SlideShare a Scribd company logo
1 of 106
Download to read offline
4
InternationalJournalofGynecology&ObstetricsVol.131(2015)S75–S172
131
S2
ELSEVIER
Official publication of FIGO
The International Federation
of Gynecology and Obstetrics
FIGO Cancer Report 2015
Guest Editors: Lynette Denny, Michael Quinn
OBSTETRICS
GYNECOLOGY
International Journal of
Volume 131 Supplement 2 October 2015 ISSN 0020-7292
International Journal of
GYNECOLOGY
& OBSTETRICS
Volume 131, Supplement 2 (2015)
Amsterdam • Boston • London • New York • Oxford • Paris • Philadelphia • San Diego • St. Louis
© 2015 Published by Elsevier Ireland Ltd. on behalf of International Federation of Gynecology and
Obstetrics. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/
licenses/by-nc-nd/4.0/)
This journal and the individual contributions contained in it are protected under copyright by International
Federation of Gynecology and Obstetrics, and the following terms and conditions apply to their use:
Photocopying
Single photocopies of single articles may be made for personal use as allowed by national copyright laws.
Permission of the Publisher and payment of a fee is required for all other photocopying, including multiple or
systematic copying, copy-ing for advertising or promotional purposes, resale, and all forms of document
delivery. Special rates are available for educational institutions that wish to make photocopies for non-profit
educational classroom use.
For information on how to seek permission visit www.elsevier.com/permissions or call: (+44) 1865 843830
(UK)/(+1) 215 239 3804 (USA).
Derivative Works
Subscribers may reproduce tables of contents or prepare lists of articles including abstracts for internal
circulation within their institutions. Permission of the Publisher is required for resale or distribution outside
the institution. Permission of the Publisher is required for all other derivative works, including compilations
and translations (please consult www.elsevier.com/permissions).
Electronic Storage or Usage
Permission of the Publisher is required to store or use electronically any material contained in this journal,
including any article or part of an article (please consult www.elsevier.com/permissions). Except as outlined
above, no part of this publication may be reproduced, stored in a retrieval system or transmitted in any form
or by any means, electronic, mechanical, photocopying, recording or otherwise, without prior written permission
of the Publisher.
Notice
No responsibility is assumed by the Publisher for any injury and/or damage to persons or property as a matter
of products liability, negligence or otherwise, or from any use or operation of any methods, products,
instructions or ideas contained in the material herein. Because of rapid advances in the medical sciences,
in particular, independent verification of diagnoses and drug dosages should be made.
Although all advertising material is expected to conform to ethical (medical) standards, inclusion in this
publication does not constitute a guarantee or endorsement of the quality or value of such product or
of the claims made of it by its manufacturer.
Language (usage and editing services)
Please write your text in good English (American or British usage is accepted, but not a mixture of these).
Authors who feel their English language manuscript may require editing to eliminate possible grammatical or
spelling errors and to conform to correct scientific English may wish to use the English Language Editing service
available from Elsevier’s WebShop http://webshop.elsevier.com/languageediting/ or visit our customer support
site http://support.elsevier.com for more information.
Illustration services
Elsevier’s WebShop (http://webshop.elsevier.com/illustrationservices) offers Illustration Services to authors
preparing to submit a manuscript but concerned about the quality of the images accompanying their article.
Elsevier’s expert illustrators can produce scientific, technical and medical-style images, as well as a full range
of charts, tables and graphs. Image ‘polishing’ is also available, where our illustrators take your image(s) and
improve them to a professional standard. Please visit the website to find out more.
Printed by Henry Ling, Dorchester
The paper used in this publication meets the requirements of ANSI/NISO Z39.48-1992 (Permanence of Paper)
Supplement to
International Journal of Gynecology & Obstetrics
Volume 131, Supplement 2
FIGO Cancer Report 2015
Guest Editors: Lynette Denny, Michael Quinn
International Federation of Gynecology and
Obstetrics (FIGO)
Officers
President:
Vice-President:
President-Elect:
Past-President:
Honorary Treasurer:
Honorary Secretary:
S. Arulkumaran (UK)
E.C. Morales (Mexico)
C.N. Purandare (India)
G. Serour (Egypt)
W. Holzgreve (Germany)
G.C. Di Renzo (Italy)
FIGO Chief Executive
H. Rushwan (Sudan/UK)
Executive Board
Argentina N.C. Garello
Australia & New Zealand C. Tippett
Belgium F. Debiève
Bolivia C. Fuchtner
Brazil N.R. de Melo
Canada J. Blake
Chile H. Munoz
Colombia J.D.VillegasEcheverri
Denmark A.T. Pedersen
Egypt N.A. Darwish
Ethiopia Y.G. Ferede
Finland S. Grénman
France B. Carbonne
Germany W. Jonat
Japan T. Kimura
Lebanon F. El-Kak
Malaysia A.A. Yahya
Paraguay A. Acosta
South Africa B.D. Goolab
Spain J. Laílla Vicens
Taiwan T.-H. Su
Uruguay J.G. Allonso Tellechea
United Kingdom T. Falconer
United States of America J.N. Martin
International Editions and Collaborations
IJGO India
Editor-in-Chief: Dr Rohit V. Bhatt (rohit.v.bhatt@gmail.com)
Editorial Office: Jaypee Brothers Medical Publishers (P) Ltd
4838/24, Ansari Road, Daryaganj
New Delhi 110 002, India
E-mail: ijgoindia@jaypeebrothers.com
IJGO China
Editor-in-Chief: Dr Zhenyu Zhang
Department of Ob/Gyn
Chaoyang Hospital
No. 8 Baijiazhung Rd
Chaoyang District
Beijing, 100020 China
E-mail: zhenyuzhang2000@yahoo.com
For information about
FIGO:
The Secretariat of FIGO is at FIGO House, Suite 3,
Waterloo Court, 10 Theed Street, London, SE1 8ST UK.
Tel: +44 20 7928-1166
Fax: +44 20 7928-7099
E-mail: figo@figo.org
Website: www.figo.org
All enquiries concerning FIGO may be sent to
the Honorary Secretary at that address.
Editor:
Editor Emeritus:
Associate Editor:
Managing Editor:
Deputy Managing Editor:
Associate Editors
Ethical and Legal Issues
in Reproductive Health:
Enabling Technologies:
FIGO Staging of Gynecologic
Cancer:
Contemporary Issues in
Women’s Health:
Statistical Consultant:
Editorial Office:
R.M. Adanu (Ghana)
T.R.B. Johnson (USA)
J. Sciarra (USA)
W. Holzgreve (Germany)
P. Serafini (Brazil)
J. Fortney (USA)
C. Addington (UK)
A. Cantor (UK)
R. Cook (Canada)
B. Dickens (Canada)
M. Hammoud (USA)
L. Denny (South Africa)
V. Boama (Qatar)
V. Guinto (Philippines)
C. Sosa (Uruguay)
A. Vahratian (USA)
FIGO Secretariat, FIGO House
Suite 3 - Waterloo Court, 10 Theed Street,
London, SE1 8ST, UK
Tel: +44 20 7928 1166
Fax: +44 20 7928 7099
E-mail: ijgo@figo.org
International Journal of
GYNECOLOGY
& OBSTETRICS
International Journal of
GYNECOLOGY
& OBSTETRICS
EDITORIAL
L. Denny, M. Quinn
South Africa, Australia
FIGO Cancer Report 2015 S75
CANCER STAGING AND CLINICAL GUIDELINES
N.F. Hacker, P.J. Eifel,
J. van der Velden
Australia, USA, The Netherlands
Cancer of the vulva S76
N.F. Hacker, P.J. Eifel,
J. van der Velden
Australia, USA, The Netherlands
Cancer of the vagina S84
A. Bermudez, N. Bhatla, E. Leung
Argentina, India, Canada
Cancer of the cervix uteri S88
F. Amant, M.R. Mirza, M. Koskas,
C.L. Creutzberg
Belgium, Denmark, France,
The Netherlands
Cancer of the corpus uteri S96
J. Prat, 'N. Mbatani
Spain, South Africa
Uterine sarcomas S105
J.S. Berek, C. Crum, M. Friedlander
USA, Australia
Cancer of the ovary, fallopian tube, and peritoneum S111
H.Y.S. Ngan, M.J. Seckl,
R.S. Berkowitz, Y. Xiang,
F. Golfier, P.K. Sekharan,
J.R. Lurain
China, UK, USA, France, India
Update on the diagnosis and management of gestational trophoblastic
disease
S123
P.S. Binder, J. Prat, D.G. Mutch
USA, Spain
The future role of molecular staging in gynecologic cancer S127
CONTENTS
Volume 131, Supplement 2, October 2015
CANCER THERAPY
E.Å. Lundqvist, K. Fujiwara,
M. Seoud
Sweden, Japan, Lebanon
Principles of chemotherapy S146
M. Seoud, E.Å. Lundqvist,
K. Fujiwara
Lebanon, Sweden, Japan
Targeted therapy in gynecologic cancers: Ready for prime time? S150
S.K. Shrivastava, U. Mahantshetty,
K. Narayan
India, Australia
Principles of radiation therapy in low-resource and well-developed
settings, with particular reference to cervical cancer
S153
PATIENT CARE
S.V. Carr
Australia
Psychosexual health in gynecological cancer S159
K. Weare
Australia
Rehabilitation after gynecological cancer treatment S164
S.M. Kibel, J.M. Cain
South Africa, USA
Palliative care in gynecological cancer S167
PATHOLOGY
J. Prat
Spain
Pathology of cancers of the female genital tract S132
EDITORIAL
FIGO Cancer Report 2015
The FIGO Gynecologic Oncology Committee is pleased to present the
second edition of the FIGO Cancer Report. Historically the report was
published at each FIGO World Congress as the “Annual Report,” going
as far back as 1937, and was composed of survival data from various
institutions around the world. Since 2009 the process of collating
international incidence and survival data stopped and a new Committee
was formed. This Committee initiated the current format of the FIGO
Cancer Report and its first edition was presented at the FIGO Congress
held in Rome, Italy, in October 2012. The Committee has since
established collaboration with the Catalan Institute of Oncology to
resurrect the data collection program. This process is now at an
advanced stage and a detailed situational analysis of institutions able
to provide care to women with gynecological cancers is being conduct-
ed as we write this editorial in May 2015. Once the situational analysis is
complete the intention is to select 20 or so institutions in low- and
middle-income countries to collect incidence, stage, treatment, and
survival data. This will give us a global picture of the multiple inequities
in cancer care internationally and enable more targeted intervention by
organizations such as FIGO.
It is our intention to ensure that this, the second edition of the FIGO
Cancer Report, is disseminated as widely as possible internationally, and
therefore the chapters are available online with Open Access. We are
also developing a mobile App of the different criteria for FIGO staging,
which will ensure that those with smart phones will have instant access.
The chapters contained in the report have either been updated or are
new chapters. The new chapters include trophoblastic disease, targeted
therapies for gynecological cancer, rehabilitation after treatment for
gynecological cancer, psychosexual health in gynecological cancers,
palliative care in gynecological cancer, and the future role of molecular
staging in gynecological cancer. These six new chapters are added
to nine others that include the new staging for ovary, fallopian tube,
and peritoneum. Each chapter presents the site specific staging, the
rationale behind staging, as well as internationally accepted evidence-
based guidelines. All chapters are fully referenced and the quality of
the evidence for intervention is graded.
The current FIGO Gynecologic Oncology Committee members are:
Lynette Denny (Chair), Gynecologic Oncologist, South Africa
Michael Quinn (Co-Chair), Gynecologic Oncologist, Australia
Sergio Pecorelli (Immediate Past Chair), Gynecologic Oncologist, Italy
Adriana Bermudez, Gynecologic Oncologist, Argentina
Joanna Cain, Gynecologic Oncologist, USA
Keiichi Fujiwara, Gynecologic Oncologist, Japan
Neville Hacker, Gynecologic Oncologist, Australia
Elisabeth Åvall Lundqvist, Medical Oncologist, Sweden
Jaime Prat, Pathologist, Spain
Shyam Shrivastava, Radiation Oncologist, India
The Committee composition is likely to change during the 2015 FIGO
World Congress when the report will be released, as members leave the
committee after serving a six-year term. The composition of the new
Committee will be announced by the incoming President of FIGO.
Lynette Denny
Groote Schuur Hospital/University of Cape Town, Cape Town, South Africa
E-mail address: lynette.denny@uct.ac.za.
Michael Quinn
Royal Women’s Hospital and University of Melbourne, Melbourne, Australia
International Journal of Gynecology and Obstetrics 131 (2015) S75
http://dx.doi.org/10.1016/j.ijgo.2015.06.024
0020-7292/© 2015 Published by Elsevier Ireland Ltd. on behalf of International Federation of Gynecology and Obstetrics. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Contents lists available at ScienceDirect
International Journal of Gynecology and Obstetrics
journal homepage: www.elsevier.com/locate/ijgo
FIGO CANCER REPORT 2015
Cancer of the vulva
Neville F. Hacker a
, Patricia J. Eifel b
, Jacobus van der Velden c
a
Gynecologic Oncology Cancer Centre, Royal Hospital for Women, Randwick, Australia
b
Department of Radiation Oncology, M.D. Anderson Cancer Center, Houston, TX, USA
c
Department of Gynecology, Academic Medical Center, Amsterdam, The Netherlands
1. Staging
1.1. Anatomy
1.1.1. Primary site
Cases should be classified as carcinoma of the vulva when the prima-
ry site of growth is in the vulva. Any lesion that involves both the vagina
and vulva (i.e. crosses the hymenal ring) should be classified as a carci-
noma of the vulva. Tumors that present in the vulva as secondary
growths, from either a genital or extragenital site, should be excluded.
Malignant melanoma should be reported separately. There must be
histologic confirmation of the cancer.
1.1.2. Nodal stations
The inguinal and femoral nodes are the first sites of regional spread.
1.1.3. Metastatic sites
Patients who have extrapelvic metastases or who have involvement
of pelvic lymph nodes (external, hypogastric, obturator, and common
iliac) are classified as having stage IVB disease.
1.2. Surgical staging classification
The staging system for vulvar cancer has been based on surgical find-
ings since 1988. The final diagnosis is dependent upon thorough histo-
pathologic evaluation of the operative specimen (vulva and lymph
nodes). Various modifications have been made over time, with a subdi-
vision of Stage I added in 1994. The FIGO staging of vulvar carcinoma
was last changed in 2009 by the FIGO Committee on Gynecologic
Oncology [1], to give better prognostic discrimination between stages
(Table 1). Table 2 compares the FIGO staging with the Union of Interna-
tional Cancer Control “TNM” classification.
1.2.1. Histopathologic types
Approximately 80% of cases are squamous cell carcinomas, and many
cases, particularly in younger women, are HPV related. Melanomas are
the second most common cancer seen in cancer centers, although
community-based studies have reported basal cell carcinomas to be the
second most common vulvar cancer [2]. The histopathologic types are:
• squamous cell carcinoma
• melanoma
• verrucous carcinoma
• Paget’s disease of vulva
• adenocarcinoma, not otherwise specified (NOS)
• basal cell carcinoma, NOS
• Bartholin’s gland carcinoma.
1.2.2. Histopathologic grades (G)
• GX: Grade cannot be assessed
• G1: Well differentiated
• G2: Moderately differentiated
• G3: Poorly or undifferentiated.
2. Introduction
Carcinoma of the vulva is an uncommon tumor, representing about
4% of gynecologic malignancies. Because of the relatively small experi-
ence of individual institutions, randomized trials of therapeutic ap-
proaches are uncommon, and most studies are based on retrospective
clinicopathologic reviews [3].
It is predominantly a disease of postmenopausal women, with the
age-specific incidence increasing with increasing age. Although the
external location of the vulva should encourage early presentation,
vulvar cancers are often advanced at the time of diagnosis.
Most squamous carcinomas occur on the labia majora, but the labia
minora, clitoris, and perineum may also be primary sites.
Vulvar intraepithelial neoplasia (VIN), a precursor lesion in some
cases, tends to occur in younger women and may be associated with
similar lesions of the cervix and vagina. A new classification of squa-
mous VIN was introduced by the International Society for the Study of
Vulvovaginal Disease (ISSVD) in 2004 [4]. The term VIN 1 is no longer
used, and VIN 2 and 3 are simply called VIN. There are two types of
VIN: (1) VIN, usual type (warty, basaloid, and mixed), which is HPV-
related in most cases; and (2) VIN, differentiated type, which is seen
particularly in older women, and is often associated with lichen
sclerosus and/or squamous hyperplasia. The incidence of VIN, usual
type, and the incidence of invasive vulvar cancer in premenopausal
women should both decrease significantly with increasing use of
HPV vaccination.
International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
http://dx.doi.org/10.1016/j.ijgo.2015.06.002
0020-7292/© 2015 Published by Elsevier Ireland Ltd. on behalf of International Federation of Gynecology and Obstetrics. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Contents lists available at ScienceDirect
International Journal of Gynecology and Obstetrics
journal homepage: www.elsevier.com/locate/ijgo
Treatment of vulvar cancer used to be primarily surgical, but radia-
tion therapy and, to a lesser extent, chemotherapy have been progres-
sively integrated into the treatment protocol over the past 30 years.
Therefore, management has evolved into an individualized multidisci-
plinary approach, and patients should be referred centrally to a gyneco-
logical cancer center where all relevant expertise is available [5,6]. Level
of Evidence B
3. Screening
There is no screening procedure for vulvar cancer. However, patients
with a past history of cervical or vaginal cancer should have inspection
of the vulva, with or without colposcopic examination, as part of their
regular follow-up. Patients with lichen sclerosus or a past history of
VIN should also be kept under regular surveillance, and taught to under-
take regular self-examination with a mirror.
4. Squamous cell carcinoma
4.1. Presenting symptoms
Vulvar cancer may be asymptomatic, but most patients present with
a vulvar lump or ulcer, which may or may not be painful. There is often a
long-standing history of pruritus, which may be due to associated
vulvar dystrophy. Bleeding or discharge is an occasional presenting
symptom, and patients with advanced disease may present with a
lump in the groin caused by metastases to groin lymph nodes.
4.2. Diagnosis
If the disease appears to be entirely intraepithelial, initial assessment
should consist of multiple biopsies to exclude an invasive focus. A 3 or
4 mm Keyes biopsy instrument is ideal for this purpose. Patients with
multifocal lesions should have biopsies taken from several lesions.
If there appears to be invasive cancer present, a wedge or Keyes bi-
opsy under local anesthesia in the office is usually sufficient to confirm
the diagnosis. The biopsy should include some underlying stroma.
For small tumors, it is preferable not to excise the entire lesion at the
time of biopsy because this makes it more difficult to plan the subse-
quent definitive resection.
If the lesion is 2 cm or less in diameter and depth of stromal inva-
sion is less than or equal to 1 mm on the initial biopsy, radical local
excision of the lesion must be undertaken to allow serial sectioning to
properly assess the depth of invasion. If there is still no focus found
with a depth of invasion greater than 1 mm, this excision will also be
the definitive treatment [3].
4.3. Investigations
(1) Cervical cytology, if applicable.
(2) Colposcopy of the cervix and vagina because of the common as-
sociation with other squamous intraepithelial lesions.
(3) For large lesions, a CT or MRI scan of the pelvis and groins is help-
ful to detect any enlarged lymph nodes in the groins or pelvis,
erosion into underlying bony structures, or other evidence of
metastatic disease.
(4) Routine full blood count, biochemical profile, and chest
X-ray preoperatively.
4.4. Clinical practice guidelines
The clinical findings should be recorded on a staging diagram
(e.g. Fig. 1). The findings according to the staging delineated in
Tables 1 and 2 are usually listed on the reverse side of the diagram.
4.5. Treatment
4.5.1. Treatment of vulvar intraepithelial neoplasia
Various treatment modalities are available for treating intraepithelial
lesions of the vulva [7,8]. Once the diagnosis has been established, super-
ficial local excision of the vulvar epithelium with a 0.5–1.0-cm margin is
considered adequate for lesions of the lateral aspect of the vulva. Lesions
involving the labia minora may also be treated by local excision but may
respond favorably to laser vaporization. Laser is also appropriate for
clitoral and perianal lesions. Laser treatment of the hair-bearing skin of
the vulva will usually produce depigmentation and destruction of hair
follicles, with subsequent loss of hair growth. Large lesions may be
treated with a skinning vulvectomy and split-thickness skin graft. Level
of Evidence C
Two randomized controlled trials have shown promising results
with the topical immune response modifier, imiquimod, with complete
response rates of 35–81% reported [9,10]. Long-term follow-up of pa-
tients in one of the studies showed good sustained benefit, although
the number of patients in the study was small [9]. Level of Evidence A
Table 1
FIGO staging of carcinoma of the vulva.
FIGO
Stage
Description
I Tumor confined to the vulva
IA Lesions ≤ 2 cm in size, confined to the vulva or perineum and with stromal
invasion ≤ 1.0 mma
, no nodal metastasis
IB Lesions N 2 cm in size or with stromal invasion N 1.0 mma
, confined to the
vulva or perineum, with negative nodes
II Tumor of any size with extension to adjacent perineal structures (lower
third of urethra, lower third of vagina, anus) with negative nodes
III Tumor of any size with or without extension to adjacent perineal
structures (lower third of urethra, lower third of vagina, anus) with
positive inguinofemoral nodes
IIIA (i) With 1 lymph node metastasis (≥5 mm), or
(ii) With 1–2 lymph node metastasis(es) (b5 mm)
IIIB (i) With 2 or more lymph node metastases (≥5 m), or
(ii) With 3 or more lymph node metastases (b5 mm)
IIIC With positive nodes with extracapsular spread.
IV Tumor invades other regional (upper 2/3 urethra, upper 2/3 vagina), or
distant structures
IVA Tumor invades any of the following:
(i) upper urethral and/or vaginal mucosa, bladder mucosa, rectal mucosa,
or fixed to pelvic bone, or
(ii) fixed or ulcerated inguinofemoral lymph nodes
IVB Any distant metastasis including pelvic lymph nodes
a
The depth of invasion is defined as the measurement of the tumor from the epithelial–
stromal junction of the adjacent most superficial dermal papilla to the deepest point of
invasion.
Table 2
Cancer of the vulva: FIGO staging compared with TNM classification.
FIGO Stage Union for International Cancer Control (UICC)
T (tumor) N (lymph nodes) M (metastasis)
I T1 N0 M0
IA T1a N0 M0
IB T1b N0 M0
II T2/T3 N0 M0
IIIA T1, T2, T3 N1a, N1b M0
IIIB T1, T2, T3 N2a, N2b M0
IIIC T1, T2, T3 N2c M0
IVA T4 N0–N2 M0
IVB Any T N3 M0
S77N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
4.5.2. Invasive vulvar cancer
Management of vulvar cancer must be individualized. There is no
standard operation and the emphasis should be on performing the
most conservative operation consistent with cure of the disease [3].
In considering surgical treatment options, it is necessary to consider
independently the most appropriate management of:
(1) The primary lesion.
(2) The groin lymph nodes.
In cases of locally advanced disease, treatment options for each site
should be considered independently and then in the context of the over-
all management of the patient, in order to select a treatment that will op-
timize the likelihood of cure and minimize treatment-related morbidity.
4.5.3. Microinvasive vulvar cancer (Stage IA)
Stage IA carcinoma of the vulva is defined as a single lesion measur-
ing 2 cm or less in diameter with a depth of invasion of 1.0 mm or less,
the depth being measured from the epithelial–stromal junction of the
most adjacent superficial dermal papilla to the deepest point of inva-
sion. Lesions of this extent should be managed with radical local
excision. Groin dissection is not necessary for lesions of this type
[11,12]. Level of Evidence C
4.5.4. Early vulvar cancer
Tumors confined to the vulva without suspicious lymph nodes, as
determined by clinical examination, with or without ultrasonic or radio-
logical assessment, may be considered early.
4.5.4.1. Management of the primary lesion (Fig. 2). To decrease psycho-
sexual morbidity, a more conservative operation than radical
vulvectomy usually is indicated. The procedure may be called a radical
local excision, and for localized lesions, this operation is as effective as
radical vulvectomy in preventing local recurrence [3,13–17].
Surgical removal should achieve lateral margins of at least 1 cm, and
the deep margin should be the inferior fascia of the urogenital dia-
phragm, which is co-planar with the fascia lata and the fascia over the
pubic symphysis [13,16–18].
If the lesion is close to the urethra, the distal 1 cm of the urethra may
be resected without jeopardizing urinary continence.
If there is associated VIN, this should be superficially excised to con-
trol symptoms, to exclude other areas of superficial invasion, and to
Fig. 1. Vulvar staging diagram.
S78 N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
prevent subsequent progression to invasive cancer. This is especially
true for differentiated VIN. Level of Evidence C
4.5.4.2. Management of groin lymph nodes. Recurrence in the groin
carries a very high mortality; therefore, appropriate groin treatment
is the single most important factor in reducing mortality from early
vulvar cancer [3].
All patients with FIGO stage 1B or stage II lesions should have at least
an ipsilateral inguinofemoral lymphadenectomy. Level of Evidence C
The incidence of positive contralateral nodes in patients with small
lateral lesions and negative ipsilateral nodes is less than 1%, so unilateral
groin dissection is appropriate for such lesions [3].
Bilateral groin dissection should be performed for midline tumors,
and for those involving the anterior labia minora [19]. Large lateral
tumors should probably also have bilateral dissection, and definitely if
the ipsilateral nodes are positive [19].
Sentinel node excision is being increasingly practiced in many
centers following the European multicenter observational study on sen-
tinel node detection [20]. This procedure detects nodal metastasis in
most patients with regional spread of disease, and is associated with a
lower rate of lymphedema than complete lymphadenectomy. The
study, (GROINSS-V), involved 403 women with a unifocal tumor con-
fined to the vulva less than 4 cm in diameter, stromal invasion more
than 1 mm, and clinically negative lymph nodes [20]. Sentinel nodes
were identified using blue dye and radiolabelled technetium. Lymphad-
enectomy was omitted in sentinel node negative women. Groin recur-
rences occurred in 2.3% of patients, with a median follow-up of 35
months. Overall disease-specific survival was 97% after 3 years and mor-
bidity was substantially reduced. Higher false-negative rates have been
reported in other studies [21–23].
Owing to the small but definite false-negative rate with sentinel
node biopsy, and the high risk of death if groin recurrence occurs,
some patients, properly informed of the risks and benefits, will elect
to have a full groin dissection, despite the greater complication rate
[24–26]. Level of Evidence B
4.5.4.3. Groin dissection. It is recommended that both inguinal and fem-
oral nodes be removed, as inguinal node dissection alone is associated
with a higher incidence of groin recurrence [27]. Level of Evidence A
The femoral nodes are situated medial to the femoral vein within the
fossa ovalis. There is no need to remove the fascia lata to dissect the
femoral nodes [28]. Groin dissection may be safely performed through
a triple incision approach, and this should improve primary healing
compared with an en bloc resection of the vulva and groins [29]. Level
of Evidence C
An en bloc approach may still be useful for clitoral or periclitoral
lesions. To avoid skin necrosis, all subcutaneous tissue above the super-
ficial fascia must be preserved.
Groin dissection (with postoperative radiation for patients with
positive groin nodes) was found to be superior to groin irradiation in
one small randomized trial [30]. Pretreatment imaging that might
have detected grossly enlarged nodes was not performed in that early
trial and the radiation technique used was considered inadequate
to cover the at-risk inguinofemoral nodes [31]. Retrospective clinical
reviews have suggested that radiation alone can control microscopic
disease in the groins if adequate coverage of the inguinofemoral nodes
is confirmed [32,33].
4.5.4.4. Management of patients with positive groin nodes. The Gynecolog-
ic Oncology Group demonstrated superior results for pelvic and inguinal
radiation compared with pelvic node dissection for patients who had an
inguinal lymph node dissection with findings of grossly positive or more
than one positive node [34]. Level of Evidence A. A recent retrospective,
multicenter German study also reported improved survival for patients
with positive groin nodes who received adjuvant radiotherapy directed
at the groins (positive/negative other fields) [35].
Several studies have emphasized the prognostic significance of the
morphology of positive groin nodes, particularly the size of the metasta-
sis and the presence or absence of extracapsular spread [36–38].
Patients with one small lymph node metastasis do not appear to
benefit from adjuvant radiation therapy; several series suggest that
their prognosis is good after inguinofemoral lymphadenectomy alone
[37–39], but the number of patients in most series is too small to draw
definitive conclusions. A multicenter Dutch study of 75 patients with
vulvar cancer and one positive lymph node of all sizes reported that
adjuvant radiation was only beneficial if extracapsular spread was
present [40].
Reasonable indications for bilateral pelvic and groin irradiation in
patients with positive groin nodes would be:
• The presence of extracapsular spread.
• Two or more positive groin nodes. Level of Evidence B
An ongoing international prospective observational trial (GROINSS-V
II) is investigating the efficacy of groin radiation, without inguinofemoral
lymphadenectomy, for patients with a single positive sentinel lymph
node 2 mm or less in diameter. Pending the results of this study, all
patients who have had a sentinel lymph node biopsy and are found
to have one or more positive nodes should be treated with a full
inguinofemoral lymph node dissection, followed by radiotherapy to
the groins and pelvis if indicated.
4.5.4.5. Radiation fields and doses. In most cases, fields should include the
inguinofemoral, external iliac, and internal iliac lymph nodes. The upper
border may be extended if there is extensive inguinal involvement or
suspicion of pelvic node metastasis.
One of a variety of radiation techniques can be selected, depending
on the patient’s body habitus and extent of disease. Treatments should
always be based on three-dimensional planning using high-quality CT
or MRI images.
Combined photon and electron techniques are often used to treat
the regional nodes, without overdosing the femoral heads. Care must
be taken to completely include both the superficial and deep inguinal
Fig. 2. Management of early vulvar cancer. * If there is associated VIN or lichen sclerosis,
these areas may be superficially excised.
S79N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
lymph nodes. In thin patients, care must be taken to avoid underdosage of
superficial inguinal nodes by high-energy photon beams. If electron beams
are used, the energy must be sufficient to cover the femoral nodes. In recent
years, some clinicians have begun to use intensity-modulated radiation
therapy (IMRT) or other inverse-planned, computer-controlled delivery
techniques to treat vulvar cancer. Although these techniques can reduce
acute radiation effects in skin and soft tissue, the treatment planning
and delivery are complex, and the opportunity for unanticipated
underdosage of the target is substantial, suggesting that these cases may
be best treated by clinicians who have considerable specialized expertise.
The dose of radiation is determined by the initial extent of regional
disease and any known residual. After a groin dissection with microscopic
inguinal metastases, 50 Gy in 1.8–2.0 Gy fractions is usually sufficient.
If there are multiple positive nodes or if there is evidence of
extracapsular extension, doses up to 60 Gy may be given to a reduced
volume. Gross residual disease usually requires 60–70 Gy to achieve a
high probability of regional disease control.
The effectiveness of concurrent chemotherapy in the treatment of
groin and pelvic lymph nodes is unknown.
4.5.5. Advanced vulvar cancer
Patients with primary tumors extending beyond the vulva, or bulky
positive groin nodes are considered to have advanced vulvar cancer. For
such patients, multimodality treatment planning is particularly important.
4.5.5.1. Management of groin lymph nodes. It is desirable to determine the
status of the groin nodes prior to planning the overall treatment [3].
Pelvic CT or MRI should be part of the patient’s initial workup. These
studies are particularly helpful in suggesting the extent of inguinal or
pelvic lymphadenopathy (Fig. 3). Pelvic MRI can also provide useful
information about the anatomical extent of the primary lesion, but is
not mandatory.
If there are no suspicious nodes in the groin on CT scan, bilateral
inguinofemoral lymphadenectomy may be performed. If final histologic
assessment reveals positive nodes, adjuvant radiation to the groin and
pelvis should follow the guidelines given for early stage disease. If the
nodes are negative, groin and pelvic radiation may be eliminated.
Alternatively, primary chemoradiation therapy may be used to treat
the primary tumor as well as the groin and pelvic nodes if surgery is
deemed inappropriate for the individual patient [32,33].
If nodes are clinically positive, a complete lymphadenectomy should
be avoided because full groin dissection together with postoperative
groin irradiation may result in severe lymphedema. Only enlarged
nodes from the groin and pelvis should be removed if feasible, and the
patient given postoperative groin and pelvic radiation [41]. Level of
Evidence C
If there are ulcerated or fixed groin nodes, they should be resected if
not infiltrating muscle or femoral vessels, as determined by imaging
studies. If nodes are not felt to be resectable, they should be biopsied
to confirm the diagnosis then treated with primary radiation, with or
without chemotherapy. If appropriate, the nodes may be resected fol-
lowing radiation if there has been an incomplete response (Fig. 4)
[42]. Level of Evidence C
4.5.5.2. Management of the primary tumor (Fig. 5). If it is possible to resect
the primary lesion with clear surgical margins and without sphincter
damage leading to urinary or fecal incontinence, primary surgical exci-
sion is usually the preferred treatment. It usually follows dissection of
the groins, although this is not mandatory.
If primary surgery would result in the need for a bowel or urinary
stoma, it is preferable to employ primary radiation therapy, some-
times followed by a more limited resection of the residual tumor or
tumor bed [43,44].
Chemoradiation has been used extensively for large lesions if surgi-
cal resection would damage central structures (anus, urethra); durable
complete responses without the need for post-treatment surgery have
been well described [45–49].
The groin nodes and pelvis may need to be included in the treatment
field depending on the status of the groin nodes, as determined initially.
Treatment with neoadjuvant cisplatin and 5-fluorouracil, or other
drug combinations, has been reported in small retrospective studies to
be effective for preservation of the anal sphincter and/or urethra in
patients with advanced vulvar cancer [50,51]. This approach deserves
further clinical research.
4.5.5.3. Radiation protocol. If the groin nodes are positive and meet the
requirements described earlier for adjuvant radiation, the initial radia-
tion treatment fields should include the pelvis, inguinal nodes, and pri-
mary site, which are treated to a total dose of at least 50 Gy. Care must
be taken to adequately cover the inguinal nodes.
Some clinicians prefer to treat in an open-leg position but care must
be taken to apply bolus to the vulva to avoid underdosage of potentially
involved skin regions.
Areas of gross disease or particularly high risk are usually boosted
either with appositional fields of electrons selected to provide an ade-
quate dose to the surface and at depth, or with conformal external
beam therapy. Gross vulvar disease probably requires 60–70 Gy to
achieve local control, although investigators are currently exploring
Fig. 3. Management of clinically suspicious groin nodes. Fig. 4. Management of clinically obvious groin nodes.
S80 N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
a wide variety of chemoradiation schedules, and the relationship be-
tween dose and local control remains somewhat uncertain. Level of
Evidence C
4.5.5.4. Close surgical margins. Most recurrences from vulvar cancer
occur on the vulva. Rouzier et al. [52] described two types of local recur-
rences, those at the primary site and those at a remote site. In an analysis
of patients with vulvar cancer from the Royal Hospital for Women in
Sydney [13], primary site recurrences occurred with a median disease-
free interval of 21 months, and were associated with a histological mar-
gin of 8 mm or less, as previously reported [16 − 18]. Remote site recur-
rences occurred with a median disease-free interval of 69 months, and
were more commonly associated with lichen sclerosus. Although
some recent papers have not found an association between margin dis-
tance and local recurrence, these papers have not distinguished primary
from remote site recurrences [53,54].
Postoperative radiation is of benefit for patients with close surgical
margins (less than 5 mm), if the margins cannot be re-excised [55]. A re-
cent study of 205 patients with vulvar cancer from Boston reported that
the highest risk of vulvar recurrence was associated with margins of
5 mm or less (P = 0.002), and that patients who received a dose of
more than or equal to 56 Gy had a lower risk of relapse than those
who received less than or equal to 50.4 Gy (P b 0.05) [56].
In some cases, the positive margin may be boosted with brachyther-
apy, although this technique requires experience to avoid an excessive
risk of necrosis. Alternatively, the operative bed may be treated with
an appositional electron field or in some cases, carefully planned confor-
mal external beam irradiation. Level of Evidence C
5. Special situations
5.1. Vulvar melanoma
Vulvar melanoma is the second most common neoplasm of the
vulva. The majority of lesions involve the clitoris or labia minora. The
Clark or Breslow modifications to the micro staging system should be
used for the staging of vulvar melanoma rather than the more common
TNM/FIGO system. These systems measure the depth of invasion in
terms of the descriptive histology of the skin.
Any pigmented lesion on the vulva should be excised for diagnosis
unless it has been known to be present and unchanged for some years.
In line with trends toward more conservative surgery for cutaneous
melanomas, there is a trend toward more conservative resection of
vulvar melanomas [57–59]. Primary lesions should be treated by radical
local excision, with margins around the lesion of at least 1 cm. Level of
Evidence C
The role of node dissection is also controversial, but the Intergroup
Surgical Melanoma Program has conducted a prospective, multi-
institutional randomized trial of elective node dissection versus obser-
vation for intermediate thickness cutaneous melanomas (1–4 mm)
[60]. There were 740 patients entered into the trial, and elective node
dissection resulted in a significantly better survival for patients
60 years of age or younger, patients with tumors 1–2 mm thick, and
patients without tumor ulceration.
5.2. Bartholin’s gland cancer
Cancers arising in the Bartholin’s gland may be either transitional or
squamous types, arising from the duct, or an adenocarcinoma from the
gland itself. Adenoid cystic and adenosquamous variants have also been
reported. Adenocarcinomas of the vulva occur, on average, approxi-
mately a decade earlier than invasive squamous cancers. Frequently,
diagnosis is made after resection of what was thought to be a persisting
Bartholin’s cyst.
The standard approach for Bartholin’s gland carcinomas has been
radical vulvectomy and bilateral groin dissection. However, ipsilateral
groin dissection and radical hemi-vulvectomy may be equally effective
for early lesions [61]. Because these lesions are deep in the ischiorectal
fossa, surgical margins are more likely to be close, particularly for
bulky lesions, and postoperative radiation to the vulva may decrease
the likelihood of local recurrence [61]. Level of Evidence C
If the ipsilateral groin nodes are positive, bilateral groin and pelvic
radiation may decrease regional recurrence.
For adenoid cystic lesions, radical local excision alone is the treat-
ment of choice, with adjuvant local radiation recommended for positive
margins or perineural invasion [62]. Level of Evidence C
5.3. Paget’s Disease
This is predominantly an intraepithelial lesion, but on occasion it
may be associated with an underlying invasive adenocarcinoma. It is
usually of primary cutaneous origin from the vulva, but may be second-
ary to an anorectal, urothelial, or noncutaneous genital tract carcinoma
(e.g. endocervical or endometrial) [63].
The disease occurs predominantly in the menopausal or postmeno-
pausal population. Most patients will present with vulvar discomfort
and itching and on examination, an eczematoid, weeping lesion is
often seen. Diagnosis is usually confirmed by biopsy, which will gener-
ally differentiate an intraepithelial from an invasive lesion [3,64].
Intraepithelial Paget’s disease requires superficial local excision. It is
difficult to obtain clear margins with this disease, as often the underly-
ing histologic change will extend far beyond the macroscopic lesion.
More recently, there has been a move to perform less radical resection
for intraepithelial lesions, with re-excision at a later date should lesions
become symptomatic or clinically visible [65]. Lesions that involve or
extend into the urethra or anus can be particularly difficult to manage,
and may require laser therapy.
If there is an underlying adenocarcinoma, the invasive component
should be treated by radical local excision with margins of at least
1 cm. At least an ipsilateral inguinofemoral lymphadenectomy should
be performed for unilateral lesions, with adjuvant radiation following
the same indications as for squamous carcinomas [66]. Level of
Evidence C
6. Pathology
The surgical specimen should be correctly orientated and photo-
graphed. Photographs should be used to indicate the origin of tissue
blocks. The size of the specimen should be measured and the
Fig. 5. Management of advanced primary tumor. Treatment should usually follow dissec-
tion of the groins. Groin and pelvic radiation should follow standard indications.
S81N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
dimensions of any visible tumor measured. The macroscopic tumor-free
surgical margins should be measured. Sections are taken through the
tumor to measure tumor depth. Sections should be taken from urethral,
anal, and vaginal resection margins.
Lymph nodes should be carefully dissected out and the site from
which they are removed recorded. A full cross-section of each lymph
node should be embedded.
The following histological points should be noted:
(a) Tumor type: keratinizing, basaloid, bowenoid.
(b) Depth of invasion: measured from the epithelial–stromal junc-
tion of the adjacent dermal papilla to the deepest point of inva-
sion by the tumor.
(c) Tumor grade.
(d) Histological measurement of tumor-free margins and statement
as to whether the tumor is completely excised.
(e) Presence or absence of perineural or vascular space invasion.
(f) Nature of the adjacent nonmalignant squamous epithelium: VIN,
lichen sclerosus, squamous hyperplasia, HPV-associated changes.
(g) Sites and number of nodes examined and number of positive
nodes. Presence or absence of extracapsular extension.
Conflict of interest
The authors declare that they have no conflicts of interest.
References
[1] Hacker NF. Revised FIGO staging for carcinoma of the vulva. Int J Gynecol Obstet
2009;105(2):105–6.
[2] Van der Velden J, van Lindert AC, Gimbrere CH, Oosting H, Heintz AP. Epidemiologic
data on vulvar cancer: Comparison of hospital with population-based data. Gynecol
Oncol 1996;62(3):379–83.
[3] Hacker NF, Cancer Vulvar. In: Berek JS, Hacker NF, editors. Berek and Hacker’s
Gynecologic Oncology. 6th ed. Philadelphia: Lippincott Williams and Wilkins;
2015. p. 560–607.
[4] Sideri M, Jones RW, Wilkinson EJ, Preti M, Heller DS, Scurry J, et al. Squamous vulvar
intraepithelial neoplasia: 2004 modified terminology, ISSVD Vulvar Oncology Sub-
committee. J Reprod Med 2005;50(11):807–10.
[5] van der Velden J, van Lindert AC, Gimbrere CH, Oosting H, Heintz AP. Epidemiologic
data on vulvar cancer: comparison of hospital with population-based data. Gynecol
Oncol 1996;62(3):379–83.
[6] Rhodes CA, Cummins C, Shafi M. The management of squamous cell vulval cancer: a
population based retrospective study of 411 cases. Br J Obstet Gynaecol 1998;
105(2):200–5.
[7] Wallbillich JJ, Rhodes HE, Milbourne AM, Munsell MF, Frumovitz M, Brown J, et al.
Vulvar intraepithelial neoplasia (VIN 2/3): comparing clinical outcomes and evaluat-
ing risk factors for recurrence. Gynecol Oncol 2012;127(2):312–5.
[8] Rodolakis A, Diakomanolis E, Vlachos G, Iconomou T, Protopappas A, Stefanidis C,
et al. Vulvar intraepithelial neoplasia (VIN)–diagnostic and therapeutic challenges.
Eur J Gynaecol Oncol 2003;24(3):317–22.
[9] Terlou A, van Seters M, Ewing PC, Aaronson NK, Gundy CM, Heijmans-Antonissen C,
et al. Treatment of vulvar intraepithelial neoplasia with topical imiquimod: Seven
years median follow-up of a randomized clinical trial. Gynecol Oncol 2011;121(1):
157–62.
[10] Mathiesen O, Buus SK, Cramers M. Topical imiquimod can reverse vulvar intraep-
ithelial neoplasia: A randomised, double-blinded study. Gynecol Oncol 2007;
107(2):219–22.
[11] Iversen T, Abeler V, Aalders J. Individualized treatment of stage I carcinoma of the
vulva. Obstet Gynecol 1981;57(1):85–90.
[12] Hacker NF, Berek JS, Lagasse LD, Nieberg RK, Leuchter RS. Individualization of treat-
ment for stage I squamous cell vulvar carcinoma. Obstet Gynecol 1984;63(4):
155–62.
[13] Tantipalakorn C, Robertson G, Marsden DE, Gebski V, Hacker NF. Outcome and pat-
terns of recurrence for International Federation of Gynecology and Obstetrics (FIGO)
stages I and II squamous cell vulvar cancer. Obstet Gynecol 2009;113(4):895–901.
[14] Farias-Eisner R, Cirisano FD, Grouse D, Leuchter RS, Karlan BR, Lagasse LD, et al. Con-
servative and individualized surgery for early squamous carcinoma of the vulva: the
treatment of choice for stage I and II (T1-2NO-1 M0) disease. Gynecol Oncol 1994;
53(1):55–8.
[15] Burke TW, Levenback C, Coleman RL, Morris M, Silva EG, Gershenson DM. Surgical
therapy of T1 and T2 vulvar carcinoma: further experience with radical wide exci-
sion and selective inguinal lymphadenectomy. Gynecol Oncol 1995;57(2):215–20.
[16] Heaps JM, Fu YS, Montz FJ, Hacker NF, Berek JS. Surgical pathological variables pre-
dictive of local recurrence in squamous cell carcinoma of the vulva. Gynecol Oncol
1990;38(3):309–14.
[17] De Hullu JA, Hollema H, Lolkema S, Boezen M, Boonstra H, Burger MP, et al. Vulvar
carcinoma: the price of less radical surgery. Cancer 2002;95(11):2331–8.
[18] Chan JK, Sugiyama V, Pham H, Gu M, Rutgers J, Osann K, et al. Margin distance and
other clinico-pathological prognostic factors in vulvar carcinoma: a multivariate
analysis. Gynecol Oncol 2007;104(3):636–41.
[19] Iversen T, Aas M. Lymph drainage from the vulva. Gynecol Oncol 1983;16(2):
179–89.
[20] Van der Zee AG, Oonk MH, De Hullu JA, Ansink AC, Vergote I, Verheijen RH, et al.
Sentinel node dissection is safe in the treatment of early stage vulvar cancer. J Clin
Oncol 2008;26(6):884–9.
[21] Hampl M, Hantschmann P, Michels W, Hillemanns P. German Multicenter Study
Group Validation of the accuracy of the sentinel lymph node procedure in patients
with vulvar cancer: results of a multicenter study in Germany. Gynecol Oncol
2008;111(2):282–8.
[22] Levenback CF, Ali S, Coleman RL, Gold MA, Fowler JM, Judson PL, et al. Lymphatic
mapping and sentinel lymph node biopsy in women with squamous cell carcinoma
of the vulva: a gynecologic oncology group study. J Clin Oncol 2012;30(31):
3786–91.
[23] Radziszewski J, Kowalewska M, Jedrzejczak T, Kozlowicz-Gudzinska I, Nasierowska-
Guttmejer A, Bidzinski M, et al. The accuracy of the sentinel lymph node concept in
early stage squamous cell vulvar carcinoma. Gynecol Oncol 2010;116(3):473–7.
[24] de Hullu JA, Ansink AC, Tymstra T, van der Zee AG. What doctors and patients think
about false-negative sentinel lymph nodes in vulvar cancer. J Psychosom Obstet
Gynaecol 2001;22(4):199–203.
[25] Gan S, Magarey C, Schwartz P, Papadatos G, Graham P, Vallentine J. Women’s choice
between sentinel lymph node biopsy and axillary clearance. ANZ J Surg 2002;72(2):
110–3.
[26] Farrell R. Gebski, Hacker NF. Quality of life after complete lymphadenectomy for vul-
var cancer: do women prefer sentinel node biopsy? Int J. Gynecol Oncol 2014;24(4):
813–9.
[27] Stehman FB, Bundy BN, Dvoretsky PM, Creasman WT. Early stage I carcinoma of the
vulva treated with ipsilateral superficial inguinal lymphadenectomy and modified
radical hemivulvectomy: a prospective study of the Gynecologic Oncology Group.
Obstet Gynecol 1992;79(4):490–7.
[28] Micheletti L, Borgno G, Barbero M, Preti M, Cavanna L, Nicolaci P, et al. Deep femoral
lymphadenectomy with preservation of the fascia lata. Preliminary report on 42
invasive vulvar carcinomas. J Reprod Med 1990;35(12):1130–4.
[29] Hacker NF, Leuchter RS, Berek JS, Castaldo TW, Lagasse LD. Radical vulvectomy and
bilateral inguinal lymphadenectomy through separate groin incisions. Obstet
Gynecol 1981;58(5):574–9.
[30] Stehman F, Bundy B, Thomas G, Varia M, Okagaki T, Roberts J, et al. Groin dissection
versus groin radiation in carcinoma of the vulva: a Gynecologic Oncology Group
study. Int J Radiat Oncol Biol Phys 1992;24(2):389–96.
[31] Koh WJ, Chiu M, Stelzer KJ, Greer BE, Mastras D, Comsia N, et al. Femoral vessel
depth and the implication for groin node radiation. Int J Radiat Oncol Biol Phys
1993;27(3):969–74.
[32] Katz A, Eifel PJ, Jhingran A, Levenback CF. The role of radiation therapy in preventing
regional recurrences of invasive squamous cell carcinoma of the vulva. Int J Radiat
Oncol Biol Phys 2003;57(2):409–18.
[33] Petereit DG, Mehta MP, Buchler DA, Kinsella TJ. A retrospective review of nodal
treatment for vulvar cancer. Am J Clin Oncol 1993;16(1):38–42.
[34] Homesley HD, Bundy BN, Sedlis A, Adcock L. Radiation therapy versus pelvic node
resection for carcinoma of the vulva with positive groin nodes. Obstet Gynecol
1986;68(6):733–40.
[35] Mahner S, Jueckstock J, Hilpert F, Neuser P, Harter P, de Gregorio N, et al. Adjuvant
therapy in lymph node positive vulvar cancer: The AGO-CaRT-1 study. J Natl Cancer
Inst 2015;107(3):dju426.
[36] Origoni M, Sideri M, Garsia S, Carinelli SG, Ferrari AG. Prognostic value of patholog-
ical patterns of lymph node positivity in squamous cell carcinoma of the vulva stage
III and IVA FIGO. Gynecol Oncol 1992;45(3):313–6.
[37] Paladini D, Cross P, Lopes A, Monaghan J. Prognostic significance of lymph node
variables in squamous cell cancer of the vulva. Cancer 1994;74(9):2491–6.
[38] van der Velden J, van Lindert AC, Lammes FB, ten Kate FJ, Sie-Go DM, Oosting H, et al.
Extracapsular growth of lymph node metastases in squamous cell cancer of the
vulva. The impact on recurrence and survival. Cancer 1995;75(12):2885–90.
[39] Hacker NF, Berek JS, Lagasse LD, Leuchter RS, Moore JG. Management of regional
lymph nodes and their prognostic influence in vulvar cancer. Obstet Gynecol
1983;61(4):408–12.the vulva. The impact on recurrence and survival. Cancer
1995;75(12):2885–90.
[40] Fons G, Groenen SM, Oonk MH, Ansink AC, van der Zee AG, Burger MP, et al. Adju-
vant radiotherapy in patients with vulvar cancer and one intracapsular lymph
node metastasis is not beneficial. Gynecol Oncol 2009;114(2):343–5.
[41] Hyde SE, Valmadre S, Hacker NF, Schilthuis MS, van der Velden J. Squamous cell car-
cinoma of the vulva with bulky positive groin nodes-nodal debulking versus full
groin dissection prior to radiation therapy. Int J Gynecol Cancer 2007;17(1):154–8.
[42] Montana GS, Thomas GM, Moore DH, Saxer A, Mangan CE, Lentz SS, et al. Preopera-
tive chemoradiation for carcinoma for the vulva with N2/N3 nodes: a Gynecologic
Oncology Group Study. Int J Radiat Oncol Biol Phys 2000;48(4):1007–13.
[43] Hacker NF, Berek JS, Juillard GJF, Lagasse LD. Preoperative radiation therapy for local-
ly advanced vulvar cancer. Cancer 1984;54(10):2056–60.
[44] Boronow RC, Hickman BT, Reagan MT, Smith RA, Steadham RE. Combined therapy as
an alternative to exenteration for locally advanced vulvovaginal cancer. II. Results,
complications and dosimetric and surgical considerations. Am J Clin Oncol 1987;
10(2):171–81.
[45] Moore DH, Ali S, Koh WJ, Michael H, Barnes MN, McCourt CK, et al. A phase II trial of
radiation therapy and weekly cisplatin chemotherapy for the treatment of locally
S82 N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
advanced squamous cell carcinoma of the vulva: a gynecologic oncology group
study. Gynecol Oncol 2012;124(3):529–33.
[46] Lupi G, Raspagliesi F, Zucali R, Fontanelli R, Paladini D, Kenda R, et al. Combined pre-
operative chemoradiotherapy followed by radical surgery in locally advanced vulvar
carcinoma. A pilot study. Cancer 1996;77(8):1472–8.
[47] Landoni F, Maneo A, Zanetta G, Colombo A, Nava S, Placa F, et al. Concurrent preop-
erative chemotherapy with 5-fluorouracil and mitomycin C and radiotherapy
(FUMIR) followed by limited surgery in locally advanced and recurrent vulvar carci-
noma. Gynecol Oncol 1996;61(3):321–7.
[48] Cunningham MJ, Goyer RP, Gibbons SK, Kredentser DC, Malfetano JH, Keys H. Prima-
ry radiation, cisplatin, and 5-fluorouracil for advanced squamous cell carcinoma of
the vulva. Gynecol Oncol 1997;66(2):258–61.
[49] Beriwal S, Coon D, Heron DE, Kelley JL, Edwards RP, Sukumvanich P, et al. Preoper-
ative intensity-modulated radiotherapy and chemotherapy for locally advanced
vulvar cancer. Gynecol Oncol 2008;109(2):291–5.
[50] Geisler JP, Manahan KJ, Buller RE. Neoadjuvant chemotherapy in vulvar cancer:
avoiding primary exenteration. Gynecol Oncol 2006;100(1):53–7.
[51] Aragona AM, Cuneo N, Soderini AH, Alcoba E, Greco A, Reyes C, et al. Tailoring the
treatment of locally advanced carcinoma of the vulva: Neoadjuvant chemotherapy
followed by radical surgery: Results from a multicenter study. Int J Gynecol Cancer
2012;22(7):1258–63.
[52] Rouzier R, Haddad B, Plantier F, Dubois P, Pelisse M, Paniel BJ. Local relapse in pa-
tients treated for squamous cell vulvar carcinoma: incidence and prognostic value.
Obstet Gynecol 2002;100(6):1159–67.
[53] Groenen SM, Timmers PJ, Burger CW. Recurrence rate in vulvar carcinoma in rela-
tion to pathological margin distance. Int J Gynecol Cancer 2010;20(5):869–73.
[54] Woelber L, Choschzick M, Eulenburg C, Hager M, Jaenicke F, Gieseking F, et al. Prog-
nostic value of pathological resection margin distance in squamous cell cancer of the
vulva. Ann Surg Oncol 2011;18(13):3811–8.
[55] Faul CM, Mirmow D, Huang O, Gerszten K, Day R, Jones MW. Adjuvant radiation for
vulvar carcinoma: improved local control. Int J Radiat Oncol Biol Phys 1997;38(2):
381–9.
[56] Viswanathan AN, Pinto AP, Schultz D, Berkowitz R, Crum CP. Relationship of margin
status and radiation dose to recurrence in postoperative vulvar carcinoma. Gynecol
Oncol 2013;130(3):545–9.
[57] Verschraegen CF, Benjapibal M, Supakarapongkul W, Levy LB, Ross M, Atkinson EN,
et al. Vulvar melanomas at the M.D. Anderson Cancer Center: 25 years later. Int J
Gynecol Cancer 2001;11(5):359–64.
[58] Mert I, Semaan A, Winer I, Morris RT, Ali-Fehmi R. Vulvar/vaginal melanoma: an up-
dated surveillance epidemiology and end results database review. Comparison with
cutaneous melanoma and significance of racial disparities. Int J Gynecol Oncol 2013;
23(6):1118–25.
[59] Heinzelmann-Schwarz VA, Nixdorf S, Valadan M, Diczbalis M, Olivier J, Otton G, et al.
A clinicopathological review of 33 patients with vulvar melanoma identifies c-KIT as
a prognostic marker. Int J Mol Med 2014;33(4):784–94.
[60] Balch CM, Soong SJ, Bartolucci AA, Urist MM, Karakousis CP, Smith TJ, et al. Efficacy of
an elective regional lymph node dissection of 1 to 4 mm thick melanomas for
patients 60 years of age and younger. Ann Surg 1996;224(3):255–63.
[61] Copeland LJ, Sneige N, Gershenson DM, McGuffee VB, Abdul-Karim F, Rutledge FN.
Bartholin gland carcinoma. Obstet Gynecol 1986;67(6):794–801.
[62] Yang SY, Lee JW, Kim WS, Jung KL, Lee SJ, Lee JH, et al. Adenoid cystic carcinoma of
the Bartholin’s gland: report of two cases and review of the literature. Gynecol Oncol
2006;100(2):422–5.
[63] Wilkinson EJ, Brown HM. Vulvar Paget disease of urothelial origin: a report of three
cases and a proposed classification of vulvar Paget disease. Hum Pathol 2002;33(5):
540–54.
[64] Fanning J, Lambert L, Hale TM, Morris PC, Schuerch C. Paget’s disease of the vulva:
prevalence of associated vulvar adenocarcinoma, invasive Paget’s disease, and recur-
rence after surgical excision. Am J Obstet Gynecol 1999;180(1 Pt 1):24–7.
[65] Black D, Tornos C, Soslow RA, Awtrey CS, Barakat RR, Chi DS. The outcomes of
patients with positive margins after excision for intraepithelial Paget’s disease of
the vulva. Gynecol Oncol 2007;104(3):547–50.
[66] Karam A, Dorigo O. Treatment outcomes in a large cohort of patients with invasive
Extramammary Paget’s disease. Gynecol Oncol 2012;125(2):346–51.
S83N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
FIGO CANCER REPORT 2015
Cancer of the vagina
Neville F. Hacker a
, Patricia J. Eifel b
, Jacobus van der Velden c
a
Gynecologic Oncology Cancer Centre, Royal Hospital for Women, Randwick, Australia
b
Department of Radiation Oncology, M.D. Anderson Cancer Center, Houston, TX, USA
c
Department of Gynecology, Academic Medical Center, Amsterdam, The Netherlands
1. Staging
A description of the staging classification for primary vaginal carci-
noma is detailed in Table 1.
1.1. Anatomy
1.1.1. Primary site
The vagina extends from the hymenal ring upward to the uterine
cervix. Cases should be classified as carcinoma of the vagina when the
primary site of the growth is in the vagina. Tumors present in the vagina
as secondary growths from either genital or extra-genital sites should
be excluded. A growth that has extended to the portio of the cervix
and reached the area of the external os should always be allotted to
carcinoma of the cervix. A growth limited to the urethra should be
classified as carcinoma of the urethra. A tumor involving the vulva
should be classified as carcinoma of the vulva. There should be histologic
verification of the disease.
1.1.2. Nodal stations
The upper two-thirds of the vagina is drained by lymphatics to the
pelvic nodes, with the lymphatics paralleling the course of the uterine
artery and the vaginal artery to the obturator, hypogastric (internal
iliac), and external iliac nodes. The distal third of the vagina drains to
the inguinal-femoral nodes. Some lesions, particularly those involving
the posterior vaginal wall, may drain via pararectal lymphatic channels
to presacral nodes.
1.1.3. Metastatic sites
The most common sites of distant spread include the lungs, liver, and
bony skeleton. The rules for staging are similar to those for carcinoma of
the cervix.
1.1.4. Histopathologic types
Approximately 90% of primary vaginal cancers are squamous
cell carcinomas. Primary adenocarcinomas of the vagina occur, but
are rare.
1.1.5. Histopathologic grades (G)
• GX: Grade cannot be assessed.
• G1: Well differentiated.
• G2: Moderately differentiated.
• G3: Poorly or undifferentiated.
2. Introduction
Carcinomas of the vagina constitute only about 2% of malignant
neoplasms of the female genital tract [1]. The vagina, however, can
be a common site of metastatic gynecological cancer, by either
direct extension of cervical or vulvar tumors, or through lymphatic or
vascular deposits, as seen in endometrial cancer and gestational tropho-
blastic disease, respectively. Metastatic or direct extension of non-
gynecologic tumors to the vagina can also occur from the urinary
bladder, urethra, periurethral glands, rectum, and rarely the breast,
lung, or other sites.
Up to 30% of patients with primary vaginal carcinoma have a history
of in situ or invasive cervical cancer treated at least 5 years earlier
[2–4]. (It is arbitrarily assumed that an invasive squamous cell carcino-
ma occurring in the vagina more than 5 years after an invasive squa-
mous cell carcinoma of the cervix is a new primary cancer.) Some
vaginal cancers are preceded by vaginal intraepithelial neoplasia
(VAIN), although the true malignant potential of VAIN is not known
[5,6]. Prior pelvic radiation has also been considered a possible cause
of vaginal cancer [7,8].
Most vaginal cancers occur in postmenopausal or elderly women [1].
When occurring in younger patients, the disease seems to be etiologi-
cally related to cervical neoplasia, and thus HPV dependent [9].
International Journal of Gynecology and Obstetrics 131 (2015) S84–S87
Table 1
FIGO staging of cancer of the vagina.
FIGO Stage Description
I The carcinoma is limited to the vaginal wall
II The carcinoma has involved the sub-vaginal tissue but has not
extended to the pelvic wall
III The carcinoma has extended to the pelvic wall
IV The carcinoma has extended beyond the true pelvis or has
involved the mucosa of the bladder or rectum; bullous edema
as such does not permit a case to be allotted to Stage IV
IVA Tumor invades bladder and/or rectal mucosa and/or direct
extension beyond the true pelvis
IVB Spread to distant organs
http://dx.doi.org/10.1016/j.ijgo.2015.06.003
0020-7292/© 2015 Published by Elsevier Ireland Ltd. on behalf of International Federation of Gynecology and Obstetrics. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Contents lists available at ScienceDirect
International Journal of Gynecology and Obstetrics
journal homepage: www.elsevier.com/locate/ijgo
3. Screening
Routine screening for vaginal cancer following hysterectomy for
benign disease is not recommended because these women are at ex-
tremely low risk of developing vaginal cancer. Women with a history
of cervical intraepithelial or invasive neoplasia are at increased risk,
but regular cytologic screening gives a low yield. The introduction of pri-
mary HPV testing will allow the screening interval to be increased,
which may make it cost-effective in this group of patients [10].
4. Vaginal intraepithelial neoplasia (VAIN)
For patients with an abnormal pap smear and no gross abnormality,
vaginal colposcopy and use of Lugol’s iodine to stain the vagina are
necessary. Biopsy of colposcopically abnormal areas is necessary,
usually under anesthesia. Excisional biopsy is useful for lesions involv-
ing the vaginal vault, where occult carcinoma may be found in up to
28% of patients with VAIN [11].
Treatment of VAIN must be individualized. Numerous treatments,
ranging from local surgical excision or ablation through to intracavitary
radiotherapy, have been used. Selection of the appropriate treatment is
usually based on a careful study of several factors, including the general
medical condition of the patient, the histology of the lesion, the location
and extent of the disease, as well as the experience and expertise of
the treating medical team. The proximity of the urethra, bladder,
and rectum to the vaginal epithelium is an important factor to be
considered. Damage or injury to these structures can occur with possi-
ble fistula formation, particularly when the patient has had prior pelvic
radiation therapy.
Laser vaporization with a carbon dioxide laser is an effective
treatment for VAIN [12]. This technique generally requires local or
general anesthesia.
The use of topical 5-fluorouracil (5-FU) is a relatively simple ambu-
latory treatment, which does not require anesthesia or complicated
equipment [13]. This approach may be especially valuable for patients
with widespread or multifocal disease, which would require an exten-
sive surgical procedure. Adverse effects are usually minimal, as long as
it is not used more than twice a week.
Imiquimod 5% cream might represent an alternative method of
management in young, HPV-positive women with multifocal high-
grade lesions (VAIN 2/3) [14].
Excisional procedures, either with electrosurgical loops or a scalpel
excision, have also been used to treat VAIN. Surgical excision is particu-
larly appropriate for vault lesions [15]. Total vaginectomy and split-
thickness skin grafting may be occasionally necessary to treat extensive
lesions that involve virtually the entire length of the vaginal tube and
where other conservative methods have been unsuccessful. Level of
Evidence C
5. Invasive carcinoma
Most patients present with painless vaginal bleeding and
discharge, and definitive diagnosis can usually be made by biopsy
of a gross lesion detected on speculum examination. This can
often be done in the office, but may be facilitated by examination
under anesthesia.
5.1. Treatment
Whenever possible, patients should be referred to tertiary referral
units because of the rarity of these lesions and the limited experience
of most practitioners with the specialized techniques used to treat
these cancers effectively. All treatment must be individualized, and
will vary depending on the stage of disease and the site of vaginal in-
volvement. Whenever possible, an effort should be made to maintain
a functional vagina, although the combination of tumor destruction
and treatment effects may cause narrowing or shortening of the vagina,
particularly in elderly women.
5.1.1. Surgery
Surgery has a limited role because of the close proximity of the blad-
der and rectum, but may be useful in the following situations [1,16,17]:
(1) In patients with Stage I disease involving the upper posterior vagina
If the uterus is still in situ, radical hysterectomy, upper
vaginectomy to achieve clearance of at least 1 cm, and pelvic
lymphadenectomy may be performed. If hysterectomy has
been performed previously, radical upper vaginectomy and pel-
vic lymphadenectomy may be appropriate.
(2) In young patients who require radiation therapy
Pretreatment laparotomy or laparoscopy may allow ovarian
transposition, or in selected cases, surgical staging and resection
of any bulky positive lymph nodes.
(3) In selected patients with Stage IVA disease, particularly if a
rectovaginal or vesicovaginal fistula is present
Primary pelvic exenteration may be a suitable treatment option
for selected patients, either combined with pelvic lymphadenec-
tomy or preoperative radiation [2]. Bilateral groin dissection
should be considered in such patients if the lower third of the va-
gina is involved.
(4) In patients with a central recurrence after radiation therapy
Surgery will usually necessitate some type of pelvic exenteration
in such patients. Level of Evidence C
5.1.2. Radiation therapy
Radiation therapy is the treatment of choice for most patients
with vaginal cancer, and usually requires careful integration of external
beam irradiation and intracavitary or interstitial brachytherapy.
External beam and brachytherapy techniques may vary widely,
depending on the precise location of the tumor and its relationship to
critical structures.
Although some authors have advocated treatment with brachyther-
apy alone for small Stage I (or even Stage II) cancers [18–21], a combi-
nation of external beam irradiation and brachytherapy probably
reduces the risk of local–regional recurrence in such cases. For larger
lesions, treatment is started with approximately 45–50 Gy external
radiation to reduce the primary tumor volume and to treat the pelvic
nodes. This is then supplemented with brachytherapy or external beam
boosts to gross disease in the primary site and involved lymph nodes.
There is evidence for improved local control when the dose to the
primary tumor exceeds 70 Gy [19,21]. This is most easily achieved
using brachytherapy if the entire tumor volume can be treated to the
necessary dose without exceeding normal tissue tolerance. Although
brachytherapy is preferred whenever possible, highly conformal exter-
nal beam boosts may achieve more homogeneous coverage of tumor in
selected patients who have massive tumors, or intimate association of
tumor with critical structures, for example the rectovaginal septum.
If the distal one-third of the vagina is involved, the groin nodes
should be treated. Level of Evidence C
There has been limited reported experience with chemoradiation
for vaginal cancer [20–22]. A recent National Cancer Data Base study
of 13 689 patients diagnosed with vaginal cancer from 1998–2011
reported that the use of chemoradiation increased from 20.8% to
59.1% over that period [23]. The median overall survival was longer in
patients receiving chemoradiation compared with radiation alone
(56.2 vs 41.2 months), and use of chemoradiation was an independent
prognostic factor for improved survival. Level of Evidence C
5.2. Prognosis
Although the reported overall five-year survival for vaginal cancer is
only about 52% [1], reports from major centers have indicated five-year
S85N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S84–S87
survival rates comparable to cervical cancer [18–20]. A study of 193
patients from the M.D. Anderson Cancer Center in Houston reported
five-year disease-specific survival rates of 85% for 50 patients with
Stage I disease, 78% for 97 patients with Stage II, and 58% for 46 patients
with Stages III–IVA [22].
6. Special situation
6.1. Adenocarcinoma
Approximately 10% of primary vaginal carcinomas are adenocarci-
nomas, and they may arise in areas of vaginal adenosis in diethylstilbes-
trol (DES) exposed patients, in Wolffian rest elements, periurethral
glands, or foci of endometriosis. DES-related clear cell carcinomas of
the vagina occurred mainly in young women, but as the DES-exposed
cohort is now over 50 years of age, DES-related tumors are now rare.
In a series of 26 patients with non-DES-related vaginal adenocarci-
nomas reported from the M.D. Anderson Cancer Center in 2007, the me-
dian age was 54 years [24].
6.1.1. Treatment
In general, adenocarcinomas are treated in a similar manner to squa-
mous lesions, although greater emphasis should be placed on combined
modality therapy, even for small tumors, because of the greater propen-
sity for local and distant recurrence [24].
6.1.2. Prognosis
Prognosis for DES-related clear cell carcinomas of the vagina is
generally good, with an overall survival of 78% [25]. Survival for non-
DES-related adenocarcinomas is significantly worse than for squamous
cancers. A recent study of 26 such patients from the M.D. Anderson
Cancer Center reported an overall five-year survival of only 34%, with
a higher rate of both local recurrences and distant metastases [24].
6.2. Vaginal melanoma
Malignant melanomas of the vagina are rare, and almost all cases
occur in white women [26,27]. They most commonly occur in the distal
vagina, particularly on the anterior vaginal wall [26,28].
Most are deeply invasive and radical surgery has been the mainstay
of treatment, often involving some type of pelvic exenteration. Recently,
more conservative local excisions have been used, with comparable sur-
vival rates reported [25–29]. This is usually combined with postopera-
tive radiation. Overall five-year survival is about 15% [27]. Level of
Evidence C
6.3. Sarcoma botryoides
Sarcoma botryoides is a highly malignant tumor of the
rhabdomyoblasts. These neoplasms are found in infants and children
and usually present with discharge, bleeding, or a visible mass at
the introitus.
In the past, exenterative surgery was used for these lesions, but
survival was poor. More recently, conservative surgery has been used
in conjunction with preoperative or postoperative chemotherapy
and radiotherapy with significantly improved survival. Most reported
chemotherapeutic experience has been with vincristine, actinomycin
D, and cyclophosphamide (VAC) [30–32].
If the lesion is small and can be resected with organ preservation,
surgery should be the initial approach. For bulkier lesions, preoperative
chemotherapy or localized teletherapy or brachytherapy may be used.
Extended radiotherapy fields are not recommended, as they may
produce significant developmental problems with the bony pelvis by
destroying or interfering with growth centers in these structures.
Conflict of interest
The authors declare that they have no conflicts of interest.
References
[1] Hacker NF. Vaginal Cancer. In: Berek JS, Hacker NF, editors. Berek and Hacker’s
Gynecologic Oncology. 6th ed. Philadelphia: Lippincott Williams and Wilkins;
2015. p. 608–24.
[2] Eddy GL, Marks RD, Miller 3rd MC, Underwood Jr PB. Primary invasive vaginal
carcinoma. Am J Obstet Gynecol 1991;165(2):292–8.
[3] Peters 3rd WA, Kumar NB, Morley GW. Carcinoma of the vagina. Factors influencing
treatment outcome. Cancer 1989;55(4):892–7.
[4] Rubin SC, Young J, Mikuta JJ. Squamous carcinoma of the vagina: treatment,
complications, and long-term follow-up. Gynecol Oncol 1985;20(3):346–53.
[5] Lenehan PM, Meff F, Lickrish GM. Vaginal intraepithelial neoplasia: biologic aspects
and management. Obstet Gynecol 1986;68(3):333–7.
[6] Rome RM, England PG. Management of vaginal intraepithelial neoplasia: a
series of 132 cases with long term follow-up. Int J Gynecol Cancer 2000;10(5):
382–90.
[7] Pride GL, Buchler DA. Carcinoma of vagina 10 or more years following pelvic
irradiation therapy. Am J Obstet Gynecol 1977;127(5):513–8.
[8] Hellman K, Lundell M, Silfversward C, Nilsson B, Hellstrom AC, Frankendal B. Clinical
and histopathological factors related to prognosis in primary squamous cell
carcinoma of the vagina. Int J Gynecol Cancer 2006;16(3):1201–11.
[9] Hellman K, Silfversward C, Nilsson B, Hellstrom AC, Frankendal B, Pettersson
F. Primary carcinoma of the vagina: factors influencing the age at diagnosis.
The Radiumhemmet series 1956–96. Int J Gynecol Cancer 2004;14(3):
491–501.
[10] Orr JM, Barnett JC, Leath 3rd CA. Incidence of subsequent abnormal cytology in
cervical cancer patients completing five-years of post treatment surveillance
without evidence of recurrence. Gynecol Oncol 2011;122(3):501–4.
[11] Hoffman MS, DeCesare SL, Roberts WS, Fiorica JV, Finan MA, Cavanagh D. Upper
vaginectomy for in situ and occult, superficially invasive carcinoma of the vagina.
Am J Obstet Gynecol 1992;166(1 Pt 1):30–3.
[12] Yalcin OT, Rutherford TJ, Chambers SK, Chambers JT, Schwartz PE. Vaginal
intraepithelial neoplasia: treatment by carbon dioxide laser and risk factors for
failure. Eur J Obstet Gynecol Reprod Biol 2003;106(1):64–8.
[13] Krebs HB. Prophylactic topical 5-fluorouracil following treatment of human
papillomavirus-associated lesions of the vulva and vagina. Obstet Gynecol 1986;
68(6):837–41.
[14] Haidopoulos D, Diakomanolis E, Rodolakis A, Voulgaris Z, Vlachos G, Intsaklis A. Can
local application of imiquimod cream be an alternative mode of therapy for patients
with high-grade intraepithelial lesions of the vagina. Int J Gynecol Cancer 2005;
15(5):898–902.
[15] Indermaur MD, Martino MA, Fiorica JV, Roberts WS, Hoffman MS. Upper
vaginectomy for the treatment of vaginal intraepithelial neoplasia. Am J Obstet
Gynecol 2005;193(2):577–81.
[16] Tjalma WA, Monaghan JM, de Barros Lopes A, Naik R, Nordin AJ, Weyler JJ. The role
of surgery in invasive squamous carcinoma of the vagina. Gynecol Oncol 2001;
81(3):360–5.
[17] Ling B, Gao Z, Sun M, Sun F, Zhang A, Zhao W, et al. Laparoscopic radical hysterecto-
my with vaginectomy and reconstruction of vagina in patients with stage 1 primary
vaginal cancer. Gynecol Oncol 2008;109(1):92–6.
[18] Chyle V, Zagars GK, Wheeler JA, Wharton JT, Delclos L. Definitive radiotherapy for
carcinoma of the vagina: outcome and prognostic factors. Int J Radiat Oncol Biol
Phys 1996;35(5):891–905.
[19] Perez CA, Grigsby PW, Garipagaoglu M, Mutch DG, Lockett MA. Factors affecting
long-term outcome of irradiation in carcinoma of the vagina. Int J Radiat Oncol
Biol Phys 1999;44(1):37–45.
[20] Kirkbride P, Fyles A, Rawlings GA, Manchul L, Levin W, Murphy KJ, et al. Carcinoma
of the vagina: experience at the Princess Margaret Hospital (1974–1989). Gynecol
Oncol 1995;56(3):435–43.
[21] Dalrymple JL, Russell AH, Lee SW, Scudder SA, Leiserowitz GS, Kinney WK, et al.
Chemoradiation for primary invasive squamous carcinoma of the vagina. Int J
Gynecol Cancer 2004;14(1):110–7.
[22] Frank SJ, Jhingran A, Levenback C, Eifel PJ. Definitive radiation therapy for squa-
mous cell carcinoma of the vagina. Int J Radiat Oncol Biol Phys 2005;62(1):
138–47.
[23] Rajagopalan MS, Xu KM, Lin JF, Sukumvanich P, Krivak TC, Beriwal S.
Adoption and impact of concurrent chemoradiation therapy for vaginal cancer:
A National Cancer Data Base (NCDB) study. Gynecol Oncol 2014;135(3):
495–502.
[24] Frank SJ, Deavers MT, Jhingran A, Bodurka DC, Eifel PJ. Primary adenocarcinoma of
the vagina not associated with diethylstilbestrol (DES) exposure. Gynecol Oncol
2007;105(2):470–4.
[25] Herbst AL, Scully RE. Adenocarcinoma of the vagina in adolescence. A report of 7
cases including 6 clear-cell carcinomas (so-called mesonephromas). Cancer 1970;
25(4):745–51.
[26] Reid GC, Schmidt RW, Roberts JA, Hopkins MP, Barrett RJ, Morley GW. Primary
melanoma of the vagina: a clinicopathologic analysis. Obstet Gynecol 1989;74(2):
190–9.
[27] Kirschner AN, Kidd EA, Dewees T, Perkins SM. Treatment approach and outcomes of
vaginal melanoma. Int J Gynecol Cancer 2013;23(8):1484–9.
S86 N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S84–S87
[28] Miner TJ, Delgado R, Zeisler J, Busam, Alektiar K, Barakat R, et al. Primary
vaginal melanoma: a critical analysis of therapy. Ann Surg Oncol 2004;11(1):
34–9.
[29] Buchanan DJ, Schlaerth J, Kurosaki T. Primary vaginal melanoma: thirteen year
disease-free survival after wide local excision and review of recent literature. Am J
Obstet Gynecol 1998;178(6):1177–84.
[30] Magné N, Haie-Meder C. Brachytherapy for genital-tract rhabdomyosarcomas in
girls: technical aspects, reports, and perspectives. Lancet Oncol 2007;8(8):
725–9.
[31] Magné N, Oberlin O, Martelli H, Gerbaulet A, Chassagne D, Haie-Meder C. Vulval and
vaginal rhabdomyosarcoma in children: The Institute Gustave Roussy brachytherapy
experience with particular attention on long-term outcomes. Int J Gynecol Cancer
2006;16(Suppl. 3):610 (Abst 0038).
[32] Arndt CA, Donaldson SS, Anderson JR, Andrassy RJ, Laurie F, Link MP, et al. What
constitutes optimal therapy for patients with rhabdomyosarcoma of the female
genital tract. Cancer 2001;91(12):2454–68.
S87N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S84–S87
FIGO CANCER REPORT 2015
Cancer of the cervix uteri
Adriana Bermudez a
, Neerja Bhatla b
, Eric Leung c
a
Gynecologic Oncology Unit, Buenos Aires University Hospital, Buenos Aires, Argentina
b
Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, New Delhi, India
c
Department of Radiation Oncology, Sunnybrook Health Sciences Center, Odette Cancer Centre, Toronto, Ontario, Canada
1. Introduction
Worldwide, cervical cancer is now the fourth most common female
malignancy in both incidence and mortality, following breast, colorectal,
and lung cancers, and results in approximately 527 600 new cases and
265 700 deaths annually [1]. It is the second most commonly diagnosed
cancer and third most common cause of cancer death among females in
low-resource countries. More than 85% of new cases are diagnosed in
economically disadvantaged people. Nearly 90% of cervical cancer
deaths occur in low-resource regions of the world.
1.1. Anatomy
The cervix is the lower aspect of the uterus. It is roughly cylindrical
in shape, projects through the superior-anterior vaginal wall, and
communicates with the vagina through the endocervical canal, which
terminates in the external os located at the top of the vagina. Cancer
of the cervix may originate from the mucosa of the surface of the cervix
or from within the canal. Carcinoma of the uterine cervix grows locally
and may extend in continuity to the uterus and paracervical tissues, and
pelvic organs.
Cervical cancer may spread to regional lymph nodes, and only later
metastasize hematogenously to distant structures. Studies on sentinel
lymph nodes show that the cervix is drained into the following first
echelon nodal stations most commonly: external iliac (43%), obturator
(26%) and parametrial (21%), from where they drain to the common
iliac nodes. From the common iliac nodes, lymph drainage goes to the
para-aortic nodes. The most common sites of distant spread include
the para-aortic, mediastinal and supraclavicular nodes, the lungs, liver,
and skeleton.
2. Staging
FIGO staging is based on clinical examination. The FIGO staging
guidelines were most recently updated in 2009 (Table 1) [2]. Stage 0
is no longer included in the FIGO 2009 staging.
A thorough pelvic examination is mandatory to provide information
for FIGO staging, and this rarely requires anesthesia. When there is
doubt as to which stage a particular cancer should be allocated, the ear-
lier stage is mandatory.
The following examinations are permitted for the determination of
FIGO staging, as indicated by presenting characteristics (see sections
below): palpation, inspection, colposcopy, endocervical curettage, hys-
teroscopy, cystoscopy, proctoscopy, intravenous pyelography, ultra-
sound of the renal tract, and X-ray examination of the lungs and
skeleton. Blood tests should include full blood count, and renal and
liver functions. Syphilis and HIV serology need to be considered, based
on discussion with the patient about risk factors.
2.1. Initial assessment of microinvasive disease
The diagnosis of both Stage IA1 and IA2 should be based on micro-
scopic examination of removed tissue, preferably a cone biopsy, which
must include the entire lesion. The depth of invasion should not be
greater than 5 mm taken from the base of the epithelium, either surface
or glandular, from which it originates. The second dimension, the hori-
zontal spread, must not exceed 7 mm. Vascular space involvement,
either venous or lymphatic, should not alter the staging, but should be
specifically recorded because it may affect treatment decisions. Macro-
scopically obvious lesions, and those with larger dimensions, should
be staged as IB. It is impossible to clinically determine if a cancer of
the cervix has extended to the corpus. Extension to the corpus should
therefore be disregarded for staging purposes.
The diagnosis of Stage IA1 or IA2 disease can only be made on the
basis of a cone biopsy with negative margins, or on a trachelectomy or
hysterectomy specimen. If the margins of the cone biopsy are positive
for cervical intraepithelial neoplasia (CIN) III or invasive cancer, a sec-
ond cone biopsy should be performed or the patient treated as for
Stage IB1 disease [3].
2.2. Initial evaluation of grossly invasive disease
Visible lesions require a biopsy to confirm a diagnosis of cervical
carcinoma. A patient with a growth apparently fixed to the pelvic wall
by a short and indurated, but not nodular, parametrium should be
allotted to Stage IIB. Stage III should be defined for cases where the
parametrium is nodular to the pelvic wall or if the growth itself extends
to the pelvic wall. The presence of hydronephrosis or non-functioning
kidney(s) resulting from obstruction of the ureter(s) by cancer also per-
mits a case to be allotted to Stage III.
International Journal of Gynecology and Obstetrics 131 (2015) S88–S95
http://dx.doi.org/10.1016/j.ijgo.2015.06.004
0020-7292/© 2015 Published by Elsevier Ireland Ltd. on behalf of International Federation of Gynecology and Obstetrics. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Contents lists available at ScienceDirect
International Journal of Gynecology and Obstetrics
journal homepage: www.elsevier.com/locate/ijgo
In cases of grossly invasive disease, a chest X-ray, and evaluation of
hydronephrosis (with renal ultrasound, intravenous pyelography, CT,
or MRI) are mandatory. The bladder and rectum are evaluated by cys-
toscopy and sigmoidoscopy only if the patient is clinically symptomatic.
Cystoscopy is also recommended in cases of endocervical growth with
a barrel-shaped surface and in cases where the growth has extended
to the anterior vaginal wall. Suspected bladder or rectal involvement
should be confirmed by biopsy and histologic evidence. The presence
of bullous edema, as such, should not permit a case to be allotted to
Stage IV.
Imaging evaluation may be of additional benefit to clinical examina-
tion in practice areas where resources allow. Imaging may allow for
identification of additional prognostic factors and help direct selection
of therapy. MRI provides the best radiologic assessment of primary
tumors greater than 10 mm, but is not mandatory [4–8]. Level of
Evidence B
CT and/or MRI and/or positron emission tomography (PET) may
provide information on nodal status or systemic spread, but are not
mandatory. Compared with CT and MRI, PET-CT is a more accurate im-
aging method for detecting nodal metastasis that are greater than
10 mm [5,9–12]. Isolated and unexpected areas of PET enhancement
should be further investigated with tissue diagnosis, if possible, to con-
firm or exclude the presence of distant metastatic disease [11,13,14].
Level of Evidence B
Compared with radiologic evaluation, surgical node dissection is
more accurate for assessment of para-aortic nodal disease [15,16]. In pa-
tients with advanced disease, laparoscopic staging of para-aortic lymph
nodes may be considered to allow treatment according to extent of
disease [17]. No impact on survival has been demonstrated; however,
surgical exclusion of para-aortic lymph node involvement portends a
better prognosis than radiographic exclusion alone [18]. Level of
Evidence B
In a review of 22 articles that evaluated the safety and impact of pre-
treatment surgical para-aortic lymph node staging (PALNS), para-aortic
lymph node metastases were found in 18% (range, 8%−42%) of
patients with cervical cancer Stage IB-IVA [19]. The mean complication
rate of PALNS was 9% (range, 4%−24%), the most common complication
being lymphocysts. PET-CT appears to be the most accurate imaging
method, with false-negative results in 4%−15% of cases. Positive para-
aortic nodes have been identified in up to 35% of Stage IIB and 20% of
Stage III tumors [19]. Knowing the status of para-aortic nodes may pro-
vide prognostic information as well as guide the extent of adjuvant or
primary radiation. It is, however, controversial and is not recommended
as a routine practice, particularly in resource-restricted environments
and in women with advanced disease associated with constitutional
symptoms.
2.3. Pathologic staging
In cases treated by surgical procedures, the pathologist’s findings in
the removed tissues can be the basis for accurate statements on the ex-
tent of disease. The findings should not be allowed to change the clinical
staging, but should be recorded in the manner described for the patho-
logic staging of disease. The TNM nomenclature is appropriate for this
purpose [20]. Unlike FIGO staging criteria, TNM staging accounts for
node positivity; however, the FIGO and TNM classifications are other-
wise virtually identical in describing the anatomical extent of disease.
Clinical staging is essential to select and evaluate therapy, while the
pathological stage provides the most precise data from which to esti-
mate prognosis and calculate end results.
Infrequently, hysterectomy may be carried out in the presence of un-
suspected invasive cervical carcinoma. Such cases cannot be clinically
staged or included in therapeutic statistics, but it is desirable that they
be reported separately. If considered appropriate, some of these patients
may be offered repeat laparotomy with full parametrectomy and pelvic
lymphadenectomy to allow potentially curative surgery and/or deter-
mine the need for adjuvant chemoradiation [21].
Staging is determined at the time of the primary diagnosis and can-
not be altered, even at recurrence. Only if the rules for clinical staging
are strictly observed is it possible to compare results among clinics
and by differing modes of therapy.
2.4. Histopathology
All tumors must be microscopically verified. Cases should be clas-
sified as carcinomas of the cervix if the primary growth is in the
cervix. All histologic types must be included. The histopathologic
types are:
• Squamous cell carcinoma (keratinizing; non-keratinizing; verrucous).
• Endometrioid adenocarcinoma.
• Clear cell adenocarcinoma.
• Adenosquamous carcinoma.
• Adenoid cystic carcinoma.
• Small cell carcinoma.
• Undifferentiated carcinoma.
Table 1
Cancer of the cervix uteri.
Stage Description
I The carcinoma is strictly confined to the cervix (extension to the uterine corpus should be disregarded).
IA Invasive cancer identified only microscopically. (All gross lesions even with superficial invasion are Stage IB cancers.) Invasion is limited to
measured stromal invasion with a maximum depth of 5 mm and no wider than 7 mm.
IA1 Measured invasion of stroma ≤ 3 mm in depth and ≤ 7 mm width.
IA2 Measured invasion of stroma N 3 mm and b 5 mm in depth and ≤ 7 mm width.
IB Clinical lesions confined to the cervix, or preclinical lesions greater than stage IA.
IB1 Clinical lesions no greater than 4 cm in size.
IB2 Clinical lesions N 4 cm in size.
II The carcinoma extends beyond the uterus, but has not extended onto the pelvic wall or to the lower third of vagina.
IIA Involvement of up to the upper 2/3 of the vagina. No obvious parametrial involvement.
IIA1 Clinically visible lesion ≤ 4 cm
IIA2 Clinically visible lesion N 4 cm
IIB Obvious parametrial involvement but not onto the pelvic sidewall.
III The carcinoma has extended onto the pelvic sidewall. On rectal examination, there is no cancer free space between the tumor and pelvic sidewall.
The tumor involves the lower third of the vagina. All cases of hydronephrosis or non-functioning kidney should be included unless they are known
to be due to other causes.
IIIA Involvement of the lower vagina but no extension onto pelvic sidewall.
IIIB Extension onto the pelvic sidewall, or hydronephrosis/non-functioning kidney.
IV The carcinoma has extended beyond the true pelvis or has clinically involved the mucosa of the bladder and/or rectum.
IVA Spread to adjacent pelvic organs.
IVB Spread to distant organs.
S89A. Bermudez et al. / International Journal of Gynecology and Obstetrics 131 (2015) S88–S95
Grading by any of several methods is encouraged, but is not a basis
for modifying the stage groupings. Histopathologic grades are as follows:
• GX: Grade cannot be assessed.
• G1: Well differentiated.
• G2: Moderately differentiated.
• G3: Poorly or undifferentiated.
When surgery is the primary treatment, the histologic findings per-
mit the case to have pathologic staging, as described above. In this situ-
ation, the TNM nomenclature may be used.
3. Cervical cancer screening
Primary prevention of cervical cancer through HPV vaccination of
girls, and secondary prevention through the detection of cervical cancer
precursors by various screening methods and their appropriate
treatment, are both known to be effective preventive measures.
Details on cervical cancer screening can be accessed via the FIGO
website (www.figo.org).
4. Management of cervical cancer
4.1. Microinvasion
4.1.1. Stage IA1
Conization is the treatment of choice for this stage. If the patient has
completed childbearing, hysterectomy (abdominal, vaginal, or laparo-
scopic) may be considered [22].
Follow-up with Pap smears every 3 months for 2 years, and
then every 6 months for a further 3 years should be performed. If
follow-up is normal at 5 years, the screening schedule may be complet-
ed according to the recommendations in each country [23,24]. Level of
Evidence C
4.1.2. Stage IA2
Since lymph nodes may be involved in this stage, lymphadenectomy
is necessary [25,26]. The recommended treatment is type 2 radical
hysterectomy (ligation of the uterine artery where it crosses the ureter,
although a vaginal cuff is not necessary) with pelvic lymphadenectomy.
If fertility is desired, options are: (1) cervical conization with extra-
peritoneal or laparoscopic pelvic lymphadenectomy; or (2) radical
abdominal, vaginal, or endoscopic trachelectomy with pelvic lymphad-
enectomy performed according to the surgical approach [27,28].
4.1.3. Post-treatment follow-up after microinvasive carcinoma
Follow-up with Pap smears every 3 months for 2 years, and
then every 6 months for a further 3 years should be performed. If
follow-up is normal at 5 years, the screening schedule may be complet-
ed according to the recommendations in each country [23,24]. Level of
Evidence C
4.2. Grossly invasive cervical carcinoma (FIGO Stage IB–IVA)
Concurrent platinum-based chemoradiation is the most indicated
treatment for this stage although neoadjuvant chemotherapy may
play a role in selected settings [29]. The treatment approach should be
decided based on the availability of resources, and tumor- and
patient-related factors.
4.2.1. Surgical management
Surgical treatment may be indicated in Stage IB1–IIA1 disease: mod-
ified radical or radical (abdominal or endoscopic) hysterectomy with
pelvic lymphadenectomy [30–32]. Level of Evidence B
Primary pelvic exenteration may be considered for Stage IVA disease
without extension to the pelvic sidewall or extra-pelvic disease [33–46].
Level of Evidence C
4.2.1.1. Sentinel lymph node assessment
Identification of sentinel lymph nodes can be performed with dual
labeling using blue dye and radiocolloid [47–49]. These procedures
may be considered in early stage cervical cancer, Stage IA and IB1
[50–52].
If lymphovascular space invasion is present, pelvic lymphadenecto-
my needs to be considered. Level of Evidence C
Sentinel lymph node assessment of pelvic lymph nodes should not
be utilized in advanced disease [53].
4.2.1.2. Trend to lesser surgery for small tumors
Stages IA2–IB1 with tumor size of less than 2 cm, cervical stromal in-
vasion of less than 50%, and node negative on MR/CT imaging have been
considered as low risk.
Simple hysterectomy or trachelectomy, with either pelvic lymphad-
enectomy or sentinel lymph node assessment, have been considered
as adequate surgical treatment for low-risk cases [54,55]. Level of
Evidence D
4.2.1.3. Adjuvant radiation/chemotherapy
The risk of recurrence after radical surgery is increased in the pres-
ence of positive nodes, positive parametria, or positive surgical margins.
Adjuvant concurrent chemoradiation (cisplatin with or without 5-
fluorouracil) improves overall survival, progression-free survival, and
both local and distant recurrences compared with pelvic irradiation
alone in such patients [42]. Level of Evidence B
Risk of pelvic recurrence is also increased in those with uninvolved
nodes but with primary associated risk factors: tumor size greater
than 4 cm, capillary-like space (CLS) involvement, and outer one-third
invasion of the cervical stroma [43,44]. Adjuvant whole pelvic chemo-
irradiation reduces the local failure rate and improves progression-
free survival compared with patients treated with surgery alone [43].
Level of Evidence B
Adjuvant radiation therapy with and without chemotherapy
may be particularly beneficial for patients with adenocarcinoma or
adenosquamous histology, given the relatively higher rates of distant
failure [42,43]. Level of Evidence C
Patients with positive common iliac or para-aortic nodes may be
treated by extended field radiation [56,57], with or without chemother-
apy. Level of Evidence C
Intensity modulated radiation therapy has been explored in the
postoperative setting. A prospective multi-institutional study has
shown acceptable toxicities with this approach and a randomized trial
(TIME-C) is underway comparing intensity-modulated radiation thera-
py (IMRT) with standard field-based radiation therapy in postoperative
cervical and endometrial cancer [58,59]. Although there is currently
insufficient evidence at the present time to recommend IMRT as a stan-
dard of care, many centers have shifted to using this technique in post-
operative cervical and endometrial cancer treatment.
4.2.2. Neoadjuvant chemotherapy and surgery
The theoretical rationale for the use of neoadjuvant chemotherapy
(NACT) includes the induction of tumor shrinkage to facilitate radical
excision, and a possible improvement in outcomes over surgery alone.
There is also a possibility of NACT sterilizing nodes and parametria,
thereby reducing risk factors for adjuvant therapy after surgery; howev-
er, the efficacy of neoadjuvant therapy in this situation is not known.
A meta-analysis of randomized trials of neoadjuvant platinum-
based chemotherapy prior to definitive surgery shows that patients
treated with NACT have better survival outcomes than those treated
with primary radiation alone, given at a relatively low dose [60]. No ran-
domized data compare the results of NACT followed by surgery with
concurrent chemoradiation. The European Organization for Research
and Treatment of Cancer is currently conducting a Phase 3 study com-
paring NACT and surgery with definitive chemoradiation in patients
with FIGO Stages IB2, IIA2, or IIB cervical cancers.
S90 A. Bermudez et al. / International Journal of Gynecology and Obstetrics 131 (2015) S88–S95
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment
FIGO Cancer Report 2015 highlights cancer staging and treatment

More Related Content

Similar to FIGO Cancer Report 2015 highlights cancer staging and treatment

WhatsApp +16575292372 original IELTS certificate, IELTS certificate
 WhatsApp +16575292372 original IELTS certificate, IELTS certificate  WhatsApp +16575292372 original IELTS certificate, IELTS certificate
WhatsApp +16575292372 original IELTS certificate, IELTS certificate IELTS Indicator | Take IELTS
 
WhatsApp +16575292372 Buy Telc Goethe IELTS certificate
WhatsApp +16575292372 Buy Telc Goethe IELTS certificate WhatsApp +16575292372 Buy Telc Goethe IELTS certificate
WhatsApp +16575292372 Buy Telc Goethe IELTS certificate IELTS Indicator | Take IELTS
 
WhatsApp +16575292372 Verified Telc Ielts Certificate without exam
 WhatsApp +16575292372 Verified Telc Ielts Certificate without exam WhatsApp +16575292372 Verified Telc Ielts Certificate without exam
WhatsApp +16575292372 Verified Telc Ielts Certificate without examIELTS Indicator | Take IELTS
 
WhatsApp +16575292372 Buy Telc Goethe ielts toefl pte gre gmat
 WhatsApp +16575292372 Buy Telc Goethe ielts toefl pte gre gmat  WhatsApp +16575292372 Buy Telc Goethe ielts toefl pte gre gmat
WhatsApp +16575292372 Buy Telc Goethe ielts toefl pte gre gmat IELTS Indicator | Take IELTS
 
WhatsApp +16575292372 How to Buy PMP certificates Exam in Saudi Arabia
WhatsApp +16575292372 How to Buy PMP certificates Exam in Saudi ArabiaWhatsApp +16575292372 How to Buy PMP certificates Exam in Saudi Arabia
WhatsApp +16575292372 How to Buy PMP certificates Exam in Saudi ArabiaIELTS Indicator | Take IELTS
 
WhatsApp +16575292372 Registered Telc TOFEL certificate for sale Switzerland
WhatsApp +16575292372 Registered Telc TOFEL certificate for sale SwitzerlandWhatsApp +16575292372 Registered Telc TOFEL certificate for sale Switzerland
WhatsApp +16575292372 Registered Telc TOFEL certificate for sale SwitzerlandIELTS Indicator | Take IELTS
 
WhatsApp +16575292372 ielts certificate online without exams
WhatsApp +16575292372 ielts certificate online without examsWhatsApp +16575292372 ielts certificate online without exams
WhatsApp +16575292372 ielts certificate online without examsIELTS Indicator | Take IELTS
 
WhatsApp +16575292372 Ielts Certificate | Verified Ielts Certificate
WhatsApp +16575292372 Ielts Certificate | Verified Ielts Certificate WhatsApp +16575292372 Ielts Certificate | Verified Ielts Certificate
WhatsApp +16575292372 Ielts Certificate | Verified Ielts Certificate IELTS Indicator | Take IELTS
 
A framework for malaria elimination.pdf
A framework for malaria elimination.pdfA framework for malaria elimination.pdf
A framework for malaria elimination.pdfEdsonFidelis5
 
Minimally Invasive Orthopaedic Trauma.pdf
Minimally Invasive Orthopaedic Trauma.pdfMinimally Invasive Orthopaedic Trauma.pdf
Minimally Invasive Orthopaedic Trauma.pdfthunmnh1
 
Cell Phones And Brain Cancer
Cell Phones And Brain CancerCell Phones And Brain Cancer
Cell Phones And Brain CancerDocJess
 
Since January Elsevier has created a COVID-19 resource center with...
Since January Elsevier has created a COVID-19 resource center with...Since January Elsevier has created a COVID-19 resource center with...
Since January Elsevier has created a COVID-19 resource center with...Valentina Corona
 
Журнал Medical Physics International, часть 1, том 2 (издатель - IOMP, Междун...
Журнал Medical Physics International, часть 1, том 2 (издатель - IOMP, Междун...Журнал Medical Physics International, часть 1, том 2 (издатель - IOMP, Междун...
Журнал Medical Physics International, часть 1, том 2 (издатель - IOMP, Междун...oncoportal.net
 
The ICU Ultrasound Pocket Book - guia rápido
The ICU Ultrasound Pocket Book - guia rápidoThe ICU Ultrasound Pocket Book - guia rápido
The ICU Ultrasound Pocket Book - guia rápidoYasminBittar
 
Continuous Update Project Overview (Conference: Diet and cancer: from prevent...
Continuous Update Project Overview (Conference: Diet and cancer: from prevent...Continuous Update Project Overview (Conference: Diet and cancer: from prevent...
Continuous Update Project Overview (Conference: Diet and cancer: from prevent...World Cancer Research Fund International
 
Workshop on Principles of Cancer Epidemiology 2018 Brochure - Christian Medic...
Workshop on Principles of Cancer Epidemiology 2018 Brochure - Christian Medic...Workshop on Principles of Cancer Epidemiology 2018 Brochure - Christian Medic...
Workshop on Principles of Cancer Epidemiology 2018 Brochure - Christian Medic...Ahmad Ozair
 

Similar to FIGO Cancer Report 2015 highlights cancer staging and treatment (20)

WhatsApp +16575292372 original IELTS certificate, IELTS certificate
 WhatsApp +16575292372 original IELTS certificate, IELTS certificate  WhatsApp +16575292372 original IELTS certificate, IELTS certificate
WhatsApp +16575292372 original IELTS certificate, IELTS certificate
 
WhatsApp +16575292372 Buy Telc Goethe IELTS certificate
WhatsApp +16575292372 Buy Telc Goethe IELTS certificate WhatsApp +16575292372 Buy Telc Goethe IELTS certificate
WhatsApp +16575292372 Buy Telc Goethe IELTS certificate
 
WhatsApp +16575292372 Verified Telc Ielts Certificate without exam
 WhatsApp +16575292372 Verified Telc Ielts Certificate without exam WhatsApp +16575292372 Verified Telc Ielts Certificate without exam
WhatsApp +16575292372 Verified Telc Ielts Certificate without exam
 
WhatsApp +16575292372 Buy Telc Goethe ielts toefl pte gre gmat
 WhatsApp +16575292372 Buy Telc Goethe ielts toefl pte gre gmat  WhatsApp +16575292372 Buy Telc Goethe ielts toefl pte gre gmat
WhatsApp +16575292372 Buy Telc Goethe ielts toefl pte gre gmat
 
WhatsApp +16575292372 How to Buy PMP certificates Exam in Saudi Arabia
WhatsApp +16575292372 How to Buy PMP certificates Exam in Saudi ArabiaWhatsApp +16575292372 How to Buy PMP certificates Exam in Saudi Arabia
WhatsApp +16575292372 How to Buy PMP certificates Exam in Saudi Arabia
 
WhatsApp +16575292372 Registered Telc TOFEL certificate for sale Switzerland
WhatsApp +16575292372 Registered Telc TOFEL certificate for sale SwitzerlandWhatsApp +16575292372 Registered Telc TOFEL certificate for sale Switzerland
WhatsApp +16575292372 Registered Telc TOFEL certificate for sale Switzerland
 
WhatsApp +16575292372 Buy PMP certificate online
WhatsApp +16575292372 Buy PMP certificate onlineWhatsApp +16575292372 Buy PMP certificate online
WhatsApp +16575292372 Buy PMP certificate online
 
WhatsApp +16575292372 ielts certificate online without exams
WhatsApp +16575292372 ielts certificate online without examsWhatsApp +16575292372 ielts certificate online without exams
WhatsApp +16575292372 ielts certificate online without exams
 
WhatsApp +16575292372 Ielts Certificate | Verified Ielts Certificate
WhatsApp +16575292372 Ielts Certificate | Verified Ielts Certificate WhatsApp +16575292372 Ielts Certificate | Verified Ielts Certificate
WhatsApp +16575292372 Ielts Certificate | Verified Ielts Certificate
 
Cranial nerves
Cranial nerves  Cranial nerves
Cranial nerves
 
HELSINKI DECLARATION ON PATIENT SAFETY IN ANAESTHESIOLOGY
HELSINKI DECLARATION ON PATIENT SAFETY IN ANAESTHESIOLOGYHELSINKI DECLARATION ON PATIENT SAFETY IN ANAESTHESIOLOGY
HELSINKI DECLARATION ON PATIENT SAFETY IN ANAESTHESIOLOGY
 
A framework for malaria elimination.pdf
A framework for malaria elimination.pdfA framework for malaria elimination.pdf
A framework for malaria elimination.pdf
 
9241541792 eng
9241541792 eng9241541792 eng
9241541792 eng
 
Minimally Invasive Orthopaedic Trauma.pdf
Minimally Invasive Orthopaedic Trauma.pdfMinimally Invasive Orthopaedic Trauma.pdf
Minimally Invasive Orthopaedic Trauma.pdf
 
Cell Phones And Brain Cancer
Cell Phones And Brain CancerCell Phones And Brain Cancer
Cell Phones And Brain Cancer
 
Since January Elsevier has created a COVID-19 resource center with...
Since January Elsevier has created a COVID-19 resource center with...Since January Elsevier has created a COVID-19 resource center with...
Since January Elsevier has created a COVID-19 resource center with...
 
Журнал Medical Physics International, часть 1, том 2 (издатель - IOMP, Междун...
Журнал Medical Physics International, часть 1, том 2 (издатель - IOMP, Междун...Журнал Medical Physics International, часть 1, том 2 (издатель - IOMP, Междун...
Журнал Medical Physics International, часть 1, том 2 (издатель - IOMP, Междун...
 
The ICU Ultrasound Pocket Book - guia rápido
The ICU Ultrasound Pocket Book - guia rápidoThe ICU Ultrasound Pocket Book - guia rápido
The ICU Ultrasound Pocket Book - guia rápido
 
Continuous Update Project Overview (Conference: Diet and cancer: from prevent...
Continuous Update Project Overview (Conference: Diet and cancer: from prevent...Continuous Update Project Overview (Conference: Diet and cancer: from prevent...
Continuous Update Project Overview (Conference: Diet and cancer: from prevent...
 
Workshop on Principles of Cancer Epidemiology 2018 Brochure - Christian Medic...
Workshop on Principles of Cancer Epidemiology 2018 Brochure - Christian Medic...Workshop on Principles of Cancer Epidemiology 2018 Brochure - Christian Medic...
Workshop on Principles of Cancer Epidemiology 2018 Brochure - Christian Medic...
 

Recently uploaded

Beyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactBeyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactPECB
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13Steve Thomason
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdfQucHHunhnh
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityGeoBlogs
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxGaneshChakor2
 
Russian Call Girls in Andheri Airport Mumbai WhatsApp 9167673311 💞 Full Nigh...
Russian Call Girls in Andheri Airport Mumbai WhatsApp  9167673311 💞 Full Nigh...Russian Call Girls in Andheri Airport Mumbai WhatsApp  9167673311 💞 Full Nigh...
Russian Call Girls in Andheri Airport Mumbai WhatsApp 9167673311 💞 Full Nigh...Pooja Nehwal
 
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxPOINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxSayali Powar
 
BAG TECHNIQUE Bag technique-a tool making use of public health bag through wh...
BAG TECHNIQUE Bag technique-a tool making use of public health bag through wh...BAG TECHNIQUE Bag technique-a tool making use of public health bag through wh...
BAG TECHNIQUE Bag technique-a tool making use of public health bag through wh...Sapna Thakur
 
1029 - Danh muc Sach Giao Khoa 10 . pdf
1029 -  Danh muc Sach Giao Khoa 10 . pdf1029 -  Danh muc Sach Giao Khoa 10 . pdf
1029 - Danh muc Sach Giao Khoa 10 . pdfQucHHunhnh
 
Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3JemimahLaneBuaron
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)eniolaolutunde
 
Introduction to Nonprofit Accounting: The Basics
Introduction to Nonprofit Accounting: The BasicsIntroduction to Nonprofit Accounting: The Basics
Introduction to Nonprofit Accounting: The BasicsTechSoup
 
Measures of Dispersion and Variability: Range, QD, AD and SD
Measures of Dispersion and Variability: Range, QD, AD and SDMeasures of Dispersion and Variability: Range, QD, AD and SD
Measures of Dispersion and Variability: Range, QD, AD and SDThiyagu K
 
microwave assisted reaction. General introduction
microwave assisted reaction. General introductionmicrowave assisted reaction. General introduction
microwave assisted reaction. General introductionMaksud Ahmed
 
Disha NEET Physics Guide for classes 11 and 12.pdf
Disha NEET Physics Guide for classes 11 and 12.pdfDisha NEET Physics Guide for classes 11 and 12.pdf
Disha NEET Physics Guide for classes 11 and 12.pdfchloefrazer622
 
Activity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdfActivity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdfciinovamais
 
Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17Celine George
 
APM Welcome, APM North West Network Conference, Synergies Across Sectors
APM Welcome, APM North West Network Conference, Synergies Across SectorsAPM Welcome, APM North West Network Conference, Synergies Across Sectors
APM Welcome, APM North West Network Conference, Synergies Across SectorsAssociation for Project Management
 

Recently uploaded (20)

Beyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactBeyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global Impact
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdf
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activity
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptx
 
Russian Call Girls in Andheri Airport Mumbai WhatsApp 9167673311 💞 Full Nigh...
Russian Call Girls in Andheri Airport Mumbai WhatsApp  9167673311 💞 Full Nigh...Russian Call Girls in Andheri Airport Mumbai WhatsApp  9167673311 💞 Full Nigh...
Russian Call Girls in Andheri Airport Mumbai WhatsApp 9167673311 💞 Full Nigh...
 
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptxPOINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
POINT- BIOCHEMISTRY SEM 2 ENZYMES UNIT 5.pptx
 
BAG TECHNIQUE Bag technique-a tool making use of public health bag through wh...
BAG TECHNIQUE Bag technique-a tool making use of public health bag through wh...BAG TECHNIQUE Bag technique-a tool making use of public health bag through wh...
BAG TECHNIQUE Bag technique-a tool making use of public health bag through wh...
 
1029 - Danh muc Sach Giao Khoa 10 . pdf
1029 -  Danh muc Sach Giao Khoa 10 . pdf1029 -  Danh muc Sach Giao Khoa 10 . pdf
1029 - Danh muc Sach Giao Khoa 10 . pdf
 
Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3Q4-W6-Restating Informational Text Grade 3
Q4-W6-Restating Informational Text Grade 3
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)
 
Introduction to Nonprofit Accounting: The Basics
Introduction to Nonprofit Accounting: The BasicsIntroduction to Nonprofit Accounting: The Basics
Introduction to Nonprofit Accounting: The Basics
 
INDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptx
INDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptxINDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptx
INDIA QUIZ 2024 RLAC DELHI UNIVERSITY.pptx
 
Measures of Dispersion and Variability: Range, QD, AD and SD
Measures of Dispersion and Variability: Range, QD, AD and SDMeasures of Dispersion and Variability: Range, QD, AD and SD
Measures of Dispersion and Variability: Range, QD, AD and SD
 
microwave assisted reaction. General introduction
microwave assisted reaction. General introductionmicrowave assisted reaction. General introduction
microwave assisted reaction. General introduction
 
Disha NEET Physics Guide for classes 11 and 12.pdf
Disha NEET Physics Guide for classes 11 and 12.pdfDisha NEET Physics Guide for classes 11 and 12.pdf
Disha NEET Physics Guide for classes 11 and 12.pdf
 
Activity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdfActivity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdf
 
Mattingly "AI & Prompt Design: Structured Data, Assistants, & RAG"
Mattingly "AI & Prompt Design: Structured Data, Assistants, & RAG"Mattingly "AI & Prompt Design: Structured Data, Assistants, & RAG"
Mattingly "AI & Prompt Design: Structured Data, Assistants, & RAG"
 
Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17
 
APM Welcome, APM North West Network Conference, Synergies Across Sectors
APM Welcome, APM North West Network Conference, Synergies Across SectorsAPM Welcome, APM North West Network Conference, Synergies Across Sectors
APM Welcome, APM North West Network Conference, Synergies Across Sectors
 

FIGO Cancer Report 2015 highlights cancer staging and treatment

  • 1. 4 InternationalJournalofGynecology&ObstetricsVol.131(2015)S75–S172 131 S2 ELSEVIER Official publication of FIGO The International Federation of Gynecology and Obstetrics FIGO Cancer Report 2015 Guest Editors: Lynette Denny, Michael Quinn OBSTETRICS GYNECOLOGY International Journal of Volume 131 Supplement 2 October 2015 ISSN 0020-7292
  • 2. International Journal of GYNECOLOGY & OBSTETRICS Volume 131, Supplement 2 (2015) Amsterdam • Boston • London • New York • Oxford • Paris • Philadelphia • San Diego • St. Louis
  • 3. © 2015 Published by Elsevier Ireland Ltd. on behalf of International Federation of Gynecology and Obstetrics. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/ licenses/by-nc-nd/4.0/) This journal and the individual contributions contained in it are protected under copyright by International Federation of Gynecology and Obstetrics, and the following terms and conditions apply to their use: Photocopying Single photocopies of single articles may be made for personal use as allowed by national copyright laws. Permission of the Publisher and payment of a fee is required for all other photocopying, including multiple or systematic copying, copy-ing for advertising or promotional purposes, resale, and all forms of document delivery. Special rates are available for educational institutions that wish to make photocopies for non-profit educational classroom use. For information on how to seek permission visit www.elsevier.com/permissions or call: (+44) 1865 843830 (UK)/(+1) 215 239 3804 (USA). Derivative Works Subscribers may reproduce tables of contents or prepare lists of articles including abstracts for internal circulation within their institutions. Permission of the Publisher is required for resale or distribution outside the institution. Permission of the Publisher is required for all other derivative works, including compilations and translations (please consult www.elsevier.com/permissions). Electronic Storage or Usage Permission of the Publisher is required to store or use electronically any material contained in this journal, including any article or part of an article (please consult www.elsevier.com/permissions). Except as outlined above, no part of this publication may be reproduced, stored in a retrieval system or transmitted in any form or by any means, electronic, mechanical, photocopying, recording or otherwise, without prior written permission of the Publisher. Notice No responsibility is assumed by the Publisher for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions or ideas contained in the material herein. Because of rapid advances in the medical sciences, in particular, independent verification of diagnoses and drug dosages should be made. Although all advertising material is expected to conform to ethical (medical) standards, inclusion in this publication does not constitute a guarantee or endorsement of the quality or value of such product or of the claims made of it by its manufacturer. Language (usage and editing services) Please write your text in good English (American or British usage is accepted, but not a mixture of these). Authors who feel their English language manuscript may require editing to eliminate possible grammatical or spelling errors and to conform to correct scientific English may wish to use the English Language Editing service available from Elsevier’s WebShop http://webshop.elsevier.com/languageediting/ or visit our customer support site http://support.elsevier.com for more information. Illustration services Elsevier’s WebShop (http://webshop.elsevier.com/illustrationservices) offers Illustration Services to authors preparing to submit a manuscript but concerned about the quality of the images accompanying their article. Elsevier’s expert illustrators can produce scientific, technical and medical-style images, as well as a full range of charts, tables and graphs. Image ‘polishing’ is also available, where our illustrators take your image(s) and improve them to a professional standard. Please visit the website to find out more. Printed by Henry Ling, Dorchester The paper used in this publication meets the requirements of ANSI/NISO Z39.48-1992 (Permanence of Paper)
  • 4. Supplement to International Journal of Gynecology & Obstetrics Volume 131, Supplement 2 FIGO Cancer Report 2015 Guest Editors: Lynette Denny, Michael Quinn
  • 5. International Federation of Gynecology and Obstetrics (FIGO) Officers President: Vice-President: President-Elect: Past-President: Honorary Treasurer: Honorary Secretary: S. Arulkumaran (UK) E.C. Morales (Mexico) C.N. Purandare (India) G. Serour (Egypt) W. Holzgreve (Germany) G.C. Di Renzo (Italy) FIGO Chief Executive H. Rushwan (Sudan/UK) Executive Board Argentina N.C. Garello Australia & New Zealand C. Tippett Belgium F. Debiève Bolivia C. Fuchtner Brazil N.R. de Melo Canada J. Blake Chile H. Munoz Colombia J.D.VillegasEcheverri Denmark A.T. Pedersen Egypt N.A. Darwish Ethiopia Y.G. Ferede Finland S. Grénman France B. Carbonne Germany W. Jonat Japan T. Kimura Lebanon F. El-Kak Malaysia A.A. Yahya Paraguay A. Acosta South Africa B.D. Goolab Spain J. Laílla Vicens Taiwan T.-H. Su Uruguay J.G. Allonso Tellechea United Kingdom T. Falconer United States of America J.N. Martin International Editions and Collaborations IJGO India Editor-in-Chief: Dr Rohit V. Bhatt (rohit.v.bhatt@gmail.com) Editorial Office: Jaypee Brothers Medical Publishers (P) Ltd 4838/24, Ansari Road, Daryaganj New Delhi 110 002, India E-mail: ijgoindia@jaypeebrothers.com IJGO China Editor-in-Chief: Dr Zhenyu Zhang Department of Ob/Gyn Chaoyang Hospital No. 8 Baijiazhung Rd Chaoyang District Beijing, 100020 China E-mail: zhenyuzhang2000@yahoo.com For information about FIGO: The Secretariat of FIGO is at FIGO House, Suite 3, Waterloo Court, 10 Theed Street, London, SE1 8ST UK. Tel: +44 20 7928-1166 Fax: +44 20 7928-7099 E-mail: figo@figo.org Website: www.figo.org All enquiries concerning FIGO may be sent to the Honorary Secretary at that address.
  • 6. Editor: Editor Emeritus: Associate Editor: Managing Editor: Deputy Managing Editor: Associate Editors Ethical and Legal Issues in Reproductive Health: Enabling Technologies: FIGO Staging of Gynecologic Cancer: Contemporary Issues in Women’s Health: Statistical Consultant: Editorial Office: R.M. Adanu (Ghana) T.R.B. Johnson (USA) J. Sciarra (USA) W. Holzgreve (Germany) P. Serafini (Brazil) J. Fortney (USA) C. Addington (UK) A. Cantor (UK) R. Cook (Canada) B. Dickens (Canada) M. Hammoud (USA) L. Denny (South Africa) V. Boama (Qatar) V. Guinto (Philippines) C. Sosa (Uruguay) A. Vahratian (USA) FIGO Secretariat, FIGO House Suite 3 - Waterloo Court, 10 Theed Street, London, SE1 8ST, UK Tel: +44 20 7928 1166 Fax: +44 20 7928 7099 E-mail: ijgo@figo.org International Journal of GYNECOLOGY & OBSTETRICS
  • 7. International Journal of GYNECOLOGY & OBSTETRICS EDITORIAL L. Denny, M. Quinn South Africa, Australia FIGO Cancer Report 2015 S75 CANCER STAGING AND CLINICAL GUIDELINES N.F. Hacker, P.J. Eifel, J. van der Velden Australia, USA, The Netherlands Cancer of the vulva S76 N.F. Hacker, P.J. Eifel, J. van der Velden Australia, USA, The Netherlands Cancer of the vagina S84 A. Bermudez, N. Bhatla, E. Leung Argentina, India, Canada Cancer of the cervix uteri S88 F. Amant, M.R. Mirza, M. Koskas, C.L. Creutzberg Belgium, Denmark, France, The Netherlands Cancer of the corpus uteri S96 J. Prat, 'N. Mbatani Spain, South Africa Uterine sarcomas S105 J.S. Berek, C. Crum, M. Friedlander USA, Australia Cancer of the ovary, fallopian tube, and peritoneum S111 H.Y.S. Ngan, M.J. Seckl, R.S. Berkowitz, Y. Xiang, F. Golfier, P.K. Sekharan, J.R. Lurain China, UK, USA, France, India Update on the diagnosis and management of gestational trophoblastic disease S123 P.S. Binder, J. Prat, D.G. Mutch USA, Spain The future role of molecular staging in gynecologic cancer S127 CONTENTS Volume 131, Supplement 2, October 2015
  • 8. CANCER THERAPY E.Å. Lundqvist, K. Fujiwara, M. Seoud Sweden, Japan, Lebanon Principles of chemotherapy S146 M. Seoud, E.Å. Lundqvist, K. Fujiwara Lebanon, Sweden, Japan Targeted therapy in gynecologic cancers: Ready for prime time? S150 S.K. Shrivastava, U. Mahantshetty, K. Narayan India, Australia Principles of radiation therapy in low-resource and well-developed settings, with particular reference to cervical cancer S153 PATIENT CARE S.V. Carr Australia Psychosexual health in gynecological cancer S159 K. Weare Australia Rehabilitation after gynecological cancer treatment S164 S.M. Kibel, J.M. Cain South Africa, USA Palliative care in gynecological cancer S167 PATHOLOGY J. Prat Spain Pathology of cancers of the female genital tract S132
  • 9. EDITORIAL FIGO Cancer Report 2015 The FIGO Gynecologic Oncology Committee is pleased to present the second edition of the FIGO Cancer Report. Historically the report was published at each FIGO World Congress as the “Annual Report,” going as far back as 1937, and was composed of survival data from various institutions around the world. Since 2009 the process of collating international incidence and survival data stopped and a new Committee was formed. This Committee initiated the current format of the FIGO Cancer Report and its first edition was presented at the FIGO Congress held in Rome, Italy, in October 2012. The Committee has since established collaboration with the Catalan Institute of Oncology to resurrect the data collection program. This process is now at an advanced stage and a detailed situational analysis of institutions able to provide care to women with gynecological cancers is being conduct- ed as we write this editorial in May 2015. Once the situational analysis is complete the intention is to select 20 or so institutions in low- and middle-income countries to collect incidence, stage, treatment, and survival data. This will give us a global picture of the multiple inequities in cancer care internationally and enable more targeted intervention by organizations such as FIGO. It is our intention to ensure that this, the second edition of the FIGO Cancer Report, is disseminated as widely as possible internationally, and therefore the chapters are available online with Open Access. We are also developing a mobile App of the different criteria for FIGO staging, which will ensure that those with smart phones will have instant access. The chapters contained in the report have either been updated or are new chapters. The new chapters include trophoblastic disease, targeted therapies for gynecological cancer, rehabilitation after treatment for gynecological cancer, psychosexual health in gynecological cancers, palliative care in gynecological cancer, and the future role of molecular staging in gynecological cancer. These six new chapters are added to nine others that include the new staging for ovary, fallopian tube, and peritoneum. Each chapter presents the site specific staging, the rationale behind staging, as well as internationally accepted evidence- based guidelines. All chapters are fully referenced and the quality of the evidence for intervention is graded. The current FIGO Gynecologic Oncology Committee members are: Lynette Denny (Chair), Gynecologic Oncologist, South Africa Michael Quinn (Co-Chair), Gynecologic Oncologist, Australia Sergio Pecorelli (Immediate Past Chair), Gynecologic Oncologist, Italy Adriana Bermudez, Gynecologic Oncologist, Argentina Joanna Cain, Gynecologic Oncologist, USA Keiichi Fujiwara, Gynecologic Oncologist, Japan Neville Hacker, Gynecologic Oncologist, Australia Elisabeth Åvall Lundqvist, Medical Oncologist, Sweden Jaime Prat, Pathologist, Spain Shyam Shrivastava, Radiation Oncologist, India The Committee composition is likely to change during the 2015 FIGO World Congress when the report will be released, as members leave the committee after serving a six-year term. The composition of the new Committee will be announced by the incoming President of FIGO. Lynette Denny Groote Schuur Hospital/University of Cape Town, Cape Town, South Africa E-mail address: lynette.denny@uct.ac.za. Michael Quinn Royal Women’s Hospital and University of Melbourne, Melbourne, Australia International Journal of Gynecology and Obstetrics 131 (2015) S75 http://dx.doi.org/10.1016/j.ijgo.2015.06.024 0020-7292/© 2015 Published by Elsevier Ireland Ltd. on behalf of International Federation of Gynecology and Obstetrics. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Contents lists available at ScienceDirect International Journal of Gynecology and Obstetrics journal homepage: www.elsevier.com/locate/ijgo
  • 10. FIGO CANCER REPORT 2015 Cancer of the vulva Neville F. Hacker a , Patricia J. Eifel b , Jacobus van der Velden c a Gynecologic Oncology Cancer Centre, Royal Hospital for Women, Randwick, Australia b Department of Radiation Oncology, M.D. Anderson Cancer Center, Houston, TX, USA c Department of Gynecology, Academic Medical Center, Amsterdam, The Netherlands 1. Staging 1.1. Anatomy 1.1.1. Primary site Cases should be classified as carcinoma of the vulva when the prima- ry site of growth is in the vulva. Any lesion that involves both the vagina and vulva (i.e. crosses the hymenal ring) should be classified as a carci- noma of the vulva. Tumors that present in the vulva as secondary growths, from either a genital or extragenital site, should be excluded. Malignant melanoma should be reported separately. There must be histologic confirmation of the cancer. 1.1.2. Nodal stations The inguinal and femoral nodes are the first sites of regional spread. 1.1.3. Metastatic sites Patients who have extrapelvic metastases or who have involvement of pelvic lymph nodes (external, hypogastric, obturator, and common iliac) are classified as having stage IVB disease. 1.2. Surgical staging classification The staging system for vulvar cancer has been based on surgical find- ings since 1988. The final diagnosis is dependent upon thorough histo- pathologic evaluation of the operative specimen (vulva and lymph nodes). Various modifications have been made over time, with a subdi- vision of Stage I added in 1994. The FIGO staging of vulvar carcinoma was last changed in 2009 by the FIGO Committee on Gynecologic Oncology [1], to give better prognostic discrimination between stages (Table 1). Table 2 compares the FIGO staging with the Union of Interna- tional Cancer Control “TNM” classification. 1.2.1. Histopathologic types Approximately 80% of cases are squamous cell carcinomas, and many cases, particularly in younger women, are HPV related. Melanomas are the second most common cancer seen in cancer centers, although community-based studies have reported basal cell carcinomas to be the second most common vulvar cancer [2]. The histopathologic types are: • squamous cell carcinoma • melanoma • verrucous carcinoma • Paget’s disease of vulva • adenocarcinoma, not otherwise specified (NOS) • basal cell carcinoma, NOS • Bartholin’s gland carcinoma. 1.2.2. Histopathologic grades (G) • GX: Grade cannot be assessed • G1: Well differentiated • G2: Moderately differentiated • G3: Poorly or undifferentiated. 2. Introduction Carcinoma of the vulva is an uncommon tumor, representing about 4% of gynecologic malignancies. Because of the relatively small experi- ence of individual institutions, randomized trials of therapeutic ap- proaches are uncommon, and most studies are based on retrospective clinicopathologic reviews [3]. It is predominantly a disease of postmenopausal women, with the age-specific incidence increasing with increasing age. Although the external location of the vulva should encourage early presentation, vulvar cancers are often advanced at the time of diagnosis. Most squamous carcinomas occur on the labia majora, but the labia minora, clitoris, and perineum may also be primary sites. Vulvar intraepithelial neoplasia (VIN), a precursor lesion in some cases, tends to occur in younger women and may be associated with similar lesions of the cervix and vagina. A new classification of squa- mous VIN was introduced by the International Society for the Study of Vulvovaginal Disease (ISSVD) in 2004 [4]. The term VIN 1 is no longer used, and VIN 2 and 3 are simply called VIN. There are two types of VIN: (1) VIN, usual type (warty, basaloid, and mixed), which is HPV- related in most cases; and (2) VIN, differentiated type, which is seen particularly in older women, and is often associated with lichen sclerosus and/or squamous hyperplasia. The incidence of VIN, usual type, and the incidence of invasive vulvar cancer in premenopausal women should both decrease significantly with increasing use of HPV vaccination. International Journal of Gynecology and Obstetrics 131 (2015) S76–S83 http://dx.doi.org/10.1016/j.ijgo.2015.06.002 0020-7292/© 2015 Published by Elsevier Ireland Ltd. on behalf of International Federation of Gynecology and Obstetrics. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Contents lists available at ScienceDirect International Journal of Gynecology and Obstetrics journal homepage: www.elsevier.com/locate/ijgo
  • 11. Treatment of vulvar cancer used to be primarily surgical, but radia- tion therapy and, to a lesser extent, chemotherapy have been progres- sively integrated into the treatment protocol over the past 30 years. Therefore, management has evolved into an individualized multidisci- plinary approach, and patients should be referred centrally to a gyneco- logical cancer center where all relevant expertise is available [5,6]. Level of Evidence B 3. Screening There is no screening procedure for vulvar cancer. However, patients with a past history of cervical or vaginal cancer should have inspection of the vulva, with or without colposcopic examination, as part of their regular follow-up. Patients with lichen sclerosus or a past history of VIN should also be kept under regular surveillance, and taught to under- take regular self-examination with a mirror. 4. Squamous cell carcinoma 4.1. Presenting symptoms Vulvar cancer may be asymptomatic, but most patients present with a vulvar lump or ulcer, which may or may not be painful. There is often a long-standing history of pruritus, which may be due to associated vulvar dystrophy. Bleeding or discharge is an occasional presenting symptom, and patients with advanced disease may present with a lump in the groin caused by metastases to groin lymph nodes. 4.2. Diagnosis If the disease appears to be entirely intraepithelial, initial assessment should consist of multiple biopsies to exclude an invasive focus. A 3 or 4 mm Keyes biopsy instrument is ideal for this purpose. Patients with multifocal lesions should have biopsies taken from several lesions. If there appears to be invasive cancer present, a wedge or Keyes bi- opsy under local anesthesia in the office is usually sufficient to confirm the diagnosis. The biopsy should include some underlying stroma. For small tumors, it is preferable not to excise the entire lesion at the time of biopsy because this makes it more difficult to plan the subse- quent definitive resection. If the lesion is 2 cm or less in diameter and depth of stromal inva- sion is less than or equal to 1 mm on the initial biopsy, radical local excision of the lesion must be undertaken to allow serial sectioning to properly assess the depth of invasion. If there is still no focus found with a depth of invasion greater than 1 mm, this excision will also be the definitive treatment [3]. 4.3. Investigations (1) Cervical cytology, if applicable. (2) Colposcopy of the cervix and vagina because of the common as- sociation with other squamous intraepithelial lesions. (3) For large lesions, a CT or MRI scan of the pelvis and groins is help- ful to detect any enlarged lymph nodes in the groins or pelvis, erosion into underlying bony structures, or other evidence of metastatic disease. (4) Routine full blood count, biochemical profile, and chest X-ray preoperatively. 4.4. Clinical practice guidelines The clinical findings should be recorded on a staging diagram (e.g. Fig. 1). The findings according to the staging delineated in Tables 1 and 2 are usually listed on the reverse side of the diagram. 4.5. Treatment 4.5.1. Treatment of vulvar intraepithelial neoplasia Various treatment modalities are available for treating intraepithelial lesions of the vulva [7,8]. Once the diagnosis has been established, super- ficial local excision of the vulvar epithelium with a 0.5–1.0-cm margin is considered adequate for lesions of the lateral aspect of the vulva. Lesions involving the labia minora may also be treated by local excision but may respond favorably to laser vaporization. Laser is also appropriate for clitoral and perianal lesions. Laser treatment of the hair-bearing skin of the vulva will usually produce depigmentation and destruction of hair follicles, with subsequent loss of hair growth. Large lesions may be treated with a skinning vulvectomy and split-thickness skin graft. Level of Evidence C Two randomized controlled trials have shown promising results with the topical immune response modifier, imiquimod, with complete response rates of 35–81% reported [9,10]. Long-term follow-up of pa- tients in one of the studies showed good sustained benefit, although the number of patients in the study was small [9]. Level of Evidence A Table 1 FIGO staging of carcinoma of the vulva. FIGO Stage Description I Tumor confined to the vulva IA Lesions ≤ 2 cm in size, confined to the vulva or perineum and with stromal invasion ≤ 1.0 mma , no nodal metastasis IB Lesions N 2 cm in size or with stromal invasion N 1.0 mma , confined to the vulva or perineum, with negative nodes II Tumor of any size with extension to adjacent perineal structures (lower third of urethra, lower third of vagina, anus) with negative nodes III Tumor of any size with or without extension to adjacent perineal structures (lower third of urethra, lower third of vagina, anus) with positive inguinofemoral nodes IIIA (i) With 1 lymph node metastasis (≥5 mm), or (ii) With 1–2 lymph node metastasis(es) (b5 mm) IIIB (i) With 2 or more lymph node metastases (≥5 m), or (ii) With 3 or more lymph node metastases (b5 mm) IIIC With positive nodes with extracapsular spread. IV Tumor invades other regional (upper 2/3 urethra, upper 2/3 vagina), or distant structures IVA Tumor invades any of the following: (i) upper urethral and/or vaginal mucosa, bladder mucosa, rectal mucosa, or fixed to pelvic bone, or (ii) fixed or ulcerated inguinofemoral lymph nodes IVB Any distant metastasis including pelvic lymph nodes a The depth of invasion is defined as the measurement of the tumor from the epithelial– stromal junction of the adjacent most superficial dermal papilla to the deepest point of invasion. Table 2 Cancer of the vulva: FIGO staging compared with TNM classification. FIGO Stage Union for International Cancer Control (UICC) T (tumor) N (lymph nodes) M (metastasis) I T1 N0 M0 IA T1a N0 M0 IB T1b N0 M0 II T2/T3 N0 M0 IIIA T1, T2, T3 N1a, N1b M0 IIIB T1, T2, T3 N2a, N2b M0 IIIC T1, T2, T3 N2c M0 IVA T4 N0–N2 M0 IVB Any T N3 M0 S77N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
  • 12. 4.5.2. Invasive vulvar cancer Management of vulvar cancer must be individualized. There is no standard operation and the emphasis should be on performing the most conservative operation consistent with cure of the disease [3]. In considering surgical treatment options, it is necessary to consider independently the most appropriate management of: (1) The primary lesion. (2) The groin lymph nodes. In cases of locally advanced disease, treatment options for each site should be considered independently and then in the context of the over- all management of the patient, in order to select a treatment that will op- timize the likelihood of cure and minimize treatment-related morbidity. 4.5.3. Microinvasive vulvar cancer (Stage IA) Stage IA carcinoma of the vulva is defined as a single lesion measur- ing 2 cm or less in diameter with a depth of invasion of 1.0 mm or less, the depth being measured from the epithelial–stromal junction of the most adjacent superficial dermal papilla to the deepest point of inva- sion. Lesions of this extent should be managed with radical local excision. Groin dissection is not necessary for lesions of this type [11,12]. Level of Evidence C 4.5.4. Early vulvar cancer Tumors confined to the vulva without suspicious lymph nodes, as determined by clinical examination, with or without ultrasonic or radio- logical assessment, may be considered early. 4.5.4.1. Management of the primary lesion (Fig. 2). To decrease psycho- sexual morbidity, a more conservative operation than radical vulvectomy usually is indicated. The procedure may be called a radical local excision, and for localized lesions, this operation is as effective as radical vulvectomy in preventing local recurrence [3,13–17]. Surgical removal should achieve lateral margins of at least 1 cm, and the deep margin should be the inferior fascia of the urogenital dia- phragm, which is co-planar with the fascia lata and the fascia over the pubic symphysis [13,16–18]. If the lesion is close to the urethra, the distal 1 cm of the urethra may be resected without jeopardizing urinary continence. If there is associated VIN, this should be superficially excised to con- trol symptoms, to exclude other areas of superficial invasion, and to Fig. 1. Vulvar staging diagram. S78 N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
  • 13. prevent subsequent progression to invasive cancer. This is especially true for differentiated VIN. Level of Evidence C 4.5.4.2. Management of groin lymph nodes. Recurrence in the groin carries a very high mortality; therefore, appropriate groin treatment is the single most important factor in reducing mortality from early vulvar cancer [3]. All patients with FIGO stage 1B or stage II lesions should have at least an ipsilateral inguinofemoral lymphadenectomy. Level of Evidence C The incidence of positive contralateral nodes in patients with small lateral lesions and negative ipsilateral nodes is less than 1%, so unilateral groin dissection is appropriate for such lesions [3]. Bilateral groin dissection should be performed for midline tumors, and for those involving the anterior labia minora [19]. Large lateral tumors should probably also have bilateral dissection, and definitely if the ipsilateral nodes are positive [19]. Sentinel node excision is being increasingly practiced in many centers following the European multicenter observational study on sen- tinel node detection [20]. This procedure detects nodal metastasis in most patients with regional spread of disease, and is associated with a lower rate of lymphedema than complete lymphadenectomy. The study, (GROINSS-V), involved 403 women with a unifocal tumor con- fined to the vulva less than 4 cm in diameter, stromal invasion more than 1 mm, and clinically negative lymph nodes [20]. Sentinel nodes were identified using blue dye and radiolabelled technetium. Lymphad- enectomy was omitted in sentinel node negative women. Groin recur- rences occurred in 2.3% of patients, with a median follow-up of 35 months. Overall disease-specific survival was 97% after 3 years and mor- bidity was substantially reduced. Higher false-negative rates have been reported in other studies [21–23]. Owing to the small but definite false-negative rate with sentinel node biopsy, and the high risk of death if groin recurrence occurs, some patients, properly informed of the risks and benefits, will elect to have a full groin dissection, despite the greater complication rate [24–26]. Level of Evidence B 4.5.4.3. Groin dissection. It is recommended that both inguinal and fem- oral nodes be removed, as inguinal node dissection alone is associated with a higher incidence of groin recurrence [27]. Level of Evidence A The femoral nodes are situated medial to the femoral vein within the fossa ovalis. There is no need to remove the fascia lata to dissect the femoral nodes [28]. Groin dissection may be safely performed through a triple incision approach, and this should improve primary healing compared with an en bloc resection of the vulva and groins [29]. Level of Evidence C An en bloc approach may still be useful for clitoral or periclitoral lesions. To avoid skin necrosis, all subcutaneous tissue above the super- ficial fascia must be preserved. Groin dissection (with postoperative radiation for patients with positive groin nodes) was found to be superior to groin irradiation in one small randomized trial [30]. Pretreatment imaging that might have detected grossly enlarged nodes was not performed in that early trial and the radiation technique used was considered inadequate to cover the at-risk inguinofemoral nodes [31]. Retrospective clinical reviews have suggested that radiation alone can control microscopic disease in the groins if adequate coverage of the inguinofemoral nodes is confirmed [32,33]. 4.5.4.4. Management of patients with positive groin nodes. The Gynecolog- ic Oncology Group demonstrated superior results for pelvic and inguinal radiation compared with pelvic node dissection for patients who had an inguinal lymph node dissection with findings of grossly positive or more than one positive node [34]. Level of Evidence A. A recent retrospective, multicenter German study also reported improved survival for patients with positive groin nodes who received adjuvant radiotherapy directed at the groins (positive/negative other fields) [35]. Several studies have emphasized the prognostic significance of the morphology of positive groin nodes, particularly the size of the metasta- sis and the presence or absence of extracapsular spread [36–38]. Patients with one small lymph node metastasis do not appear to benefit from adjuvant radiation therapy; several series suggest that their prognosis is good after inguinofemoral lymphadenectomy alone [37–39], but the number of patients in most series is too small to draw definitive conclusions. A multicenter Dutch study of 75 patients with vulvar cancer and one positive lymph node of all sizes reported that adjuvant radiation was only beneficial if extracapsular spread was present [40]. Reasonable indications for bilateral pelvic and groin irradiation in patients with positive groin nodes would be: • The presence of extracapsular spread. • Two or more positive groin nodes. Level of Evidence B An ongoing international prospective observational trial (GROINSS-V II) is investigating the efficacy of groin radiation, without inguinofemoral lymphadenectomy, for patients with a single positive sentinel lymph node 2 mm or less in diameter. Pending the results of this study, all patients who have had a sentinel lymph node biopsy and are found to have one or more positive nodes should be treated with a full inguinofemoral lymph node dissection, followed by radiotherapy to the groins and pelvis if indicated. 4.5.4.5. Radiation fields and doses. In most cases, fields should include the inguinofemoral, external iliac, and internal iliac lymph nodes. The upper border may be extended if there is extensive inguinal involvement or suspicion of pelvic node metastasis. One of a variety of radiation techniques can be selected, depending on the patient’s body habitus and extent of disease. Treatments should always be based on three-dimensional planning using high-quality CT or MRI images. Combined photon and electron techniques are often used to treat the regional nodes, without overdosing the femoral heads. Care must be taken to completely include both the superficial and deep inguinal Fig. 2. Management of early vulvar cancer. * If there is associated VIN or lichen sclerosis, these areas may be superficially excised. S79N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
  • 14. lymph nodes. In thin patients, care must be taken to avoid underdosage of superficial inguinal nodes by high-energy photon beams. If electron beams are used, the energy must be sufficient to cover the femoral nodes. In recent years, some clinicians have begun to use intensity-modulated radiation therapy (IMRT) or other inverse-planned, computer-controlled delivery techniques to treat vulvar cancer. Although these techniques can reduce acute radiation effects in skin and soft tissue, the treatment planning and delivery are complex, and the opportunity for unanticipated underdosage of the target is substantial, suggesting that these cases may be best treated by clinicians who have considerable specialized expertise. The dose of radiation is determined by the initial extent of regional disease and any known residual. After a groin dissection with microscopic inguinal metastases, 50 Gy in 1.8–2.0 Gy fractions is usually sufficient. If there are multiple positive nodes or if there is evidence of extracapsular extension, doses up to 60 Gy may be given to a reduced volume. Gross residual disease usually requires 60–70 Gy to achieve a high probability of regional disease control. The effectiveness of concurrent chemotherapy in the treatment of groin and pelvic lymph nodes is unknown. 4.5.5. Advanced vulvar cancer Patients with primary tumors extending beyond the vulva, or bulky positive groin nodes are considered to have advanced vulvar cancer. For such patients, multimodality treatment planning is particularly important. 4.5.5.1. Management of groin lymph nodes. It is desirable to determine the status of the groin nodes prior to planning the overall treatment [3]. Pelvic CT or MRI should be part of the patient’s initial workup. These studies are particularly helpful in suggesting the extent of inguinal or pelvic lymphadenopathy (Fig. 3). Pelvic MRI can also provide useful information about the anatomical extent of the primary lesion, but is not mandatory. If there are no suspicious nodes in the groin on CT scan, bilateral inguinofemoral lymphadenectomy may be performed. If final histologic assessment reveals positive nodes, adjuvant radiation to the groin and pelvis should follow the guidelines given for early stage disease. If the nodes are negative, groin and pelvic radiation may be eliminated. Alternatively, primary chemoradiation therapy may be used to treat the primary tumor as well as the groin and pelvic nodes if surgery is deemed inappropriate for the individual patient [32,33]. If nodes are clinically positive, a complete lymphadenectomy should be avoided because full groin dissection together with postoperative groin irradiation may result in severe lymphedema. Only enlarged nodes from the groin and pelvis should be removed if feasible, and the patient given postoperative groin and pelvic radiation [41]. Level of Evidence C If there are ulcerated or fixed groin nodes, they should be resected if not infiltrating muscle or femoral vessels, as determined by imaging studies. If nodes are not felt to be resectable, they should be biopsied to confirm the diagnosis then treated with primary radiation, with or without chemotherapy. If appropriate, the nodes may be resected fol- lowing radiation if there has been an incomplete response (Fig. 4) [42]. Level of Evidence C 4.5.5.2. Management of the primary tumor (Fig. 5). If it is possible to resect the primary lesion with clear surgical margins and without sphincter damage leading to urinary or fecal incontinence, primary surgical exci- sion is usually the preferred treatment. It usually follows dissection of the groins, although this is not mandatory. If primary surgery would result in the need for a bowel or urinary stoma, it is preferable to employ primary radiation therapy, some- times followed by a more limited resection of the residual tumor or tumor bed [43,44]. Chemoradiation has been used extensively for large lesions if surgi- cal resection would damage central structures (anus, urethra); durable complete responses without the need for post-treatment surgery have been well described [45–49]. The groin nodes and pelvis may need to be included in the treatment field depending on the status of the groin nodes, as determined initially. Treatment with neoadjuvant cisplatin and 5-fluorouracil, or other drug combinations, has been reported in small retrospective studies to be effective for preservation of the anal sphincter and/or urethra in patients with advanced vulvar cancer [50,51]. This approach deserves further clinical research. 4.5.5.3. Radiation protocol. If the groin nodes are positive and meet the requirements described earlier for adjuvant radiation, the initial radia- tion treatment fields should include the pelvis, inguinal nodes, and pri- mary site, which are treated to a total dose of at least 50 Gy. Care must be taken to adequately cover the inguinal nodes. Some clinicians prefer to treat in an open-leg position but care must be taken to apply bolus to the vulva to avoid underdosage of potentially involved skin regions. Areas of gross disease or particularly high risk are usually boosted either with appositional fields of electrons selected to provide an ade- quate dose to the surface and at depth, or with conformal external beam therapy. Gross vulvar disease probably requires 60–70 Gy to achieve local control, although investigators are currently exploring Fig. 3. Management of clinically suspicious groin nodes. Fig. 4. Management of clinically obvious groin nodes. S80 N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
  • 15. a wide variety of chemoradiation schedules, and the relationship be- tween dose and local control remains somewhat uncertain. Level of Evidence C 4.5.5.4. Close surgical margins. Most recurrences from vulvar cancer occur on the vulva. Rouzier et al. [52] described two types of local recur- rences, those at the primary site and those at a remote site. In an analysis of patients with vulvar cancer from the Royal Hospital for Women in Sydney [13], primary site recurrences occurred with a median disease- free interval of 21 months, and were associated with a histological mar- gin of 8 mm or less, as previously reported [16 − 18]. Remote site recur- rences occurred with a median disease-free interval of 69 months, and were more commonly associated with lichen sclerosus. Although some recent papers have not found an association between margin dis- tance and local recurrence, these papers have not distinguished primary from remote site recurrences [53,54]. Postoperative radiation is of benefit for patients with close surgical margins (less than 5 mm), if the margins cannot be re-excised [55]. A re- cent study of 205 patients with vulvar cancer from Boston reported that the highest risk of vulvar recurrence was associated with margins of 5 mm or less (P = 0.002), and that patients who received a dose of more than or equal to 56 Gy had a lower risk of relapse than those who received less than or equal to 50.4 Gy (P b 0.05) [56]. In some cases, the positive margin may be boosted with brachyther- apy, although this technique requires experience to avoid an excessive risk of necrosis. Alternatively, the operative bed may be treated with an appositional electron field or in some cases, carefully planned confor- mal external beam irradiation. Level of Evidence C 5. Special situations 5.1. Vulvar melanoma Vulvar melanoma is the second most common neoplasm of the vulva. The majority of lesions involve the clitoris or labia minora. The Clark or Breslow modifications to the micro staging system should be used for the staging of vulvar melanoma rather than the more common TNM/FIGO system. These systems measure the depth of invasion in terms of the descriptive histology of the skin. Any pigmented lesion on the vulva should be excised for diagnosis unless it has been known to be present and unchanged for some years. In line with trends toward more conservative surgery for cutaneous melanomas, there is a trend toward more conservative resection of vulvar melanomas [57–59]. Primary lesions should be treated by radical local excision, with margins around the lesion of at least 1 cm. Level of Evidence C The role of node dissection is also controversial, but the Intergroup Surgical Melanoma Program has conducted a prospective, multi- institutional randomized trial of elective node dissection versus obser- vation for intermediate thickness cutaneous melanomas (1–4 mm) [60]. There were 740 patients entered into the trial, and elective node dissection resulted in a significantly better survival for patients 60 years of age or younger, patients with tumors 1–2 mm thick, and patients without tumor ulceration. 5.2. Bartholin’s gland cancer Cancers arising in the Bartholin’s gland may be either transitional or squamous types, arising from the duct, or an adenocarcinoma from the gland itself. Adenoid cystic and adenosquamous variants have also been reported. Adenocarcinomas of the vulva occur, on average, approxi- mately a decade earlier than invasive squamous cancers. Frequently, diagnosis is made after resection of what was thought to be a persisting Bartholin’s cyst. The standard approach for Bartholin’s gland carcinomas has been radical vulvectomy and bilateral groin dissection. However, ipsilateral groin dissection and radical hemi-vulvectomy may be equally effective for early lesions [61]. Because these lesions are deep in the ischiorectal fossa, surgical margins are more likely to be close, particularly for bulky lesions, and postoperative radiation to the vulva may decrease the likelihood of local recurrence [61]. Level of Evidence C If the ipsilateral groin nodes are positive, bilateral groin and pelvic radiation may decrease regional recurrence. For adenoid cystic lesions, radical local excision alone is the treat- ment of choice, with adjuvant local radiation recommended for positive margins or perineural invasion [62]. Level of Evidence C 5.3. Paget’s Disease This is predominantly an intraepithelial lesion, but on occasion it may be associated with an underlying invasive adenocarcinoma. It is usually of primary cutaneous origin from the vulva, but may be second- ary to an anorectal, urothelial, or noncutaneous genital tract carcinoma (e.g. endocervical or endometrial) [63]. The disease occurs predominantly in the menopausal or postmeno- pausal population. Most patients will present with vulvar discomfort and itching and on examination, an eczematoid, weeping lesion is often seen. Diagnosis is usually confirmed by biopsy, which will gener- ally differentiate an intraepithelial from an invasive lesion [3,64]. Intraepithelial Paget’s disease requires superficial local excision. It is difficult to obtain clear margins with this disease, as often the underly- ing histologic change will extend far beyond the macroscopic lesion. More recently, there has been a move to perform less radical resection for intraepithelial lesions, with re-excision at a later date should lesions become symptomatic or clinically visible [65]. Lesions that involve or extend into the urethra or anus can be particularly difficult to manage, and may require laser therapy. If there is an underlying adenocarcinoma, the invasive component should be treated by radical local excision with margins of at least 1 cm. At least an ipsilateral inguinofemoral lymphadenectomy should be performed for unilateral lesions, with adjuvant radiation following the same indications as for squamous carcinomas [66]. Level of Evidence C 6. Pathology The surgical specimen should be correctly orientated and photo- graphed. Photographs should be used to indicate the origin of tissue blocks. The size of the specimen should be measured and the Fig. 5. Management of advanced primary tumor. Treatment should usually follow dissec- tion of the groins. Groin and pelvic radiation should follow standard indications. S81N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
  • 16. dimensions of any visible tumor measured. The macroscopic tumor-free surgical margins should be measured. Sections are taken through the tumor to measure tumor depth. Sections should be taken from urethral, anal, and vaginal resection margins. Lymph nodes should be carefully dissected out and the site from which they are removed recorded. A full cross-section of each lymph node should be embedded. The following histological points should be noted: (a) Tumor type: keratinizing, basaloid, bowenoid. (b) Depth of invasion: measured from the epithelial–stromal junc- tion of the adjacent dermal papilla to the deepest point of inva- sion by the tumor. (c) Tumor grade. (d) Histological measurement of tumor-free margins and statement as to whether the tumor is completely excised. (e) Presence or absence of perineural or vascular space invasion. (f) Nature of the adjacent nonmalignant squamous epithelium: VIN, lichen sclerosus, squamous hyperplasia, HPV-associated changes. (g) Sites and number of nodes examined and number of positive nodes. Presence or absence of extracapsular extension. Conflict of interest The authors declare that they have no conflicts of interest. References [1] Hacker NF. Revised FIGO staging for carcinoma of the vulva. Int J Gynecol Obstet 2009;105(2):105–6. [2] Van der Velden J, van Lindert AC, Gimbrere CH, Oosting H, Heintz AP. Epidemiologic data on vulvar cancer: Comparison of hospital with population-based data. Gynecol Oncol 1996;62(3):379–83. [3] Hacker NF, Cancer Vulvar. In: Berek JS, Hacker NF, editors. Berek and Hacker’s Gynecologic Oncology. 6th ed. Philadelphia: Lippincott Williams and Wilkins; 2015. p. 560–607. [4] Sideri M, Jones RW, Wilkinson EJ, Preti M, Heller DS, Scurry J, et al. Squamous vulvar intraepithelial neoplasia: 2004 modified terminology, ISSVD Vulvar Oncology Sub- committee. J Reprod Med 2005;50(11):807–10. [5] van der Velden J, van Lindert AC, Gimbrere CH, Oosting H, Heintz AP. Epidemiologic data on vulvar cancer: comparison of hospital with population-based data. Gynecol Oncol 1996;62(3):379–83. [6] Rhodes CA, Cummins C, Shafi M. The management of squamous cell vulval cancer: a population based retrospective study of 411 cases. Br J Obstet Gynaecol 1998; 105(2):200–5. [7] Wallbillich JJ, Rhodes HE, Milbourne AM, Munsell MF, Frumovitz M, Brown J, et al. Vulvar intraepithelial neoplasia (VIN 2/3): comparing clinical outcomes and evaluat- ing risk factors for recurrence. Gynecol Oncol 2012;127(2):312–5. [8] Rodolakis A, Diakomanolis E, Vlachos G, Iconomou T, Protopappas A, Stefanidis C, et al. Vulvar intraepithelial neoplasia (VIN)–diagnostic and therapeutic challenges. Eur J Gynaecol Oncol 2003;24(3):317–22. [9] Terlou A, van Seters M, Ewing PC, Aaronson NK, Gundy CM, Heijmans-Antonissen C, et al. Treatment of vulvar intraepithelial neoplasia with topical imiquimod: Seven years median follow-up of a randomized clinical trial. Gynecol Oncol 2011;121(1): 157–62. [10] Mathiesen O, Buus SK, Cramers M. Topical imiquimod can reverse vulvar intraep- ithelial neoplasia: A randomised, double-blinded study. Gynecol Oncol 2007; 107(2):219–22. [11] Iversen T, Abeler V, Aalders J. Individualized treatment of stage I carcinoma of the vulva. Obstet Gynecol 1981;57(1):85–90. [12] Hacker NF, Berek JS, Lagasse LD, Nieberg RK, Leuchter RS. Individualization of treat- ment for stage I squamous cell vulvar carcinoma. Obstet Gynecol 1984;63(4): 155–62. [13] Tantipalakorn C, Robertson G, Marsden DE, Gebski V, Hacker NF. Outcome and pat- terns of recurrence for International Federation of Gynecology and Obstetrics (FIGO) stages I and II squamous cell vulvar cancer. Obstet Gynecol 2009;113(4):895–901. [14] Farias-Eisner R, Cirisano FD, Grouse D, Leuchter RS, Karlan BR, Lagasse LD, et al. Con- servative and individualized surgery for early squamous carcinoma of the vulva: the treatment of choice for stage I and II (T1-2NO-1 M0) disease. Gynecol Oncol 1994; 53(1):55–8. [15] Burke TW, Levenback C, Coleman RL, Morris M, Silva EG, Gershenson DM. Surgical therapy of T1 and T2 vulvar carcinoma: further experience with radical wide exci- sion and selective inguinal lymphadenectomy. Gynecol Oncol 1995;57(2):215–20. [16] Heaps JM, Fu YS, Montz FJ, Hacker NF, Berek JS. Surgical pathological variables pre- dictive of local recurrence in squamous cell carcinoma of the vulva. Gynecol Oncol 1990;38(3):309–14. [17] De Hullu JA, Hollema H, Lolkema S, Boezen M, Boonstra H, Burger MP, et al. Vulvar carcinoma: the price of less radical surgery. Cancer 2002;95(11):2331–8. [18] Chan JK, Sugiyama V, Pham H, Gu M, Rutgers J, Osann K, et al. Margin distance and other clinico-pathological prognostic factors in vulvar carcinoma: a multivariate analysis. Gynecol Oncol 2007;104(3):636–41. [19] Iversen T, Aas M. Lymph drainage from the vulva. Gynecol Oncol 1983;16(2): 179–89. [20] Van der Zee AG, Oonk MH, De Hullu JA, Ansink AC, Vergote I, Verheijen RH, et al. Sentinel node dissection is safe in the treatment of early stage vulvar cancer. J Clin Oncol 2008;26(6):884–9. [21] Hampl M, Hantschmann P, Michels W, Hillemanns P. German Multicenter Study Group Validation of the accuracy of the sentinel lymph node procedure in patients with vulvar cancer: results of a multicenter study in Germany. Gynecol Oncol 2008;111(2):282–8. [22] Levenback CF, Ali S, Coleman RL, Gold MA, Fowler JM, Judson PL, et al. Lymphatic mapping and sentinel lymph node biopsy in women with squamous cell carcinoma of the vulva: a gynecologic oncology group study. J Clin Oncol 2012;30(31): 3786–91. [23] Radziszewski J, Kowalewska M, Jedrzejczak T, Kozlowicz-Gudzinska I, Nasierowska- Guttmejer A, Bidzinski M, et al. The accuracy of the sentinel lymph node concept in early stage squamous cell vulvar carcinoma. Gynecol Oncol 2010;116(3):473–7. [24] de Hullu JA, Ansink AC, Tymstra T, van der Zee AG. What doctors and patients think about false-negative sentinel lymph nodes in vulvar cancer. J Psychosom Obstet Gynaecol 2001;22(4):199–203. [25] Gan S, Magarey C, Schwartz P, Papadatos G, Graham P, Vallentine J. Women’s choice between sentinel lymph node biopsy and axillary clearance. ANZ J Surg 2002;72(2): 110–3. [26] Farrell R. Gebski, Hacker NF. Quality of life after complete lymphadenectomy for vul- var cancer: do women prefer sentinel node biopsy? Int J. Gynecol Oncol 2014;24(4): 813–9. [27] Stehman FB, Bundy BN, Dvoretsky PM, Creasman WT. Early stage I carcinoma of the vulva treated with ipsilateral superficial inguinal lymphadenectomy and modified radical hemivulvectomy: a prospective study of the Gynecologic Oncology Group. Obstet Gynecol 1992;79(4):490–7. [28] Micheletti L, Borgno G, Barbero M, Preti M, Cavanna L, Nicolaci P, et al. Deep femoral lymphadenectomy with preservation of the fascia lata. Preliminary report on 42 invasive vulvar carcinomas. J Reprod Med 1990;35(12):1130–4. [29] Hacker NF, Leuchter RS, Berek JS, Castaldo TW, Lagasse LD. Radical vulvectomy and bilateral inguinal lymphadenectomy through separate groin incisions. Obstet Gynecol 1981;58(5):574–9. [30] Stehman F, Bundy B, Thomas G, Varia M, Okagaki T, Roberts J, et al. Groin dissection versus groin radiation in carcinoma of the vulva: a Gynecologic Oncology Group study. Int J Radiat Oncol Biol Phys 1992;24(2):389–96. [31] Koh WJ, Chiu M, Stelzer KJ, Greer BE, Mastras D, Comsia N, et al. Femoral vessel depth and the implication for groin node radiation. Int J Radiat Oncol Biol Phys 1993;27(3):969–74. [32] Katz A, Eifel PJ, Jhingran A, Levenback CF. The role of radiation therapy in preventing regional recurrences of invasive squamous cell carcinoma of the vulva. Int J Radiat Oncol Biol Phys 2003;57(2):409–18. [33] Petereit DG, Mehta MP, Buchler DA, Kinsella TJ. A retrospective review of nodal treatment for vulvar cancer. Am J Clin Oncol 1993;16(1):38–42. [34] Homesley HD, Bundy BN, Sedlis A, Adcock L. Radiation therapy versus pelvic node resection for carcinoma of the vulva with positive groin nodes. Obstet Gynecol 1986;68(6):733–40. [35] Mahner S, Jueckstock J, Hilpert F, Neuser P, Harter P, de Gregorio N, et al. Adjuvant therapy in lymph node positive vulvar cancer: The AGO-CaRT-1 study. J Natl Cancer Inst 2015;107(3):dju426. [36] Origoni M, Sideri M, Garsia S, Carinelli SG, Ferrari AG. Prognostic value of patholog- ical patterns of lymph node positivity in squamous cell carcinoma of the vulva stage III and IVA FIGO. Gynecol Oncol 1992;45(3):313–6. [37] Paladini D, Cross P, Lopes A, Monaghan J. Prognostic significance of lymph node variables in squamous cell cancer of the vulva. Cancer 1994;74(9):2491–6. [38] van der Velden J, van Lindert AC, Lammes FB, ten Kate FJ, Sie-Go DM, Oosting H, et al. Extracapsular growth of lymph node metastases in squamous cell cancer of the vulva. The impact on recurrence and survival. Cancer 1995;75(12):2885–90. [39] Hacker NF, Berek JS, Lagasse LD, Leuchter RS, Moore JG. Management of regional lymph nodes and their prognostic influence in vulvar cancer. Obstet Gynecol 1983;61(4):408–12.the vulva. The impact on recurrence and survival. Cancer 1995;75(12):2885–90. [40] Fons G, Groenen SM, Oonk MH, Ansink AC, van der Zee AG, Burger MP, et al. Adju- vant radiotherapy in patients with vulvar cancer and one intracapsular lymph node metastasis is not beneficial. Gynecol Oncol 2009;114(2):343–5. [41] Hyde SE, Valmadre S, Hacker NF, Schilthuis MS, van der Velden J. Squamous cell car- cinoma of the vulva with bulky positive groin nodes-nodal debulking versus full groin dissection prior to radiation therapy. Int J Gynecol Cancer 2007;17(1):154–8. [42] Montana GS, Thomas GM, Moore DH, Saxer A, Mangan CE, Lentz SS, et al. Preopera- tive chemoradiation for carcinoma for the vulva with N2/N3 nodes: a Gynecologic Oncology Group Study. Int J Radiat Oncol Biol Phys 2000;48(4):1007–13. [43] Hacker NF, Berek JS, Juillard GJF, Lagasse LD. Preoperative radiation therapy for local- ly advanced vulvar cancer. Cancer 1984;54(10):2056–60. [44] Boronow RC, Hickman BT, Reagan MT, Smith RA, Steadham RE. Combined therapy as an alternative to exenteration for locally advanced vulvovaginal cancer. II. Results, complications and dosimetric and surgical considerations. Am J Clin Oncol 1987; 10(2):171–81. [45] Moore DH, Ali S, Koh WJ, Michael H, Barnes MN, McCourt CK, et al. A phase II trial of radiation therapy and weekly cisplatin chemotherapy for the treatment of locally S82 N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
  • 17. advanced squamous cell carcinoma of the vulva: a gynecologic oncology group study. Gynecol Oncol 2012;124(3):529–33. [46] Lupi G, Raspagliesi F, Zucali R, Fontanelli R, Paladini D, Kenda R, et al. Combined pre- operative chemoradiotherapy followed by radical surgery in locally advanced vulvar carcinoma. A pilot study. Cancer 1996;77(8):1472–8. [47] Landoni F, Maneo A, Zanetta G, Colombo A, Nava S, Placa F, et al. Concurrent preop- erative chemotherapy with 5-fluorouracil and mitomycin C and radiotherapy (FUMIR) followed by limited surgery in locally advanced and recurrent vulvar carci- noma. Gynecol Oncol 1996;61(3):321–7. [48] Cunningham MJ, Goyer RP, Gibbons SK, Kredentser DC, Malfetano JH, Keys H. Prima- ry radiation, cisplatin, and 5-fluorouracil for advanced squamous cell carcinoma of the vulva. Gynecol Oncol 1997;66(2):258–61. [49] Beriwal S, Coon D, Heron DE, Kelley JL, Edwards RP, Sukumvanich P, et al. Preoper- ative intensity-modulated radiotherapy and chemotherapy for locally advanced vulvar cancer. Gynecol Oncol 2008;109(2):291–5. [50] Geisler JP, Manahan KJ, Buller RE. Neoadjuvant chemotherapy in vulvar cancer: avoiding primary exenteration. Gynecol Oncol 2006;100(1):53–7. [51] Aragona AM, Cuneo N, Soderini AH, Alcoba E, Greco A, Reyes C, et al. Tailoring the treatment of locally advanced carcinoma of the vulva: Neoadjuvant chemotherapy followed by radical surgery: Results from a multicenter study. Int J Gynecol Cancer 2012;22(7):1258–63. [52] Rouzier R, Haddad B, Plantier F, Dubois P, Pelisse M, Paniel BJ. Local relapse in pa- tients treated for squamous cell vulvar carcinoma: incidence and prognostic value. Obstet Gynecol 2002;100(6):1159–67. [53] Groenen SM, Timmers PJ, Burger CW. Recurrence rate in vulvar carcinoma in rela- tion to pathological margin distance. Int J Gynecol Cancer 2010;20(5):869–73. [54] Woelber L, Choschzick M, Eulenburg C, Hager M, Jaenicke F, Gieseking F, et al. Prog- nostic value of pathological resection margin distance in squamous cell cancer of the vulva. Ann Surg Oncol 2011;18(13):3811–8. [55] Faul CM, Mirmow D, Huang O, Gerszten K, Day R, Jones MW. Adjuvant radiation for vulvar carcinoma: improved local control. Int J Radiat Oncol Biol Phys 1997;38(2): 381–9. [56] Viswanathan AN, Pinto AP, Schultz D, Berkowitz R, Crum CP. Relationship of margin status and radiation dose to recurrence in postoperative vulvar carcinoma. Gynecol Oncol 2013;130(3):545–9. [57] Verschraegen CF, Benjapibal M, Supakarapongkul W, Levy LB, Ross M, Atkinson EN, et al. Vulvar melanomas at the M.D. Anderson Cancer Center: 25 years later. Int J Gynecol Cancer 2001;11(5):359–64. [58] Mert I, Semaan A, Winer I, Morris RT, Ali-Fehmi R. Vulvar/vaginal melanoma: an up- dated surveillance epidemiology and end results database review. Comparison with cutaneous melanoma and significance of racial disparities. Int J Gynecol Oncol 2013; 23(6):1118–25. [59] Heinzelmann-Schwarz VA, Nixdorf S, Valadan M, Diczbalis M, Olivier J, Otton G, et al. A clinicopathological review of 33 patients with vulvar melanoma identifies c-KIT as a prognostic marker. Int J Mol Med 2014;33(4):784–94. [60] Balch CM, Soong SJ, Bartolucci AA, Urist MM, Karakousis CP, Smith TJ, et al. Efficacy of an elective regional lymph node dissection of 1 to 4 mm thick melanomas for patients 60 years of age and younger. Ann Surg 1996;224(3):255–63. [61] Copeland LJ, Sneige N, Gershenson DM, McGuffee VB, Abdul-Karim F, Rutledge FN. Bartholin gland carcinoma. Obstet Gynecol 1986;67(6):794–801. [62] Yang SY, Lee JW, Kim WS, Jung KL, Lee SJ, Lee JH, et al. Adenoid cystic carcinoma of the Bartholin’s gland: report of two cases and review of the literature. Gynecol Oncol 2006;100(2):422–5. [63] Wilkinson EJ, Brown HM. Vulvar Paget disease of urothelial origin: a report of three cases and a proposed classification of vulvar Paget disease. Hum Pathol 2002;33(5): 540–54. [64] Fanning J, Lambert L, Hale TM, Morris PC, Schuerch C. Paget’s disease of the vulva: prevalence of associated vulvar adenocarcinoma, invasive Paget’s disease, and recur- rence after surgical excision. Am J Obstet Gynecol 1999;180(1 Pt 1):24–7. [65] Black D, Tornos C, Soslow RA, Awtrey CS, Barakat RR, Chi DS. The outcomes of patients with positive margins after excision for intraepithelial Paget’s disease of the vulva. Gynecol Oncol 2007;104(3):547–50. [66] Karam A, Dorigo O. Treatment outcomes in a large cohort of patients with invasive Extramammary Paget’s disease. Gynecol Oncol 2012;125(2):346–51. S83N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S76–S83
  • 18. FIGO CANCER REPORT 2015 Cancer of the vagina Neville F. Hacker a , Patricia J. Eifel b , Jacobus van der Velden c a Gynecologic Oncology Cancer Centre, Royal Hospital for Women, Randwick, Australia b Department of Radiation Oncology, M.D. Anderson Cancer Center, Houston, TX, USA c Department of Gynecology, Academic Medical Center, Amsterdam, The Netherlands 1. Staging A description of the staging classification for primary vaginal carci- noma is detailed in Table 1. 1.1. Anatomy 1.1.1. Primary site The vagina extends from the hymenal ring upward to the uterine cervix. Cases should be classified as carcinoma of the vagina when the primary site of the growth is in the vagina. Tumors present in the vagina as secondary growths from either genital or extra-genital sites should be excluded. A growth that has extended to the portio of the cervix and reached the area of the external os should always be allotted to carcinoma of the cervix. A growth limited to the urethra should be classified as carcinoma of the urethra. A tumor involving the vulva should be classified as carcinoma of the vulva. There should be histologic verification of the disease. 1.1.2. Nodal stations The upper two-thirds of the vagina is drained by lymphatics to the pelvic nodes, with the lymphatics paralleling the course of the uterine artery and the vaginal artery to the obturator, hypogastric (internal iliac), and external iliac nodes. The distal third of the vagina drains to the inguinal-femoral nodes. Some lesions, particularly those involving the posterior vaginal wall, may drain via pararectal lymphatic channels to presacral nodes. 1.1.3. Metastatic sites The most common sites of distant spread include the lungs, liver, and bony skeleton. The rules for staging are similar to those for carcinoma of the cervix. 1.1.4. Histopathologic types Approximately 90% of primary vaginal cancers are squamous cell carcinomas. Primary adenocarcinomas of the vagina occur, but are rare. 1.1.5. Histopathologic grades (G) • GX: Grade cannot be assessed. • G1: Well differentiated. • G2: Moderately differentiated. • G3: Poorly or undifferentiated. 2. Introduction Carcinomas of the vagina constitute only about 2% of malignant neoplasms of the female genital tract [1]. The vagina, however, can be a common site of metastatic gynecological cancer, by either direct extension of cervical or vulvar tumors, or through lymphatic or vascular deposits, as seen in endometrial cancer and gestational tropho- blastic disease, respectively. Metastatic or direct extension of non- gynecologic tumors to the vagina can also occur from the urinary bladder, urethra, periurethral glands, rectum, and rarely the breast, lung, or other sites. Up to 30% of patients with primary vaginal carcinoma have a history of in situ or invasive cervical cancer treated at least 5 years earlier [2–4]. (It is arbitrarily assumed that an invasive squamous cell carcino- ma occurring in the vagina more than 5 years after an invasive squa- mous cell carcinoma of the cervix is a new primary cancer.) Some vaginal cancers are preceded by vaginal intraepithelial neoplasia (VAIN), although the true malignant potential of VAIN is not known [5,6]. Prior pelvic radiation has also been considered a possible cause of vaginal cancer [7,8]. Most vaginal cancers occur in postmenopausal or elderly women [1]. When occurring in younger patients, the disease seems to be etiologi- cally related to cervical neoplasia, and thus HPV dependent [9]. International Journal of Gynecology and Obstetrics 131 (2015) S84–S87 Table 1 FIGO staging of cancer of the vagina. FIGO Stage Description I The carcinoma is limited to the vaginal wall II The carcinoma has involved the sub-vaginal tissue but has not extended to the pelvic wall III The carcinoma has extended to the pelvic wall IV The carcinoma has extended beyond the true pelvis or has involved the mucosa of the bladder or rectum; bullous edema as such does not permit a case to be allotted to Stage IV IVA Tumor invades bladder and/or rectal mucosa and/or direct extension beyond the true pelvis IVB Spread to distant organs http://dx.doi.org/10.1016/j.ijgo.2015.06.003 0020-7292/© 2015 Published by Elsevier Ireland Ltd. on behalf of International Federation of Gynecology and Obstetrics. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Contents lists available at ScienceDirect International Journal of Gynecology and Obstetrics journal homepage: www.elsevier.com/locate/ijgo
  • 19. 3. Screening Routine screening for vaginal cancer following hysterectomy for benign disease is not recommended because these women are at ex- tremely low risk of developing vaginal cancer. Women with a history of cervical intraepithelial or invasive neoplasia are at increased risk, but regular cytologic screening gives a low yield. The introduction of pri- mary HPV testing will allow the screening interval to be increased, which may make it cost-effective in this group of patients [10]. 4. Vaginal intraepithelial neoplasia (VAIN) For patients with an abnormal pap smear and no gross abnormality, vaginal colposcopy and use of Lugol’s iodine to stain the vagina are necessary. Biopsy of colposcopically abnormal areas is necessary, usually under anesthesia. Excisional biopsy is useful for lesions involv- ing the vaginal vault, where occult carcinoma may be found in up to 28% of patients with VAIN [11]. Treatment of VAIN must be individualized. Numerous treatments, ranging from local surgical excision or ablation through to intracavitary radiotherapy, have been used. Selection of the appropriate treatment is usually based on a careful study of several factors, including the general medical condition of the patient, the histology of the lesion, the location and extent of the disease, as well as the experience and expertise of the treating medical team. The proximity of the urethra, bladder, and rectum to the vaginal epithelium is an important factor to be considered. Damage or injury to these structures can occur with possi- ble fistula formation, particularly when the patient has had prior pelvic radiation therapy. Laser vaporization with a carbon dioxide laser is an effective treatment for VAIN [12]. This technique generally requires local or general anesthesia. The use of topical 5-fluorouracil (5-FU) is a relatively simple ambu- latory treatment, which does not require anesthesia or complicated equipment [13]. This approach may be especially valuable for patients with widespread or multifocal disease, which would require an exten- sive surgical procedure. Adverse effects are usually minimal, as long as it is not used more than twice a week. Imiquimod 5% cream might represent an alternative method of management in young, HPV-positive women with multifocal high- grade lesions (VAIN 2/3) [14]. Excisional procedures, either with electrosurgical loops or a scalpel excision, have also been used to treat VAIN. Surgical excision is particu- larly appropriate for vault lesions [15]. Total vaginectomy and split- thickness skin grafting may be occasionally necessary to treat extensive lesions that involve virtually the entire length of the vaginal tube and where other conservative methods have been unsuccessful. Level of Evidence C 5. Invasive carcinoma Most patients present with painless vaginal bleeding and discharge, and definitive diagnosis can usually be made by biopsy of a gross lesion detected on speculum examination. This can often be done in the office, but may be facilitated by examination under anesthesia. 5.1. Treatment Whenever possible, patients should be referred to tertiary referral units because of the rarity of these lesions and the limited experience of most practitioners with the specialized techniques used to treat these cancers effectively. All treatment must be individualized, and will vary depending on the stage of disease and the site of vaginal in- volvement. Whenever possible, an effort should be made to maintain a functional vagina, although the combination of tumor destruction and treatment effects may cause narrowing or shortening of the vagina, particularly in elderly women. 5.1.1. Surgery Surgery has a limited role because of the close proximity of the blad- der and rectum, but may be useful in the following situations [1,16,17]: (1) In patients with Stage I disease involving the upper posterior vagina If the uterus is still in situ, radical hysterectomy, upper vaginectomy to achieve clearance of at least 1 cm, and pelvic lymphadenectomy may be performed. If hysterectomy has been performed previously, radical upper vaginectomy and pel- vic lymphadenectomy may be appropriate. (2) In young patients who require radiation therapy Pretreatment laparotomy or laparoscopy may allow ovarian transposition, or in selected cases, surgical staging and resection of any bulky positive lymph nodes. (3) In selected patients with Stage IVA disease, particularly if a rectovaginal or vesicovaginal fistula is present Primary pelvic exenteration may be a suitable treatment option for selected patients, either combined with pelvic lymphadenec- tomy or preoperative radiation [2]. Bilateral groin dissection should be considered in such patients if the lower third of the va- gina is involved. (4) In patients with a central recurrence after radiation therapy Surgery will usually necessitate some type of pelvic exenteration in such patients. Level of Evidence C 5.1.2. Radiation therapy Radiation therapy is the treatment of choice for most patients with vaginal cancer, and usually requires careful integration of external beam irradiation and intracavitary or interstitial brachytherapy. External beam and brachytherapy techniques may vary widely, depending on the precise location of the tumor and its relationship to critical structures. Although some authors have advocated treatment with brachyther- apy alone for small Stage I (or even Stage II) cancers [18–21], a combi- nation of external beam irradiation and brachytherapy probably reduces the risk of local–regional recurrence in such cases. For larger lesions, treatment is started with approximately 45–50 Gy external radiation to reduce the primary tumor volume and to treat the pelvic nodes. This is then supplemented with brachytherapy or external beam boosts to gross disease in the primary site and involved lymph nodes. There is evidence for improved local control when the dose to the primary tumor exceeds 70 Gy [19,21]. This is most easily achieved using brachytherapy if the entire tumor volume can be treated to the necessary dose without exceeding normal tissue tolerance. Although brachytherapy is preferred whenever possible, highly conformal exter- nal beam boosts may achieve more homogeneous coverage of tumor in selected patients who have massive tumors, or intimate association of tumor with critical structures, for example the rectovaginal septum. If the distal one-third of the vagina is involved, the groin nodes should be treated. Level of Evidence C There has been limited reported experience with chemoradiation for vaginal cancer [20–22]. A recent National Cancer Data Base study of 13 689 patients diagnosed with vaginal cancer from 1998–2011 reported that the use of chemoradiation increased from 20.8% to 59.1% over that period [23]. The median overall survival was longer in patients receiving chemoradiation compared with radiation alone (56.2 vs 41.2 months), and use of chemoradiation was an independent prognostic factor for improved survival. Level of Evidence C 5.2. Prognosis Although the reported overall five-year survival for vaginal cancer is only about 52% [1], reports from major centers have indicated five-year S85N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S84–S87
  • 20. survival rates comparable to cervical cancer [18–20]. A study of 193 patients from the M.D. Anderson Cancer Center in Houston reported five-year disease-specific survival rates of 85% for 50 patients with Stage I disease, 78% for 97 patients with Stage II, and 58% for 46 patients with Stages III–IVA [22]. 6. Special situation 6.1. Adenocarcinoma Approximately 10% of primary vaginal carcinomas are adenocarci- nomas, and they may arise in areas of vaginal adenosis in diethylstilbes- trol (DES) exposed patients, in Wolffian rest elements, periurethral glands, or foci of endometriosis. DES-related clear cell carcinomas of the vagina occurred mainly in young women, but as the DES-exposed cohort is now over 50 years of age, DES-related tumors are now rare. In a series of 26 patients with non-DES-related vaginal adenocarci- nomas reported from the M.D. Anderson Cancer Center in 2007, the me- dian age was 54 years [24]. 6.1.1. Treatment In general, adenocarcinomas are treated in a similar manner to squa- mous lesions, although greater emphasis should be placed on combined modality therapy, even for small tumors, because of the greater propen- sity for local and distant recurrence [24]. 6.1.2. Prognosis Prognosis for DES-related clear cell carcinomas of the vagina is generally good, with an overall survival of 78% [25]. Survival for non- DES-related adenocarcinomas is significantly worse than for squamous cancers. A recent study of 26 such patients from the M.D. Anderson Cancer Center reported an overall five-year survival of only 34%, with a higher rate of both local recurrences and distant metastases [24]. 6.2. Vaginal melanoma Malignant melanomas of the vagina are rare, and almost all cases occur in white women [26,27]. They most commonly occur in the distal vagina, particularly on the anterior vaginal wall [26,28]. Most are deeply invasive and radical surgery has been the mainstay of treatment, often involving some type of pelvic exenteration. Recently, more conservative local excisions have been used, with comparable sur- vival rates reported [25–29]. This is usually combined with postopera- tive radiation. Overall five-year survival is about 15% [27]. Level of Evidence C 6.3. Sarcoma botryoides Sarcoma botryoides is a highly malignant tumor of the rhabdomyoblasts. These neoplasms are found in infants and children and usually present with discharge, bleeding, or a visible mass at the introitus. In the past, exenterative surgery was used for these lesions, but survival was poor. More recently, conservative surgery has been used in conjunction with preoperative or postoperative chemotherapy and radiotherapy with significantly improved survival. Most reported chemotherapeutic experience has been with vincristine, actinomycin D, and cyclophosphamide (VAC) [30–32]. If the lesion is small and can be resected with organ preservation, surgery should be the initial approach. For bulkier lesions, preoperative chemotherapy or localized teletherapy or brachytherapy may be used. Extended radiotherapy fields are not recommended, as they may produce significant developmental problems with the bony pelvis by destroying or interfering with growth centers in these structures. Conflict of interest The authors declare that they have no conflicts of interest. References [1] Hacker NF. Vaginal Cancer. In: Berek JS, Hacker NF, editors. Berek and Hacker’s Gynecologic Oncology. 6th ed. Philadelphia: Lippincott Williams and Wilkins; 2015. p. 608–24. [2] Eddy GL, Marks RD, Miller 3rd MC, Underwood Jr PB. Primary invasive vaginal carcinoma. Am J Obstet Gynecol 1991;165(2):292–8. [3] Peters 3rd WA, Kumar NB, Morley GW. Carcinoma of the vagina. Factors influencing treatment outcome. Cancer 1989;55(4):892–7. [4] Rubin SC, Young J, Mikuta JJ. Squamous carcinoma of the vagina: treatment, complications, and long-term follow-up. Gynecol Oncol 1985;20(3):346–53. [5] Lenehan PM, Meff F, Lickrish GM. Vaginal intraepithelial neoplasia: biologic aspects and management. Obstet Gynecol 1986;68(3):333–7. [6] Rome RM, England PG. Management of vaginal intraepithelial neoplasia: a series of 132 cases with long term follow-up. Int J Gynecol Cancer 2000;10(5): 382–90. [7] Pride GL, Buchler DA. Carcinoma of vagina 10 or more years following pelvic irradiation therapy. Am J Obstet Gynecol 1977;127(5):513–8. [8] Hellman K, Lundell M, Silfversward C, Nilsson B, Hellstrom AC, Frankendal B. Clinical and histopathological factors related to prognosis in primary squamous cell carcinoma of the vagina. Int J Gynecol Cancer 2006;16(3):1201–11. [9] Hellman K, Silfversward C, Nilsson B, Hellstrom AC, Frankendal B, Pettersson F. Primary carcinoma of the vagina: factors influencing the age at diagnosis. The Radiumhemmet series 1956–96. Int J Gynecol Cancer 2004;14(3): 491–501. [10] Orr JM, Barnett JC, Leath 3rd CA. Incidence of subsequent abnormal cytology in cervical cancer patients completing five-years of post treatment surveillance without evidence of recurrence. Gynecol Oncol 2011;122(3):501–4. [11] Hoffman MS, DeCesare SL, Roberts WS, Fiorica JV, Finan MA, Cavanagh D. Upper vaginectomy for in situ and occult, superficially invasive carcinoma of the vagina. Am J Obstet Gynecol 1992;166(1 Pt 1):30–3. [12] Yalcin OT, Rutherford TJ, Chambers SK, Chambers JT, Schwartz PE. Vaginal intraepithelial neoplasia: treatment by carbon dioxide laser and risk factors for failure. Eur J Obstet Gynecol Reprod Biol 2003;106(1):64–8. [13] Krebs HB. Prophylactic topical 5-fluorouracil following treatment of human papillomavirus-associated lesions of the vulva and vagina. Obstet Gynecol 1986; 68(6):837–41. [14] Haidopoulos D, Diakomanolis E, Rodolakis A, Voulgaris Z, Vlachos G, Intsaklis A. Can local application of imiquimod cream be an alternative mode of therapy for patients with high-grade intraepithelial lesions of the vagina. Int J Gynecol Cancer 2005; 15(5):898–902. [15] Indermaur MD, Martino MA, Fiorica JV, Roberts WS, Hoffman MS. Upper vaginectomy for the treatment of vaginal intraepithelial neoplasia. Am J Obstet Gynecol 2005;193(2):577–81. [16] Tjalma WA, Monaghan JM, de Barros Lopes A, Naik R, Nordin AJ, Weyler JJ. The role of surgery in invasive squamous carcinoma of the vagina. Gynecol Oncol 2001; 81(3):360–5. [17] Ling B, Gao Z, Sun M, Sun F, Zhang A, Zhao W, et al. Laparoscopic radical hysterecto- my with vaginectomy and reconstruction of vagina in patients with stage 1 primary vaginal cancer. Gynecol Oncol 2008;109(1):92–6. [18] Chyle V, Zagars GK, Wheeler JA, Wharton JT, Delclos L. Definitive radiotherapy for carcinoma of the vagina: outcome and prognostic factors. Int J Radiat Oncol Biol Phys 1996;35(5):891–905. [19] Perez CA, Grigsby PW, Garipagaoglu M, Mutch DG, Lockett MA. Factors affecting long-term outcome of irradiation in carcinoma of the vagina. Int J Radiat Oncol Biol Phys 1999;44(1):37–45. [20] Kirkbride P, Fyles A, Rawlings GA, Manchul L, Levin W, Murphy KJ, et al. Carcinoma of the vagina: experience at the Princess Margaret Hospital (1974–1989). Gynecol Oncol 1995;56(3):435–43. [21] Dalrymple JL, Russell AH, Lee SW, Scudder SA, Leiserowitz GS, Kinney WK, et al. Chemoradiation for primary invasive squamous carcinoma of the vagina. Int J Gynecol Cancer 2004;14(1):110–7. [22] Frank SJ, Jhingran A, Levenback C, Eifel PJ. Definitive radiation therapy for squa- mous cell carcinoma of the vagina. Int J Radiat Oncol Biol Phys 2005;62(1): 138–47. [23] Rajagopalan MS, Xu KM, Lin JF, Sukumvanich P, Krivak TC, Beriwal S. Adoption and impact of concurrent chemoradiation therapy for vaginal cancer: A National Cancer Data Base (NCDB) study. Gynecol Oncol 2014;135(3): 495–502. [24] Frank SJ, Deavers MT, Jhingran A, Bodurka DC, Eifel PJ. Primary adenocarcinoma of the vagina not associated with diethylstilbestrol (DES) exposure. Gynecol Oncol 2007;105(2):470–4. [25] Herbst AL, Scully RE. Adenocarcinoma of the vagina in adolescence. A report of 7 cases including 6 clear-cell carcinomas (so-called mesonephromas). Cancer 1970; 25(4):745–51. [26] Reid GC, Schmidt RW, Roberts JA, Hopkins MP, Barrett RJ, Morley GW. Primary melanoma of the vagina: a clinicopathologic analysis. Obstet Gynecol 1989;74(2): 190–9. [27] Kirschner AN, Kidd EA, Dewees T, Perkins SM. Treatment approach and outcomes of vaginal melanoma. Int J Gynecol Cancer 2013;23(8):1484–9. S86 N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S84–S87
  • 21. [28] Miner TJ, Delgado R, Zeisler J, Busam, Alektiar K, Barakat R, et al. Primary vaginal melanoma: a critical analysis of therapy. Ann Surg Oncol 2004;11(1): 34–9. [29] Buchanan DJ, Schlaerth J, Kurosaki T. Primary vaginal melanoma: thirteen year disease-free survival after wide local excision and review of recent literature. Am J Obstet Gynecol 1998;178(6):1177–84. [30] Magné N, Haie-Meder C. Brachytherapy for genital-tract rhabdomyosarcomas in girls: technical aspects, reports, and perspectives. Lancet Oncol 2007;8(8): 725–9. [31] Magné N, Oberlin O, Martelli H, Gerbaulet A, Chassagne D, Haie-Meder C. Vulval and vaginal rhabdomyosarcoma in children: The Institute Gustave Roussy brachytherapy experience with particular attention on long-term outcomes. Int J Gynecol Cancer 2006;16(Suppl. 3):610 (Abst 0038). [32] Arndt CA, Donaldson SS, Anderson JR, Andrassy RJ, Laurie F, Link MP, et al. What constitutes optimal therapy for patients with rhabdomyosarcoma of the female genital tract. Cancer 2001;91(12):2454–68. S87N.F. Hacker et al. / International Journal of Gynecology and Obstetrics 131 (2015) S84–S87
  • 22. FIGO CANCER REPORT 2015 Cancer of the cervix uteri Adriana Bermudez a , Neerja Bhatla b , Eric Leung c a Gynecologic Oncology Unit, Buenos Aires University Hospital, Buenos Aires, Argentina b Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, New Delhi, India c Department of Radiation Oncology, Sunnybrook Health Sciences Center, Odette Cancer Centre, Toronto, Ontario, Canada 1. Introduction Worldwide, cervical cancer is now the fourth most common female malignancy in both incidence and mortality, following breast, colorectal, and lung cancers, and results in approximately 527 600 new cases and 265 700 deaths annually [1]. It is the second most commonly diagnosed cancer and third most common cause of cancer death among females in low-resource countries. More than 85% of new cases are diagnosed in economically disadvantaged people. Nearly 90% of cervical cancer deaths occur in low-resource regions of the world. 1.1. Anatomy The cervix is the lower aspect of the uterus. It is roughly cylindrical in shape, projects through the superior-anterior vaginal wall, and communicates with the vagina through the endocervical canal, which terminates in the external os located at the top of the vagina. Cancer of the cervix may originate from the mucosa of the surface of the cervix or from within the canal. Carcinoma of the uterine cervix grows locally and may extend in continuity to the uterus and paracervical tissues, and pelvic organs. Cervical cancer may spread to regional lymph nodes, and only later metastasize hematogenously to distant structures. Studies on sentinel lymph nodes show that the cervix is drained into the following first echelon nodal stations most commonly: external iliac (43%), obturator (26%) and parametrial (21%), from where they drain to the common iliac nodes. From the common iliac nodes, lymph drainage goes to the para-aortic nodes. The most common sites of distant spread include the para-aortic, mediastinal and supraclavicular nodes, the lungs, liver, and skeleton. 2. Staging FIGO staging is based on clinical examination. The FIGO staging guidelines were most recently updated in 2009 (Table 1) [2]. Stage 0 is no longer included in the FIGO 2009 staging. A thorough pelvic examination is mandatory to provide information for FIGO staging, and this rarely requires anesthesia. When there is doubt as to which stage a particular cancer should be allocated, the ear- lier stage is mandatory. The following examinations are permitted for the determination of FIGO staging, as indicated by presenting characteristics (see sections below): palpation, inspection, colposcopy, endocervical curettage, hys- teroscopy, cystoscopy, proctoscopy, intravenous pyelography, ultra- sound of the renal tract, and X-ray examination of the lungs and skeleton. Blood tests should include full blood count, and renal and liver functions. Syphilis and HIV serology need to be considered, based on discussion with the patient about risk factors. 2.1. Initial assessment of microinvasive disease The diagnosis of both Stage IA1 and IA2 should be based on micro- scopic examination of removed tissue, preferably a cone biopsy, which must include the entire lesion. The depth of invasion should not be greater than 5 mm taken from the base of the epithelium, either surface or glandular, from which it originates. The second dimension, the hori- zontal spread, must not exceed 7 mm. Vascular space involvement, either venous or lymphatic, should not alter the staging, but should be specifically recorded because it may affect treatment decisions. Macro- scopically obvious lesions, and those with larger dimensions, should be staged as IB. It is impossible to clinically determine if a cancer of the cervix has extended to the corpus. Extension to the corpus should therefore be disregarded for staging purposes. The diagnosis of Stage IA1 or IA2 disease can only be made on the basis of a cone biopsy with negative margins, or on a trachelectomy or hysterectomy specimen. If the margins of the cone biopsy are positive for cervical intraepithelial neoplasia (CIN) III or invasive cancer, a sec- ond cone biopsy should be performed or the patient treated as for Stage IB1 disease [3]. 2.2. Initial evaluation of grossly invasive disease Visible lesions require a biopsy to confirm a diagnosis of cervical carcinoma. A patient with a growth apparently fixed to the pelvic wall by a short and indurated, but not nodular, parametrium should be allotted to Stage IIB. Stage III should be defined for cases where the parametrium is nodular to the pelvic wall or if the growth itself extends to the pelvic wall. The presence of hydronephrosis or non-functioning kidney(s) resulting from obstruction of the ureter(s) by cancer also per- mits a case to be allotted to Stage III. International Journal of Gynecology and Obstetrics 131 (2015) S88–S95 http://dx.doi.org/10.1016/j.ijgo.2015.06.004 0020-7292/© 2015 Published by Elsevier Ireland Ltd. on behalf of International Federation of Gynecology and Obstetrics. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Contents lists available at ScienceDirect International Journal of Gynecology and Obstetrics journal homepage: www.elsevier.com/locate/ijgo
  • 23. In cases of grossly invasive disease, a chest X-ray, and evaluation of hydronephrosis (with renal ultrasound, intravenous pyelography, CT, or MRI) are mandatory. The bladder and rectum are evaluated by cys- toscopy and sigmoidoscopy only if the patient is clinically symptomatic. Cystoscopy is also recommended in cases of endocervical growth with a barrel-shaped surface and in cases where the growth has extended to the anterior vaginal wall. Suspected bladder or rectal involvement should be confirmed by biopsy and histologic evidence. The presence of bullous edema, as such, should not permit a case to be allotted to Stage IV. Imaging evaluation may be of additional benefit to clinical examina- tion in practice areas where resources allow. Imaging may allow for identification of additional prognostic factors and help direct selection of therapy. MRI provides the best radiologic assessment of primary tumors greater than 10 mm, but is not mandatory [4–8]. Level of Evidence B CT and/or MRI and/or positron emission tomography (PET) may provide information on nodal status or systemic spread, but are not mandatory. Compared with CT and MRI, PET-CT is a more accurate im- aging method for detecting nodal metastasis that are greater than 10 mm [5,9–12]. Isolated and unexpected areas of PET enhancement should be further investigated with tissue diagnosis, if possible, to con- firm or exclude the presence of distant metastatic disease [11,13,14]. Level of Evidence B Compared with radiologic evaluation, surgical node dissection is more accurate for assessment of para-aortic nodal disease [15,16]. In pa- tients with advanced disease, laparoscopic staging of para-aortic lymph nodes may be considered to allow treatment according to extent of disease [17]. No impact on survival has been demonstrated; however, surgical exclusion of para-aortic lymph node involvement portends a better prognosis than radiographic exclusion alone [18]. Level of Evidence B In a review of 22 articles that evaluated the safety and impact of pre- treatment surgical para-aortic lymph node staging (PALNS), para-aortic lymph node metastases were found in 18% (range, 8%−42%) of patients with cervical cancer Stage IB-IVA [19]. The mean complication rate of PALNS was 9% (range, 4%−24%), the most common complication being lymphocysts. PET-CT appears to be the most accurate imaging method, with false-negative results in 4%−15% of cases. Positive para- aortic nodes have been identified in up to 35% of Stage IIB and 20% of Stage III tumors [19]. Knowing the status of para-aortic nodes may pro- vide prognostic information as well as guide the extent of adjuvant or primary radiation. It is, however, controversial and is not recommended as a routine practice, particularly in resource-restricted environments and in women with advanced disease associated with constitutional symptoms. 2.3. Pathologic staging In cases treated by surgical procedures, the pathologist’s findings in the removed tissues can be the basis for accurate statements on the ex- tent of disease. The findings should not be allowed to change the clinical staging, but should be recorded in the manner described for the patho- logic staging of disease. The TNM nomenclature is appropriate for this purpose [20]. Unlike FIGO staging criteria, TNM staging accounts for node positivity; however, the FIGO and TNM classifications are other- wise virtually identical in describing the anatomical extent of disease. Clinical staging is essential to select and evaluate therapy, while the pathological stage provides the most precise data from which to esti- mate prognosis and calculate end results. Infrequently, hysterectomy may be carried out in the presence of un- suspected invasive cervical carcinoma. Such cases cannot be clinically staged or included in therapeutic statistics, but it is desirable that they be reported separately. If considered appropriate, some of these patients may be offered repeat laparotomy with full parametrectomy and pelvic lymphadenectomy to allow potentially curative surgery and/or deter- mine the need for adjuvant chemoradiation [21]. Staging is determined at the time of the primary diagnosis and can- not be altered, even at recurrence. Only if the rules for clinical staging are strictly observed is it possible to compare results among clinics and by differing modes of therapy. 2.4. Histopathology All tumors must be microscopically verified. Cases should be clas- sified as carcinomas of the cervix if the primary growth is in the cervix. All histologic types must be included. The histopathologic types are: • Squamous cell carcinoma (keratinizing; non-keratinizing; verrucous). • Endometrioid adenocarcinoma. • Clear cell adenocarcinoma. • Adenosquamous carcinoma. • Adenoid cystic carcinoma. • Small cell carcinoma. • Undifferentiated carcinoma. Table 1 Cancer of the cervix uteri. Stage Description I The carcinoma is strictly confined to the cervix (extension to the uterine corpus should be disregarded). IA Invasive cancer identified only microscopically. (All gross lesions even with superficial invasion are Stage IB cancers.) Invasion is limited to measured stromal invasion with a maximum depth of 5 mm and no wider than 7 mm. IA1 Measured invasion of stroma ≤ 3 mm in depth and ≤ 7 mm width. IA2 Measured invasion of stroma N 3 mm and b 5 mm in depth and ≤ 7 mm width. IB Clinical lesions confined to the cervix, or preclinical lesions greater than stage IA. IB1 Clinical lesions no greater than 4 cm in size. IB2 Clinical lesions N 4 cm in size. II The carcinoma extends beyond the uterus, but has not extended onto the pelvic wall or to the lower third of vagina. IIA Involvement of up to the upper 2/3 of the vagina. No obvious parametrial involvement. IIA1 Clinically visible lesion ≤ 4 cm IIA2 Clinically visible lesion N 4 cm IIB Obvious parametrial involvement but not onto the pelvic sidewall. III The carcinoma has extended onto the pelvic sidewall. On rectal examination, there is no cancer free space between the tumor and pelvic sidewall. The tumor involves the lower third of the vagina. All cases of hydronephrosis or non-functioning kidney should be included unless they are known to be due to other causes. IIIA Involvement of the lower vagina but no extension onto pelvic sidewall. IIIB Extension onto the pelvic sidewall, or hydronephrosis/non-functioning kidney. IV The carcinoma has extended beyond the true pelvis or has clinically involved the mucosa of the bladder and/or rectum. IVA Spread to adjacent pelvic organs. IVB Spread to distant organs. S89A. Bermudez et al. / International Journal of Gynecology and Obstetrics 131 (2015) S88–S95
  • 24. Grading by any of several methods is encouraged, but is not a basis for modifying the stage groupings. Histopathologic grades are as follows: • GX: Grade cannot be assessed. • G1: Well differentiated. • G2: Moderately differentiated. • G3: Poorly or undifferentiated. When surgery is the primary treatment, the histologic findings per- mit the case to have pathologic staging, as described above. In this situ- ation, the TNM nomenclature may be used. 3. Cervical cancer screening Primary prevention of cervical cancer through HPV vaccination of girls, and secondary prevention through the detection of cervical cancer precursors by various screening methods and their appropriate treatment, are both known to be effective preventive measures. Details on cervical cancer screening can be accessed via the FIGO website (www.figo.org). 4. Management of cervical cancer 4.1. Microinvasion 4.1.1. Stage IA1 Conization is the treatment of choice for this stage. If the patient has completed childbearing, hysterectomy (abdominal, vaginal, or laparo- scopic) may be considered [22]. Follow-up with Pap smears every 3 months for 2 years, and then every 6 months for a further 3 years should be performed. If follow-up is normal at 5 years, the screening schedule may be complet- ed according to the recommendations in each country [23,24]. Level of Evidence C 4.1.2. Stage IA2 Since lymph nodes may be involved in this stage, lymphadenectomy is necessary [25,26]. The recommended treatment is type 2 radical hysterectomy (ligation of the uterine artery where it crosses the ureter, although a vaginal cuff is not necessary) with pelvic lymphadenectomy. If fertility is desired, options are: (1) cervical conization with extra- peritoneal or laparoscopic pelvic lymphadenectomy; or (2) radical abdominal, vaginal, or endoscopic trachelectomy with pelvic lymphad- enectomy performed according to the surgical approach [27,28]. 4.1.3. Post-treatment follow-up after microinvasive carcinoma Follow-up with Pap smears every 3 months for 2 years, and then every 6 months for a further 3 years should be performed. If follow-up is normal at 5 years, the screening schedule may be complet- ed according to the recommendations in each country [23,24]. Level of Evidence C 4.2. Grossly invasive cervical carcinoma (FIGO Stage IB–IVA) Concurrent platinum-based chemoradiation is the most indicated treatment for this stage although neoadjuvant chemotherapy may play a role in selected settings [29]. The treatment approach should be decided based on the availability of resources, and tumor- and patient-related factors. 4.2.1. Surgical management Surgical treatment may be indicated in Stage IB1–IIA1 disease: mod- ified radical or radical (abdominal or endoscopic) hysterectomy with pelvic lymphadenectomy [30–32]. Level of Evidence B Primary pelvic exenteration may be considered for Stage IVA disease without extension to the pelvic sidewall or extra-pelvic disease [33–46]. Level of Evidence C 4.2.1.1. Sentinel lymph node assessment Identification of sentinel lymph nodes can be performed with dual labeling using blue dye and radiocolloid [47–49]. These procedures may be considered in early stage cervical cancer, Stage IA and IB1 [50–52]. If lymphovascular space invasion is present, pelvic lymphadenecto- my needs to be considered. Level of Evidence C Sentinel lymph node assessment of pelvic lymph nodes should not be utilized in advanced disease [53]. 4.2.1.2. Trend to lesser surgery for small tumors Stages IA2–IB1 with tumor size of less than 2 cm, cervical stromal in- vasion of less than 50%, and node negative on MR/CT imaging have been considered as low risk. Simple hysterectomy or trachelectomy, with either pelvic lymphad- enectomy or sentinel lymph node assessment, have been considered as adequate surgical treatment for low-risk cases [54,55]. Level of Evidence D 4.2.1.3. Adjuvant radiation/chemotherapy The risk of recurrence after radical surgery is increased in the pres- ence of positive nodes, positive parametria, or positive surgical margins. Adjuvant concurrent chemoradiation (cisplatin with or without 5- fluorouracil) improves overall survival, progression-free survival, and both local and distant recurrences compared with pelvic irradiation alone in such patients [42]. Level of Evidence B Risk of pelvic recurrence is also increased in those with uninvolved nodes but with primary associated risk factors: tumor size greater than 4 cm, capillary-like space (CLS) involvement, and outer one-third invasion of the cervical stroma [43,44]. Adjuvant whole pelvic chemo- irradiation reduces the local failure rate and improves progression- free survival compared with patients treated with surgery alone [43]. Level of Evidence B Adjuvant radiation therapy with and without chemotherapy may be particularly beneficial for patients with adenocarcinoma or adenosquamous histology, given the relatively higher rates of distant failure [42,43]. Level of Evidence C Patients with positive common iliac or para-aortic nodes may be treated by extended field radiation [56,57], with or without chemother- apy. Level of Evidence C Intensity modulated radiation therapy has been explored in the postoperative setting. A prospective multi-institutional study has shown acceptable toxicities with this approach and a randomized trial (TIME-C) is underway comparing intensity-modulated radiation thera- py (IMRT) with standard field-based radiation therapy in postoperative cervical and endometrial cancer [58,59]. Although there is currently insufficient evidence at the present time to recommend IMRT as a stan- dard of care, many centers have shifted to using this technique in post- operative cervical and endometrial cancer treatment. 4.2.2. Neoadjuvant chemotherapy and surgery The theoretical rationale for the use of neoadjuvant chemotherapy (NACT) includes the induction of tumor shrinkage to facilitate radical excision, and a possible improvement in outcomes over surgery alone. There is also a possibility of NACT sterilizing nodes and parametria, thereby reducing risk factors for adjuvant therapy after surgery; howev- er, the efficacy of neoadjuvant therapy in this situation is not known. A meta-analysis of randomized trials of neoadjuvant platinum- based chemotherapy prior to definitive surgery shows that patients treated with NACT have better survival outcomes than those treated with primary radiation alone, given at a relatively low dose [60]. No ran- domized data compare the results of NACT followed by surgery with concurrent chemoradiation. The European Organization for Research and Treatment of Cancer is currently conducting a Phase 3 study com- paring NACT and surgery with definitive chemoradiation in patients with FIGO Stages IB2, IIA2, or IIB cervical cancers. S90 A. Bermudez et al. / International Journal of Gynecology and Obstetrics 131 (2015) S88–S95