous
we found none in 300 cases reviewed of women under
Intraepithelial Lesion
age 25 (Crum & Lee, unpublished). However, it should
Patients with HSIL (CIN2 and CIN3) are treated with be emphasized that nearly 50% of young women with a
either an excisional procedure (such as, cone biopsy or consensus diagnosis of CIN2 are HPV-16 positive; if their
LEEP) or by an ablative procedure (such as, cryotherapy concurrent cytology is HSIL, the rate is over 60%.125
or CO2 laser). An acceptable alternative for HSIL in ado- Therefore, the decision to spare some of these women a
lescents and patients in their early 20s would be follow-up LEEP or cone biopsy must be made not only based on
for up to 24 months and treatment only if the HSIL persists. age alone but also in the context of their cytologic and
Ablative procedures are only acceptable in patients who colposcopic findings and the degree of certainty that the
have an adequate coloscopy and in whom invasive cancer patients can be safely followed.
has been ruled out. Some authors also suggest that ablative
procedures be limited to patients with a negative ECC. Ablative Procedures (Electrical Excision, Loop
Cold knife cone biopsy has long been the gold standard Electrosurgical Excision Procedure or Large Loop
for the treatment of HSIL, because it can reliably rule out Excision of the Transformation Zone, Cone Biopsy,
an occult invasive carcinoma. Because it is generally or Cryocautery)
performed in an operating room with general or regional Management of the Loop Electrosurgical Excision
anesthesia, conization is more expensive than a LEEP, Procedure Specimen
which can be performed as an office procedure with local Processing and Interpretation
anesthesia. If the margin status is critical (e.g., a patient LEEP specimens may come in one or several fragments,
with known microinvasive carcinoma), a cold knife cone well oriented or impossible to reconstruct. The prosector
is preferred. Between ablative procedures, cryotherapy is must identify the mucosal surface and section each fragment
preferred because the equipment is less expensive than perpendicular to the long axis. Margins should be desig-
for a CO2 laser, and it can be performed without anesthesia. nated only where a cautery artifact can be identified (Fig.
Cryotherapy was widely employed in the 1970s and 1980s 13.37A and B). Thus, there are basically three options in
but has largely been replaced in the United States by the reporting margins: (1) involved, (2) uninvolved, or (3)
LEEP. CO2 laser vaporization is appropriate for the same not amenable to evaluation because of distortion or exces-
patients in whom cryotherapy might be considered. Because sive cautery artifact. If the lesion extends to an edge but
of equipment costs and the need for anesthesia, laser the edge does not contain thermal artifact, margin involve-
vaporization is more expensive than cryotherapy. Laser ment is not confirmed (Fig. 13.38A-C).

345
Diagnostic Gynecologic and Obstetric Pathology

A B
Fig. 13.37.   A, Sectioning of loop electrosurgical excision procedure (LEEP) specimens. Intact specimen

and, B, following serial sectioning in a clockwise orientation.

A B C
Fig. 13.38.   A, Margins seen in loop electrosurgical excision procedure (LEEP) specimens include

normal cauterized edge. B, Cauterized edge with high-grade squamous intraepithelial lesion (HSIL).
C, Edge with HSIL that has no cautery present. The latter is not interpreted as a margin.

Interpretation of the Endocervical Curettage to manage follow-up. Salient points concerning the ECC
The purpose of performing an immediate posttreatment include:
endocervical evaluation is to assist in determining the risk • When performed at the time of excisional surgery, ECC
for recurrent or persistent disease and to delineate endo- has been shown to have excellent correlation with
cervical pathology. The magnitude of its contribution to conization histopathologic results and negative predic-
management has diminished with the advent of LEEP, tive value, particularly when conization margins are
more conservative management, and the use of HPV testing negative as well.126,127
346
Cervical Squamous Neoplasia 13

• A positive ECC at the time of conization is also predictive ties but also thermal artifacts following LEEP and
of residual disease in the hysterectomy specimen. spurious glandular cell atypias because of sampling of
• The main limitation of ECC in this application is its the lower uterine segment. The evidence indicates that,
potential for contamination with ectocervical cells, yield- for most studies, post-procedural cytology is a sensitive
ing a false-positive result and decreasing its specificity. test for recurrent disease but carries a small false-negative
• A positive ECC at conization increases the risk of invasive rate. Specificity may vary depending on the experience
carcinoma in the residual cervix of patients whose of the cytopathologist and variables (cautery artifact,
conization revealed high-grade intraepithelial neoplasia changes in anatomy) that alter the nature of the post-
at the margin. The risk is greatest in women over 50 or procedure cytologic specimen.
whose colposcopic evaluation is unsatisfactory. In summary, when considering HPV testing as an adjunct
to the Pap smear, the following can be expected:
The Status of the Margins • The sensitivity and negative predictive value of HPV
Margins are a predictor for recurrent disease regardless of testing is high and can be used either as an adjunct to
whether conization was performed. Eight major studies cytology or, more important, as a tool to exclude
were reported in the 1970s and 1980s that encompassed nonspecific atypias developing postablation.
3894 cones. In this group, there were 3066 with cleared • HPV testing done prior to 6 months’ post-ablation is
margins and an overall 2.9% rate of recurrence or persis- not recommended unless the patient has a cytologic
tence. There were 828 cones (21.3%) with involved margins, abnormality.
with a 22% rate of recurrence or persistence. In a review • A negative HPV test at 1 year, combined with a negative
of 20 articles, positive margins varied from 13% to 45%. smear, carries a very high negative predictive value.
Recurrences with positive margins ranged from 10% to
69% with negative margin from 5% to 38%. Although Diagnosis and Management of Invasive
the presence of involved margins does not necessarily Squamous Cell Carcinoma
indicate the need for immediate further action other than
careful observation in most cases, patients should be Introduction
counseled on increased risk for persistent or recurrent
disease.128 Since the late 1960s, pathologists have witnessed a dramatic
In summary, the assessment of surgical margins should reduction in the number of cervical cancers and, most
be attempted on all specimens sent for pathologic review. significantly, the proportion that are advanced. The case
Endocervical evaluation and margin status may predict example of a cervical cancer with parametrial spread, nodal
risk for recurrent or persistent disease. For the endocervical involvement, and hydronephrosis is a rarity in the United
evaluation, it is acceptable to use either an ECC or an States, albeit more common in low-resource settings where
endobrush, although the latter has limited specificity when women are seldom screened. Currently fewer than 11,000
employed immediately following excision. women/year develop carcinoma of the cervix, and practi-
tioners are noting a higher proportion of adenocarcinomas,
which are not infrequently seen in women in their third
Postexcision Follow-Up
and fourth decades.131 This group is discussed in Chapter
Parameters that influence outcome following excisional 14. The majority of squamous carcinomas occur in women
or ablative therapy include (1) size of lesion treated, (2) who are between ages 40 and 55, with approximately 10%
presence or absence of a positive endocervical curetting younger than 30 years old. At least 50% are stage I when
during or following the cone biopsy, (3) presence or absence diagnosed. Of stage I tumors, 10% to 15% will be less
of positive ecto- or endocervical margins in excised speci- than 5 mm in depth (stage IA). Survival is close to 100%
mens, (4) post-therapy cytology, (5) HPV status post-therapy for stage IA and diminishes with invasion beyond 5 mm
and in some studies, and (6) the age of the patient. and extension beyond the cervix (stage IIA) (Tables 13.8
Regarding recurrence risk the following should be noted: and 13.9).132
• The percentage of patients with abnormal histologic
outcome that are preceded by an abnormal cytology. Colposcopic, Cytologic, and Histologic Predictors
Most, if not all, positive histologic outcomes are associ- of Malignancy
ated with abnormal cytology.129 Sensitivity and Specificity of the Papanicolaou Test
• Likelihood of detecting HPV diminishes with time from for Invasion
nearly 50% at 6 months to 1.1% at 24 months, and The clinician will either suspect the diagnosis of invasion
the odds ratio for recurrence if HPV positive at 24 based on (1) symptomatology, (2) the cytologic report,
months is 26.130 One study showed that the follow-up (3) colposcopic examination, or (4) discover the invasion
risk for CIN3 a cytologically negative, HPV negative following pathologic examination. The biopsy will cor-
case was zero.87 roborate a Pap diagnosis of invasive cancer in nearly 90%
• Most recurrences following an interval of normal cytol- of cases.133 In contrast, when the Pap is “suspicious” for
ogy are either infections by other HPVs or infections invasion or microinvasion, the predicted value drops to
by the same HPV but confined to the ectocervix. This 22% and 17%, respectively.134 Sensitivity of the Pap for
underscores the shift in recurrence location once the microinvasion increases as a function of lesion depth,
SCJ is removed.86 ranging from 14% to 88% for lesions less than 1 mm,
• Causes of reported abnormalities in Pap smears following and greater than 2 mm in depth.135 The cytologic features
cone biopsy include not only squamous cell abnormali- of invasive cervical cancer is discussed in detail later in
347
Diagnostic Gynecologic and Obstetric Pathology

Table 13.8  Staging of Cervical Squamous Carcinoma Table 13.9  Stage-Specific Frequency, Mean Age, and
(International Federation of Gynecology and Obstetrics) Outcome of Cervical Carcinoma

Stage Description Stage Percentage Mean Age 5-Year Survival (%)

Stage I Tumor Is Strictly Confined to Cervix IA1 4.4 44.5 99

IA Preclinical carcinomas of cervix diagnosed only by IA2 3.2 44.6 97


microscopy; all gross lesions even with superficial invasion
are stage IB cancers, invasion is limited to measured IB 39 48.6 80
stromal invasion with maximum depth of 5 mm and no
wider than 7 mm IIA 6.9 55.5 66

IA1 Stromal invasion no greater than 3 mm and no wider IIB 2.2 53.7 64
than 7 mma
IIIA 1.5 63.0 33
IA2 Maximum depth of invasion of stroma greater than 3 mm
and no greater than 5 mm taken from base of epithelium, IIIB 19.8 56.4 39
either surface or glandular, from which it originates;
horizontal invasion not more than 7 mm IVA 2.5 60.1 17

IB Clinical lesions confined to the cervix or preclinical lesions IVB 1.7 58.1 9
greater than stage IA From Eddy GL, Bundy BN, Creasman WT, et al: Treatment of (“bulky”) stage IB cervical
cancer with or without neoadjuvant vincristine and cisplatin prior to radical
IB1 Clinical lesion no longer than 4 cm in size hysterectomy and pelvic/para-aortic lymphadenectomy: a phase III trial of the
Gynecologic Oncology Group. Gynecol Oncol 106:362-369, 2007.
IB2 Clinical lesion greater than 4 cm in size

Stage II Extension Beyond Cervix but Not to Pelvic Wall;


Involves Vagina but Not the Lower Third

IIA Involves vagina, but not lower third; no obvious extension


to parametria

IIB Involves vagina, but not lower third; obvious parametrial


involvement

Stage III Extension to Pelvic Wall; on Rectal Examination, No


Cancer-Free Space Between Tumor and Pelvic Wall;
Involves Lower Third of Vagina

IIIA No extension to pelvic side wall

IIIB Extension to pelvic side wall

Stage IV Extension Beyond True Pelvis or Involvement of


Bladder or Rectal Mucosa

Bullous edema does not permit a case to be assigned to


stage IV Fig. 13.39.   Tumor diathesis in a cervical cytology preparation.
a
Definition of microinvasion by the Society of Gynecologic Oncologists (SGO) includes
these criteria plus absence of capillary-lymphatic space invasion.
Adapted from Pecorelli S: Revised FIGO staging for carcinoma of the vulva, cervix,
and endometrium. Int J Gynaecol Obstet 105(2):103-104, 2009.
Role of Human Papillomavirus Testing
Although HPV testing is not considered a front-line
diagnostic test for cancer per se, one recent study empha-
sized its value in low-resource settings. In this study, a
single HPV test proved to be the most effective at detecting
the chapter. As a rule, approximately 1 in 1000, 650, and cervical cancers earlier and in a more treatable form than
150 ASCUS, LSIL, and HSIL Paps, respectively, will be visual inspection only and cytologic screening.138 Although
followed by a diagnosis of invasive squamous cell carci- such results may not be translated to populations with
noma on biopsy.136 access to multiple lifetime Pap tests, they further reinforce
Tumor diathesis is traditionally associated with invasive the potential advantages of HPV testing when the options
squamous cell carcinoma of the cervix but is not invariably limit the number of tests.
present (Fig. 13.39). Rushing and Cibas examined 28
smears from 19 patients with squamous carcinoma and
Cytologic Alterations Most Commonly Associated With
reported diathesis in 54%, with diathesis correlating with
“Missed” Invasive Carcinomas
depth of invasion. They concluded that it may not always
be possible on cervicovaginal smears to distinguish between The following changes are uncommonly associated with
an intraepithelial lesion and a shallow invasive cancer.137 cancer, but based on experience derived principally from
348
Cervical Squamous Neoplasia 13

medical-legal consultation, the five major pitfalls in rec- the more difficult to identify and appreciate. With the
ognizing invasive squamous or glandular carcinomas and higher index of suspicion seen in current practice, the
their precursors include the misclassification (or under- overdiagnosis of innocuous immature metaplasia is also
appreciation) of the following (Fig. 13.40): likely, particularly on liquid-based preparations.
• Keratinizing lesions: Abnormal keratinizing cells must • HCGs of cells (see Fig. 13.40C): Invasive squamous
be approached with caution (see Fig. 13.40A), and the or adenocarcinomas may present with sheets of cells
diagnosis of LSIL to describe such lesions without some with variable degrees of nuclear atypia. Typically, these
qualification is not recommended. cell aggregates overlap and exhibit nuclear enlargement
• Immature squamous cells with a metaplastic-cell and prominent nucleoli. They may be misinterpreted as
phenotype (see Fig. 13.40B): Atypical immature squa- endometrial, lower uterine segment, and endocervical
mous cells with a metaplastic phenotype are among cells. These are discussed in greater detail in Chapter 14.

A B C

D E
Fig. 13.40.  Cytologic findings associated with cancers or their precursors that may be missed or
underappreciated include abnormal keratinizing cells interpreted as low-grade squamous intraepithelial
lesion (LSIL) (A), small neoplastic cells confused with metaplasia (B), hyperchromatic clumped groups
of tumor cells interpreted as endometrial cells or reactive endocervical cells (C), and extremely rare,
koilocytosis (LSIL) (D) followed by invasive carcinoma (E).
349
Diagnostic Gynecologic and Obstetric Pathology

• HSIL or carcinoma associated with “classic” koilocytosis identified features more commonly seen in HSILs associated
(rare): It is estimated that approximately 0.1% of LSIL with invasive carcinoma. These include the following:
smears will be followed by a diagnosis of squamous • Extensive involvement of surface epithelium and deep
cell carcinoma. The authors have encountered two cases endocervical crypts by expansile HSIL must be distin-
of this type, one of which contained cells raising the guished from a nested pattern of invasion.
possibility of a higher-grade SIL in addition to LSIL • Luminal necrosis with keratin debris in the centers of
(see Fig. 13.40D and E). what appear to be crypts must also be carefully evaluated
• “Reactive appearing” but atypical endocervical cells: to exclude nested pattern of invasion.
Adenocarcinomas of the cervix may occasionally present • Intraepithelial squamous maturation (Fig. 13.42A and
with small clusters of cells closely resembling reactive B) is not diagnostic but a worrisome finding. Other
changes. features of concern include disorganized epithelial
growth in which the epithelium contains discrete
independent units of proliferating cells (see Fig. 13.42C
Clinical Detection of Invasion
and D), papillary architecture (see Fig. 13.42E), and,
When a mass lesion is present, the diagnosis of invasion possibly, the presence of other forms of differentiation
is not difficult (Fig. 13.41A-C). Colposcopy will detect a in the form of conspicuous mucin production (see Fig.
proportion of these lesions (see Fig. 13.41D and E). 13.42F).143 Tidbury et al. found that the extent of HSIL
However, the colposcopist can expect to miss a significant was sevenfold higher in cases with invasion.144
proportion of early invasive squamous cell carcinomas • Unusual fibrous proliferations in the stroma should
of the cervix. In one study, the sensitivity of colposcopy also alert the pathologist to the possible presence of
for microinvasion was 50%, but specificity was 91%. In invasion, although the same may be produced by biopsy
one-third of cases, neither the colposcopic examination artifacts. Under certain circumstances, a diagnosis of
nor the Pap smear identified invasion.139 Atypical vessels invasion may be suspected in the evaluation of an ECC
were found in only one-third of cases in the study by based on the previously described criteria.
Liu et al.140
Diagnosis of Invasion
Percentage of High-Grade Squamous Intraepithelial
Introduction
Lesions Associated With (Micro) Invasion
Before classifying a cervical tissue sample as invasive
The proportion of biopsy-proven HSILs associated with carcinoma, the pathologist must manage four tasks, which
early invasion varies from less than 1% to 7%. In a recent comprise (1) determining whether invasion is possibly
clinical trial of 125 women with documented HSIL who present, (2) excluding mimics, (3) applying the criteria
were followed for 6 months, one (0.8%) was found to for superficially invasive (or microinvasive) carcinoma,
have early invasion on cone biopsy.141 In a follow-up study, and (4) advising the clinician with the use of appropriate
we found no cancers in 300 women younger than 25 years reporting.
old with HSIL on biopsy.141a As mentioned earlier, abnormal differentiation, increased
epithelial thickness, loss of polarity, altered differentiation,
and mucin production may herald a greater risk of invasion.
Risk of Invasion Following Treatment for High-Grade
The latter is often associated with adenocarcinoma in situ,
Squamous Intraepithelial Lesion
and not infrequently is associated with invasive carci-
Soutter et al. observed more than 2000 women with more noma.115,145 However, ultimately, the diagnosis rests on
than 44,000 woman-years of follow-up. A total of 33 criteria specific for stromal invasion.
developed invasive cancer: 14 microinvasive with a cumula- How does one approach “borderline microinvasion”?
tive rate of invasion 8 years after treatment of 5.8 per 1000 Although it is not an accepted category, Wilkinson and
women and 85 per 100,000 woman-years.142 The risk of Komorowski defined borderline microinvasion (less than
developing cancer did not change throughout the follow-up 1 mm) and assessed its risk.146 The authors noted that
period, and the authors concluded that conservative 4.8% and 27% of in situ and microinvasive lesions were
outpatient therapy reduced the risk of invasive cancer by reclassified as “borderline invasion.” They found no
95% during the first 8 years after treatment. However, even metastases in the 29 cases they followed. The implication
with careful, long-term follow-up, the risk of invasive is that a diagnosis of suspicious for, or diagnostic of, very
cervical cancer among these women is about fivefold the early stromal invasion (or that which is suspicious for
general population of women. The authors recommended invasion) can be managed conservatively. Another term
continued follow-up for at least 10 years after conservative likely applied to this form of invasion is budding invasion,
treatment of CIN. Given the negative predictive value of where altered differentiation is appreciated at the epithelial
HPV testing, it is likely that the risk is less in cases docu- stromal interface but no stromal infiltration is present.
mented as HPV negative.
Criteria for Invasive Squamous Cell Carcinoma
Features of High-Grade Squamous Intraepithelial
The most commonly used criteria for invasion include
Lesion Portending Stromal Invasion
the following:
In general, there is no method that predicts which HSIL • A desmoplastic response in the adjacent stroma
will progress to invasion. However, some authors have (Fig. 13.43A)
350
Cervical Squamous Neoplasia 13

A B

E
Fig. 13.41.   Gross appearance of invasive squamous cell carcinoma (A) seen as a fungating mass at

the os in the opened cervix (B). C, A smaller carcinoma is situated at the squamocolumnar junction
(SCJ). Low (D) and higher power (E) colposcopic images of a squamous cell carcinoma. E, Note the
abnormal vessels. (B and E, Courtesy Dr. Quek Swee Chong.)

351
Diagnostic Gynecologic and Obstetric Pathology

A B

C D

E F
Fig. 13.42.   Features that increase the risk of associated invasive carcinoma include aberrant differentiation

(A and B) with or without disorganized epithelial growth (C and D), papillary architecture (E), and
intraepithelial mucin production associated with stratified variants of adenocarcinoma in situ (F).

• Focal conspicuous maturation of the neoplastic epithe- irregularities, which are not typically seen with gland
lium with prominent nucleal (see Fig. 13.43A and B) (crypt) involvement or tangential sectioning through
• Blurring of or lack of a sharp contrast between epithelium gland involvement.
and stroma, often seen at lower magnification • The appearance of pseudocrypt involvement (see Fig.
• Loss of polarity of the nuclei at the epithelial-stromal 13.43D and E): Pseudocrypt involvement is defined as
border with absence of the palisaded pattern charac- discrete circumscribed nests of invasive carcinoma,
teristic of CIN (see Fig. 13.43B) usually with central necrosis, that may mimic crypt
Three additional features include: involvement. In contrast to crypts, this form of invasive
• Scalloping of the margins at the epithelial-stromal carcinoma does not exhibit glandular epithelium, is
interface (see Fig. 13.43C): Scalloping refers to fine often composed of multiple circumscribed nests, often
352
Cervical Squamous Neoplasia 13

C D

E
F
Fig. 13.43.   A and B, Diagnosis of invasion includes loss of polarization, maturation, and desmoplasia;

C, scalloped contours; D, pseudoglands with desmoplasia or, E, retraction artifacts; and complex interlacing
growth patterns (F, “epithelial duplication”).
353
Diagnostic Gynecologic and Obstetric Pathology

contains central necrosis, and may display a loss of mined that approximately one-third were overdiagnosed
polarity. intraepithelial lesions, underscoring the potential problems
• Apparent “folding or duplication” of the neoplastic in lesion interpretation.147 The most important mimics of
epithelium (see Fig. 13.43F): Duplication of epithelium refers microinvasion include the following:
to the presence of vascular structures within a sheet of • Tangentially sectioned epithelium, benign or neoplastic
neoplastic epithelial cells, producing an image of incom- (Fig. 13.44A)
pletely formed papillae. These features aid in recognizing • Prior biopsy sites (see Fig. 13.44B)
invasion in the presence of an intense inflammatory • Inflammatory or reparative changes in CIN, including
response, which may obscure desmoplasia on the one pseudoepitheliomatous changes (see Fig. 13.44C)
hand and blur the epithelial-stromal interface on the other. • Obscuring of the epithelial-stromal interface by inflam-
mation or other artifacts (see Fig. 13.44D)
• Crypt (gland) involvement that is inflamed or tangentially
Immunohistochemistry sectioned (see Fig. 13.44E and F): Crypt involvement
Differential Diagnosis of Invasion is characterized by preservation of epithelial polarity, a
A previous review of 265 cases of presumed microinvasion smooth epithelial-stromal interface, and, in contrast to
sent to the Gynecologic Oncology Group (GOG) deter- the parameter of duplication, each nest of epithelium

A C

B D
Fig. 13.44.  Mimics of invasion include tangential section of metaplasia (A), displaced benign epithelium
from a biopsy artifact (B), and pseudoepitheliomatous hyperplasia in a squamous intraepithelial lesion
(SIL) (C, because of underlying submucosal leiomyoma). Disrupted benign mucosa with inflammation
(D), and extensive crypt involvement with inflammation (E). F, The latter is typically distinguished from
invasive carcinoma by the preservation of polarity and a sharply defined epithelial stromal interface.
G, Large aggregates of activated lymphoid cells in ill-defined germinal centers can mimic malignancy.
354
Cervical Squamous Neoplasia 13

F G
Fig. 13.44, cont’d.

is discrete and separate from the adjacent one. Artifacts management include whether the areas in question are in
are usually produced by prior biopsy, in which a stromal continuity with, or within 1 mm of, the surface epithelium;
response may be present. They should not be associated are limited to one or two foci; are not associated with other
with the other parameters of invasion. parameters of invasion, including capillary–lymphatic space
• Cautery or crush artifact: Cautery or crush artifact should invasion (CLSI); and are associated with clear margins.
be recognized and reported if it hinders diagnosis. Intraepithelial lesions associated with underlying inflam-
Inflammatory changes are responsible for the greatest mation, either from secondary infection or previous biopsy,
diagnostic difficulty, because these alterations may blur must be evaluated carefully to avoid the ovediagnosis of
the epithelial-stromal interface or, in combination with invasion when the epithelial-stromal interface is disrupted
prior trauma, be associated with small nests of epithe- by the inflammatory process. Another mimic of invasion
lium in the inflammatory cell infiltrates. is placental implantation site, which was discussed earlier.
• Misinterpretation of cancer in the ECC, including
implantation site and floaters from other sites Defining Superficially Invasive (Microinvasive)
The laboratory management of these findings is to obtain
levels to determine whether additional features of invasive
Squamous Cell Carcinoma
carcinoma are present and obtain consultation if there is a In recent years, the proportion of invasive cervical carci-
question of invasion in a cone specimen. Factors influencing noma diagnosed at an early stage (less than 5 mm in
355
Diagnostic Gynecologic and Obstetric Pathology

• It is not clearly deeper than 3 mm in the original biopsy


Table 13.10  Follow-Up of Stage IA Cervical Cancer
and does not exhibit CLSI.
• The amount of tissue in the sample is small.
Positive
Stage Definition Nodes Recurrences Deaths (%)
Measurement of Invasion
1A1 <1 mm 0.07 0.38 0.07
Once the cone biopsy is performed, the measurement of
1 to 2.9 mm 1.9 1.5 0.5 depth of invasion should be made from the most superficial
epithelial-stromal interface of the adjacent intraepithelial
1A2 3 to 5 mm 7.8 4.5 2.4 process (Fig. 13.45A). This is not always possible if the
From Eddy GL, Bundy BN, Creasman WT, et al: Treatment of (“bulky”) stage IB cervical tissue is distorted or the epithelium is absent, in which
cancer with or without neoadjuvant vincristine and cisplatin prior to radical
hysterectomy and pelvic/para-aortic lymphadenectomy: a phase III trial of the
case the thickness of the tumor should be measured (see
Gynecologic Oncology Group. Gynecol Oncol 106:362-369, 2007. Fig. 13.45B and C). This is best accomplished using an
ocular micrometer or a method of measurement that makes
it possible to identify with certainty if the lesion has invaded
Table 13.11  Frequency of Capillary-Lymphatic Space to a depth of over 3 mm. If the microscope stage has a
Invasion, Nodal Involvement, and Recurrence Risk micrometer scale, the epithelial-stromal interface or top
of the lesion can be placed at the upper edge of the field.
Capillary-Lymphatic
Depth of Invasion Taking note of the scale, the stage can be moved until the
Space Invasion <1 mm 1 to 2.9 mm 3 to 5 mm top of the field reaches the lower limits of invasion.
The distance traveled can be determined by examining
Frequency 4.4% 16.4% 19.7% the scale (see Fig. 13.45D).
Recurrence risk Present 3.1% 15.7%
Parameters of Importance
Absent 0.6% 1.7%
The following issues require attention when considering
Nodal involvement Present 8.2% 7.5% microinvasion:
• Tumor type: This discussion is limited to squamous
Absent 0.8% 8.3%
carcinomas. However, certain subsets of squamous
From Lecuru F, Neji K, Robin F, et al: Microinvasive carcinoma of the cervix: rationale carcinoma, such as papillary squamous carcinomas,
for conservative treatment in early squamous cell carcinoma. Eur J Gynaecol Oncol
1:465-470, 1997.
may not be as easily evaluated without complete exci-
sion. Neuroendocrine carcinomas and adenocarcinomas
naturally will be evaluated using different criteria.
depth) has increased more than tenfold and currently is • Tumor dimension (depth and length): The risk of pelvic
approximately 21%. Staging systems for early invasive lymph node metastases increases significantly between
cervical cancer are designed to integrate biologic behavior 1 and 5 mm of invasion and is estimated as high as
and clinical management (Tables 13.10 and 13.11). 4.3% for lesions invading between 3.1 and 5 mm (see
Squamous carcinoma of the cervix was previously staged Table 13.10).148-151
under two systems endorsed by the Society of Gynecologic However, a diagnosis of microinvasion (as defined by
Oncologists (SGO) and International Federation of the therapeutic alternatives of conization or simple hys-
Gynecology and Obstetrics (FIGO), respectively. The terectomy) in the United States requires that the carcinoma
principal difference between the two systems was in the invade less than 3 mm into the stroma, based on recom-
staging of early invasion. The SGO-endorsed system defined mendations of the SGO. Burghardt et al. proposed that
stage IA as equal to or less than 3 mm in depth by 7 mm tumor volume be taken into account as a more precise
in length without CLSI. This cut-off distinguished cases predictor of recurrence and metastases. Lesions smaller
amenable to conservative (excision-only) therapy from than 420 mm rarely recur.152 A more simplified approach
those requiring lymph node dissection. The FIGO system places the cut-off at 7 mm in length.153
originally subdivided stage I into “minimal invasion” (less
than 1 mm) and invasion less than 5 mm in depth, making
Capillary-Lymphatic Space Invasion
no recommendation as to management. The two systems
have since come into near agreement, using the SGO The frequency of CLSI has varied widely but increases as
definition to identify stage IA squamous cell carcinoma a function of depth. Van Nagell et al. and Ostor, Rome,
of the cervix (see Table 13.8). and Quinn reported CLSI in 24% of superficially invasive
carcinomas based on Ulex europaeus agglutinin I (UEAI)
stains.151,154 They noted that six of 12 (50%) tumors with
When Cone Biopsy Should Be Considered
lymph node metastases were associated with CLSI, not-
If either superficial invasion or a growth pattern character- withstanding the fact that 19% of cases with negative lymph
izing invasion (confluent neoplastic growth or papillary nodes had CLSI. Similarly, 50% of invasive recurrences
carcinoma) is identified in a biopsy specimen, cone biopsy were associated with CLSI in the original tumor versus
is necessary in the following situations: 14% of patients with no recurrence. CLSI has been associ-
• The lesion does not appear grossly invasive clinically ated with an adverse prognosis in carcinomas exceeding
or colposcopically. 3 mm in depth in most, if not all, reports.155,156 The
356
Cervical Squamous Neoplasia 13

A B

C
Fig. 13.45.  A, Views of different patterns of stromal invasion. Conventional infiltration is measured
from the highest epithelial-stromal interface, seen here as a cleavage plane where the epithelium has
lifted from the stroma. B, Invasive patterns mimicking gland involvement are best measured from the
surface. C, When the distinction of intraepithelial and invasive components cannot be made, a measure
of thickness is most appropriate. D, Microscope stage scale can be used to determine distance in millimeters
from one point in the slide to another, measuring the distance traveled across a single reference point
(microscopic field edge).

implication from these multiple studies is that the majority are salvaged, prudence dictates that the nodes be removed
of cases with CLSI do not present with histologically or sampled if CLSI is unequivocal.
positive lymph nodes, but CLSI is associated more com- In practice, oncologists request that the presence of
monly with an adverse outcome. CLSI be reported and usually opt for radical therapy if it
In a review, Benedet and Anderson computed a tenfold is seen in a lesion less than 3 mm in depth. For these
increased risk (8.3% vs. 0.8%) of nodal metastases in stage reasons, over- and under-interpretation of CLSI must be
IA1 (less than 3 mm invasion) tumors with CLSI, conferring avoided.
a risk of metastases of over 8% and recurrence of 15% CLSI typically exhibits the following features: (1) rounded
(see Tables 13.10 and 13.11).157 Thus, there is sufficient nests of tumor cells, (2) enclosed within a sharply defined
evidence of increased risk to warrant careful counseling space, (3) molding of the tumor nests to the vascular
of patients whose tumors contain CLSI and serious con- space, and (4) an absence of a surrounding stromal
sideration of lymph node dissection, while recognizing response (Fig. 13.46A and B).
that more than 85% of stage IA1 tumors with CLSI will
not have nodal metastases. It is important to emphasize
Conditions Mimicking Vascular Space Invasion
that not all risk can be eliminated by lymph node dissec-
tion. One study found a relationship between CLSI and • Retraction artifacts may produce confusion in the
extrapelvic recurrences, whereas another found that a worse interpretation of CLSI. To avoid this pitfall, it is best
prognosis associated with CLSI was not influenced by to evaluate CLSI at the periphery of the lesion and to
lymph node dissection. Nevertheless, because an appre- avoid the diagnosis of CLSI in the proximity of desmo-
ciable number of patients with positive pelvic lymph nodes plasia (Figs. 13.46A and 13.47A and B).
357
Diagnostic Gynecologic and Obstetric Pathology

A B
Fig. 13.46.  A, Capillary-lymphatic space invasion (CLSI) is best evaluated at the periphery of the
invasive tumor and should be devoid of tissue reaction (desmoplasia). B, CLSI is characterized typically
by nests of differentiated neoplastic epithelium within discrete spaces. Endothelial cells may or may not
be conspicuous.

• Neoplastic epithelium displaced into vascular spaces information that will allow the clinician to come to a
during injection of anesthetic or during specimen conclusion regarding therapy. This is essentially the same
handling may also be confused with CLSI.158 This approach that is found in the World Health Organization
condition is characterized by the following features: (WHO) monograph.159 The purpose of this strategy is
• Prominent vascular dilatation in the area because twofold. First, clinicians should not assume that a diagnosis
of the injection of microinvasion identifies an entity that is easily repro-
• Intravascular neoplastic epithelium that closely duced between pathologists. Determining which patients
resembles CIN are amenable to cone biopsy versus requiring lymph node
• Variable contouring of the epithelium dissection is a subjective process, and clinicians should
• Evidence of trauma, including surface disruption be discouraged from presuming that a report stating
resulting from the needle tract (see Fig. 13.47C-E) “microinvasion” does not require further review, to avoid
A diagnosis of CLSI should be made with caution both overestimation and underestimation of invasion. As
when invasion is not seen. Conversely, if the a rule, all cases falling into the range of those defined as
characteristic features of CLSI are present, a microinvasion should prompt a dialogue between the
compulsive search for invasion should be made. pathologist and clinician and, ideally, be reviewed by two
In discohesive tumors with abundant neoplastic or more pathologists. When reviewing the biopsy, we report
epithelium, transfer of tumor into larger vascular the largest dimensions of the lesion to aid the clinician
spaces may occur by the process of “buttering,” in deciding the next step in management (i.e., cone biopsy
wherein sectioning spreads the cells into available vs. radical hysterectomy), particularly if there is no visible
spaces. Such artifacts should be obvious by the mass on clinical examination. In cone biopsies, the fol-
presence of loose aggregates elsewhere in the lowing should be reported:
specimen and the absence of the characteristic • Depth
rounded clusters of tumor cells. • Length of the entire lesion
• Other forms of pseudovascular space invasion include • Whether length constitutes continuous tumor or is
menstrual endometrium within small vessels (see Fig. composed of multiple small foci
13.47F). Recognition of this pitfall requires attention • The presence or absence of CLSI
to the nature of the cells within the vessels. • The status of endocervical, ectocervical, and deep margins
and, if negative, distance (in millimeters) from invasive
tumor to these margins
Reporting Superficially Invasive Squamous Carcinoma
• Intraepithelial disease and its relationship to the margins
We do not use the term “microinvasive” carcinoma, prefer- • Glandular differentiation if present (criteria for “micro-
ring to report such tumors as “superficially invasive invasive adenocarcinoma” exist, but experience with
squamous cell carcinomas” and providing appropriate this entity is less extensive, and the oncologist must
358
Cervical Squamous Neoplasia 13

A B C

D E F
Fig. 13.47.   Differential diagnosis of capillary–lymphatic space invasion (CLSI) includes retraction

artifacts (A and B), anesthetic needle tracts (C), and displaced neoplastic epithelium in vascular spaces
(D and E). This phenomenon is often accompanied by generalized submucosal vascular dilatation.
F, Menstrual endometrium may occasionally be present in vessels and may confuse the unwary.

always be made aware of glandular differentiation in aggregate distance from one another. This finding should
any invasive lesion) be reported in a narrative. In the authors’ experience,
it is unusual to encounter multiple foci, each of which
measures less than 3 mm by less than 7 mm in dimen-
Common Dilemmas and Their Management
sion. The latter circumstances would be more likely to
• Multifocal lesions, none of which exceed the criteria mandate lymph node dissection than the finding of
for microinvasion: These should be described in detail. small foci of early stromal invasion.
It is not uncommon to identify several small foci of • Invasion originating from an endocervical crypt: This
early invasion in multiple areas of different sections. occasionally occurs and will elicit the favorite question
These foci may be technically greater than 7 mm in of many pathologists who manage this disease, which
359
Diagnostic Gynecologic and Obstetric Pathology

is whether to measure the depth from the surface or microinvasion have been exceeded. If not, the appropri-
the crypt (Fig. 13.48). The most important distinction ate practice is to measure the depth from the epithelial-
to make when faced with this problem is to ensure that stromal interface of the crypt. Nevertheless, these findings
the “crypts” are not actually cohesive nests of invasive should be detailed in a narrative on the report and
carcinoma. If they are, it is likely that the criteria for taken into account when planning therapy.
• Suspicion of CLSI in a single space that is not confirmed
on levels: The diagnosis of CLSI—and with it the almost
certain decision to perform lymph node dissection—
should be based on more than a single focus in a single
tissue section. If a single focus is seen or suspected,
multiple tissue sections should be reviewed. If one
suspicious focus cannot be confirmed on additional
sections, it should be reported in a narrative.
• Papillary lesions without stromal invasion (Fig. 13.49):
Depending on the degree to which the papillae are
well formed, the differential diagnosis includes a ver-
rucopapillary HSIL and a papillary squamous (or
squamo-transitional) carcinoma. If either diagnosis is
proposed, the report must specify that squamous cell
carcinoma cannot be excluded and that depth of invasion
cannot be assessed. Further diagnostic strategies (cone
biopsy) may be required if a mass lesion is not appreci-
ated before deciding whether a radical hysterectomy is
indicated.
• CLSI in association with SIL only (as noted earlier):
When CLSI is present, two causes must be excluded,
both of which we have encountered in practice. The
first is artifactual introduction of neoplastic epithelium
into the vascular space, usually during the injection of
local anesthetic before the surgical procedure takes
place.158 The second is that an underlying invasive
Fig. 13.48.   Invasion from a crypt. When present as an isolated focus,

the tumor can be measured from the epithelial-stromal interface of the carcinoma is present but has not been sampled. The
crypt. We also give the distance from the highest epithelial–stromal latter is confirmed by additional sectioning or further
interface. tissue biopsies.

A B C
Fig. 13.49.  A and B, Papillary squamo-transitional carcinoma may show minimal or no invasion in
limited samples. C, Confluence of epithelial growth with duplication of epithelium and multiple stromal
cores characterizes these tumors.
360
Cervical Squamous Neoplasia 13

Management of Superficially Invasive Squamous Pathology of Invasive Squamous Carcinoma


Cell Carcinoma (Stage IA)
Grading
Occult invasive cancers that are not visible to the naked Grading of squamous cell carcinoma (Box 13.2) has rela-
eye are staged as IA. Cone biopsy is an acceptable option tively little prognostic significance. There are three classically
of definitive management if the lesion fulfills the criteria described categories of differentiation. Predominantly
for microinvasion and ample margins are identified. These keratinizing tumors (grade 1) are mainly differentiated
lesions constitute stage IA1. Studies of cone biopsy versus with conspicuous keratin pearls (Fig. 13.50A). Large cell
hysterectomy have not shown a significant difference in nonkeratinizing carcinomas (grade 2) exhibit greater
the risk of subsequent metastatic spread. Hysterectomy nuclear pleomorphism, infiltrative borders, and inflam-
(by any route) is the preferred approach for women who mation (see Fig. 13.50B), and small cell nonkeratinizing
do not desire to maintain their fertility. Cancers that do carcinomas (grade 3) are characterized by sheets of smaller
not meet the criteria for microinvasion (i.e., extending nonkeratinized cells with high N/C ratio (see Fig. 13.50C).161
deeper than 3 mm into the cervical stroma, exceeding Four variants of large cell keratinizing and nonkeratiniz-
7 mm in length, or showing unequivocal CLSI) are staged ing carcinomas are as follows:
as IA2. Modified radical hysterectomy and lymph node • Lymphoepithelial-like carcinomas, which consist of
dissection are the standards for these lesions. Cases with poorly defined aggregates of nonkeratinized tumors
multifocal disease that collectively span more than 7 mm,
or cohesive lesions with a blunt epithelial-stromal interface,
may be treated on a case-by-case basis.
Radical trachelectomy—combining removal of the cervix,
BOX 13.2  Categories of Squamous Carcinoma
proximal parametrium, and a 1- to 2-cm vaginal margin
along with a pelvic lymph node dissection—has been Squamous cell carcinoma
employed in recent years as a fertility-sparing alternative Large cell keratinizing (well-differentiated)
to hysterectomy and pelvic lymph node dissection for the Large cell nonkeratinizing (moderately differentiated)
Small cell nonkeratinizing (poorly differentiated)
management of stage IA2 and selected stage IB1 carcinomas. Lymphoepithelial-like carcinoma
The trachelectomy can be performed vaginally, abdominally, Spindle cell (sarcomatoid) carcinoma
or with laparoscopy. A cerclage suture is generally placed Verrucopapillary carcinomas
at the time of the trachelectomy. A recent review of more Papillary (squamo-transitional) carcinoma
than 900 cases reports disease-free survival rates comparable Verrucous carcinoma (rare)a
Condylomatous carcinomaa
to those seen with radical hysterectomy.160 There have been Basaloid carcinomasb
more than 300 pregnancies with 196 live births. Approxi- a
In young women, an extensive (giant) condyloma must be excluded.
mately 10% of the pregnancies have resulted in delivery b
May be associated with adenoid basal and adenoid cystic carcinomas, as well as
before 32 weeks. carcinosarcomas.

A B C
Fig. 13.50.   Three common patterns of squamous cell carcinoma of the cervix are A, large cell keratinizing

(well-differentiated); B, large cell nonkeratinizing (moderately differentiated); C, and small cell nonke-
ratinizing (poorly differentiated). Differentiation has minimal influence on prognosis for squamous
carcinoma.
361
Diagnostic Gynecologic and Obstetric Pathology

cells, often with indistinct cytoplasmic borders, inter- • Spindle cell squamous cell carcinoma, also termed
mixed with abundant lymphoid cells, similar to their “sarcomatoid squamous cell carcinoma,” has been
morphologic counterparts in the pharynx: The term is described in the vulva and cervix and consists of an
an adaptation of that used to define similar tumors of expansile tumor mass with a spindled cell component.163
the nasopharynx. The differential diagnosis includes The “sarcoma” is typically not differentiated, but tumors
lymphoid hyperplasia and lymphoma, both of which with osteoclast giant cells have been reported.164 The
can be easily distinguished with stains for epithelial tumors frequently contain both squamous carcinoma
cells (keratin, epithelial membrane antigen) or squamous and spindle cell components, either separate or
cells (p63) (Fig. 13.51A-C).162 subtly blending (Fig. 13.52A-C). Ultrastructural and

A B C
Fig. 13.51.   A and B, Lymphoepithelial-like carcinoma of the cervix exhibits an indistinct blending of

a monomorphic nonkeratinizing tumor with a prominent lymphoid infiltrate. C, Strong positivity for
p63 is consistent with a squamous origin.

A B
Fig. 13.52.  A, Spindle cell squamous carcinoma displaying the interface between epithelioid (center)
and spindled (sarcomatoid) elements. B, A stain for cytokeratin highlights the epithelial component.
362
Cervical Squamous Neoplasia 13

immunohistochemical studies indicate that these tumors of this entity. In both, the invasive component was
are derived from squamous cell carcinoma and several either focal or unrecognized prior to hysterectomy.165
features will aid in the correct diagnosis of these tumors: • Verrucous carcinomas are essentially unheard of in
• True carcinosarcomas of the cervix almost always the cervix, although we have encountered rare lesions
arise in association with basaloid neoplasms. Thus, reminiscent of this entity (see Fig. 13.53C and D).
if the pathologist encounters a spindle cell neoplasm One review of reported cases reclassified most as
of the cervix not associated with a basaloid squamous other forms of verrucopapillary neoplasia.166
tumor, the differential diagnosis narrows to a pure • Conventional condylomatous lesions with invasion
sarcoma versus a spindle cell carcinoma. and metastasis: We have encountered one case of
• If an epithelial component is appreciated, it will be HPV-11 positive verruciform neoplasm of the cervix
a typical large cell nonkeratinizing squamous cell that was indistinguishable from a large condyloma,
carcinoma. yet was associated with regional lymph node metas-
• A spindle cell squamous carcinoma is also likely if tases (see Fig. 13.53E and F).
a precursor lesion (CIN) is also present. In some • Papillary squamotransitional carcinomas of the
cases, a transition from CIN or squamous carcinoma uterine cervix are characterized by filiform papillae
(superficial) to spindle cell morphology (deep) is lined by neoplastic cells identical to high-grade CIN
present. and may have features resembling transitional cell
• Special stains for keratins will usually be positive carcinoma of urothelial origin (see Fig. 13.49A-C).167
but may vary in intensity. The term transitional is based principally on the light
• Verrucopapillary neoplasms of the cervix that either are microscopic appearance, inasmuch as special stains
malignant or can mimic malignancy are addressed as for squamous (CK7) are typically positive and
a group for clarity (Table 13.12). They include the transitional cell differentiation (CK20) is present in
following (Fig. 13.53A-F): less than 10% of cases. Because these tumors
• Extensive (giant) condylomata are rare and are more are predominantly papillary, invasion may not be
commonly reported in the anogenital mucosa, confirmed and may require wedge biopsy or LEEP
particularly in men (see Chapter 10) (see Fig. 13.19). cone. However, the majority (90%) will demonstrate
The authors have seen two cases in the cervix that invasion with sufficient sampling.
were initially misinterpreted as verrucous carcinomas, • Condylomatous carcinomas consisting of well-
primarily because they were accompanied by extensive differentiated exophytic squamous cell carcinomas
hyperkeratosis and inflammation and lacked con- with conspicuous superficial and basal cell atypias
spicuous koilocytotic atypia. Both were HPV-11 are occasionally encountered. The most compelling
positive. One telltale finding is the extension of the reason for subclassifying these tumors is to distinguish
process into crypts with preservation of the overlying them from verrucous carcinomas and condylomata.168
columnar cells and lack of stromal replacement (see • Basaloid squamous carcinomas are extremely rare vari-
Fig. 13.19). ants of cervical neoplasia that exhibit a mixture of
• Well-differentiated squamous neoplasms with basaloid and variable mature squamous differentiation
conventional invasion constitute a unique subset (Fig. 13.54A-C). These tumors are technically squamous,
of tumors in which the papillary component fails but some have been associated with adenoid basal
to fulfill the criteria for carcinoma but is associated carcinomas and may actually arise from these tumors
with frankly infiltrative squamous carcinoma (see Fig. early in their evolution. They may also be associated
13.53A and B). The authors have seen rare examples with a sarcomatous element.

Table 13.12  Differential Diagnosis of Papillary Cervical Neoplasia

Tumor Differentiation Koilocytosis Basal Atypia Infiltration

Condyloma (LSIL) Mature Yes No None

Extensive (giant) condyloma Mature Variable No None

Immature condyloma Immature squamous Minimal No None

Papillary squamotransitional carcinoma Immature squamous No Yes Variable

Condylomatous carcinoma Mature squamous Variable Yes Present

Verrucous carcinoma Mature squamous No Minimal Blunt

Papillary undifferentiated carcinoma None No Yes Present


LSIL, Low-grade squamous intraepithelial lesion.
363
Diagnostic Gynecologic and Obstetric Pathology

A B

C D

E F
Fig. 13.53.   Verrucopapillary neoplasia. In addition to occasional large condylomata (see Fig. 13.19)

and papillary squamous carcinomas (see Fig. 13.49), there are other, even rarer lesions that might be
encountered. A and B, A lesion resembling a condyloma that abruptly transitions to an invasive squamous
carcinoma. C and D, More rare lesions that resemble in part a verrucous carcinoma. E, Rarely, a lesion
indistinguishable from a condyloma (upper and upper right inset) merges with a less well-differentiated
component (lower and lower left inset). F, Invasion was also demonstrated. This picture is rarely encountered
and can be associated with “low-risk” human papillomaviruses (HPVs).

364
Cervical Squamous Neoplasia 13

A B

C
Fig. 13.54.  Carcinomas with basaloid phenotype may present as solid (A) or interlacing (B) cordlike
growth patterns. C, Strong staining for p63 confirms the basal (versus columnar) pattern of differentiation.

Reporting Squamous Carcinomas


Significance of Columnar Cell Differentiation
Histologic reporting should include the following:
A significant proportion of cervical squamous carcinomas • Grade (well, moderately, or poorly differentiated)
are associated with columnar cell differentiation. The • Cell type
prognostic significance of columnar differentiation is not • Depth of invasion or thickness
clear. In one study, 27% of randomly selected squamous • Extent of tumor: The extent of invasion into extra-cervical
carcinomas contained some degree of mucin positivity. tissues and metastases to both pelvic and extrapelvic
Approximately 5% contained sufficient mucin to warrant organs should be recorded.
reclassification as adenosquamous carcinomas, suggesting • Angiolymphatic vascular space invasion: The presence
that mucin stains may in some cases aid in classification.169 of tumor within blood vessels or lymphatic vessels
In a second study, 87 stage I cervical carcinomas treated should be noted and an attempt made to distinguish,
by radical hysterectomy were studied. A total of 39% were when possible, between them.
mucin positive, and this group (and pure adenocarcinomas) • Status of lymph nodes: The presence or absence of
was significantly associated with a higher risk of lymph metastases in each submitted group of lymph nodes
node metastases.170 Markers used to distinguish squamous should be reported, recording the total number of
carcinomas from other tumors, including neuroendocrine involved lymph nodes in relation to the total number
carcinomas, are discussed in Chapter 15. of lymph nodes identified.
365
Diagnostic Gynecologic and Obstetric Pathology

• Status of resection margins: The adequacy of local patients.175,176 Conversely, approximately 3% of all cancer
excision should be assessed by careful examination of patients are pregnant at the time of diagnosis. The two
resection margins, the latter preferably marked by the important clinical issues are (1) influence of pregnancy
use of ink; the distance from the deepest point of stromal on outcome and (2) the risk of delaying delivery until
invasion to the closest (inked) margin of resection may fetal maturity permits viability and the potential negative
be noted in the report. impact of cone biopsy on the pregnancy.

Influence of Pregnancy on Outcome


Treatment and Outcome of Several studies of cervical cancer during pregnancy noted
Squamous Carcinoma no significant worsening of outcome.175,177,178 Adverse
Clinical Factors Influencing Outcome/Presentation outcome was directly related to tumor extent.
Presenting Signs/Symptoms
Pretorius et al. evaluated a cohort of 81 women with cervical Safety of Delays in Treatment
cancer and correlated presentation with outcome. Abnormal In general, management of pregnant patients with cervical
vaginal bleeding and an abnormal Pap smear were the cancer is as follows:
dominant presenting findings (56% and 28%). Other • Patients under gestational age of 20 weeks undergo
symptoms, such as pain and discharge, were seen in less immediate surgery or irradiation with fetal loss. In some
than 10%. An abnormal Pap smear was associated with patients with advanced disease, hysterotomy is required
stage I disease and a significantly longer disease-free survival to evacuate the pregnancy.
(96%) than vaginal bleeding (51%). Both exceeded the • Patients at gestational age of 20 weeks or older with
disease-free survival of those presenting with pain (29%). low-stage disease may be treated with observation of
Thus, early detection (by cytology) is associated with a up to 4 months.
significantly higher disease-free survival and significantly • Conization can be performed safely for cases of early
smaller tumor volume.171 (microinvasive) carcinoma. The safety of longer follow-
up intervals is unclear based on relatively few data.
Age of Presentation Sorosky et al. followed seven stage IB pregnant patients
Patients younger than 35 years old constitute slightly more who desired pregnancy retention from 21 to 207 days
than 10% of women with cervical cancer and a disproportion- (median 109 days), with no adverse outcomes and no
ate number of those who have previously been screened. apparent worsening of clinical stage.179
Whether the under-35 age group is at greater risk of an Inasmuch as chemoradiation is now being used to
adverse outcome is doubtful. Jennings et al. performed a manage women with more advanced cervical cancer, this
retrospective cohort study comparing women younger and approach has been successfully used in selected women
older than 35 years old.172 They found no significant differ- in an effort to preserve the fetus and in small series without
ence in the incidence of nonsquamous tumors, tumor grade, adverse effect.180,181
lymph node involvement, HPV status, tumor recurrence, or
survival between the two groups. Young patients appeared Viral Factors and Outcome
to enter the study at significantly earlier stages of the disease, HPV-16 predominates in squamous neoplasia in contrast
and a greater proportion of them underwent surgical treat- to HPV-18, which is seen more frequently in glandular
ment.172 Yang et al. studied stage IB to IIA cervical cancers and neuroendocrine neoplasms. The role of HPV type as
among women 35 years old or younger and older and an independent prognostic indicator has been addressed
reported a similar overall survival (71.2% vs. 72.4%) for in several studies. Many, but not all, indicate a worse
both groups. Nonsquamous carcinomas, including adeno- outcome for patients whose tumors harbor HPV-16 or
carcinoma and small cell carcinoma, were both associated -18. Lombard et al. studied 297 patients for which HPV
with HPV-18 and the younger patients and conferred a slightly typing was available (83%), with a median follow-up of
higher but not significant risk of recurrence/persistence; 38 months.182 Although they reported no significant
however, these differences were not significant, and 71% of relationship between virologic data and tumor stage/node
the recurrences were squamous cell carcinomas.173 status, the 5-year disease-free survival rate was 100% for
Early studies of HPV prevalence in cervical cancer sug- patients with intermediate-risk HPV-associated tumors,
gested the existence of an HPV-negative older population, 58% for patients with HPV-16–positive tumors, and 38%
possibly with a worse prognosis. Notwithstanding occa- for patients with HPV-18–positive tumors (p = 0.02). In
sional tumors in older women, such as minimal deviation multivariate analysis, the RR of death conferred by HPV-18
adenocarcinoma and verrucous carcinoma, that may not was 2.4 times that of HPV-16, and it was 4.4 times that
be associated with HPV, a relationship between age and for patients with a viral type different from HPV-16/18.182
HPV prevalence has not been substantiated in cervical Schwartz et al. similarly found the risk of tumor-related
cancer. Baay et al. found no statistically significant differ- mortality to be 2.2-fold higher in HPV-18– versus HPV-16–
ence in either the prevalence of HPV DNA or distribution positive tumors with the associations strongest for those
of genotypes between women with cervical cancer who with FIGO stage IB/IIA disease. The HPV-18 associations
were younger or older than 65 years old.174 were strongest for patients with FIGO stage IB or IIA
disease.183 The increased risk was also seen in the subset
Presentation During Pregnancy of patients with squamous carcinomas, with HPV-18
Carcinoma of the cervix during pregnancy is rare and is conferring increased risk, particularly in earlier-stage disease.
diagnosed in approximately 1 per 3000 to 10,000 pregnant Lai et al. noted a significantly worse outcome for
366
Cervical Squamous Neoplasia 13

HPV-18–positive tumors.184 Other studies have not found retrospectively applying the FIGO 1994 definition, reported
a relationship between these HPV types and poor stage IB2 cervical cancer patients had a significantly higher
prognosis.185,186 incidence of nodal metastasis (21% vs. 44%) and poorer
survival (73% vs. 90%) following radical hysterectomy
than patients with stage IB1, despite the more frequent
Management of Squamous Cell Carcinoma use of postoperative radiation therapy (38% vs. 72%).193
Improvements in Prognosis Since the Late 1970s Similarly, Trattner et al. found that stage IB1 patients had
Covens et al. summarized their experience in a large practice an overall survival of 90% compared with the 40% 5-year
and noted a consistent reduction in comorbidity (e.g., survival rate for stage IB2 patients.194
blood loss, length of stay, transfusion, infections) associated Recently, some investigators have questioned the need
with cervical cancer therapy over a 16-year period.187 for resection of the parametrium with lesions of less than
However, parameters linked to early detection and prog- 2 cm size (stages IA2 and IB1). They have proposed pelvic
nosis (age, tumor size, CLSI, and pelvic lymph node lymphadenectomy combined with conization or simple
involvement) did not change. They also noted a significant trachelectomy for patients who wish to maintain their fertil-
increase in the proportion of adenocarcinomas (28%) and ity and pelvic lymphadenectomy with simple hysterectomy
a decrease in the proportion of grade 3 tumors (28%). for patients who have completed childbearing.195
They concluded that most of the progress made was in There is controversy about the best approach to a patient
the realm of operative management than survival.185 with a stage IB2 cervical cancer. Often these are endophytic
tumors, and patients are described as having a barrel-shaped
cervix. If these patients undergo primary surgery, a large
Standard Treatment Options
percentage will require adjuvant radiation or chemoradia-
Patients with invasive cervical cancer are staged clinically tion. High-risk patients are those who have positive lymph
based on a FIGO system that dates back to the 1930s.188 nodes, involvement of the parametrium, or involvement
The system allows comparisons to be made between of a surgical margin.196 All of these patients will be treated
patients treated primarily by surgery and those treated with chemoradiation. An intermediate risk group has also
primarily with radiation therapy. It does not allow the been defined by the GOG based on a scoring system that
stage to be changed by information obtained from a surgical incorporates clinical lesion size, depth of stromal invasion,
procedure. It also prohibits the inclusion of information and involvement of the capillary-lymphatic space.197 For
from “high-tech” imaging studies such as computed these intermediate-risk patients, postoperative radiation
tomography (CT), magnetic resonance imaging (MRI), or lowers the pelvic failure rate but has not been shown to
positron emission tomography (PET) scans. Because have a statistically significant impact on overall survival.198
patients will be treated based on information not included Chemoradiation is now being studied in a randomized
in assigning a clinical stage, in practice treatment will be trial for this intermediate-risk group.
much more heterogenous than described here. Primary chemoradiation is also an option for stage IB2
• Stage IA1: Management is detailed in the preceding tumors. The survival is roughly comparable to that obtained
discussion of microinvasion. with radical hysterectomy followed by adjuvant irradiation
• Stage IA2: Standard treatment would be a modified or chemoradiation. The complication rate is thought to
radical hysterectomy with lymphadenectomy or radiation be less for patients who receive primary radiation therapy
therapy. Radical trachelectomy is an option for patients without hysterectomy. For patients with a bulky stage IB
wishing fertility preservation. tumor, simple hysterectomy was advocated following
• Stage IB: Both surgery and radiation therapy can be completion of the radiation, but a GOG trial showed it
utilized for the management of stage IB1 cervical cancer did not improve survival.199 Neoadjuvant chemotherapy
with similar-appearing results. However, significant followed by radical hysterectomy and selective postoperative
selection bias exists in determining who is selected for radiation was advocated for a time. A recent GOG trial
surgery or radiation therapy.189 Radical hysterectomy is showed this approach did not improve survival over that
the preferred option for smaller tumors and carries a obtained with radiation therapy alone.200
nearly 100% disease-specific survival rate for tumors
under 2 cm.190 Because of controversy regarding the Stage IIA
appropriate management of early-stage disease, Landoni For tumors that extend to the vagina but are limited to
et al. performed a randomized study of radical surgery the upper two-thirds, without involvement of the para-
versus radiotherapy for stage IB to IIA cervical cancer.191 metria, both radical hysterectomy and radiation therapy
These authors found no difference in overall or disease- have been utilized. Radical surgery would be suggested
free survival. However, 46 of 55 (84%) patients with for a younger patient if an adequate margin can be obtained
bulky tumors in the surgery group received adjuvant and adjuvant radiation can be avoided. However, if radia-
radiation for additional risk factors. tion is likely to be required, primary radiation alone with
It has long been recognized that, among tumors limited both external and intracavitary is usually preferable.
to the cervix, tumor size is predictive of nodal involvement
and survival.192 Although opinion over the appropriate Stage IIB
treatment has varied widely, the designation by FIGO of For tumors that extend into the parametria (stage IIB),
stages IB1 and IB2 in 1994 established criteria differentiat- the standard treatment in the United States has been
ing the clinical management of smaller from larger gross radiation therapy. In contrast, many European and Japanese
cervical tumors limited to the cervix. Finan et al., gynecologic oncologists have preferred radical hysterectomy
367
Diagnostic Gynecologic and Obstetric Pathology

and lymphadenectomy followed by adjuvant radiation Recurrent cervical cancer represents a particularly difficult
therapy. More recently, the introduction of chemotherapy problem, with few long-term survivors. The majority of
into a multimodality treatment for localized cervical cancer patients with recurrent disease present initially with locally
has been extensively studied. Treatment schemas have advanced disease and, as demonstrated in recent random-
utilized chemotherapy administered before radical surgery ized trials, their disease-free and overall survival curves
or radiation and concurrently with radiation. Because tumor are almost identical. For patients who have a localized
size is an important predictor of response to radiation central recurrence following hysterectomy, radiation therapy
therapy and eventual survival, the use of chemotherapy to can be utilized. More commonly, localized recurrences
reduce tumor size before the onset of definitive radiation may follow radiation, and radical surgery (usually exentera-
therapy is attractive. However, despite significant responses tion) is required, with survival rates of 25% to 50%,
to neoadjuvant chemotherapy, randomized trials have failed depending on patient selection criteria. Recent surgical
to demonstrate improvement in survival, possibly because advances, including continent urinary diversion, anal
of the development of crossresistance or tumor repopulation. preservation, and vaginal reconstruction, can significantly
In contrast, the concurrent administration of cisplatin-based improve the patient’s quality of life. Patients with metastatic
chemotherapy has resulted in reproducible benefit in disease who receive chemotherapy generally fare poorly,
randomized trials of cervical cancer patients with a variety with median survivals of 6 to 8 months. The addition of
of stages and indications for radiation therapy.201-205 These the antiangiogenic agent bevacizumab to multi-agent
trials demonstrated improvements in disease-free and overall chemotherapy has been shown to increase survival in
survival of 30% to 50%, respectively, with concurrent patients with metastatic and recurrent cervical cancer.210
chemotherapy. Although the chemotherapy regimens varied,
the unifying theme was that all were cisplatin based. Based Prevention
on the favorable results of five large randomized trials in Vaccines
cervical cancer, the National Cancer Institute issued a clinical Prophylactic HPV vaccines derived from viral-like particles
announcement advocating the concurrent use of cisplatin- have been available for over 10 years and those targeting
based chemotherapy with radiation for cervical cancer HPV-16 and HPV-18 have been estimated to reduce the
patients who required radiation therapy. risk of cervical cancer by 70% in a population of preme-
Neoadjuvant chemotherapy has been studied for patients narchal women (Table 13.13; Fig. 13.55). The new non-
who have locally advanced disease and are not thought avalent vaccine targets nine types and is estimated to reduce
to be candidates for primary surgery. Various combina- the rate of cervical cancer by 90% in an optimally vaccinated
tions of chemotherapy agents have been studied.206-208 population.211 Future challenges will entail recalibrating
In some protocols, patients who responded to chemo- the frequency with which cytologic screening will need
therapy then had a radical hysterectomy, and in other to be performed in the vaccinated population.212 A perpetual
protocols those patients received a standard course of
radiation therapy. A Cochran review of randomized trials
suggested a benefit to neoadjuvant chemotherapy when
compared with standard radiation therapy.209 However,
neoadjuvant chemotherapy has not been compared with the
now-accepted regimens of concurrent chemotherapy and
radiation.

Stage IIIA
Tumors with involvement of the lower third of the vagina
in patients who do not have extension to the pelvic side
wall are relatively rare. Concurrent chemotherapy and
radiation therapy would be appropriate for most of these
patients.

Stage IIIB
These patients have fixation to one or both pelvic side
walls or have obstruction of a ureter. Concurrent chemo-
therapy and radiation therapy is the accepted treatment.

Stage IVA
These patients have involvement of the bladder or rectum
without evidence of distant metastasis. A few of these
patients are candidates for pelvic exenteration, but most
will have pelvic side wall involvement and are treated with
concurrent chemotherapy and radiation therapy.
Fig. 13.55.   Viral-like particles similar to those currently used in vaccine
Stage IVB trials. (Viral-like particles produced by the laboratory of Robert Rose,
These patients present with metastatic disease and are usually University of Rochester, Rochester, NY. The micrograph is courtesy of
treated for palliation only. Treatment is individualized. Linda Stannard, University of Cape Town, Cape Town, South Africa.)
368
Cervical Squamous Neoplasia 13

Table 13.13  Outcome of Vaccination for Human Papillomavirus

End Point Vaccine Population Vaccine Efficacy (%) Reference

Persistent infection HPV-16 Unex 100.0 11

Transient infection HPV-16 Unex 91.2 11

HPV-16 + CIN HPV-16 Unex 100.0 11

HPV-16/18 + CIN2 HPV-16/18 Unex 92.9 to 98.1 331

All CIN2 HPV-16/18 Ex 30.4 331

All CIN2 HPV-16/18 Unex 70.2 331

Other high-risk HPV + CIN2 HPV-16/18 Unex 54.0 331

All CIN3 HPV-16/18 Ex 33.4 331

All CIN3 HPV-16/18 Unex 87.0 331


CIN, Cervical intraepithelial neoplasia; CIN2, cervical intraepithelial neoplasia grade 2; Ex, exposed to HPV; HPV, human papillomavirus; Unex, unexposed to HPV.

epithelium, which is 10 to 20 times less vulnerable to


malignancy. Several studies, each offering a different vantage
point, suggest that a prophylactic procedure that removed
the cells in or near the SCJ would significantly reduce the
risk of subsequent cervical cancer.
• Studies employing cautery to the cervix have reported sig-
nificant reductions in subsequent cervical carcinoma.214,215
• One study found that cryotherapy to the cervix reduced
the rate of subsequent HPV positivity to 50% of
untreated patients.216
• A study of cervical pathology and HPV status following
cone biopsy showed that delayed recurrences (presum-
ably signifying new vs. persistent untreated lesions)
were invariably on the ectocervix and low grade, with
frequent regressions. This implies that re-infections of
the ectocervical squamous epithelium by the same or
a different HPV type are far less likely to manifest as
Fig. 13.56.  A new squamocolumnar junction (SCJ) following excision. HSIL.86
There is no evidence of metaplasia. It is postulated that following excision • We have found that SCJ-negative LSILs confer a very
of the original SCJ, the unique SCJ cells are not regenerated, and this is low risk of subsequent HSIL.91
one reason for the diminished risk of squamous cell carcinoma. The aformentioned reports do not confirm with absolute
certainty that prophylactic cryotherapy of the cervix would
sharply reduce cancer risk, but the prospect that HPV
issue that must be addressed is maximizing the cost- infection rates would diminish is an intriguing one. It
effectiveness of HPV vaccination in the societies that are begs the question of whether removing the SCJ cells from
most vulnerable to cervical cancer.213 What can be antici- an HPV-positive/cytologically-negative woman would
pated, however, is a continued reduction in the incidence reduce the likelihood of HPV detection in follow-up
and death rates for cervical cancer. and—by association—the risk of cervical neoplasia. Studies
of “see and treat” where acetowhite lesions of the cervix
Targeting the Squamocolumnar Junction are treated by ablation have shown a significant reduction
The SCJ remains a largely unexplored primary target for in CIN2-3 outcome.217,218 Studies that have recently identi-
cervical cancer prevention, despite its tantalizing association fied morphologically latent HPV within SCJ cells have
with the origins of cervical cancer.3 It is clear that removal further supported this concept.219 In fact a study of “screen
of the SCJ by cone biopsy or LEEP alters both subsequent and treat” HPV-positive women were managed in three
cervical cancer risk and the morphology of “late recur- groups: (1) cryotherapy, (2) cryotherapy if acetowhite
rences,” lesions that emerge after a period of normal cervical epithelium was seen, and (3) no therapy for 6 months.
cytology (Fig. 13.56). The theoretical basis for this would Outcome CIN2+ was observed in 1.5%, 3.8%, and 5.6%,
be that removal of the most vulnerable cells brings the respectively, underscoring the value of cryotherapy when
risk of new HSILs closer to that expected for vaginal no visible lesion was observed.220 Similar albeit slightly
369
Diagnostic Gynecologic and Obstetric Pathology

Table 13.14  Outcome Risk of Cervical Neoplasia Following Prophylactic Ablation of the Squamocolumnar Junction

Reference Population Group Method Outcome Measure Change in Risk Versus Controls

214 General All Cautery SCC 6-fold reductiona

215 General All Cautery SCC 54-fold reduction a

216 South Africa HIV−/HPV− Cryotherapy HPV+ 55% reduction

221 Sourth Africa HPV+ Cryotherapy CIN2+ 70% reduction


a
Some differences noted between cases and controls in terms of socioeconomic status, age, and screening suggesting confounding variables.
CIN2, Cervical intraepithelial neoplasia grade 2; HIV, human immunodeficiency virus; HPV, human papillomavirus; SCC, squamous cell carcinoma.

less reductions were shown in HIV-infected women.221 progressively higher index of associated HPV-16. This is
These studies are summarized in Table 13.14. What should reflected indirectly in the high efficacy rates for all CIN2
be clear from these studies is that strategies that target the and CIN3 in recent vaccine studies and suggests the
source of cervical cancers have considerable promise and vaccines will have a major impact on clinically
in some settings could offer greater cost effectiveness than significant CIN2 and CIN3.
complex screening guidelines.222 • There is evidence that pre-emptive ablation of the SCJ in
women who are HPV positive will reduce their risk of an
HSIL or cancer outcome.

KEY POINTS
• The squamocolumnar junction (SCJ) has a unique
Please see the Appendix for suggested ICD-10 codes.
morphology and immunophenotype, emblematic of
cells capable of multi-potential differentiation, and the
SCJ immunophenotype is reproduced in a high References
percentage of high-grade squamous intraepithelial
lesions (HSILs). 1. Darragh TM, Colgan TJ, Cox JT, et al: The lower anogenital squamous
terminology standardization project for HPV-associated lesions:
• The diagnosis of cervical intraepithelial neoplasia grade background and consensus recommendations from the College of
2 (CIN2) is poorly reproduced across observers and p16 American Pathologists and the American Society for Colposcopy and
Cervical Pathology. Arch Pathol Lab Med 136:1266–1297, 2012.
immunostaining, like high-risk human papillomaviruses 2. Schiffman MH, Brinton LA: The epidemiology of cervical carcinogenesis.
(HPVs), and has a broad range of expression across both Cancer 76(10 Suppl):1888–1901, 1995.
low-grade squamous intraepithelial lesions (LSILs) and 3. Herfs M, Yamamoto Y, Laury A, et al: A discrete population of
squamocolumnar junction cells implicated in the pathogenesis of cervical
HSILs. Moreover p16 does not reliably predict an HSIL cancer. Proc Natl Acad Sci USA 109:10516–10521, 2012.
outcome in LSILs. It has the most value (like HPV 4. Rosenfeld WD, Rose E, Vermund SH, et al: Follow-up evaluation of
testing) when it is negative. cervicovaginal human papillomavirus infection in adolescents. J Pediatr
121(2):307–311, 1992.
• Temporal histologic “progression” from CIN1 to CIN2 5. Kjær SK, Frederiksen K, Munk C, et al: Long-term absolute risk of
or CIN3, although a time-honored concept, must be cervical intraepithelial neoplasia grade 3 or worse following human
papillomavirus infection: role of persistence. J Natl Cancer Inst 102(19):
viewed critically, because it can be falsely implied by 1478–1488, 2010.
separate sequential or concurrent viral infections, 6. Saslow D, Andrews KS, Manassaram-Baptiste D, et al: Human
sampling errors, and histologic misclassification of the papillomavirus vaccination guideline update: American Cancer Society
guideline endorsement. American Cancer Society Guideline Development
index and outcome biopsy. Group. CA Cancer J Clin 66:375–385, 2016.
• Lesions falling into the gray zone between LSIL and 7. Khan MJ, Castle PE, Lorincz AT, et al: The elevated 10-year risk of cervical
HSIL (CIN1-2) are lesions of indeterminate (or precancer and cancer in women with human papillomavirus (HPV) type
16 or 18 and the possible utility of type-specific HPV testing in clinical
intermediate) grade. If the pathologist is uncertain, this practice. J Natl Cancer Inst 97:1072–1079, 2005.
should be communicated to the clinician. 8. Castle PE, Rodríguez AC, Burk RD, et al: Proyecto Epidemiológico
Guanacaste (PEG) Group: short term persistence of human papillomavirus
• Always consider an extensive (giant) condyloma when and risk of cervical precancer and cancer: population based cohort study.
faced with a large, well-differentiated verrucous cervical Br Med J 339:b2569, 2009.
lesion in a young woman. 9. Castle PE, Cox JT, Schiffman M, et al: Factors influencing histologic
confirmation of high-grade squamous intraepithelial lesion cytology.
• A formal diagnosis of “microinvasive” squamous Obstet Gynecol 112:637–645, 2008.
carcinomas is best replaced with the diagnosis of 10. Munoz N, Bosch FX, de Sanjose S, et al: Epidemiologic classification of
“superficially invasive” squamous carcinoma, with human papillomavirus types associated with cervical cancer. N Engl J Med
348(6):518–527, 2003.
appropriate information provided to facilitate 11. Smith JS, Lindsay L, Hoots B, et al: Human papillomavirus type
management. distribution in invasive cervical cancer and high-grade cervical lesions: a
meta-analysis update. Int J Cancer 121(3):621–632, 2007.
• Increasing the stringency of the criteria (in terms of 12. Sherman ME, Wang SS, Wheeler CM, et al: Determinants of human
interobserver agreement, association with high-grade, papillomavirus load among women with histological cervical
cytology, etc.) for defining CIN2 is associated with a intraepithelial neoplasia 3: dominant impact of surrounding low-grade
lesions. Cancer Epidemiol Biomarkers Prev 12:1038–1044, 2003.
370
Cervical Squamous Neoplasia 13
13. Sherman ME, Wang SS, Wheeler CM, et al: Determinants of human 39. Kjaer SK, Engholm G, Dahl C, et al: Case-control study of risk factors for
papillomavirus load among women with histological cervical cervical squamous cell neoplasia in Denmark. IV: role of smoking habits.
intraepithelial neoplasia 3: dominant impact of surrounding low-grade Eur J Cancer Prev 5:359–365, 1996.
lesions. Cancer Epidemiol Biomarkers Prev 12:1038–1044, 2003. 40. Winer RL, Hughes JP, Feng Q, et al: Condom use and the risk of genital
14. Ylitalo N, Josefsson A, Melbye M, et al: A prospective study showing human papillomavirus infection in young women. N Engl J Med 354:
long-term infection with human papillomavirus 16 before the 2645–2654, 2006.
development of cervical carcinoma in situ. Cancer Res 60(21):6027–6032, 41. Tobian AA, Serwadda D, Quinn TC, et al: Male circumcision for the
2000. prevention of HSV-2 and HPV infections and syphilis. N Engl J Med
15. Hopman EH, Rozendaal L, Voorhorst FJ, et al: High-risk human 360(13):1298–1309, 2009.
papillomavirus in women with normal cervical cytology prior to the 42. International Collaboration of Epidemiological Studies of Cervical Cancer,
development of abnormal cytology and colposcopy. Br J Obstet Gynaecol Appleby P, Beral V, et al: Cervical cancer and hormonal contraceptives:
107:600–604, 2000. collaborative reanalysis of individual data for 16,573 women with cervical
16. Wang SS, Wheeler CM, Hildesheim A, et al: Human leukocyte antigen cancer and 35,509 women without cervical cancer from 24
class I and II alleles and risk of cervical neoplasia: results from a epidemiological studies. Lancet 370(9599):1609–1621, 2007.
population-based study in Costa Rica. J Infect Dis 184(10):1310–1314, 43. Ali S, Astley SB, Sheldon TA, et al: Detection and measurement of DNA
2001. adducts in the cervix of smokers and non-smokers. Int J Gynecol Cancer
17. Herrero R, Hildesheim A, Bratti C, et al: Population-based study of human 4(3):188–193, 1994.
papillomavirus infection and cervical neoplasia in rural Costa Rica. J Natl 44. Kapeu AS, Luostarinen T, Jellum E, et al: Is smoking an independent risk
Cancer Inst 92(6):464–474, 2000. factor for invasive cervical cancer? A nested case-control study within
18. Klencke BJ, Palefsky JM: Anal cancer: an HIV-associated cancer. Hematol Nordic biobanks. Am J Epidemiol 169(4):480–488, 2009.
Oncol Clin North Am 17:859–872, 2003. 45. Lee PN, Thornton AJ, Hamling JS: Epidemiological evidence on
19. ter Haar-van Eck SA, Rischen-Vos J, Chadha-Ajwani S, et al: The incidence environmental tobacco smoke and cancers other than lung or breast. Regul
of cervical intraepithelial neoplasia among women with renal transplant Toxicol Pharmacol 80:134–163, 2016.
in relation to cyclosporine. Br J Obstet Gynaecol 102(1):58–61, 1995. 46. Hildesheim A, Herrero R, Castle PE, et al: HPV co-factors related to the
20. Alloub MI, Barr BB, McLaren KM, et al: Human papillomavirus infection development of cervical cancer: results from a population-based study in
and cervical intraepithelial neoplasia in women with renal allografts. Costa Rica. Br J Cancer 84(9):1219–1226, 2001.
Br Med J 298(6667):153–156, 1989. 47. Reesink-Peters N, Ossewaarde JM, van der Zee AG, et al: No association of
21. Halpert R, Fruchter RG, Sedlis A, et al: Human papillomavirus and lower anti-Chlamydia trachomatis antibodies and severity of cervical neoplasia.
genital neoplasia in renal transplant patients. Obstet Gynecol 68:251–258, Sex Transm Infect 77(2):101–102, 2001.
1986. 48. Nonato DR, Alves RR, Ribeiro AA, et al: Prevalence and factors associated
22. Wright TC, Jr, Sun XW: Anogenital papillomavirus infection and neoplasia with coinfection of human papillomavirus and Chlamydia trachomatis in
in immunodeficient women. Obstet Gynecol Clin North Am 23(4):861–893, adolescents and young women. Am J Obstet Gynecol 215(6):753.e1–753.
1996. e9, 2016.
23. Moscicki AB, Ellenberg JH, Vermund SH, et al: Prevalence of and risks for 49. Marrazzo JM, Stine K, Koutsky LA: Genital human papillomavirus
cervical human papillomavirus infection and squamous intraepithelial infection in women who have sex with women: a review. Am J Obstet
lesions in adolescent girls: impact of infection with human Gynecol 183(3):770–774, 2000.
immunodeficiency virus. Arch Pediatr Adolesc Med 154:127–134, 2000. 50. Fethers K, Marks C, Mindel A, et al: Sexually transmitted infections and
24. Ahdieh L, Munoz A, Vlahov D, et al: Cervical neoplasia and repeated risk behaviours in women who have sex with women. Sex Transm Infect
positivity of human papillomavirus infection in human 76(5):345–349, 2000.
immunodeficiency virus-seropositive and -seronegative women. Am J 51. Wright TC, Jr: Cervical cancer screening in the 21st century: is it time to
Epidemiol 151(12):1148–1157, 2000. retire the PAP smear? Clin Obstet Gynecol 50:313–323, 2007.
25. Ellerbrock TV, Chiasson MA, Bush TJ, et al: Incidence of cervical 52. McKenna M, McMenamin M, McDowell A: HPV16 and HPV18 genotyping
squamous intraepithelial lesions in HIV-infected women. J Am Med Assoc triage in young women with borderline cytology or mild dyskaryosis:
283(8):1031–1037, 2000. effect of age on genotype-specific risk of high-grade CIN. Cytopathology
26. Cardillo M, Hagan R, Abadi J, et al: CD4 T-Cell count, viral load, 27:261–268, 2016.
squamous in-traepithelial lesions in women infected with the human 53. Lazcano-Ponce E, Herrero R, Munoz N, et al: Epidemiology of HPV
immunodeficiency virus. Cancer 93(2):111–114, 2001. infection among Mexican women with normal cervical cytology. Int J
27. Heard I, Tassie JM, Schmitz V, et al: Increased risk of cervical disease Cancer 91:412–420, 2001.
among human immunodeficiency virus-infected women with severe 54. Gyllensten U, Gustavsson I, Lindell M, et al: Primary high-risk HPV
immunosuppression and high human papillomavirus load (1). Obstet screening for cervical cancer in post-menopausal women. Gynecol Oncol
Gynecol 96(3):403–409, 2000. 125(2):343–345, 2012.
28. La Ruche G, You B, Mensah-Ado I, et al: Human papillomavirus and 55. Smelov V, Elfström KM, Johansson AL, et al: Long-term HPV type-specific
human immunodeficiency virus infections: relation with cervical risks of high-grade cervical intraepithelial lesions: a 14-year follow-up of a
dysplasia-neoplasia in African women. Int J Cancer 76(4):480–486, randomized primary HPV screening trial. Int J Cancer 136:1171–1180,
1998. 2015.
29. Parkin DM, Wabinga H, Nambooze S, et al: AIDS-related cancers in 56. Schiffman M, Burk RD, Boyle S, et al: A study of genotyping for
Africa: maturation of the epidemic in Uganda. AIDS 13(18):2563–2570, management of human papillomavirus-positive, cytology-negative cervical
1999. screening results. J Clin Microbiol 53:52–59, 2015.
30. Franceschi S, Dal Maso L, Arniani S, et al: Risk of cancer other than 57. Bryans FE, Boyes DA, Fidler HK: The influence of a cytological screening
Kaposi’s sarcoma and non-Hodgkin’s lymphoma in persons with AIDS in program upon the incidence of invasive squamous cell carcinoma of the
Italy. Cancer and AIDS Registry Linkage Study. Br J Cancer 78(7):966–997, cervix in British Columbia. Am J Obstet Gynecol 88:898–906, 1964.
1998. 58. Miller AB: The (in)efficiency of cervical screening in Europe. Eur J Cancer
31. Kahesa C, Mwaiselage J, Wabinga HR: Association between invasive cancer 38(3):321–326, 2002.
of the cervix and HIV-1 infection in Tanzania: the need for dual screening. 59. Van de Velde N, Boily MC, Drolet M, et al: Population-level impact
BMC Public Health 8:262, 2008. of the bivalent, quadrivalent, and nonavalent human papillomavirus
32. Chokunonga E, Levy LM, Bassett MT, et al: Aids and cancer in Africa: the vaccines: a model-based analysis. J Natl Cancer Inst 21(104):1712–1723,
evolving epidemic in Zimbabwe. AIDS 13(18):2583–2588, 1999. 2012.
33. La Ruche G, Leroy V, Mensah-Ado I, et al: Short-term follow up of cervical 60. The 1988 Bethesda System for reporting cervical/vaginal cytologic
squamous intraepithelial lesions associated with HIV and human diagnoses. Developed and approved at the National Cancer Institute
papillomavirus infections in Africa. Int J STD AIDS 10(6):363–368, 1999. Workshop, Bethesda, Maryland, USA, December 12-13, 1988. Acta Cytol
34. Lomalisa P, Smith T, Guidozzi F: Human immunodeficiency virus 33(5):567–574, 1989.
infection and invasive cervical cancer in South Africa. Gynecol Oncol 61. Solomon D, Davey D, Kurman R, et al: The 2001 Bethesda System:
77(3):460–463, 2000. terminology for reporting results of cervical cytology. JAMA 287(16):
35. Karlsson R, Jonsson M, Edlund K, et al: Lifetime number of partners as 2114–2119, 2002.
the only independent risk factor for human papillomavirus infection: a 62. Nayar R, Wilbur DC, editors: The Bethesda System for reporting cervical
population-based study. Sex Transm Dis 22(2):119–127, 1995. cytology: definitions, criteria, and explanatory notes, ed 3, New York, 2015,
36. Thomas DB, Ray RM, Kuypers J, et al: Human papillomaviruses and Springer.
cervical cancer in Bangkok. III. The role of husbands and commercial sex 63. Nayar R, Wilbur DC: The Pap test and Bethesda 2014. Acta Cytol 59:
workers. Am J Epidemiol 153(8):740–748, 2001. 121–132, 2015.
37. Agarwal SS, Sehgal A, Sardana S, et al: Role of male behavior in cervical 64. Nitschmann C, May T, Mirkovic J, et al: Screening history among women
carcinogenesis among women with one lifetime sexual partner. Cancer with invasive cervical cancer in an academic medical center: will we miss
72(5):1666–1669, 1993. cancers following updated guidelines? J Womens Health (Larchmt)
38. Krebs HB, Helmkamp BF: Treatment failure of genital condylomata 25:826–831, 2016.
acuminata in women: role of the male sexual partner. Am J Obstet Gynecol 65. Genest DR, Dean B, Lee KR, et al: Qualifying the cytologic diagnosis of
165(2):337–339, 1991. “atypical squamous cells of undetermined significance” affects the
371
Diagnostic Gynecologic and Obstetric Pathology

predictive value of a squamous intraepithelial lesion on subsequent 92. Huang EC, Tomic MM, Hanamornroongruang S, et al: p16ink4 and
biopsy. Arch Pathol Lab Med 122:338–341, 1998. cytokeratin 7 immunostaining in predicting HSIL outcome for low-grade
66. Human papillomavirus testing for triage of women with cytologic squamous intraepithelial lesions: a case series, literature review and
evidence of low-grade squamous intraepithelial lesions: baseline data commentary. Mod Pathol 29(12):1501–1510, 2016.
from a randomized trial. The Atypical Squamous Cells of Undetermined 93. Mills AM, Paquette C, Castle PE, et al: Risk stratification by p16
Significance/Low-Grade Squamous Intraepithelial Lesions Triage Study immunostaining of CIN1 biopsies: a retrospective study of patients from
(ALTS) Group. J Natl Cancer Inst 92:397–402, 2000. the quadrivalent HPV vaccine trials. Am J Surg Pathol 39:611–617, 2015.
67. Cibas ES, Browne TJ, Bassichis MH, et al: Enlarged squamous cell nuclei 94. Richart RM: Cervical intraepithelial neoplasia. Pathol Annu 8:301–328,
in cervical cytologic specimens from perimenopausal women: a cause of 1973.
ASC overdiagnosis. Am J Clin Pathol 124:58–61, 2005. 95. Burghardt E: Early histologic diagnosis of cervical cancer, Stuttgart, 1973,
68. Sorosky JI, Kaminski PF, Wheelock JB, et al: Clinical significance of Georg Thieme.
hyperkeratosis and parakeratosis in otherwise negative Papanicolaou 96. Koss LG: From koilocytosis to molecular biology: the impact of cytology
smears. Gynecol Oncol 39(2):132–134, 1990. on concepts of early human cancer. The 32nd Maude Abbott lecture. Mod
69. Kurman RJ, Henson DE, Herbst AL, et al: Interim guidelines for Pathol 2:526–535, 1989.
management of abnormal cervical cytology. The 1992 National Cancer 97. Purola E, Savia E: Cytology of gynecologic condyloma acuminatum. Acta
Institute Workshop. JAMA 271(23):1866–1869, 1994. Cytol 21:26–31, 1977.
70. Kiviat NB, Paavonen JA, Brockway J, et al: Cytologic manifestations of 98. Meisels A, Fortin R, Roy M: Condylomatous lesions of the cervix. II.
cervical and vaginal infections. I. Epithelial and inflammatory cellular Cytologic, colposcopic and histopathologic study. Acta Cytol 21:379–390,
changes. JAMA 253:989–996, 1985. 1977.
71. Kiviat NB, Peterson M, Kinney-Thomas E, et al: Cytologic manifestations 99. Meisels A, Roy M, Fortier M, et al: Human papillomavirus infection of the
of cervical and vaginal infections. II. Confirmation of Chlamydia cervix: the atypical condyloma. Acta Cytol 25:7–16, 1981.
trachomatis infection by direct immunofluorescence using monoclonal 100. Anderson MC, Brown CL, Buckley CH, et al: Current views on cervical
antibodies. JAMA 253:997–1000, 1985. intraepithelial neoplasia. J Clin Pathol 44:969–978, 1991.
72. Symmans F, Mechanic L, MacConnell P, et al: Correlation of cervical 101. Prendiville W, Cullimore J, Norman S: Large loop excision of the
cytology and human papillomavirus DNA detection in postmenopausal transformation zone (LLETZ): a new method of management for women
women. Int J Gynecol Pathol 11:204–209, 1992. with cervical intraepithelial neoplasia. Br J Obstet Gynaecol 96:1054–1060,
73. Kaminski PF, Sorosky JI, Wheelock JB, et al: The significance of atypical 1989.
cervical cytology in an older population. Obstet Gynecol 73(1):13–15, 102. Moscicki AB, Shiboski S, Hills NK, et al: Regression of low-grade
1989. squamous intra-epithelial lesions in young women. Lancet
74. Kaminski PF, Stevens CW, Jr, Wheelock JB: Squamous atypia on cytology: 364:1678–1683, 2004.
the influence of age. J Reprod Med 34(9):617–620, 1989. 103. Crane JM: Pregnancy outcome after loop electrosurgical excision
75. Wilbur DC: False negatives in focused rescreening of Papanicolaou procedure: a systematic review. Obstet Gynecol 102(5 Pt 1):1058–1062,
smears: how frequently are “abnormal” cells detected in retrospective 2003.
review of smears preceding cancer or high-grade intraepithelial neoplasia? 104. Cai B, Ronnett BM, Stoler M, et al: Longitudinal evaluation of
Arch Pathol Lab Med 121:273–276, 1997. interobserver and intraobserver agreement of cervical intraepithelial
76. Saito K, Saito A, Fu YS, et al: Topographic study of cervical condyloma neoplasia diagnosis among an experienced panel of gynecologic
and intraepithelial neoplasia. Cancer 59:2064–2070, 1987. pathologists. Am J Surg Pathol 31:1854–1860, 2007.
77. Lee KR, Minter LJ, Crum CP: Koilocytotic atypia in Papanicolaou smears: 105. Castle PE, Stoler MH, Solomon D, et al: The relationship of community
reproducibility and biopsy correlations. Cancer 81:10–15, 1997. biopsy-diagnosed cervical intraepithelial neoplasia grade 2 to the quality
78. van der Marel J, van Baars R, Alonso I, et al: Oncogenic human control pathology-reviewed diagnoses: an ALTS report. Am J Clin Pathol
papillomavirus-infected immature metaplastic cells and cervical neoplasia. 127:805–815, 2007.
Am J Surg Pathol 38:470–479, 2014. 106. Carreon JD, Sherman ME, Guillén D, et al: CIN2 is a much less
79. Miyatake T, Ueda Y, Yoshino K, et al: Clonality analysis and human reproducible and less valid diagnosis than CIN3: results from a
papillomavirus infection in squamous metaplasia and atypical immature histological review of population-based cervical samples. Int J Gynecol
metaplasia of uterine cervix: is atypical immature metaplasia a precursor Pathol 26:441–446, 2007.
to cervical intraepithelial neoplasia 3? Int J Gynecol Pathol 26:180–187, 107. Sano T, Oyama T, Kashiwabara K, et al: Expression status of p16 protein is
2007. associated with human papillomavirus oncogenic potential in cervical and
80. Cox JT, Schiffman M, Solomon D, et al: Prospective follow-up suggests genital lesions. Am J Pathol 153:1741–1748, 1998.
similar risk of subsequent cervical intraepithelial neoplasia grade 2 or 3 108. Klaes R, Benner A, Friedrich T, et al: p16ink4a immunohistochemistry
among women with cervical intraepithelial neoplasia grade 1 or negative improves interobserver agreement in the diagnosis of cervical
colposcopy and directed biopsy. Am J Obstet Gynecol 188:1406–1412, intraepithelial neoplasia. Am J Surg Pathol 26:1389–1399, 2002.
2003. 109. Nishio S, Fujii T, Nishio H, et al: p16(INK4a) immunohistochemistry
81. Chen EY, Tran A, Raho CJ, et al: Histological ‘progression’ from low (LSIL) is a promising biomarker to predict the outcome of low grade cervical
to high (HSIL) squamous intraepithelial lesion is an uncommon event intraepithelial neoplasia: comparison study with HPV genotyping.
and an indication for quality assurance review. Mod Pathol 23:1045–1051, J Gynecol Oncol 24:215–221, 2013.
2010. 110. Hariri J, Øster A: The negative predictive value of p16ink4a to assess the
82. Coleman DV: Cytodiagnosis of viral infections: recent results. Cancer Res outcome of cervical intraepithelial neoplasia 1 in the uterine cervix. Int J
133:33–44, 1993. Gynecol Pathol 26:223–228, 2007.
83. Laverty CR, Russell P, Black J, et al: Adenovirus infection of the cervix. Acta 111. Sagasta A, Castillo P, Saco A, et al: p16 staining has limited value in
Cytol 21:114–117, 1977. predicting the outcome of histological low-grade squamous intraepithelial
84. Paavonen J, Vesterinen E, Meyer B, et al: Colposcopic and histologic lesions of the cervix. Mod Pathol 29:51–59, 2016.
findings in cervical chlamydial infecion. Obstet Gynecol 59:712–715, 112. Paquette C, Mills AM, Stoler MH: Predictive value of cytokeratin 7
1982. immunohistochemistry in cervical low-grade squamous intraepithelial
85. Herfs M, Vargas SO, Yamamoto Y, et al: A novel blueprint for ‘top down’ lesion as a marker for risk of progression to a high-grade lesion. Am J Surg
differentiation defines the cervical squamocolumnar junction during Pathol 40:236–243, 2016.
development, reproductive life, and neoplasia. J Pathol 229:460–468, 113. Park JJ, Sun D, Quade BJ, et al: Stratified mucin-producing intraepithelial
2013. lesions of the cervix: adenosquamous or columnar cell neoplasia? Am J
86. Herfs M, Somja J, Howitt BE, et al: Unique recurrence patterns of cervical Surg Pathol 24:1414–1419, 2000.
intraepithelial neoplasia after excision of the squamocolumnar junction. 114. Ma L, Fisk JM, Zhang RR, et al: Eosinophilic dysplasia of the cervix: a
Int J Cancer 136:1043–1052, 2015. newly recognized variant of cervical squamous intraepithelial neoplasia.
87. Kocken M, Helmerhorst TJ, Berkhof J, et al: Risk of recurrent high-grade Am J Surg Pathol 28:1474–1484, 2004.
cervical intraepithelial neoplasia after successful treatment: a long-term 115. Mittal K: Utility of proliferation-associated marker MIB-1 in evaluating
multi-cohort study. Lancet Oncol 12:441–450, 2011. lesions of the uterine cervix. Adv Anat Pathol 6:177–185, 1999.
88. Yang A, Kaghad M, Wang Y, et al: p63, a p53 homolog at 3q27-29, 116. Roh MH, Agoston E, Birch C, et al: p16 Immunostaining patterns in
encodes multiple products with transactivating, death-inducing, and microglandular hyperplasia of the cervix and their significance. Int J
dominant-negative activities. Mol Cell 2:305–316, 1998. Gynecol Pathol 28(2):107–113, 2009.
89. Quade BJ, Yang A, Wang Y, et al: Expression of the p53 homologue p63 in 117. Garzetti GG, Ciavattini A, Goteri G, et al: Vaginal micropapillary lesions
early cervical neoplasia. Gynecol Oncol 80:24–29, 2001. are not related to human papillomavirus infection: in situ hybridization
90. Witkiewicz AK, Hecht JL, Cviko A, et al: Microglandular hyperplasia: a and polymerase chain reaction detection techniques. Gynecol Obstet Invest
model for the de novo emergence and evolution of endocervical reserve 38:134–139, 1994.
cells. Hum Pathol 36:154–161, 2005. 118. Jovanovic AS, McLachlin CM, Shen L, et al: Postmenopausal squamous
91. Herfs M, Parra-Herran C, Howitt BE, et al: Cervical squamocolumnar atypia: a spectrum including “pseudo-koilocytosis.” Mod Pathol 8:
junction-specific markers define distinct, clinically relevant subsets 408–412, 1995.
of low-grade squamous intraepithelial lesions. Am J Surg Pathol 37: 119. Huettner PC, Gersell DJ: Placental site nodule: a clinicopathologic study
1311–1318, 2013. of 38 cases. Int J Gynecol Pathol 13:191–198, 1994.
372
Cervical Squamous Neoplasia 13
120. Kobak WH, Roman LD, Felix JC, et al: The role of endocervical curettage 149. Lecuru F, Neji K, Robin F, et al: Microinvasive carcinoma of the cervix:
at cervical conization for high-grade dysplasia. Obstet Gynecol 85:197–201, rationale for conservative treatment in early squamous cell carcinoma.
1995. Eur J Gynaecol Oncol 1:465–470, 1997.
121. Fine BA, Feinstein GI, Sabella V: The pre- and postoperative value of 150. Marana HR, de Andrade JM, Matthes AC, et al: Microinvasive carcinoma
endocervical curettage in the detection of cervical intraepithelial neoplasia of the cervix: analysis of prognostic factors. Eur J Gynaecol Oncol 22:64–66,
and invasive cervical cancer. Gynecol Oncol 71:46–49, 1998. 2001.
122. Andersen W, Frierson H, Barber S, et al: Sensitivity and specificity of 151. Ostor A, Rome R, Quinn M: Microinvasive adenocarcinoma of the
endocervical curettage and the endocervical brush for the evaluation of cervix: a clinicopathologic study of 77 women. Obstet Gynecol 89:88–93,
the endocervical canal. Am J Obstet Gynecol 159:702–707, 1988. 1997.
123. Cox JT, Schiffman M, Solomon D: Prospective follow-up suggests similar 152. Burghardt E, Winter R, Tamussino K, et al: Diagnosis and surgical
risk of subsequent cervical intraepithelial neoplasia grade 2 or 3 among treatment of cervical cancer. Crit Rev Oncol Hematol 17:181–231, 1994.
women with cervical intraepithelial neoplasia grade 1 or negative 153. Creasman WT, Zaino RJ, Major FJ, et al: Early invasive carcinoma of the
colposcopy and directed biopsy. Am J Obstet Gynecol 188:1406–1412, 2003. cervix (3 to 5 mm invasion): risk factors and prognosis. A Gynecologic
124. Kyrgiou M, Koliopoulos G, Martin-Hirsch P, et al: Obstetric outcomes after Oncology Group Study. Am J Obstet Gynecol 178:62–65, 1998.
conservative treatment for intraepithelial or early invasive cervical lesions: 154. van Nagell JR, Jr, Greenwell N, Powell DF, et al: Microinvasive carcinoma
systematic review and meta-analysis. Lancet 367:489–498, 2006. of the cervix. Am J Obstet Gynecol 145:981–991, 1983.
125. Crum CP: Laboratory management of CIN 2: the consensus is consensus. 155. Buckley SL, Tritz DM, Van Le L, et al: Lymph node metastases and
Am J Clin Pathol 130:162–164, 2008. prognosis in patients with stage IA2 cervical cancer. Gynecol Oncol 63:4–9,
126. Fine BA, Feinstein GI, Sabella V: The pre- and postoperative value of 1996.
endocervical curettage in the detection of cervical intraepithelial neoplasia 156. Delgado G, Bundy BN, Fowler WC, et al: Prospective surgical pathological
and invasive cervical cancer. Gynecol Oncol 71:46–49, 1998. study of stage I squamous carcinoma of the cervix: a Gynecologic
127. Vierhout ME, de Planque PM: Concomitant endocervical curettage and Oncology Group Study. Gynecol Oncol 35:314–320, 1989.
cervical conization. Acta Obstet Gynecol Scand 70:359–361, 1991. 157. Benedet JL, Anderson GH: Stage IA carcinoma of the cervix revisited.
128. Wright TC, Cox JT, Massad LS, et al: 2001 consensus guidelines for the Obstet Gynecol 87:1052–1059, 1996.
management of women with cervical intraepithelial neoplasia. Am J Obstet 158. McLachlin CM, Devine P, Muto M, et al: Pseudoinvasion of vascular
Gynecol 189:295–304, 2003. spaces: report of an artifact caused by cervical lidocaine injection prior to
129. Gardeil F, Barry-Walsh C, Prendiville W, et al: Persistent intraepithelial loop diathermy. Hum Pathol 25:208–211, 1994.
neoplasia after excision for cervical intraepithelial neoplasia grade III. 159. Kurman RJ, Carcangiu ML, Herrington S, et al: WHO classification of
Obstet Gynecol 89:419–422, 1997. tumours of female reproductive organs, Lyon, 2014, IARC.
130. Kim YT, Lee JM, Hur SY, et al: Clearance of human papillomavirus 160. Milliken DA, Shepherd JH: Fertility preserving surgery for carcinoma of
infection after successful conization in patients with cervical the cervix. Curr Opin Oncol 20:575–580, 2008.
intraepithelial neoplasia. Int J Cancer 126(8):1903–1909, 2010. 161. Reagan JW, Fu YS: Histologic types and prognosis of cancers of the uterine
131. Castanon A, Landy R, Sasieni PD: Is cervical screening preventing cervix. Int J Radiat Oncol Biol Phys 5:1015–1020, 1979.
adenocarcinoma and adenosquamous carcinoma of the cervix? Int J 162. Wang TY, Chen BF, Yang YC, et al: Histologic and immunophenotypic
Cancer 139(5):1040–1045, 2016. classification of cervical carcinomas by expression of the p53 homologue
132. Benedetti-Panici P, Maneschi F, D’Andrea G, et al: Early cervical carcinoma: p63: a study of 250 cases. Hum Pathol 32:479–486, 2001.
the natural history of lymph node involvement redefined on the basis of 163. Steeper TA, Piscioli F, Rosai J: Squamous cell carcinoma with sarcoma-like
thorough parametrectomy and giant section study. Cancer 88:2267–2274, stroma of the female genital tract: clinicopathologic study of four cases.
2000. Cancer 52:890–898, 1983.
133. Uyar DS, Eltabbakh GH, Mount SL: Positive predictive value of 164. Pang LC: Sarcomatoid squamous cell carcinoma of the uterine cervix with
liquid-based and conventional cervical Papanicolaou smears reported as osteoclast-like giant cells: report of two cases. Int J Gynecol Pathol 17:
malignant. Gynecol Oncol 89:227–232, 2003. 174–177, 1998.
134. Johnson SJ, Wadehra V: How predictive is a cervical smear suggesting 165. Koenig C, Turnicky RP, Kankam CF, et al: Papillary squamotransitional cell
invasive squamous cell carcinoma? Cytopathology 12:144–150, 2001. carcinoma of the cervix: a report of 32 cases. Am J Surg Pathol 21:915–921,
135. Nguyen GK, Nguyen-Ho P, Husain M, et al: Cervical squamous cell 1997.
carcinoma and its precursor lesions: cytodiagnostic criteria and pitfalls. 166. Robertson DI, Maung R, Duggan MA: Verrucous carcinoma of the genital
Anat Pathol 1:139–164, 1996. tract: is it a distinct entity? Can J Surg 36:147–151, 1993.
136. Cox JT: Management of atypical squamous cells of undetermined 167. Al Nafussi A, Al Yusif R: Papillary squamotransitional cell carcinoma of
significance and low-grade squamous intra-epithelial lesion by human the uterine cervix: an advanced stage disease despite superficial location:
papillomavirus testing. Best Pract Res Clin Obstet Gynaecol 15:715–741, report of two cases and review of the literature. Eur J Gynaecol Oncol
2001. 19:455–457, 1998.
137. Rushing L, Cibas ES: Frequency of tumor diathesis in smears from 168. Mittal KR, Demopoulos RI, Goswami S: Patterns of keratin 19 expression
women with squamous cell carcinoma of the cervix. Acta Cytol 41: in normal, metaplastic, condylomatous, atrophic, dysplastic, malignant
781–785, 1997. cervical squamous epithelium. Am J Clin Pathol 98:419–423, 1992.
138. Sankaranarayanan R, Nene BM, Dinshaw KA, et al: A cluster randomized 169. Husniy Dilek F, Kucukali T: Mucin production in carcinomas of the
controlled trial of visual, cytology and human papillomavirus screening uterine cervix. Eur J Obstet Gynecol Reprod Biol 79:149–151, 1998.
for cancer of the cervix in rural India. Int J Cancer 116:617–623, 2005. 170. Benda JA: Histopathologic prognostic factors in early stage cervical
139. Paraskevaidis E, Kitchener HC, Miller ID, et al: A population-based study carcinoma. J Natl Cancer Inst Monogr 21:27–34, 1996.
of microinvasive disease of the cervix—a colposcopic and cytologic 171. Pretorius R, Semrad N, Watring W, et al: Presentation of cervical cancer.
analysis. Gynecol Oncol 45:9–12, 1992. Gynecol Oncol 42:48–53, 1991.
140. Liu WM, Chao KC, Wang KI, et al: Colposcopic assessment in 172. Jennings OG, Soeters RP, Tiltman AJ, et al: The natural history of
microinvasive carcinoma of the cervix. Zhonghua Yi Xue Za Zhi (Taipei) carcinoma of the cervix in young women. S Afr Med J 82:351–354, 1992.
43:171–176, 1989. 173. Yang YC, Shen J, Tate JE, et al: Cervical cancer in young women in Taiwan:
141. Crum CP, Beach KJ, Hedley ML, et al: Dynamics of human papillomavirus prognosis is independent of papillomavirus or tumor cell type. Gynecol
infection between biopsy and excision of cervical intraepithelial neoplasia: Oncol 64:59–63, 1997.
results from the ZYC101a protocol. J Infect Dis 189:1348–1354, 2004. 174. Baay MF, Tjalma WA, Weyler J, et al: Prevalence of human papillomavirus
141a.  Crum CP, Lee: Unpublished data. in elderly women with cervical cancer. Gynecol Obstet Invest 52:248–251,
142. Soutter WP, Haidopoulos D, Gornall RJ, et al: Is conservative treatment 2001.
for adenocarcinoma in situ of the cervix safe? Br J Obstet Gynaecol 108: 175. Duggan B, Muderspach LI, Roman LD, et al: Cervical cancer in pregnancy:
1184–1189, 2001. reporting on planned delay in therapy. Obstet Gynecol 82(4 Pt 1):598–602,
143. Al Nafussi AI, Hughes DE: Histological features of CIN3 and their value 1993.
in predicting invasive microinvasive squamous carcinoma. J Clin Pathol 176. Allen DG, Planner RS, Tang PT, et al: Invasive cervical cancer in pregnancy.
47:799–804, 1994. Aust N Z J Obstet Gynaecol 35:408–412, 1995.
144. Tidbury P, Singer A, Jenkins D: CIN 3: the role of lesion size in invasion. 177. Monk BJ, Montz FJ: Invasive cervical cancer complicating intrauterine
Br J Obstet Gynaecol 99(7):583–586, 1992. pregnancy: treatment with radical hysterectomy. Obstet Gynecol 80(2):
145. Onishi J, Sato Y, Sawaguchi A, et al: Stratified mucin-producing 199–203, 1992.
intraepithelial lesion with invasive carcinoma: 12 cases with 178. Sivanesaratnam V, Jayalakshmi P, Loo C: Surgical management of early
immunohistochemical and ultrastructural findings. Hum Pathol 55: invasive cancer of the cervix associated with pregnancy. Gynecol Oncol
174–181, 2016. 48:68–75, 1993.
146. Wilkinson EJ, Komorowski RA: Borderline microinvasive carcinoma of the 179. Sorosky JI, Squatrito R, Ndubisi BU, et al: Stage I squamous cell cervical
cervix. Obstet Gynecol 51:472–476, 1978. carcinoma in pregnancy: planned delay in therapy awaiting fetal maturity.
147. Sedlis A, Sall S, Tsukada Y, et al: Microinvasive carcinoma of the uterine Gynecol Oncol 59:207–210, 1995.
cervix: a clinical-pathologic study. Am J Obstet Gynecol 133:64–74, 1979. 180. Fruscio R, Villa A, Chiari S, et al: Delivery delay with neoadjuvant
148. Hopkins MP, Morley GW: Microinvasive squamous cell carcinoma of the chemotherapy for cervical cancer patients during pregnancy: a series of
cervix. J Reprod Med 39:671–673, 1994. nine cases and literature review. Gynecol Oncol 126:192–197, 2012.
373
Diagnostic Gynecologic and Obstetric Pathology

181. Ricci C, Scambia G, De Vincenzo R: Locally advanced cervical cancer in lymph nodes: a Gynecologic Oncology Group and Southwest Oncology
pregnancy: overcoming the challenge. A case series and review of the Group Study. J Clin Oncol 17(5):1339–1348, 1999.
literature. Int J Gynecol Cancer 26(8):1490–1496, 2016. 202. Morris M, Gershenson DM, Burke TW, et al: A phase II study of
182. Lombard I, Vincent-Salomon A, Validire P, et al: Human papillomavirus carboplatin and cisplatin in advanced or recurrent squamous carcinoma
genotype as a major determinant of the course of cervical cancer. J Clin of the uterine cervix. Gynecol Oncol 53(2):234–238, 1994.
Oncol 16:2613–2619, 1998. 203. Rose PG, Bundy BN, Watkins EB, et al: Concurrent cisplatin-based
183. Schwartz SM, Daling JR, Shera KA, et al: Human papillomavirus and radiotherapy and chemotherapy for locally advanced cervical cancer.
prognosis of invasive cervical cancer: a population-based study. J Clin N Engl J Med 340(15):1144–1153, 1999.
Oncol 19:1906–1915, 2001. 204. Keys HM, Bundy BN, Stehman FB, et al: Cisplatin, radiation, adjuvant
184. Lai CH, Chang CJ, Huang HJ, et al: Role of human papillomavirus hysterectomy compared with radiation and adjuvant hysterectomy for
genotype in prognosis of early-stage cervical cancer undergoing primary bulky stage IB cervical carcinoma. N Engl J Med 340(15):1154–1161,
surgery. J Clin Oncol 25:3628–3634, 2007. 1999.
185. Dahlgren L, Erlandsson F, Lindquist D, et al: Differences in human 205. Peters WA, III, Liu PY, Barrett RJ, et al: Concurrent chemotherapy and
papillomavirus type may influence clinical outcome in early stage cervical pelvic radiation therapy compared with pelvic radiation therapy alone as
cancer. Anticancer Res 26:829–832, 2006. adjuvant therapy after radical surgery in high-risk early-stage cancer of the
186. Tong SY, Lee YS, Park JS, et al: Human papillomavirus genotype as a cervix. J Clin Oncol 18:1606–1613, 2000.
prognostic factor in carcinoma of the uterine cervix. Int J Gynecol Cancer 206. Stewart LA, Parmar MK, Tierney JF: Meta-analyses and large randomized,
17:1307–1313, 2007. controlled trials. N Engl J Med 338:61–62, 1998.
187. Covens A, Rosen B, Murphy J, et al: Changes in the demographics and 207. Sardi JE, Giaroli A, Sananes C, et al: Long-term follow-up of the first
perioperative care of stage IA(2)/IB(1) cervical cancer over the past 16 randomized trial using neoadjuvant chemotherapy in stage Ib squamous
years. Gynecol Oncol 81:133–137, 2001. carcinoma of the cervix: the final results. Gynecol Oncol 67:61–69, 1997.
188. Quinn MA, Benedet JL, Odicino F, et al: Carcinoma of the cervix uteri. 208. Benedetti-Panici P, Greggi S, Colombo A, et al: Neoadjuvant
FIGO 6th Annual Report on the Results of Treatment in Gynecological chemotherapy and radical surgery versus exclusive radiotherapy in locally
Cancer. Int J Gynaecol Obstet 95(Suppl 1):S43–S103, 2006. advanced squamous cell cervical cancer: results from the Italian
189. Zola P, Ferrero A, Fuso L, et al: Different types of hysterectomy in the Multicenter Randomized Study. J Clin Oncol 20:179–188, 2002.
radio-surgical treatment of early cervical cancer (FIGO Ib-IIa). Eur J 209. Symonds P, Kirwan J, Williams C, et al: Concomitant hydroxyurea plus
Gynaecol Oncol 23:236–242, 2002. radiotherapy versus radiotherapy for carcinoma of the uterine cervix.
190. Hopkins MP, Peters WA, III, Andersen W, et al: Invasive cervical cancer Cochrane Database Syst Rev (1):CD003918, 2004.
treated initially by standard hysterectomy. Gynecol Oncol 36:7–12, 1990. 210. Bizzarri N, Ghirardi V, Alessandri F, et al: Bevacizumab for the treatment
191. Landoni F, Maneo A, Colombo A, et al: Randomised study of radical of cervical cancer. Expert Opin Biol Ther 16:407–419, 2016.
surgery versus radiotherapy for stage Ib-IIa cervical cancer. Lancet 211. Riethmuller D, Jacquard AC, Lacau St Guily J, et al: Potential impact of a
350(9077):535–540, 1997. nonavalent HPV vaccine on the occurrence of HPV-related diseases in
192. Piver MS, Chung WS: Prognostic significance of cervical lesion size and France. BMC Public Health 15:453, 2015.
pelvic node metastases in cervical carcinoma. Obstet Gynecol 46:507–510, 212. Simms KT, Smith MA, Lew JB, et al: Will cervical screening remain
1975. cost-effective in women offered the next generation nonavalent HPV
193. Finan MA, DeCesare S, Fiorica JV, et al: Radical hysterectomy for stage IB1 vaccine? Results for four developed countries. Int J Cancer 139(12):
versus IB2 carcinoma of the cervix: does the new staging system predict 2771–2780, 2016.
morbidity and survival? Gynecol Oncol 62:139–147, 1996. 213. Canfell K, Chesson H, Kulasingam SL, et al: Modeling preventative
194. Trattner M, Graf AH, Lax S, et al: Prognostic factors in surgically treated strategies against human papillomavirus-related disease in developed
stage Ib-I1b cervical carcinomas with special emphasis on the importance countries. Vaccine 30(Suppl 5):F157–F167, 2012.
of tumor volume. Gynecol Oncol 82:11–16, 2001. 214. Kauraniemi T, Räsänen-Virtanen U, Hakama M: Risk of cervical cancer
195. Biliatis I, Kucukmetin A, Patel A, et al: Small volume stage 1B1 cervical among an electrocoagulated population. Am J Obstet Gynecol 1 31 :
cancer: is radical surgery still necessary? Gynecol Oncol 126:73–77, 2012. 533–538, 1978.
196. Delgado G, Bundy BN, Fowler WC, et al: Prospective surgical pathological 215. Peyton FW, Peyton RR, Anderson VL, et al: The importance of
study of stage I squamous carcinoma of the cervix: a Gynecologic cauterization to maintain a healthy cervix: long-term study from a private
Oncology Group Study. Gynecol Oncol 35:314–320, 1989. gynecologic practice. Am J Obstet Gynecol 131:374–380, 1978.
197. Fuller AF, Jr, Elliott N, Kosloff C, et al: Determinants of increased risk for 216. Taylor S, Wang C, Wright TC, et al: Reduced acquisition and reactivation
recurrence in patients undergoing radical hysterectomy for stage IB and of human papillomavirus infections among older women treated with
IIA carcinoma of the cervix. Gynecol Oncol 33:34–39, 1989. cryotherapy: results from a randomized trial in South Africa. BMC Med
198. Sedlis A, Bundy BN, Rotman MZ, et al: A randomized trial of pelvic 8:40, 2010.
radiation therapy versus no further therapy in selected patients with stage 217. Gaffikin L, Blumenthal PD, Emerson M, et al: Safety, acceptability, and
IB carcinoma of the cervix after radical hysterectomy and pelvic feasibility of a single-visit approach to cervical-cancer prevention in rural
lymphadenectomy: a Gynecologic Oncology Group Study. Gynecol Oncol Thailand: a demonstration project. Lancet 361:814–820, 2003.
73:177–183, 1999. 218. Chumworathayi B, Blumenthal PD, Limpaphayom KK, et al: Effect of
199. Keys HM, Bundy BN, Stehman FB, et al: Radiation therapy with and single-visit VIA and cryotherapy cervical cancer prevention program in Roi
without extrafascial hysterectomy for bulky stage IB cervical carcinoma: Et, Thailand: a preliminary report. J Obstet Gynaecol Res 36:79–85, 2010.
a randomized trial of the Gynecologic Oncology Group. Gynecol Oncol 219. Mirkovic J, Howitt BE, Roncarati P, et al: Carcinogenic HPV infection in
89:343–353, 2003. the cervical squamo-columnar junction. J Pathol 236:265–271, 2015.
200. Eddy GL, Bundy BN, Creasman WT, et al: Treatment of (“bulky”) stage IB 220. Denny L, Kuhn L, Hu CC, et al: Human papillomavirus-based cervical
cervical cancer with or without neoadjuvant vincristine and cisplatin prior cancer prevention: long-term results of a randomized screening trial.
to radical hysterectomy and pelvic/para-aortic lymphadenectomy: a phase J Natl Cancer Inst 102:1557–1567, 2010.
III trial of the Gynecologic Oncology Group. Gynecol Oncol 106:362–369, 221. Kuhn L, Wang C, Tsai WY, et al: Efficacy of human papillomavirus-based
2007. screen-and-treat for cervical cancer prevention among HIV-infected
201. Whitney CW, Sause W, Bundy BN, et al: Randomized comparison of women. AIDS 24:2553–2561, 2010.
fluorouracil plus cisplatin versus hydroxyurea as an adjunct to radiation 222. Herfs M, Crum CP: Cervical cancer: squamocolumnar junction ablation—
therapy in stage IIB-IVA carcinoma of the cervix with negative para-aortic tying up loose ends? Nat Rev Clin Oncol 12:378–380, 2015.

374

You might also like