Anda di halaman 1dari 10

Current Concepts in Cervical Pathology

Kay J. Park, MD; Robert A. Soslow, MD

Context.The correct diagnosis and reporting of cervical lated to prognosis that should be included in the pathology
in situ and invasive carcinoma are essential for the appropri- report.
ate clinical management of patients with human papilloma- Data Sources.Review of current literature.
virusassociated disease. Conclusions.There are many mimics and variants of
Objectives.To review common mistakes made in the cervical squamous and glandular lesions that can be re-
diagnosis of cervical dysplasia and invasive carcinoma, de- solved with ancillary studies and careful histologic exami-
scribe variants and benign mimics of high-grade squamous nation. Prognostically important features, such as tumor
intraepithelial lesion and adenocarcinoma in situ, and dis- size, presence of vascular invasion, and margin status,
cuss available ancillary studies that can be useful in making should always be included in the pathology report.
the distinctions as well as to review important factors re- (Arch Pathol Lab Med. 2009;133:729738)

C ervical cancer is one of the most common cancers in


women worldwide with about 493 000 new cases
each year resulting in 274 000 deaths globally.1 In the Unit-
HUMAN PAPILLOMAVIRUS
Human papillomavirus preferentially infects cells in the
cervical transformation zone, an area of active cell turn-
ed States it is the 14th most common cancer in women over. Basal cells, which feature the HPV receptor, are a
and affects nearly 12 000 women each year causing ap- natural target for infection. The virus remains in the cell
proximately 4000 deaths.2 Most cervical carcinomas are in a latent state until activation causes viral replication and
etiologically related to the human papillomavirus (HPV) squamous dysplasia. Low-grade squamous intraepithelial
and about 70% of all cervical carcinomas are caused by 2 lesion/cervical intraepithelial neoplasia 1 (LSIL/CIN 1) is
types of high-risk HPV, 16 and 18.3 Once infected by HPV, characterized by koilocytotic atypia, nuclear enlargement,
cervical neoplasia undergoes a stepwise progression start- hyperchromasia, and perinuclear cytoplasmic clearing.
ing from preinvasive lesions that can be detected by These features are the result of viral proteins that affect
screening and cured with complete excision. Therefore, DNA synthesis and the structure of intermediate filaments
the detection of such lesions is critical in the appropriate in the host cell cytoplasm. Human papillomavirus gene
management of patients. Although it is important not to expression is tightly controlled in LSILs, which result
miss lesions, the overdiagnosis of benign atypias also has from productive infections of cells that have begun the
important clinical implications and can result in undue process of differentiation. High-grade squamous intraep-
physical and psychologic distress, as well as waste valu- ithelial lesions/cervical intraepithelial neoplasia 2 to 3
able resources and funds. Pathologists are faced with (HSILs/CIN 23) do not support HPV genomic replica-
many diagnostic difficulties when assessing cervical tis- tion and, therefore, do not usually have the nuclear or cy-
sue, particularly in distinguishing benign from in situ dis- toplasmic characteristics of an active viral infection.
ease and in situ from invasive carcinoma. This review fo- Traditionally, cervical squamous lesions were thought to
cuses on variants and mimics of in situ squamous and progress in a stepwise fashion from low grade to high
glandular lesions, important prognostic factors in invasive grade, the theory being that a focus of HSIL arises in,
disease that must be reported by the pathologist, and an- gradually expands, and finally replaces LSIL as a mono-
cillary tests that are available to assist in the diagnosis of clonal event. However, studies have shown that HSIL may
HPV-related lesions. also develop independently without progression or trans-
formation from LSIL; instead, they likely arise de novo
from epithelium adjacent to LSIL.4
Accurate histologic and cytologic classification of HPV-
Accepted for publication December 11, 2008.
From the Department of Pathology, Memorial Sloan-Kettering Cancer mediated lesions has important implications for guiding
Center, New York, New York. patient management, but studies have shown that inter-
The authors have no relevant financial interest in the products or observer and intraobserver reproducibility of diagnoses of
companies described in this article. cervical lesions is suboptimal. The atypical cells of unde-
Presented in part at the Surgical Pathology of Neoplastic Diseases termined significance (ASCUS)/LSIL Triage Study
course, Memorial Sloan-Kettering Cancer Center, New York, New York,
(ALTS)5 showed that variability of histologic diagnoses on
May 1216, 2008.
Reprints: Kay J. Park, MD, Department of Pathology, Memorial Sloan- biopsy is as great as with cytologic smears, even though
Kettering Cancer Center, 1275 York Ave, New York, NY 10065 histology is still considered the gold standard. Between
(e-mail: parkk@mskcc.org). the clinical center and central quality control groups, there
Arch Pathol Lab MedVol 133, May 2009 Current Concepts in Cervical PathologyPark & Soslow 729
was only moderate concordance, with the quality control range of proliferative rates with Mib-1810 (see section on an-
group more frequently rendering low-grade diagnoses. cillary studies on page 735). Follow-up studies have reported
CIN 1 had only 42% concordance, whereas the diagnostic that HPV-associated AIM showing p16 expression and high
extremes had much better concordance, 91% for normal proliferative rates subsequently recur as a bone fide dys-
and 77% for high grade. Most LSILs and even HSILs un- plastic lesion in many instances. It has been hypothesized
dergo spontaneous regression within 6 to 12 months due that AIM lesions that are HPV and p16 positive but hypo-
to host cell immune response. However, there is no way proliferative with Mib-1 may be early or regressing lesions.
to prospectively differentiate among lesions that are des- Atypical immature metaplasia is not commonly used as a
tined to regress, persist, or progress to carcinoma. Over- diagnostic term in surgical pathology reports. Our group
diagnosis of benign lesions as LSIL adds unnecessary cost currently diagnoses HSIL when we encounter AIM lesions
and wastes resources, as clinicians will often proceed di- that diffusely express p16 and are hyperproliferative with
rectly to excisional biopsy for LSIL, especially in older pa- Mib-1. Immunohistochemical stains that do not support a
tients. The patients physical and psychologic distress proliferative, HPV-associated lesion usually result in a report
should also not be underestimated. of AIM with a note describing the significance of the finding.
Eosinophilic dysplasia (ED), another descriptive term, has
SQUAMOUS LESIONS been used for intraepithelial lesions showing both squa-
The most common diagnostic challenges occur in dis- mous metaplasia and dysplasia.11 Eosinophilic dysplasia
tinguishing LSIL from benign reactive atypia, CIN 2 from is commonly associated with HPV infection and adjacent
CIN 3, and CIN 3 from atrophy and immature metaplasia. foci of classic HSIL. Most ED lesions are diffusely and
Mild nuclear cytologic changes, especially those that oc- strongly positive for p16, hyperproliferative with Mib-1,
cur in a background of inflammation, can be misinter- and positive for HPV DNA by polymerase chain reaction,
preted as LSIL. Normal squamous epithelium, with prom- all high or intermediate risk. Therefore, ED likely repre-
inent cytoplasmic glycogen or vacuolization, is also com- sents a variant of HSIL with misleading morphologic fea-
monly mistaken for koilocytosis. But true koilocytosis is tures that suggest a benign diagnosis. Eosinophilic dys-
often focal, whereas in normal epithelium there is no clear plasia is another term that is not commonly used in sur-
demarcation between abnormal and normal-appearing gical pathology reports. Our group currently diagnoses
epithelium. Benign epithelium lacks nuclear enlargement, HSIL when we encounter ED lesions that diffusely express
multinucleation (although binucleate cells are sometimes p16 and are hyperproliferative with Mib-1. Immunohis-
encountered), cellular disorganization, and maturation tochemical stains that do not support a proliferative, HPV-
disturbances. Probably the most important feature is the associated lesion result in a report of atypical metaplasia
lack of true nuclear atypia (Figure 1, A and B). with a note describing the significance of the finding.
The distinction between CIN 2 and CIN 3 is not as crucial,
because both lesions are in the high-grade category. Studies GLANDULAR LESIONS
have shown that the subjective criteria used for separating Adenocarcinoma in situ (AIS) is much less common
the different grades and the fact that the thickness of the than squamous intraepithelial lesion (SIL). Unlike squa-
immature basaloid cells can vary within the same lesion lead mous neoplasms in which precursor lesions predominate
to failures in reproducible diagnosis of CIN 2.6 over invasive carcinomas, the opposite is true for glan-
The importance of the difference between CIN 3 and dular lesions. It is more difficult to screen effectively for
atrophy or metaplasia is obvious. Atrophy and immature AIS than for SIL, so AIS may not be detected before the
metaplasia do not have the mitotic activity, nuclear pleo- development of invasive adenocarcinoma. Histologic fea-
morphism, loss of cell polarity, nuclear hyperchromasia, tures of AIS include preservation of normal glandular ar-
or clumped chromatin of HSIL. Reparative change can chitecture and involvement of part or all of the epithelium
also sometimes mimic HSIL with atypical basal cells oc- by enlarged, hyperchromatic, stratified nuclei with coarse
cupying the lower half of the epithelium. These cells have chromatin, small nucleoli, mitoses, and apoptosis. The cy-
regular nuclear contours, prominent nucleoli, and distinct toplasm can be depleted or abundant, vacuolated, granu-
cell borders, often accompanied by dense acute or chronic lar, and basophilic or eosinophilic (Figure 2). At least 90%
inflammation. of AIS cases are associated with HPV (most commonly
HPV 18 and 16). It coexists with SIL or squamous cell
SQUAMOUS VARIANT LESIONS carcinoma in 50% of cases.1223 Part of the difficulty in
Variants of squamous dysplasia that appear reactive or identifying AIS is that the lesion is not well visualized
metaplastic may also cause diagnostic dilemmas. These colposcopically as it can arise high up in the endocervical
include atypical immature metaplasia (AIM) and eosino- canal,17 and the cytologic criteria for identifying neoplastic
philic dysplasia (ED). glandular lesions are not as well defined as for SIL. Ad-
Atypical immature metaplasia is a loosely defined term enocarcinoma in situ has been described as multifocal,
for immature metaplastic cells with mild cytologic atypia, though the definitions of multifocality are often unclear.
first described by Crum et al in 1983.7 By their description, Various studies report that AIS can be multicentric (13%)19
AIM was similar to condyloma, that is, low-grade lesion. But and multiquadrant (53%).21 This is important because
since then, there has been a shift in the diagnosis to include multifocality may affect the meaning of a negative margin
both high-grade and low-grade lesions.810 This attests to the on cone excisions. Studies have shown that cone biopsies
fact that this entity spans a morphologic spectrum and likely for AIS with negative margins result in residual disease
includes many diagnostically challenging cases with an im- in a hysterectomy 6% to 44% of the time, whereas cone
mature appearance. There is lack of a uniform definition, biopsies for AIS with positive margins are associated with
which has resulted in inconsistencies in morphologic criteria. residual disease 44% to 75% of the time.24 Therefore, the
Several studies have shown that AIM can be HPV positive standard treatment for AIS is simple hysterectomy or a
or negative, be variably positive for p16, and display a large cold knife cone excision (not loop electrosurgical excision
730 Arch Pathol Lab MedVol 133, May 2009 Current Concepts in Cervical PathologyPark & Soslow
Figure 1. A, Benign squamous mucosa with glycogenization of cytoplasm forming pseudokoilocytes. The process involves the entire epithelium
(hematoxylin-eosin, original magnification 40). B, Low-grade squamous intraepithelial lesion (cervical intraepithelial neoplasia 1) with true
cytologic atypia, koilocytosis, and parabasal proliferation (hematoxylin-eosin, original magnification 400).
Figure 2. Adenocarcinoma in situ. The cells are pseudostratified and hyperchromatic with some cytoplasmic mucin. Note the luminal mitoses
and apoptotic bodies (hematoxylin-eosin, original magnification 200).
Figure 3. Tubal metaplasia. The cells are pseudostratified and appear somewhat atypical, but close evaluation reveals cells resembling fallopian
tube epithelium with ciliated and secretory type cells (hematoxylin-eosin, original magnification 200).
Figure 4. Tuboendometrioid metaplasia (hematoxylin-eosin, original magnification 200).
Figure 5. Tubal metaplasia with irregularly shaped glands and stromal reaction mimicking invasive adenocarcinoma (hematoxylin-eosin, original
magnification 100).

Arch Pathol Lab MedVol 133, May 2009 Current Concepts in Cervical PathologyPark & Soslow 731
procedure [LEEP]) with widely clear margins (10 mm ide- GD; 69, adenocarcinoma in situ). Although this method
al according to Goldstein and Mani25). Therefore, it is im- improves interobserver discordance, the biologic and clin-
portant to always mention the status of the margins and ical significance of GD remains to be determined. The as-
give the closest distance from the lesion if possible. Some sociation between GD and HPV infection is variable, and,
AISs exhibit intestinal differentiation with goblet cells and as it only infrequently coexists with AIS, it has been sug-
sometimes Paneth or argentaffin cells.26 Other related le- gested that GD might not represent an AIS precursor at
sions include adenosquamous carcinoma in situ27 and all. It is therefore imprudent to make an unqualified di-
stratified mucin-producing intraepithelial lesion (SMILE), agnosis of GD.24 Application of ancillary tests, discussed
described subsequently. subsequently, can be contributory.
AIS MIMICS GLANDULAR VARIANT LESIONS
Glandular lesions that can mimic AIS include reactive SMILE, first described by Park et al in 2000,27 is an HPV-
glandular atypia, tubal/tuboendometrioid metaplasia, associated intraepithelial columnar cell neoplasm that is
and so-called glandular dysplasia (GD). thought to arise from the reserve cells of the transforma-
Reactive glandular atypias can be secondary to inflam- tion zone. Morphologically, SMILEs are composed of im-
mation, radiation, or viral infections. Glandular lesions mature stratified cells that, although appearing similar to
showing cytologic atypia can also be found in the context SILs, display intracytoplasmic mucin or cytoplasmic vac-
of Arias-Stella reaction, microglandular hyperplasia, en- uoles that separate the cells in the mid to lower layers of
dometriosis, and mesonephric remnants.28 In reactive the epithelium (Figure 6). Overt gland formation, seen in
changes, the nuclei are typically enlarged, have prominent typical AIS, is not found in SMILE. The most consistent
nucleoli, but lack nuclear hyperchromasia. Mitotic activity feature of SMILE is the spacing of nuclei by mucin, which
is absent or minimal, as is pseudostratification. Papillary can be highlighted with a mucicarmine stain. SMILEs are
endocervicitis, featuring exaggerated papillary projections commonly associated with HSIL, AIS, adenocarcinoma,
and a dense infiltrate of acute and/or chronic inflamma- squamous carcinoma, and adenosquamous carcinoma.
tion, can also occur.29 Radiation change causes the nuclei The natural history of these lesions has not been fully elu-
and cytoplasm to enlarge, often with vacuolization, but cidated, but it has been reported that lesions may recur in
with only a slight increase in the nuclear to cytoplasmic the form of SIL or AIS. As is the case with AIM and ED,
ratio. The nuclei can look round, look vesicular with prom- SMILE should not be used as a stand-alone term in sur-
inent nucleoli, and rarely have a smudged hyperchromatic gical pathology reports. Either AIS or adenosquamous car-
appearance.30 cinoma in situ can be used, depending on the context and
Tubal metaplasia tends to occur in the upper endocer- the pathologists preference.
vix. It is usually not difficult to distinguish from AIS be-
cause ciliated, intercalated (peg), and secretory cells, more INVASIVE CARCINOMA
characteristically found in the fallopian tube, are present The diagnosis of invasive squamous or adenocarcinoma
at least focally in tubal metaplasia. Atypical tubal meta- is usually not challenging. Stage is the most important
plasia is a form of tubal metaplasia in which the glands prognostic factor in cervical cancer, but unlike the practice
are lined by similar tubal type cells, but these are crowded in other organ systems, staging is primarily assessed by
and have larger, more hyperchromatic nuclei and are physical examination. In developed countries, imaging
pseudostratified. Mitosis and apoptosis are not common modalities may also be used, but physical examination is
like they are in AIS, but the degree of atypia can be mis- still the gold standard in countries where these tests are
taken for AIS (Figure 3). Tuboendometrioid metaplasia is not widely available. Grossly inapparent carcinoma (car-
essentially tubal metaplasia without ciliated cells, al- cinoma detected by Papanicolaou test and biopsy not by
though it commonly displays more pseudostratification physical examination) is assessed on LEEP or cone biopsy,
and higher nuclear to cytoplasmic ratios than other tubal where stage is assigned by microscopic measurements of
metaplasia lesions; it lacks the architectural complexity, the extent of invasive carcinoma. Occult spread of disease
mitoses, apoptosis, and nuclear irregularity of AIS (Figure may be found at the time of surgery, but this does not
4). Tubal and tuboendometrioid metaplasia can involve change the initial stage (though it may change the treat-
the deep cervical wall with dilated irregular gland for- ment plan). Although various staging systems exist, the
mation and stromal reaction around the glands; this latter International Federation of Gynecology and Obstetrics
feature, in particular, may erroneously suggest a diagnosis (FIGO) system is almost universally used (Table). There is
of invasive adenocarcinoma31 (Figure 5). Careful exami- a clear difference in survival depending on stage with
nation of the nuclear features should allow one to properly near 99% 5-year survival for stage IA1 to 65% for IIB and
differentiate between benign and malignant. 43% for IIIB.37
Glandular dysplasia has been defined in a variety of In pathology reports for LEEPs and cone biopsies, the
ways, but in general, GD reportedly displays a lesser de- size of the invasive tumor, including depth of stromal in-
gree of abnormality than AIS.3235 The diagnostic criteria vasion and horizontal tumor spread, should always be in-
remain largely subjective. The most objective criteria for cluded, as well as the presence of lymphovascular invasion
diagnosing noninvasive glandular lesions of the cervix (LVI) and margin status. LEEP and cone specimens should
were presented in a report by Ioffe et al36 in which a nu- be inked with differential coloring of the endocervical
merical scoring system was proposed. In this scoring margin, if possible.
scheme, separate scores are assigned for the presence and Lymphovascular invasion has been shown to be an in-
degree of (1) nuclear atypia, (2) stratification, and (3) mi- dependent risk factor in relation to disease-free survival
toses/apoptosis (counted in the 2 most active glands and in a Gynecologic Oncology Group (GOG) prospective
the average number used). These 3 scores are then added study of 645 patients (disease-free survival, 89% vs 77%
to result in the total score (03, benign; 45, endocervical without and with LVI, respectively).38 However, reproduc-
732 Arch Pathol Lab MedVol 133, May 2009 Current Concepts in Cervical PathologyPark & Soslow
Figure 6. Stratified mucin-producing intraepithelial lesion (hematoxylin-eosin, original magnification 200).
Figure 7. Superficial invasive squamous carcinoma arising from endocervical gland involved by high-grade squamous intraepithelial lesion
(cervical intraepithelial neoplasia 3) (hematoxylin-eosin, original magnification 100).
Figure 8. A, High-grade squamous intraepithelial lesion (cervical intraepithelial neoplasia 2 [CIN 2]) (hematoxylin-eosin, original magnification
40). B, p16 immunohistochemical stain of CIN 2 (original magnification 40).

ible and accurate diagnosis of LVI can be problematic. The recurrences than controls. Surgical pathology reports of
most difficult issue concerns artifactual retraction or radical hysterectomy specimens should therefore include
shrinkage of stroma around invasive nests that results in the same information as reported for LEEP and cone bi-
spaces that resemble vessels. Immunostains for vascular opsies in addition to the percentage invasion into the cer-
markers, such as CD31, CD34, and lymphatic marker D2- vical wall, the gross size of the tumor, and the status of
40 (podoplanin) can help if the focus is present in sub- the parametrium.
sequent sections.39 Despite problems with recognizing The distinction between squamous and adenocarcinoma
LVI, most studies have shown that there is a relationship might have clinical relevance, as it has been postulated
between the depth of invasion and the presence of LVI and that adenocarcinomas and adenosquamous carcinomas
that LVI is a predictor of residual invasive disease in the have a worse prognosis than pure squamous carcinomas,
hysterectomy specimen.40 For some gynecologists, the with higher rates of lymph node metastases. However, a
presence of LVI in the context of microinvasive carcinoma GOG study looking at 626 patients with locally advanced
(FIGO stage IA1) triggers radical resection instead of con- disease treated with radiation showed no difference based
servative management. on histologic cell type.42 The impact of tumor grade on
In a prospective study from the GOG by Sedlis et al,41 prognosis has also been debated. Another GOG study by
patients with stage IB cervical carcinomas were random- Zaino et al40 showed that in squamous carcinomas grade
ized to pelvic radiotherapy following radical hysterectomy has little prognostic value. On the other hand, tumor dif-
and lymphadenectomy if their tumors fulfilled 2 of the ferentiation may have a significant role in adenocarcino-
following 3 criteria: large clinical tumor size, deep (greater ma.43 There are many different types and variants of pri-
than one-third) stromal invasion, and LVI. They found that mary cervical carcinomas, but this review is limited to
patients receiving adjuvant radiotherapy suffered fewer commonly encountered squamous cell and adenocarcino-
Arch Pathol Lab MedVol 133, May 2009 Current Concepts in Cervical PathologyPark & Soslow 733
International Federation of Gynecologists and invasion originated from the basal cells of the overlying
Obstetricians Staging SIL. Horizontal extent of the invasive carcinoma only
needs to be measured in cases in which the tumor depth
Stage Iconfined to the cervix
is 5 mm or less because once the tumor invades greater
IAdiagnosed only by microscopy; no visible lesions
IA1stromal invasion less than 3 mm in depth and 7 mm than 5 mm, it is stage IB regardless of the extent of hori-
or less in horizontal spread zontal spread. FIGO includes horizontal measurement in
IA2stromal invasion between 3 and 5 mm with horizon- staging but does not delineate how to measure it. When
tal spread of 7 mm or less there is only unifocal invasion, measuring horizontal ex-
IBvisible lesion or a microscopic lesion with more than 5 tent is not difficult. However, invasive carcinoma can often
mm of depth or horizontal spread of more than 7 mm be multifocal. There is no absolute method for how to best
IB1visible lesion 4 cm or less in greatest dimension
IB2visible lesion more than 4 cm measure the horizontal extent of invasion in these circum-
stances, but because tumor volume is thought to be an
Stage IIinvades beyond cervix
important factor in prognosis45,46 with higher rates of
IIAwithout parametrial invasion, but involves upper two- lymph node metastases and recurrence in women with
thirds of vagina
IIBwith parametrial invasion greater tumor volume, it may be best to measure in a way
that most accurately approximates volume. In an article
Stage IIIextends to pelvic side wall or lower one-third of the
by Reich and Pickel,47 they describe 3 patterns of micro-
vagina, causes hydronephrosis or nonfunctioning kid-
ney invasion and how each of those should be measured:
IIIAinvolves lower third of vagina Type Imultiple discrete foci of invasion are contiguous
IIIBextends to pelvic wall and/or causes hydronephrosis or with surface epithelium and the origin of invasion can
nonfunctioning kidney
be detected. In this case, measure each invasive focus
Stage IVextends beyond true pelvis or clinically involves the separately and then add together for a total horizontal
mucosa of bladder or rectum length. The HSIL in between the invasive foci is not
IVAinvades mucosa of bladder or rectum and/or extends measured.
beyond true pelvis
Type IIthe origin of invasion cannot be seen in all of the
IVBdistant metastasis
early invasive foci but the greatest lateral extent can be
seen in a single section. The breadth of the lesion is
considered to lie between the 2 most lateral foci count-
mas. A partial list of other carcinomas, some of which
ing everything, including normal and HSIL, in between.
have important clinical differences with the tumors dis-
Type IIIlike type II, except that the lesion is present in
cussed herein, include verrucous carcinoma, lymphoepi-
multiple contiguous sequential sections; the greatest
thelial-like carcinoma, intestinal adenocarcinoma, gastric-
horizontal length cannot be measured in a single slide.
like adenocarcinoma, endometrioid adenocarcinoma, clear
In this case, the width must be calculated by measuring
cell and serous adenocarcinoma, minimal deviation ade-
the distance between the sections; this is usually as-
nocarcinoma, mesonephric adenocarcinoma, glassy cell
sumed to measure 2 to 3 mm per section.
carcinoma, mucoepidermoid carcinoma, adenoid cystic
carcinoma, adenoid basal carcinoma, large cell and small Therefore, the growth pattern of the invasive foci is im-
cell neuroendocrine carcinoma, and mixed epithelial and portant and the entire cervix must be embedded and eval-
mesenchymal tumors. uated when the lesion is microscopic. Examining multiple
Correctly measuring the extent of invasion is crucial for levels may also be useful.
staging cervical cancer, especially in early-stage (micro- The nomenclature used to describe bona fide examples
scopically detected) disease because stage dictates treat- of superficially invasive carcinomas is, unfortunately, not
ment and prognosis. However, superficial invasion is fre- standardized. Proposals for definitions of microinvasion
quently overcalled. Of 265 purported cases of microinva- or microinvasive carcinoma include examples showing in-
sion submitted to a group of reference pathologists of the vasion less than 3 mm deep or those with invasion less
GOG, about 50% were rejected.44 Pitfalls of misdiagnosing than 5 mm deep, with or without LVI. The horizontal ex-
SIL with microinvasion include SIL involving endocervical tent of the invasive front is also inconsistently mentioned
glands and areas of prior biopsy site with entrapped SIL. as a criterion. The term has also been used, historically,
Areas of prior biopsy will have associated acute and more frequently in reference to squamous cell carcinoma
chronic inflammatory infiltrate and may have entrapped than adenocarcinoma, as discussed subsequently. In prac-
neoplastic epithelium buried within the stroma as a result tice, therefore, we do not advocate the use of the term
of disrupted SIL from a punch biopsy. Definitive examples microinvasive carcinoma. Instead, we report the depth of
of invasive squamous carcinoma should demonstrate stro- invasion, measure the horizontal extent of the invasive
mal desmoplasia, irregular or scalloped margins, and/or component, and mention whether LVI is present. If it is a
abundant, paradoxically mature eosinophilic cytoplasm. pathologists practice to use the term microinvasive carci-
Once the presence of invasion is confirmed, measuring noma it is recommended that the term only be used when
the depth of invasive squamous carcinoma is the next im- definitive measurements can be made (and surgical mar-
portant step. For consistency, the depth of stromal invasion gins are negative for invasive carcinoma) and the patients
should be measured following these guidelines: the depth gynecologist is informed about the criteria the pathologist
of neoplastic projections should be measured from the ini- uses for such tumors.
tial site of invasion, either from the basal lamina of the Although the FIGO staging system for cervical cancer ap-
surface epithelium or from the endocervical glands re- plies to both squamous and adenocarcinoma, there is con-
placed by intraepithelial lesions (Figure 7); in cases in siderable controversy as to whether microinvasive adenocar-
which a direct histologic continuum between invasive foci cinoma exists as a histologically recognizable entity. One rea-
and SIL cannot be demonstrated, it is assumed that the son is that the irregular distribution and architecture of the
734 Arch Pathol Lab MedVol 133, May 2009 Current Concepts in Cervical PathologyPark & Soslow
normal endocervical crypts in the cervical stroma make it
difficult to differentiate between early stromal invasion and
AIS. There are 2 unequivocal features of invasion in adeno-
carcinoma: individual cells/fragmented glands/incomplete
glands lined by malignant cells at a stromal interface and
malignant glands with desmoplastic stromal response. Other
less definitive soft features that are suggestive of early in-
vasion include complex architectural growth with papillae,
cribriforming and irregular glands growing in a confluent
or labyrinthine pattern, malignant glands adjacent to medi-
um- or thick-walled vessels, and the presence of malignant
glands below the level of normal glands. Normal cervical
glands extend to the inner one-third of the cervical wall and
can measure up to 1 cm. Nabothian cysts, tunnel clusters,
endocervical hyperplasia, deep endocervical glands, and me-
sonephric duct remnants can all extend to the outer third of
the cervical wall.
Admittedly, in about 20% of the cases, it is impossible
to distinguish between AIS and early invasive adenocar-
cinoma.48 Because there is no overlying basement mem-
brane from which to measure the starting point of inva-
sion, as there is in squamous carcinoma, measuring the
exact depth of invasion of cervical adenocarcinoma is
problematic. Most sources, including the World Health Or-
ganization,49 recommend measuring the thickness of the
tumor from the mucosal surface, rather than depth of in-
vasion as defined previously.
Some cervical adenocarcinomas, particularly villoglan-
dular adenocarcinomas, and many papillary squamous
cell carcinomas can grow as exophytic, polypoid masses
that are obvious at physical examination. Substantial ar-
chitectural complexity and the presence of a gross, mass-
forming lesion qualify for invasive carcinoma (as opposed
to HSIL or AIS), but measurement of the depth of invasion
can be problematic because these lesions frequently grow
up into the endocervical canal, instead of down into the
cervical stroma. The depth of stromal invasion here should
not reflect thickness, rather it should be measured from a
plane that underlies the exophytic tumor. Regardless of
this measurement, these tumors are regarded as at least
FIGO stage IB because they are easily detected on physical
examination.
ANCILLARY STUDIES
Various immunohistochemical and molecular assays are
available as ancillary studies in the workup of difficult
squamous and glandular lesions.
p16INK4a is a tumor suppressor gene that encodes a pro-
tein involved in cell cycle regulation. It acts in a negative
feedback loop with another tumor suppressor, retinoblasto-
ma protein, so that cell proliferation is held in check. When
high-risk HPV DNA integrates into the host cell genome, the
viral oncoprotein E7 binds to retinoblastoma protein render-
ing it inactive. The feedback loop is lost and p16 is overex- Figure 9. Tubal metaplasia showing focal patchy staining with p16.
pressed as a result. This manifests as diffuse, strong, cyto- This is not the diffuse staining pattern seen in true adenocarcinoma in
plasmic and/or nuclear staining in squamous and glandular situ (original magnification 200).
lesions associated with high-risk HPV infection (Figure 8, A Figure 10. A, Reactive squamous epithelium with inflammatory cells
and B).5053 Interpreting p16 immunostaining, however, can and pseudokoilocytes (hematoxylin-eosin, original magnification 40).
be complicated, as both the context in which p16 is applied B, Mib-1 stain in reactive epithelium. Notice staining of lymphocytes
and parabasal cells that extend to the surface of the epithelium. This
and also the distribution and intensity of its staining pattern should not be interpreted as evidence of dysplasia (original magnifi-
must be considered before using the stain to confirm the cation 40).
presence of an HPV-associated neoplasm. A variety of le-
sions unassociated with HPV can show focal or patchy p16
staining, including tubal metaplasia, squamous metaplasia,
and endometrioid adenocarcinoma (Figure 9). Serous carci-
Arch Pathol Lab MedVol 133, May 2009 Current Concepts in Cervical PathologyPark & Soslow 735
nomas can show diffuse, strong staining as well.51,54,55 Atyp- In situ hybridization (ISH) is a direct signal detection as-
ical immature metaplasia lesions that are p16 positive have say that allows visualization of HPV DNA within infected
been shown to have a higher incidence of subsequent HSILs cells. Unlike other molecular detection methods (eg, poly-
and are likely HSIL lesions that have an immature meta- merase chain reaction), the ISH assay can be automated and
plastic appearance.9 Eosinophilic dysplasias are also usually does not require the skills of highly trained laboratory per-
p16 positive, supporting the contention that these are vari- sonnel or the stringent laboratory setup required to prevent
ants of dysplastic lesions arising from cervical reserve cells.11 contamination. The interpretation of ISH is similar to that of
Atypical, proliferative glandular lesions that are diffusely immunohistochemistry staining in tissue sections (look for
and strongly p16 positive are also likely HPV-associated ad- positive nuclear staining). The ISH signal patterns of HPV
enocarcinomas as opposed to one of the reactive lesions dis- DNA have been associated with the physical status of HPV
cussed earlier. The diffuse staining pattern of HPV-associated in infected cells, that is, episomal or integrated forms.71,72 Epi-
glandular lesions can be especially helpful in curettage spec- somal forms result in blocklike nuclear labeling, whereas in-
imens where the differential is often with endometrial en- tegrated forms result in punctate, nuclear signals. Punctate
dometrioid adenocarcinomas. A useful panel in this situation signals are most frequently found in HSILs and invasive car-
includes p16, estrogen and progesterone receptors, monoclo- cinomas but not LSILs. Because the integration of oncogenic
nal carcinoembryonic antigen (CEAM), and vimentin.56,57 Al- HPV into the human genome is a critical step for cervical
though overlapping patterns are seen, the general trend is cancer carcinogenesis, the signal pattern could be a useful
for cervical adenocarcinomas to be positive for CEA(m) and marker for predicting precancerous lesion progression. In a
p16 and negative for estrogen and progesterone receptors recent study by Guo et al,73 the authors showed that the
and vimentin, whereas the opposite is likelier in endometrial Inform HPV III (Ventana Medical Systems, Tuscon, Arizona)
endometrioid adenocarcinomas. in situ hybridization assay is comparable to polymerase
Mib-1 is a monoclonal antibody that reacts with Ki-67, a chain reaction and may be a useful adjunct in detecting
nuclear cell proliferationassociated antigen that is expressed HPV-positive lesions and carcinomas. However, ISH has also
in all active parts of the cell cycle (G1, S, G2/M). Positive been shown to have low sensitivity so its utility has been
staining in parabasal cells is found under normal conditions, questioned.74
so interpreting the results of this stain rests on examination
of the middle and upper thirds of the squamous epithelium. CONCLUSIONS
High-grade squamous intraepithelial lesion (CIN 2 and CIN There are many common difficulties that arise in the
3) usually shows diffuse nuclear positivity scattered daily assessment of cervical specimens. Practicing pathol-
throughout all layers of the epithelium,58 whereas the distri- ogists should not overcall reactive squamous atypia as
bution of Mib-1 staining in CIN 1 can be less diffuse with squamous dysplasia and they should be aware of variants
only small clusters of squamous cells staining in the upper of squamous lesions and AIS that may appear metaplastic
two-thirds of the epithelium. One should be careful not to or reactive (AIM, ED, SMILE). Stage is the most important
overinterpret positive staining in the presence of inflamma- prognostic factor for cervical cancer, but other pathologic
tion because intraepithelial lymphocytes will be positive for indices are also important. One should include all prog-
Mib-1 and inflammation can also cause reactive proliferation. nostically relevant information in pathology reports for
It should also be noted that tangential sectioning sometimes best patient care, including the extent of invasion, margin
results in the impression that Mib-1positive parabasal cells status, and the presence of LVI. The depth of invasion
are superficially placed, leading to confusion with a SIL59,60 should be measured from the nearest overlying basement
(Figure 10, A and B). Adenocarcinoma and AIS should show membrane in squamous carcinoma, but the thickness of
strong diffuse staining with Mib-1, whereas benign mimics tumor (rather than depth of invasion) should be reported
such as tubal metaplasia, radiation change, and reactive atyp- in adenocarcinoma. Radical hysterectomy reports should
ia should show only focal or patchy staining at most.61,62 also contain information about the greatest tumor dimen-
SMILE should also show diffuse positivity of the entire sion and the status of the parametria. It may be difficult
thickness of the lesion, including the mucin-producing cells.27 to distinguish AIS from early invasive adenocarcinoma,
Atypical immature metaplasia, when diffusely positive for but using the criteria discussed previously should be help-
Mib-1, is likely an immature variant of HSIL.9 ful. Finally, ancillary studies (p16, MIB-1, ProEx C, HPV
ProEx C is a recently developed immunohistochemical in situ) can be useful in difficult cases, but they must be
assay that targets the expression of topoisomerase II- and interpreted in the proper context.
minichromosome maintenance protein-2, two genes that References
have been shown to be overexpressed in cervical can- 1. Ferlay J, Bray F, Pisani P, Parkin DM. GLOBOCAN 2002: Cancer Incidence,
cers.6370 The assay is a nuclear stain that is positive in Mortality and Prevalence Worldwide. IARC CancerBase No. 5, version 2.0. Lyon,
cervical dysplasia and has been validated in cytologic France: IARC; 2004.
2. US Cancer Statistics Working Group. United States Cancer Statistics: 2004
specimens for the detection of HSIL.66,67 Studies have also Incidence and Mortality. Atlanta, GA: Department of Health and Human Services,
shown that ProEx C is comparable to p16 and Mib-1 in Centers for Disease Control and Prevention, and National Cancer Institute; 2007.
the detection of high-grade lesions in formalin-fixed tissue 3. Munoz N, Bosch FX, de Sanjose S, et al. Epidemiologic classification of
human papillomavirus types associated with cervical cancer. N Engl J Med.
sections and in distinguishing them from benign mim- 20036;348(6):518527.
ics.6870 The staining pattern of ProEx C in histologic sec- 4. Robertson JH, Woodend B, Elliott H. Cytological changes preceding cervical
tions is similar to that of Mib-1 in that only nuclear pos- cancer. J Clin Pathol. 1994;47(3):278279.
itivity above the basal one-third layer is considered posi- 5. Schiffman M, Solomon D. Findings to date from the ASCUS-LSIL Triage
Study (ALTS). Arch Pathol Lab Med. 2003;127(8):946949.
tive. In a recent study by Pinto et al70 it was shown that 6. Ismail SM, Colclough AB, Dinnen JS, et al. Observer variation in histopath-
the combination of p16 and ProEx C predicted more non- ological diagnosis and grading of cervical intraepithelial neoplasia. BMJ. 1989;
SILs than p16 combined with Mib-1. This suggests that 298(6675):707710.
7. Crum CP, Egawa K, Fu YS, et al. Atypical immature metaplasia (AIM): a
ProEx C may be more efficient than Mib-1 for distinguish- subset of human papillomavirus virus infection of the cervix. Cancer. 1983;
ing reactive epithelial changes from squamous lesions. 51(12):22142219.

736 Arch Pathol Lab MedVol 133, May 2009 Current Concepts in Cervical PathologyPark & Soslow
8. Duggan MA, Akbari M, Magliocco AM. Atypical immature cervical meta- 40. Zaino RJ, Ward S, Delgado G, et al. Histopathologic predictors of the
plasia: immunoprofiling and longitudinal outcome. Hum Pathol. 2006;37(11): behavior of surgically treated stage IB squamous cell carcinoma of the cervix: a
14731481. Gynecologic Oncology Group study. Cancer. 1992;69(7):17501758.
9. Iaconis L, Hyjek E, Ellenson LH, Pirog EC. p16 and Ki-67 immunostaining 41. Sedlis A, Bundy BN, Rotman MZ, Lentz SS, Muderspach LI, Zaino RJ. A
in atypical immature squamous metaplasia of the uterine cervix: correlation with randomized trial of pelvic radiation therapy versus no further therapy in selected
human papillomavirus detection [published correction appears in Arch Pathol Lab patients with stage IB carcinoma of the cervix after radical hysterectomy and
Med. 2008;132(1):13]. Arch Pathol Lab Med. 2007;131(9):13431349. pelvic lymphadenectomy: a Gynecologic Oncology Group Study. Gynecol On-
10. Geng L, Connolly DC, Isacson C, Ronnett BM, Cho KR. Atypical immature col. 1999;73(2):177183.
metaplasia (AIM) of the cervix: is it related to high-grade squamous intraepithelial 42. Stehman FB, Bundy BN, DiSaia PJ, Keys HM, Larson JE, Fowler WC. Car-
lesion (HSIL)? Hum Pathol. 1999;30(3):345351. cinoma of the cervix treated with radiation therapy: a multi-variate analysis of
11. Ma L, Fisk JM, Zhang RR, Ulukus EC, Crum CP, Zheng W. Eosinophilic prognostic variables in the Gynecologic Oncology Group. Cancer. 1991;67(11):
dysplasia of the cervix: a newly recognized variant of cervical squamous intra- 27762785.
epithelial neoplasia. Am J Surg Pathol. 2004;28(11):14741484. 43. Shingleton HM, Orr JW, eds. Cancer of the Cervix. Philadelphia, PA: JB
12. Jaworski RC, Pacey NF, Greenberg ML, Osborn RA. The histologic diag- Lippincott; 1995.
nosis of adenocarcinoma in situ and related lesions of the cervix uteri: adeno- 44. Sedlis A, Sall S, Tsukada Y, et al. Microinvasive carcinoma of the uterine
carcinoma in situ. Cancer. 1988;61:11711181. cervix: a clinical-pathologic study. Am J Obstet Gynecol. 1979;133(1):6474.
13. Ostor AG, Duncan A, Quinn M, Rome R. Adenocarcinoma in situ of the 45. Burghardt E, Pickel H, Haas J, Lahousen M. Prognostic factors and oper-
uterine cervix: an experience with 100 cases. Gynecol Oncol. 2000;79:207210. ative treatment of stages IB to IIB cervical cancer. Am J Obstet Gynecol. 1987;
14. Denehy TR, Gregori CA, Breen JL. Endocervical curettage, cone margins, 156(4):988996.
and residual adenocarcinoma in situ of the cervix. Obstet Gynecol. 1997;90: 46. Kaspar HG, Dinh TV, Doherty MG, Hannigan EV, Kumar D. Clinical im-
16. plications of tumor volume measurement in stage I adenocarcinoma of the cervix.
15. Poynor EA, Barakat RR, Hoskins WJ. Management and follow-up of pa- Obstet Gynecol. 1993;81(2):296300.
tients with adenocarcinoma in situ of the uterine cervix. Gynecol Oncol. 1995; 47. Reich O, Pickel H. Multifocal stromal invasion in microinvasive squamous
57:158164. cell carcinoma of the cervix: how to measure and stage these lesions. Int J Gy-
16. Andersen ES, Arffmann E. Adenocarcinoma in situ of the uterine cervix: a necol Pathol. 2002;21(4):416417.
clinicopathologic study of 36 cases. Gynecol Oncol. 1989;35:17. 48. Ostor AG. Early invasive adenocarcinoma of the uterine cervix. Int J Gy-
17. Bertrand M, Lickrish GM, Colgan TJ. The anatomic distribution of cervical necol Pathol. 2000;19(1):2938.
adenocarcinoma in situ: implications for treatment. Am J Obstet Gynecol. 1987; 49. Tavassoli FA, Devilee P, eds. Tumours of the Breast and Female Genital
157:2125. Organs. Lyon, France: IARC Press; 2003. World Health Organization Classifica-
18. Muntz HG, Bell DA, Lage JM, Goff BA, Feldman S, Rice LW. Adenocar- tion of Tumours.
cinoma in situ of the uterine cervix. Obstet Gynecol. 1992;80:935939. 50. Ishikawa M, Fujii T, Masumoto N, et al. Correlation of p16INK4A over-
19. Ostor AG, Pagano R, Davoren RA, Fortune DW, Chanen W, Rome R. Ad- expression with human papillomavirus infection in cervical adenocarcinomas. Int
enocarcinoma in situ of the cervix. Int J Gynecol Pathol. 1984;3:179190. J Gynecol Pathol. 2003;22(4):378385.
20. Qizilbash AH. In situ and microinvasive adenocarcinoma of the uterine 51. Riethdorf L, Riethdorf S, Lee KR, Cviko A, Loning T, Crum CP. Human
cervix: a clinical, cytologic and histologic study of 14 cases. Am J Clin Pathol. papillomaviruses, expression of p16, and early endocervical glandular neoplasia.
1975;64:155170. Hum Pathol. 2002;33(9):899904.
21. Shin CH, Schorge JO, Lee KR, Sheets EE. Conservative management of
52. Ansari-Lari MA, Staebler A, Zaino RJ, Shah KV, Ronnett BM. Distinction
adenocarcinoma in situ of the cervix. Gynecol Oncol. 2000;79:610.
of endocervical and endometrial adenocarcinomas: immunohistochemical p16
22. Tobon H, Dave H. Adenocarcinoma in situ of the cervix: clinicopathologic
expression correlated with human papillomavirus (HPV) DNA detection. Am J
observations of 11 cases. Int J Gynecol Pathol. 1988;7:139151.
Surg Pathol. 2004;28(2):160167.
23. Wolf JK, Levenback C, Malpica A, Morris M, Burke T, Mitchell MF. Ade-
53. Keating JT, Cviko A, Riethdorf S, et al. Ki-67, cyclin E, and p16INK4 are
nocarcinoma in situ of the cervix: significance of cone biopsy margins. Obstet
complimentary surrogate biomarkers for human papillomavirus-related cervical
Gynecol. 1996;88:8286.
neoplasia. Am J Surg Pathol. 2001;25:884891.
24. Zaino RJ. Symposium part I: adenocarcinoma in situ, glandular dysplasia,
54. Horree N, Heintz AP, Sie-Go DM, van Diest PJ. p16 is consistently ex-
and early invasive adenocarcinoma of the uterine cervix. Int J Gynecol Pathol.
pressed in endometrial tubal metaplasia. Cell Oncol. 2007;29(1):3745.
2002;21(4):314326.
55. ONeill CJ, McCluggage WG. p16 expression in the female genital tract
25. Goldstein NS, Mani A. The status and distance of cone biopsy margins as
a predictor of excision adequacy for endocervical adenocarcinoma in situ. Am J and its value in diagnosis. Adv Anat Pathol. 2006;13(1):815.
Clin Pathol. 1998;109:727732. 56. Dabbs DJ, Sturtz K, Zaino RJ. The immunohistochemical discrimination of
26. Ostor AG, Pagano R, Davoren RAM, Fortune DW, Chanen W, Rome R. endometrioid adenocarcinomas. Hum Pathol. 1996;27(2):172177.
Adenocarcinoma in situ of the cervix. Int J Gynecol Pathol. 1984;3:179190. 57. Alkushi A, Irving J, Hsu F, et al. Immunoprofile of cervical and endometrial
27. Park JJ, Sun D, Quade BJ, et al. Stratified mucin-producing intraepithelial adenocarcinomas using a tissue microarray. Virchows Arch. 2003;442(3):271
lesions of the cervix: adenosquamous or columnar cell neoplasia? Am J Surg 277.
Pathol. 2000;24(10):14141419. 58. Bulten J, van der Laak JA, Gemmink JH, Pahlplatz MM, de Wilde PC,
28. Kurman RJ, ed. Blausteins Pathology of the Female Genital Tract. 5th ed. Hanselaar AG. MIB1, a promising marker for the classification of cervical intra-
New York, NY: Springer-Verlag; 2002:235241. epithelial neoplasia. J Pathol. 1996;178(3):268273.
29. Young RH, Clement PB. Pseudoneoplastic glandular lesions of the uterine 59. Pirog EC, Baergen RN, Soslow RA, et al. Diagnostic accuracy of cervical
cervix. Semin Diagn Pathol. 1991;8(4):234249. low-grade squamous intraepithelial lesions is improved with MIB-1 immunostain-
30. Lesack D, Wahab I, Gilks CB. Radiation-induced atypia of endocervical ing. Am J Surg Pathol. 2002;26(1):7075.
epithelium: a histological, immunohistochemical and cytometric study. Int J Gy- 60. Young RH, Clement PB. Endocervical adenocarcinoma and its variants:
necol Pathol. 1996;15(3):242247. their morphology and differential diagnosis. Histopathology. 2002;41:185207.
31. Oliva E, Clement PB, Young RH. Tubal and tubo-endometrioid metaplasia 61. McCluggage WG, Maxwell P, McBride HA, Hamilton PW, Bharucha H.
of the uterine cervix: unemphasized features that may cause problems in differ- Monoclonal antibodies Ki-67 and MIB1 in the distinction of tuboendometrial
ential diagnosis: a report of 25 cases. Am J Clin Pathol. 1995;103(5):618623. metaplasia from endocervical adenocarcinoma and adenocarcinoma in situ in
32. Jaworski R. Endocervical glandular dysplasia, adenocarcinoma in situ, and formalin-fixed material. Int J Gynecol Pathol. 1995;14(3):209216.
early invasive (microinvasive) adenocarcinoma of the uterine cervix. Semin Diagn 62. Pirog EC, Isacson C, Szabolcs MJ, Kleter B, Quint W, Richart RM. Prolif-
Pathol. 1990;7:190205. erative activity of benign and neoplastic endocervical epithelium and correlation
33. Brown LJ, Wells M. Cervical glandular atypia associated with squamous with HPV DNA detection. Int J Gynecol Pathol. 2002;21(1):2226.
intraepithelial neoplasia: a premalignant lesion? J Clin Pathol. 1986;39:2228. 63. Chen Y, Miller C, Mosher R, et al. Identification of cervical cancer markers
34. Casper G, Ostor A, Quinn M. A clinicopathologic study of glandular dys- by cDNA and tissue microarrays. Cancer Res. 2003;63:19271935.
plasia of the cervix. Gynecol Oncol. 1997;64:166170. 64. Malinowski DP. Multiple biomarkers in molecular oncology: molecular
35. Gompel C, Silverberg S. Pathology in Gynecology and Obstetrics. Phila- diagnostics applications in cervical cancer detection. Expert Rev Mol Diagn.
delphia, PA: JB Lippincott; 1994. 2007;7:117131.
36. Ioffe OB, Sagae S, Moritani S, Dahmoush L, Chen TT, Silverberg SG. Pro- 65. Santin AD, Zhan F, Bignotti E, et al. Gene expression profiles of primary
posal of a new scoring scheme for the diagnosis of noninvasive endocervical HPV 16- and HPV 18-infected early stage cervical cancers and normal cervical
glandular lesions. Am J Surg Pathol. 2003;27(4):452460. epithelium: identification of novel candidate biomarkers for cervical cancer di-
37. Rock JA, Jones HW, eds. Te Lindes Operative Gynecology. In press. agnosis and therapy. Virology. 2005;331:269291.
38. Delgado G, Bundy B, Zaino R, Sevin BU, Creasman WT, Major F. Pro- 66. Kelly D, Kincaid E, Fansler Z, Rosenthal DL, Clark DP. Detection of cer-
spective surgical-pathological study of disease-free interval in patients with stage vical high-grade squamous intraepithelial lesions from cytologic samples using a
IB squamous cell carcinoma of the cervix: a Gynecologic Oncology Group study. novel immunocytochemical assay (ProEx C). Cancer. 2006;108:494500.
Gynecol Oncol. 1990;38(3):352357. 67. Shroyer KR, Homer P, Heinz D, Singh M. Validation of a novel immuno-
39. Urabe A, Matsumoto T, Kimura M, Sonoue H, Kinoshita K. Grading system cytochemical assay for topoisomerase II-alpha and minichromosome mainte-
of lymphatic invasion according to D2-40 immunostaining is useful for the pre- nance protein 2 expression in cervical cytology. Cancer. 2006;108:324330.
diction of nodal metastasis in squamous cell carcinoma of the uterine cervix. 68. Shi J, Liu H, Wilkerson M, et al. Evaluation of p16NK4a, minichromosome
Histopathology. 2006;49(5):493497. maintenance protein 2, DNA topoisomerase II alpha, ProE(X) C, and p16INK4a/

Arch Pathol Lab MedVol 133, May 2009 Current Concepts in Cervical PathologyPark & Soslow 737
ProE(X) C in cervical squamous intraepithelial lesions. Hum Pathol. 2007;38: 72. Evans MF, Mount SL, Beatty BG, Cooper K. Biotinyl-tyramide-based in situ
13351344. hybridization signal patterns distinguish human papillomavirus type and grade of
69. Badr RE, Walts AE, Chung F, Bose S. BD ProEX C: a sensitive and specific cervical intraepithelial neoplasia. Mod Pathol. 2002;15:13391347.
marker of HPV-associated squamous lesions of the cervix. Am J Surg Pathol. 2008; 73. Guo M, Gong Y, Deavers M, et al. Evaluation of a commercialized in situ
32:899906. hybridization assay for detecting human papillomavirus DNA in tissue specimens
70. Pinto AP, Schlecht NF, Woo TYC, Crum CP, Cibas ES. Biomarker (ProEx C, from patients with cervical intraepithelial neoplasia and cervical carcinoma. J
p16INK4a, and MiB-1) distinction of high-grade squamous intraepithelial lesion Clin Microbiol. 2008;46(1):274280.
from its mimics. Mod Pathol. 2008;21:10671074. 74. Dabic MM, Hlupic L, Babic D, Jukic S, Seiwerth S. Comparison of poly-
71. Cooper K, Herrington CS, Stickland JE, Evans MF, McGee JO. Episomal merase chain reaction and catalyzed signal amplification in situ hybridization
and integrated human papillomavirus in cervical neoplasia shown by non-isoto- methods for human papillomavirus detection in paraffin-embedded cervical pre-
pic in situ hybridisation. J Clin Pathol. 1991;44:990996. neoplastic and neoplastic lesions. Arch Med Res. 2004;35:511516.

738 Arch Pathol Lab MedVol 133, May 2009 Current Concepts in Cervical PathologyPark & Soslow

Anda mungkin juga menyukai