You are on page 1of 128

143 Volume 143 Supplement 2 October 2018 ISSN 0020-7292

S2

International Journal of Gynecology & Obstetrics Vol. 143/S2 (2018) 1–158


International Journal of

GYNECOLOGY
&OBSTETRICS
International Journal of

EDITORIAL BOARD GYNECOLOGY


OBSTETRICS
&
Editor: R.M. Adanu (Ghana)
Editors Emeritus: T.R.B. Johnson (USA)
J.J. Sciarra (USA)

Regional representa ves:


Africa–Middle East: V. Boama (Qatar)
Asia–Oceania: V. Guinto (Philippines)
P.P. Fogarty (Malaysia)
Europe: M. Geary (Ireland)
Laࢼn America: C. Sosa (Uruguay)
North America: E. Stringer (USA)

Public Health Obstetric Consultant: E. Stringer (USA)

Ethical and Legal Issues in B. Dickens (Canada)


Reproduc ve Health: R. Cook (Canada)
C. Nwenga (South Africa)

Contemporary Issues in V. Boama (Qatar)


Women’s Health: M. Geary (Ireland)
V. Guinto (Philippines)
C. Sosa (Uruguay)

FIGO Cancer Staging: N. Bhatla (India)

Sta s cal Consultant: A. Vahra an (USA)

Managing Editor: Z. Nyakoojo (UK)


Manuscript Editor: S. Fitzpatrick (UK)
Editorial & Marke ng Assistant: H. Roberts (UK)

Editorial Office: FIGO Secretariat, FIGO House


Suite 3 - Waterloo Court, FIGO Cancer Report 2018
10 Theed Street,
London, SE1 8ST, UK Guest Editors: Neerja Bhatla and Lynette Denny
Tel: +44 20 7928 1166
Fax: +44 20 7928 7099
E-mail: ijgo@figo.org

This journal is available online at Wiley Online Library. Visit


wileyonlinelibrary.com to search the articles and register
for table of contents and e-mail alerts.

IJGO_v143_s2_cover.indd 1 31-08-2018 20:06:13


143 Volume 143 Supplement 2 October 2018 ISSN 0020-7292
S2

International Journal of Gynecology & Obstetrics Vol. 143/S2 (2018) 1–158


International Journal of

GYNECOLOGY
&OBSTETRICS
International Journal of

EDITORIAL BOARD GYNECOLOGY


OBSTETRICS
&
Editor: R.M. Adanu (Ghana)
Editors Emeritus: T.R.B. Johnson (USA)
J.J. Sciarra (USA)

Regional representa ves:


Africa–Middle East: V. Boama (Qatar)
Asia–Oceania: V. Guinto (Philippines)
P.P. Fogarty (Malaysia)
Europe: M. Geary (Ireland)
Laࢼn America: C. Sosa (Uruguay)
North America: E. Stringer (USA)

Public Health Obstetric Consultant: E. Stringer (USA)

Ethical and Legal Issues in B. Dickens (Canada)


Reproduc ve Health: R. Cook (Canada)
C. Nwenga (South Africa)

Contemporary Issues in V. Boama (Qatar)


Women’s Health: M. Geary (Ireland)
V. Guinto (Philippines)
C. Sosa (Uruguay)

FIGO Cancer Staging: N. Bhatla (India)

Sta s cal Consultant: A. Vahra an (USA)

Managing Editor: Z. Nyakoojo (UK)


Manuscript Editor: S. Fitzpatrick (UK)
Editorial & Marke ng Assistant: H. Roberts (UK)

Editorial Office: FIGO Secretariat, FIGO House


Suite 3 - Waterloo Court, FIGO Cancer Report 2018
10 Theed Street,
London, SE1 8ST, UK Guest Editors: Neerja Bhatla and Lynette Denny
Tel: +44 20 7928 1166
Fax: +44 20 7928 7099
E-mail: ijgo@figo.org

This journal is available online at Wiley Online Library. Visit


wileyonlinelibrary.com to search the articles and register
for table of contents and e-mail alerts.

IJGO_v143_s2_cover.indd 1 31-08-2018 20:06:13


SUMMARY OF AUTHOR GUIDELINES
(Full guidelines are available online at h p://obgyn.onlinelibrary.wiley.com/hub/journal/10.1002/(ISSN)1879-3479/about/author-guidelines.html)

FIGO Officers
President: C.N. Purandare (India)
INTRODUCTION LAYOUT OF MANUSCRIPTS
Vice-President: S. Grenman (Finland) Only original manuscripts that are submi ed Please refer to the online instruc ons for full
President-Elect: C. Füchtner (Bolivia) exclusively to IJGO will be considered for publica on. details on how to structure your manuscript. All
Honorary Treasurer: R. Hale (USA)
The requirements of IJGO are in accordance with the submissions must be accompanied by a cover
Honorary Secretary: G.C. Di Renzo (Italy)
Officer: Yirgu G. Ferede (Ethiopia) “Uniform Requirements for Manuscripts Submi ed to le er addressed to the Editor. The first page of the
FIGO Chief Execu ve J. Vos (UK)
Biomedical Journals” published by the Interna onal manuscript should contain the following: (1) tle; (2)
Commi ee of Medical Journal Editors (ICMJE). full names of authors; (3) affilia ons of authors; (4)
FIGO Execu ve Board
Australia & New Zealand C. Tippe India H. Divakar Editorial processes will adhere to the Commi ee of full contact details of the corresponding author; (5)
Belgium J. Van Wiemeersch Japan T. Kimura Publica on Ethics guidelines. a list of up to 8 keywords for indexing and retrieval;
Bolivia D. Mostajo Flores Kenya R.J. Kosgie (6) a brief synopsis.
Brazil C.E. Fernandes Lebanon F. El-Kak SUBMISSION Clinical Ar cles and Review Ar cles require
Canada J. Blake Mexico E.C. Morales
Chile M. Cuello Fredes Norway M. Steinholt
IJGO will consider for publica on unsolicited an abstract of 200 words or fewer; Brief
Colombia J.D. Villegas Echeverri Peru A. Celis submissions of the following: Clinical Ar cles; Communica ons do not require an abstract.
Denmark A.T. Pedersen South Korea Y.T. Kim Review Ar cles; and Brief Communica ons Full length ar cles should be structured with
Egypt N.A. Darwish Sri Lanka H. Perera
United Kingdom D. Richmond
(including Case Reports). All manuscripts should the following headings: Introduc on; Materials
Ethiopia D. Negussie
France P. Descamps United States of America J.N. Mar n be prepared according to the author guidelines and methods; Results; Discussion; Author
Germany F. Louwen Uruguay J.G. Alonso Tellechea available online. Manuscripts are submi ed to the contribu ons; Acknowledgments; Conflicts of
For informa on 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; journal via the online service Editorial Manager interest; and References. Brief Communica ons
fax: +44 20 7928 7099. Email: figo@figo.org. Website: www.figo.org. All enquiries concerning FIGO may be sent to the Honorary Secretary at that address. (www.editorialmanager.com/IJG). Authors must first do not require headings for the main text, but
register for an account on Editorial Manager, ensuring should include the following sec ons: Author
INTERNATIONAL JOURNAL OF GYNECOLOGY & OBSTETRICS (Print ISSN: 0020-7292; Online Disclaimer
that the details provided for the corresponding contribu ons; Acknowledgments; Conflicts of
ISSN: 1879-3479), is published monthly. US mailing agent: Mercury Media Processing, LLC, The Publisher, FIGO: The Interna onal Federa on of Gynecology and Obstetrics and Editors author are up-to-date so that all manuscript status interest; and References. Tables, figures, and
1850 Elizabeth Avenue, Suite #C, Rahway, NJ 07065 USA. Periodical postage paid at Rahway, cannot be held responsible for errors or any consequences arising from the use of informa on
NJ. contained in this journal; the views and opinions expressed do not necessarily reflect those of
no fica ons are successfully received. Authors can suppor ng informa on may also be submi ed.
the Publisher, FIGO: The Interna onal Federa on of Gynecology and Obstetrics and Editors, check the status of their manuscript during the
Postmaster: Send all address changes to INTERNATIONAL JOURNAL OF GYNECOLOGY & OB-
STETRICS, John Wiley & Sons Inc., C/O The Sheridan Press, PO Box 465, Hanover, PA 17331
neither does the publica on of adver sements cons tute any endorsement by the Publisher,
FIGO: The Interna onal Federa on of Gynecology and Obstetrics and Editors of the products
review process by logging into Editorial Manager. MANUSCRIPT CONTENTS
USA. adver sed. Queries rela ng to the submission process or to Where appropriate, statements on clinical trial
Copyright and Copying Publisher journal procedures can be directed to the editorial registra on, Ethics Commi ee approval, and
Copyright © 2018 International Federation of Gynecology and Obstetrics. All rights INTERNATIONAL JOURNAL OF GYNECOLOGY & OBSTETRICS is published by John Wiley &
office at ijgo@figo.org. informed consent should be included in the
reserved. No part of this publication may be reproduced, stored or transmitted in Sons Ltd., 9600 Garsington Road, Oxford, OX4 2DQ, UK. Tel: +44 (0)1865 776868; fax: +44
any form or by any means without the prior permission in writing from the copyright (0)1865 714591. manuscript. Details of any sta s cal analysis
holder. Authorization to copy items for internal and personal use is granted by the Journal Customer Services: For ordering informa on, claims and any enquiry concerning your
JOURNAL REQUIREMENTS conducted should be given in the methods. In line
copyright holder for libraries and other users registered with their local Reproduction journal subscrip on please go to www.wileycustomerhelp.com/ask or contact your nearest Studies of pa ents, pa ent records, or volunteers with ICMJE standards, author contribu ons must be
Rights Organisation (RRO), e.g. Copyright Clearance Center (CCC), 222 Rosewood office.
Drive, Danvers, MA 01923, USA (www.copyright.com), provided the appropriate fee Americas: Email: cs-journals@wiley.com; Tel: +1 781 388 8598 or +1 800 835 6770 (toll free
require Ethics Commi ee approval and informed listed and a conflicts of interest statement be made.
is paid directly to the RRO. This consent does not extend to other kinds of copying in the USA & Canada). consent. In compliance with ICMJE requirements, References should be presented as per the journal’s
such as copying for general distribution, for advertising or promotional purposes, for
creating new collective works or for resale. Special requests should be addressed to:
Europe, Middle East and Africa: Email: cs-journals@wiley.com; Tel: +44 (0) 1865 778315.
all clinical trials must be prospec vely registered style. The editors reserve the right to make any
Asia Pacific: Email: cs-journals@wiley.com; Tel: +65 6511 8000.
permissions@wiley.com Japan: For Japanese speaking support, Email: cs-japan@wiley.com. in a public trials registry. Purely observa onal necessary editorial changes.
Delivery Terms and Legal Title Visit our Online Customer Help available in 7 languages at www.wileycustomerhelp.com/ask studies (those in which the assignment of the
Where the subscrip on price includes print issues and delivery is to the recipient’s address,
delivery terms are Delivered at Place (DAP); the recipient is responsible for paying any import
Produc on Editor: Ika Lestari (email: ijgo@wiley.com).
medical interven on is not at the discre on of the LICENSING
Wiley’s Corporate Ci zenship ini a ve seeks to address the environmental, social, economic,
duty or taxes. Title to all issues transfers Free of Board (FOB) our shipping point, freight pre- and ethical challenges faced in our business and which are important to our diverse stakeholder
inves gator) will not require registra on. Authors On acceptance, authors have the op on of
paid. We will endeavour to fulfil claims for missing or damaged copies within six months of groups. Since launching the ini a ve, we have focused on sharing our content with those in wishing to use material that has already been publishing their manuscript under the terms of the
publica on, within our reasonable discre on and subject to availability. need, enhancing community philanthropy, reducing our carbon impact, crea ng global guide-
published must first obtain the permission of the journal’s standard copyright transfer agreement, or
Informa on for subscribers lines and best prac ces for paper use, establishing a vendor code of ethics, and engaging our
Internaࢼonal Journal of Gynecology & Obstetrics is published in 12 issues per year. Ins tu onal colleagues and other stakeholders in our efforts. Follow our progress at www.wiley.com/go/ copyright holder(s). Authors will be expected to under open access terms made available via Wiley
subscrip on prices for 2018 are: ci zenship provide the relevant documenta on on request. OnlineOpen.
Print & Online: US$4,792 (US and Rest of World), €4,358 (Europe), £3,217 (UK). Prices are ISSN 0020-7292 (Print)
exclusive of tax. Asia-Pacific GST, Canadian GST/HST and European VAT will be applied at the ISSN 1879-3479 (Online)
appropriate rates. For more informa on on current tax rates, please go to www.wileyonlineli- Wiley is a founding member of the UN-backed HINARI, AGORA, and OARE ini a ves. They Typeset in India by SPS.
brary.com/tax-vat. The price includes online access to the current and all online back files to are now collec vely known as Research4Life, making online scien fic content available free Printed in the UK by Hobbs the Printers Ltd.
January 1st 2014, where available. For other pricing op ons, including access informa on or at nominal cost to researchers in developing countries. Please visit Wiley’s Content Ac- For submission instruc ons, subscrip on and all other informa on visit: wileyonlinelibrary.com/journal/ijgo
and terms and condi ons, please visit www.wileyonlinelibrary.com/access. cess – Corporate Ci zenship site: h p://www.wiley.com/WileyCDA/Sec on/id-390082.html
Back issues: Single issues from current and recent volumes are available at the current sin- OnlineOpen
INTERNATIONAL JOURNAL OF GYNECOLOGY & OBSTETRICS is indexed by Academic OneFile (GALE Cengage); CAB Abstracts® (CABI); Centre for
gle issue price from cs-journals@wiley.com. Earlier issues may be obtained from Periodicals INTERNATIONAL JOURNAL OF GYNECOLOGY & OBSTETRICS accepts ar cles for Open Access Agriculture and Biosciences Interna onal (CABI); CINAHL: Cumula ve Index to Nursing & Allied Health Literature (EBSCO Publishing); Current
Service Company, 351 Fairview Avenue – Ste 300, Hudson, NY 12534, USA. Tel: +1 518 822- publica on. Please visit h p://olabout.wiley.com/WileyCDA/Sec on/id-828081.html for fur- Contents: Clinical Medicine (Clarivate Analy cs); Embase (Elsevier); Family & Society Studies Worldwide (EBSCO Publishing); Gender Studies
9300, Fax: +1 518 822-9305, Email: psc@periodicals.com ther informa on about OnlineOpen. Collec on (GALE Cengage); Gender Studies Database (EBSCO Publishing); InfoTrac (GALE Cengage); MEDLINE/PubMed (NLM); Science Cita on
Index Expanded (Clarivate Analy cs); SCOPUS (Elsevier); Web of Science (Clarivate Analy cs); Women’s Studies Interna onal (EBSCO Publishing).

IJGO_v143_s2_cover.indd 2 31-08-2018 20:06:14


SUMMARY OF AUTHOR GUIDELINES
(Full guidelines are available online at h p://obgyn.onlinelibrary.wiley.com/hub/journal/10.1002/(ISSN)1879-3479/about/author-guidelines.html)

FIGO Officers
President: C.N. Purandare (India)
INTRODUCTION LAYOUT OF MANUSCRIPTS
Vice-President: S. Grenman (Finland) Only original manuscripts that are submi ed Please refer to the online instruc ons for full
President-Elect: C. Füchtner (Bolivia) exclusively to IJGO will be considered for publica on. details on how to structure your manuscript. All
Honorary Treasurer: R. Hale (USA)
The requirements of IJGO are in accordance with the submissions must be accompanied by a cover
Honorary Secretary: G.C. Di Renzo (Italy)
Officer: Yirgu G. Ferede (Ethiopia) “Uniform Requirements for Manuscripts Submi ed to le er addressed to the Editor. The first page of the
FIGO Chief Execu ve J. Vos (UK)
Biomedical Journals” published by the Interna onal manuscript should contain the following: (1) tle; (2)
Commi ee of Medical Journal Editors (ICMJE). full names of authors; (3) affilia ons of authors; (4)
FIGO Execu ve Board
Australia & New Zealand C. Tippe India H. Divakar Editorial processes will adhere to the Commi ee of full contact details of the corresponding author; (5)
Belgium J. Van Wiemeersch Japan T. Kimura Publica on Ethics guidelines. a list of up to 8 keywords for indexing and retrieval;
Bolivia D. Mostajo Flores Kenya R.J. Kosgie (6) a brief synopsis.
Brazil C.E. Fernandes Lebanon F. El-Kak SUBMISSION Clinical Ar cles and Review Ar cles require
Canada J. Blake Mexico E.C. Morales
Chile M. Cuello Fredes Norway M. Steinholt
IJGO will consider for publica on unsolicited an abstract of 200 words or fewer; Brief
Colombia J.D. Villegas Echeverri Peru A. Celis submissions of the following: Clinical Ar cles; Communica ons do not require an abstract.
Denmark A.T. Pedersen South Korea Y.T. Kim Review Ar cles; and Brief Communica ons Full length ar cles should be structured with
Egypt N.A. Darwish Sri Lanka H. Perera
United Kingdom D. Richmond
(including Case Reports). All manuscripts should the following headings: Introduc on; Materials
Ethiopia D. Negussie
France P. Descamps United States of America J.N. Mar n be prepared according to the author guidelines and methods; Results; Discussion; Author
Germany F. Louwen Uruguay J.G. Alonso Tellechea available online. Manuscripts are submi ed to the contribu ons; Acknowledgments; Conflicts of
For informa on 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; journal via the online service Editorial Manager interest; and References. Brief Communica ons
fax: +44 20 7928 7099. Email: figo@figo.org. Website: www.figo.org. All enquiries concerning FIGO may be sent to the Honorary Secretary at that address. (www.editorialmanager.com/IJG). Authors must first do not require headings for the main text, but
register for an account on Editorial Manager, ensuring should include the following sec ons: Author
INTERNATIONAL JOURNAL OF GYNECOLOGY & OBSTETRICS (Print ISSN: 0020-7292; Online Disclaimer
that the details provided for the corresponding contribu ons; Acknowledgments; Conflicts of
ISSN: 1879-3479), is published monthly. US mailing agent: Mercury Media Processing, LLC, The Publisher, FIGO: The Interna onal Federa on of Gynecology and Obstetrics and Editors author are up-to-date so that all manuscript status interest; and References. Tables, figures, and
1850 Elizabeth Avenue, Suite #C, Rahway, NJ 07065 USA. Periodical postage paid at Rahway, cannot be held responsible for errors or any consequences arising from the use of informa on
NJ. contained in this journal; the views and opinions expressed do not necessarily reflect those of
no fica ons are successfully received. Authors can suppor ng informa on may also be submi ed.
the Publisher, FIGO: The Interna onal Federa on of Gynecology and Obstetrics and Editors, check the status of their manuscript during the
Postmaster: Send all address changes to INTERNATIONAL JOURNAL OF GYNECOLOGY & OB-
STETRICS, John Wiley & Sons Inc., C/O The Sheridan Press, PO Box 465, Hanover, PA 17331
neither does the publica on of adver sements cons tute any endorsement by the Publisher,
FIGO: The Interna onal Federa on of Gynecology and Obstetrics and Editors of the products
review process by logging into Editorial Manager. MANUSCRIPT CONTENTS
USA. adver sed. Queries rela ng to the submission process or to Where appropriate, statements on clinical trial
Copyright and Copying Publisher journal procedures can be directed to the editorial registra on, Ethics Commi ee approval, and
Copyright © 2018 International Federation of Gynecology and Obstetrics. All rights INTERNATIONAL JOURNAL OF GYNECOLOGY & OBSTETRICS is published by John Wiley &
office at ijgo@figo.org. informed consent should be included in the
reserved. No part of this publication may be reproduced, stored or transmitted in Sons Ltd., 9600 Garsington Road, Oxford, OX4 2DQ, UK. Tel: +44 (0)1865 776868; fax: +44
any form or by any means without the prior permission in writing from the copyright (0)1865 714591. manuscript. Details of any sta s cal analysis
holder. Authorization to copy items for internal and personal use is granted by the Journal Customer Services: For ordering informa on, claims and any enquiry concerning your
JOURNAL REQUIREMENTS conducted should be given in the methods. In line
copyright holder for libraries and other users registered with their local Reproduction journal subscrip on please go to www.wileycustomerhelp.com/ask or contact your nearest Studies of pa ents, pa ent records, or volunteers with ICMJE standards, author contribu ons must be
Rights Organisation (RRO), e.g. Copyright Clearance Center (CCC), 222 Rosewood office.
Drive, Danvers, MA 01923, USA (www.copyright.com), provided the appropriate fee Americas: Email: cs-journals@wiley.com; Tel: +1 781 388 8598 or +1 800 835 6770 (toll free
require Ethics Commi ee approval and informed listed and a conflicts of interest statement be made.
is paid directly to the RRO. This consent does not extend to other kinds of copying in the USA & Canada). consent. In compliance with ICMJE requirements, References should be presented as per the journal’s
such as copying for general distribution, for advertising or promotional purposes, for
creating new collective works or for resale. Special requests should be addressed to:
Europe, Middle East and Africa: Email: cs-journals@wiley.com; Tel: +44 (0) 1865 778315.
all clinical trials must be prospec vely registered style. The editors reserve the right to make any
Asia Pacific: Email: cs-journals@wiley.com; Tel: +65 6511 8000.
permissions@wiley.com Japan: For Japanese speaking support, Email: cs-japan@wiley.com. in a public trials registry. Purely observa onal necessary editorial changes.
Delivery Terms and Legal Title Visit our Online Customer Help available in 7 languages at www.wileycustomerhelp.com/ask studies (those in which the assignment of the
Where the subscrip on price includes print issues and delivery is to the recipient’s address,
delivery terms are Delivered at Place (DAP); the recipient is responsible for paying any import
Produc on Editor: Ika Lestari (email: ijgo@wiley.com).
medical interven on is not at the discre on of the LICENSING
Wiley’s Corporate Ci zenship ini a ve seeks to address the environmental, social, economic,
duty or taxes. Title to all issues transfers Free of Board (FOB) our shipping point, freight pre- and ethical challenges faced in our business and which are important to our diverse stakeholder
inves gator) will not require registra on. Authors On acceptance, authors have the op on of
paid. We will endeavour to fulfil claims for missing or damaged copies within six months of groups. Since launching the ini a ve, we have focused on sharing our content with those in wishing to use material that has already been publishing their manuscript under the terms of the
publica on, within our reasonable discre on and subject to availability. need, enhancing community philanthropy, reducing our carbon impact, crea ng global guide-
published must first obtain the permission of the journal’s standard copyright transfer agreement, or
Informa on for subscribers lines and best prac ces for paper use, establishing a vendor code of ethics, and engaging our
Internaࢼonal Journal of Gynecology & Obstetrics is published in 12 issues per year. Ins tu onal colleagues and other stakeholders in our efforts. Follow our progress at www.wiley.com/go/ copyright holder(s). Authors will be expected to under open access terms made available via Wiley
subscrip on prices for 2018 are: ci zenship provide the relevant documenta on on request. OnlineOpen.
Print & Online: US$4,792 (US and Rest of World), €4,358 (Europe), £3,217 (UK). Prices are ISSN 0020-7292 (Print)
exclusive of tax. Asia-Pacific GST, Canadian GST/HST and European VAT will be applied at the ISSN 1879-3479 (Online)
appropriate rates. For more informa on on current tax rates, please go to www.wileyonlineli- Wiley is a founding member of the UN-backed HINARI, AGORA, and OARE ini a ves. They Typeset in India by SPS.
brary.com/tax-vat. The price includes online access to the current and all online back files to are now collec vely known as Research4Life, making online scien fic content available free Printed in the UK by Hobbs the Printers Ltd.
January 1st 2014, where available. For other pricing op ons, including access informa on or at nominal cost to researchers in developing countries. Please visit Wiley’s Content Ac- For submission instruc ons, subscrip on and all other informa on visit: wileyonlinelibrary.com/journal/ijgo
and terms and condi ons, please visit www.wileyonlinelibrary.com/access. cess – Corporate Ci zenship site: h p://www.wiley.com/WileyCDA/Sec on/id-390082.html
Back issues: Single issues from current and recent volumes are available at the current sin- OnlineOpen
INTERNATIONAL JOURNAL OF GYNECOLOGY & OBSTETRICS is indexed by Academic OneFile (GALE Cengage); CAB Abstracts® (CABI); Centre for
gle issue price from cs-journals@wiley.com. Earlier issues may be obtained from Periodicals INTERNATIONAL JOURNAL OF GYNECOLOGY & OBSTETRICS accepts ar cles for Open Access Agriculture and Biosciences Interna onal (CABI); CINAHL: Cumula ve Index to Nursing & Allied Health Literature (EBSCO Publishing); Current
Service Company, 351 Fairview Avenue – Ste 300, Hudson, NY 12534, USA. Tel: +1 518 822- publica on. Please visit h p://olabout.wiley.com/WileyCDA/Sec on/id-828081.html for fur- Contents: Clinical Medicine (Clarivate Analy cs); Embase (Elsevier); Family & Society Studies Worldwide (EBSCO Publishing); Gender Studies
9300, Fax: +1 518 822-9305, Email: psc@periodicals.com ther informa on about OnlineOpen. Collec on (GALE Cengage); Gender Studies Database (EBSCO Publishing); InfoTrac (GALE Cengage); MEDLINE/PubMed (NLM); Science Cita on
Index Expanded (Clarivate Analy cs); SCOPUS (Elsevier); Web of Science (Clarivate Analy cs); Women’s Studies Interna onal (EBSCO Publishing).

IJGO_v143_s2_cover.indd 2 31-08-2018 20:06:14


DOI: 10.1002/ijgo.12617

FIGO CANCER REPORT 2018

Pathology of cancers of the female genital tract including


molecular pathology

Jaime Prat1,* | David G. Mutch2

1
Department of Pathology, Autonomous
University of Barcelona, Barcelona, Spain Abstract
2
Division of Gynecologic To better understand pathology reports, gynecologic oncologists must be familiar with
Oncology, Department of Obstetrics and
the terminology used in gynecologic pathology. This chapter of the FIGO Cancer
Gynecology, Washington University School of
Medicine, St Louis, MO, USA Report 2018 summarizes the clinical and pathological features of the most common
cancers of the female genital tract, as well as their main molecular genetic alterations.
*Correspondence
Jaime Prat, Autonomous University of In selected cases, an approach for processing surgical specimens is also included.
Barcelona, Medical School - UD Sant Pau,
Barcelona, Spain. KEYWORDS
Email: jpratdl@gmail.com
Cervical cancer; Endometrial cancer; FIGO Cancer Report; Ovarian and fallopian tube cancer;
Vulvar cancer

divided into two groups: keratinizing squamous cell carcinomas


1 |  INTRODUCTION
unrelated to HPV (>70% of cases), and warty and basaloid carci-
nomas, which are strongly associated with high-­risk HPV (<25% of
Pathology reports include not only histopathologic diagnoses, but
cases), mainly HPV16.1,2
also specific information relating to prognosis and treatment. Thus,
pathologists must have sufficient familiarity with the staging classi-
2.1.1.1 | Etiologic factors and precursor lesions
fication and management of gynecologic cancers to assure that their
Keratinizing squamous carcinomas frequently develop in older women
reports communicate clinically relevant information. Likewise, full
(mean age, 76 years), sometimes in the context of long-­standing lichen
comprehension of the pathology report by the gynecologic oncologist
sclerosus. The precursor lesion is referred to as differentiated vulvar
requires familiarity with the terminology used in gynecologic pathol-
intraepithelial neoplasia (dVIN) or VIN simplex (Fig. 1A), which carries
ogy. This chapter summarizes the pathological features of the most
a high risk of cancer development. In contrast, the HPV-­associated
common gynecologic malignancies, as well as an approach for pro-
warty and basaloid carcinomas develop from a precursor lesion called
cessing selected gynecologic surgical specimens.
squamous intraepithelial lesion (SIL) VIN comprising a spectrum of
alterations ranging from low-­grade SIL VIN (VIN1) to high-­grade SIL
VIN (VIN 2–3) (Fig. 1B). Recent proposals from both the International
2 |  VULVA Society for the Study of Vulvovaginal Disease (ISVVD) and the College
of American Pathologists (CAP)/American Society for Colposcopy and
Cervical Pathology (ASCCP) have recommended replacement of the
2.1 | Malignant tumors and premalignant conditions
older three-­tiered system (VIN 1–3) used to describe these lesions
with a two-­tiered system3 (see Table 1 in Rogers and Cuello4—this
2.1.1 | Squamous cell carcinoma
Supplement). HPV-­associated SIL VIN lesions have a low risk of pro-
Carcinoma of the vulva accounts for 4% of all female genital can- gression to invasive carcinomas (approximately 6%), except in older
cers and occurs mainly in women aged over 60 years. Squamous or immunosuppressed women.1,2 These tumors tend to develop in
cell carcinoma is the most common type (86%). These tumors are younger women.

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

Int J Gynecol Obstet 2018; 143 (Suppl. 2): 93–108 wileyonlinelibrary.com/journal/ijgo  |  93
|
94       Prat and Mutch

(A)

F I G U R E   2   Keratinizing squamous cell carcinoma of vulva. Nests


of neoplastic squamous cells, some with keratin pearls, are evident.
(B)

vulva as Stage I carcinomas; tumors extending to adjacent perineal


structures (lower one-­third of the urethra, lower one-­third of the
vagina, or anus) as Stage II; tumors with positive inguinofemoral
lymph nodes as Stage III; and tumors invading the upper two-­thirds
of the urethra, upper two-­thirds of the vagina, distal structures,
or distant metastasis as Stage IV. Tumor grade and number, size,
and location of lymph node metastases determine survival. Well-­
differentiated tumors have a better mean survival, approaching
90% if nodes are negative. Two-­thirds of women with inguinal node
metastases survive 5 years, but only one-­fourth of those with pelvic
node metastases live that long.5
F I G U R E   1   Squamous intraepithelial lesions of the vulva (VIN).
Prognosis correlates with stage of disease and lymph node sta-
(A) VIN, differentiated (simplex) type. The atypia is accentuated in
tus. The number of inguinal lymph nodes with metastases is the most
the basal and parabasal layers. There is striking epithelial maturation
in the superficial layers. (B) HPV-­related high-­grade squamous important single factor. The prognosis of patients with vulvar cancer is
intraepithelial lesion VIN (VIN3). Beneath a hyperkeratotic surface good because presentation in modern times is generally early, with an
there is severe full-­thickness dysplasia. There are numerous mitoses. overall 5-­year survival of 70%.1

2.1.1.2 | Pathology
2.1.2 | Verrucous carcinoma
Squamous intraepithelial lesion VIN may be single or multiple, and
macular, papular, or plaque-­like. Histologic grades are labeled low-­ Vulvar verrucous carcinoma is a distinct variety of squamous cell
grade SIL (VIN 1) corresponding to mild dysplasia, and high-­grade SIL carcinoma that manifests as a large fungating mass resembling a
(VIN 2–3) corresponding to moderate, and severe dysplasia, respec- giant condyloma acuminatum. HPV, usually type 6 or 11, is com-
tively. However, high-­grade SIL (VIN 3)—which includes squamous monly identified. The tumor invades with broad tongues. Verrucous
cell carcinoma in situ [CIS]—is by far the most common. This lesion is ­carcinomas rarely metastasize. Wide local surgical excision is the
treated with wide excision. treatment of choice.
Most tumors are exophytic, but some may be ulcerative.
Microscopically, the tumor is composed of invasive nests of malig-
nant squamous epithelium with central keratin pearls (Fig. 2). The 2.1.3 | Basal cell carcinoma
tumors generally grow slowly, extending to contiguous skin, vagina, Basal cell carcinomas of the vulva are identical to their counterparts
and rectum. Typically, they initially metastasize to superficial ingui- in the skin. They are not associated with HPV, rarely metastasize, and
nal lymph nodes, and then to deep inguinal, femoral, and pelvic are usually cured by surgical excision.
lymph nodes.1,2

2.1.4 | Malignant melanoma
2.1.1.3 | Clinical features
The International Federation of Gynecology and Obstetrics (FIGO) Although uncommon, malignant melanoma is the second most fre-
staging of vulvar cancer defines tumors of any size limited to the quent cancer of the vulva (5%). It occurs in the sixth and seventh
Prat and Mutch |
      95

decades, but occasionally is found in younger women. It is highly 4.2 | Cervical squamous intraepithelial
aggressive, and the prognosis is poor. Management should be accord- neoplasia (SIL)
ing to guidelines for melanoma treatment elsewhere.
Cervical squamous intraepithelial neoplasia SIL (CIN) is a spectrum
of intraepithelial changes that begins with minimal atypia and pro-
2.1.5 | Extramammary Paget disease gresses through stages of greater intraepithelial abnormalities to
invasive squamous cell carcinoma. The terms CIN, dysplasia, CIS,
This disorder usually occurs on the labia majora in older women. The
and squamous intraepithelial lesion (SIL) are commonly used inter-
lesion is large, red, moist, and apparently sharply demarcated. The ori-
changeably1,2 (Fig. 3).
gin of the diagnostic cells (Paget cells) is controversial: they may arise
in the epidermis or epidermally derived adnexal structures.
Intraepidermal Paget disease may have been present for many 4.2.1 | Epidemiology and molecular pathogenesis
years and is often far more extensive throughout the epidermis
HPV infection with persistent expression leads to CIN and cervical
than preoperative biopsies indicate. Unlike Paget disease of the
cancer (Fig. 4). Low-­grade SIL (CIN1) is a permissive infection (i.e. HPV
breast, which is almost always associated with underlying duct
is episomal, freely replicates, and thereby causes cell death). Huge
carcinoma, extramammary Paget disease is less commonly associ-
numbers of virus must accumulate in the cytoplasm before being vis-
ated with carcinoma of the skin adnexa (20%–30% of the time).
ible as a koilocyte (Fig. 3). In most cases of higher-­grade SIL (CIN2-­3),
Metastases rarely occur, so treatment requires wide local excision
viral DNA integrates into the cell genome. Proteins encoded by E6
or simple vulvectomy.1,2
and E7 genes of HPV 16 respectively bind and inactivate p53 and
Rb proteins, thereby invalidating their tumor suppressor functions.

3 | VAGINA After HPV integrates into host DNA, copies of the whole virus do not
accumulate and koilocytes are absent in many cases of high-­grade

3.1 | Malignant tumors of the vagina dysplasia and all invasive cancers. Cells in high-­grade CIN usually con-
tain HPV types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, and 68.
Primary malignant tumors of the vagina are uncommon, constituting Globally, HPV types 16 and 18 are found in 70% of invasive cancers
about 2% of all genital tract tumors. Most (80%) vaginal malignan- with some variation from region to region; the other high-­risk types
cies represent metastatic spread. Tumors confined to the vagina are account for another 25%.7
usually treated by radical hysterectomy and vaginectomy or with
radiation. Squamous cell carcinomas account for over 90% of primary
vaginal malignancies. Prognosis is related to the extent of spread of 4.2.2 | Pathology
the tumor at the time of its discovery. The 5-­year survival rate for
SIL CIN is nearly always a disease of metaplastic squamous epithe-
tumors confined to the vagina (Stage I) is 80%, whereas it is only 20%
lium in the transformation zone. The normal process by which cervi-
for those with extensive spread (Stages III/IV).1
cal squamous epithelium matures is disturbed in CIN, as evidenced
morphologically by changes in cellularity, differentiation, polarity,
nuclear features, and mitotic activity. High-­grade SIL (CIN3) is syn-
3.1.1 | Embryonal rhabdomyosarcoma (sarcoma
onymous with severe dysplasia and CIS. The sequence of histologic
botryoides)
changes from low-­grade SIL (CIN1) to high-­grade SIL (CIN2-­3) is
Embryonal rhabdomyosarcoma occurs almost exclusively in girls shown in Figure 3.1,2
under 4 years of age. It arises in the lamina propria of the vagina and
consists of primitive spindle rhabdomyoblasts, some of which show
cross-­striations. Tumors less than 3 cm in greatest dimension tend to
4.2.3 | Clinical features
be localized and may be cured by wide excision and chemotherapy. The mean age at which women develop SIL (CIN) is 24–27 years for
Larger tumors have often spread to adjacent structures, regional CIN1 and CIN2, and 35–42 years for CIN3. Based on morphologic
lymph nodes, or distant sites. Even in advanced cases, half of patients criteria, half of cases of low-­grade SIL (CIN1) regress, 10% progress
survive with radical surgery and chemotherapy.1,2 to high-­grade SIL (CIN3), and less than 2% become invasive cancer.
The average time for all grades of dysplasia to progress to high-­grade
SIL (CIN3) is about 10 years. At least 20% of cases of high-­grade SIL
4 | CERVIX
(CIN3) progress to invasive carcinoma in that time.1
When SIL (CIN) is discovered, colposcopy delineates the extent of
4.1 | Squamous cell neoplasia
the lesion and indicates areas to be biopsied. Diagnostic endocervical
Cytological screening in high-­resource countries has decreased cervi- curettage also helps to determine if there is endocervical involvement.
cal carcinoma by 50% to 85%; however, worldwide cervical cancer Women with low-­grade SIL (CIN1) are often followed conservatively
remains the fourth most common cancer in women.6 (i.e. repeat Pap smears plus close follow-­up). High-­grade lesions are
|
96       Prat and Mutch

F I G U R E   3   Interrelations of naming systems in precursor cervical lesions. This chart integrates multiple aspects of the disease. It illustrates the
changes in progressively more abnormal disease states and provides translation terminology for the dysplasia/carcinoma in situ (CIS) system, cervical
intraepithelial neoplasia (CIN) system, and The Bethesda system. The scheme also illustrates the corresponding cytologic smear resulting from
exfoliation of the most superficial cells as well as the equivalent histopathologic lesions (top). Abbreviation: SIL, squamous intraepithelial lesion.

treated with excision LEEP (loop electrosurgical excision procedure), that invade less than or equal to 3 mm (Stage IA1) have less than a
cervical conization (removal of a cone of tissue around the external os), 1%–2% risk of lymph node metastases. Conization or simple hyster-
ablation such as laser, cryosurgery, thermal coagulation, or rarely, hys- ectomy generally cures microinvasive cancers less than 3 mm deep.1,5
1
terectomy may be done. The role of LVSI as a prognostic indicator is more controversial than
that of the depth of invasion and lateral extent of tumor. The stroma in
which foci of invasion lie can retract during preparation of the tissue
4.3 | Microinvasive (superficially invasive) squamous
sections for microscopic examination. A clear space can easily be mis-
cell carcinoma
taken for LVSI. Whereas some studies concluded that the presence of
This is the earliest stage (IA) of invasive cervical cancer. In this set- tumor in lymphatic spaces was of no value by itself in predicting which
ting, stromal invasion usually arises from overlying SIL (CIN) (Fig. 5). patients are likely to have lymph node metastases, other studies have
Presently, staging of microinvasive disease is based on width and reported that the presence of LVSI is an important prognostic indicator.1
depth of invasion, defined as follows:

4.4 | Invasive squamous cell carcinoma


1. Invasion less than 3 mm (Stage IA1) or 5 mm (stage IA2) below
the basement membrane.
4.4.1 | Pathology
2. 7-mm maximum lateral extension.
Early stages of cervical cancer are often poorly defined lesions or
The earliest invasive changes (“early stromal invasion” or ESI) nodular and exophytic masses. If the tumor is within the endocervical
appear as tiny irregular epithelial buds emanating from the base of canal, it can be an endophytic mass, which can infiltrate the stroma
CIN3 lesions. These small (<1 mm) tongues of neoplastic epithelial and cause diffuse cervical enlargement. Most tumors are nonkerati-
cells do not affect the prognosis of CIN3 lesions. In the 2009 FIGO nizing, with solid nests of large malignant squamous cells and no more
classification, ESI was excluded from Stage IA1. Some gynecologic than individual cell keratinization. Most remaining cancers show nests
oncologists further limit microinvasive carcinoma to tumors lacking of keratinized cells in concentric whorls, so-­called keratin pearls.
lymphovascular space invasion (LVSI). Stage IA2 tumors are associ- Cervical cancer spreads: (1) by direct extension; (2) by contiguity;
ated with lymph node metastases in about 8% of cases whereas those (3) through lymphatic vessels; and (4) only rarely by the hematogenous
Prat and Mutch |
      97

F I G U R E   5   Superficially invasive squamous cell carcinoma. The


tumor invades less than 5 mm deep and 4 mm wide. This tumor is
Stage IA2 according to FIGO’s classification.

II, 75%; III, 35%; and IV, 10%. About 15% of patients develop recur-
rences on the vaginal vault, bladder, pelvis, or rectum within 2 years
of therapy. Radical hysterectomy is favored for localized tumor, espe-
cially in younger women; radiation therapy, chemotherapy, or combi-
nations of the two are used for more advanced tumors.1,5 (See Bhatla
et al.8 in this Supplement).

F I G U R E   4   Role of HPV in the pathogenesis of cervical neoplasia.


4.5 | Endocervical adenocarcinoma
route. Local extension into surrounding tissues (parametrium) can result This tumor makes up 20% of cervical cancers. The incidence of cer-
in ureteral compression (Stage IIIB); the corresponding clinical com- vical adenocarcinoma has increased recently, with a mean age of
plications are hydroureter, hydronephrosis, and renal failure second- 56 years at presentation. Most tumors (70%) are HPV-­associated
ary to ureteric obstruction—the most common cause (50%) of death adenocarcinomas of endocervical cell (usual) type that may exhibit
in untreated patients. Bladder and rectal involvement (Stage IVA) may foci of adenocarcinoma in situ.1,2 HPV-­unrelated types (gastric-­type
lead to fistula formation. Metastases develop generally in an organized and clear cell carcinoma) are less common and behave more aggres-
manner. First to regional lymph nodes: (1) paracervical; (2) internal iliac sively than HPV-­associated tumors.
(hypogastric) and obturator; (3) external iliac; and (4) finally para-­aortic Adenocarcinoma in situ (AIS) generally arises at the squamocolum-
nodes. Overall, progression to cancer, tumor growth, and spread are rela- nar junction and extends into the endocervical canal. This lesion may
tively slow, since the average age for patients with CIN3 is 35–40 years; not always be contiguous and can be multifocal. Invasive adenocarci-
for Stage IA carcinoma, 43 years; and for Stage IV, 57 years.1,5 noma typically presents as a polypoid or papillary mass. It is often not
visible and presents somewhat later than its squamous counterpart.
Adenocarcinoma of the endocervix spreads by local invasion and lym-
4.4.2 | Clinical features
phatic metastases, and overall survival is somewhat worse than for
HPV testing is the most reliable screening test for detecting cervical squamous carcinoma.
cancer, and is supplanting cytology in some screening algorithms in
women aged over 25 years. Co-­testing with HPV testing and cytol-
4.6 | Cervical cone biopsy/excision and
ogy is also recommended in women aged over 30 years. Where HPV
trachelectomy
testing is not available, the Pap smear remains the most commonly
used screening test, but quality assurance is a vital component of such Cone biopsy is the standard procedure performed for women with
screening programs. high-­grade CIN and glandular lesions (Fig. 6). The conventional
The clinical stage of cervical cancer is the best predictor of sur- cone biopsy is obtained using a scalpel (“cold knife”), but today it is
vival. Overall 5-­year survival is 60%, and by each stage it is: I, 90%; more often used for glandular lesions. Ectocervical lesions are best
|
98       Prat and Mutch

5.1.1.1 | Hyperplasia without atypia


This is an exaggerated proliferation of glands of irregular size and shape
with increase in the gland-­to-­stroma ratio compared with prolifera-
tive endometrium, but without significant nuclear atypia. Risk factors
include obesity, polycystic ovarian disease, and diabetes. Hyperplasia
without atypia is the result of unopposed estrogenic stimulation.
Patients have a 3–4-­fold increased endometrial carcinoma risk, ris-
ing to 10-­fold after 10 years. Progression to endometrial carcinoma
occurs in 1%–3% of women with hyperplasia without atypia.

5.1.1.2 | Atypical hyperplasia/endometrial intraepithelial


neoplasia (EIN)
This lesion shows marked glandular crowding, often as back-­to-­back
glands, with little intervening stroma and cytologic atypia. Epithelial
cell nuclei are large and hyperchromatic with prominent nucleoli. One-­
quarter to one-­third of these women will be diagnosed with endome-
F I G U R E   6   Cervical cone biopsy/excision. The craniocaudal
trioid carcinoma at immediate hysterectomy or during the first year
length (A) and diameter (B) should be measured; and both the
radial (A) and endocervical (C) margins need to be assessed (with of follow-­up.2 Trimble et al.,9 on behalf of the Society of Gynecologic
differential inking). Use a stitch to designate 12 o’clock, open cone Oncology Clinical Practice Committee, demonstrated up to a 42.6%
and pin out to fix, and then block in the entire specimen sequentially. (123/289) risk of associated cancers in this group.
Reproduced with permission (copyright Elsevier) from Mutter and EIN refers to a monoclonal neoplastic growth of genetically altered
Prat,1 Chapter 35.
cells with greatly increased risk of becoming the endometrioid type of
endometrial carcinoma. The main diagnostic criterion of EIN is that
excised by LEEP using a blend-­current or by laser where available. gland area exceeds that of stroma (volume percentage stroma <55%).
Trachelectomy may also be performed as a therapeutic procedure for Atypical hyperplasia/EIN contains many of the genetic changes seen
Stage 1A invasive carcinomas of the cervix. A trachelectomy is a more in endometrioid endometrial carcinoma, i.e. microsatellite instability,
extensive version of a cone excision, as the entire cervix is removed, and PTEN, KRAS, and CTNNB1 (beta-­catenin) mutation.1,2
with or without a vaginal cuff.
The specimen obtained on cone biopsy or LEEP is a roughly cone-­
5.1.2 | Clinical features
shaped excision of the uterine cervix to include a portion of exocervix,
external os with the entire transformation zone, and endocervical canal Hysterectomy is usually the therapy of choice if a woman does not
with varying amounts of deep tissue. The surgeon should note the 12 want more children. Women who want more children or those with
o’clock position with a black suture. For a trachelectomy specimen, the high operative risks may be treated with progestins in selected cases.
presence of vaginal cuff should be documented and measured.
Fixation for 3 hours before cutting is usually adequate. Serially cut
5.2 | Endometrial carcinoma
sections should be sequentially submitted. Submit the entire specimen
in a clockwise direction beginning at 1 o’clock (Fig. 6). Both the ecto- Endometrial carcinoma is the sixth most frequent cancer diagnosed
cervical and endocervical margins of the cone specimen need to be in women globally, with an age standardized incidence rate of 8.2 per
assessed and reported.1 100 000. It is the fourth most common cancer in women in indus-
trialized countries and the most common gynecologic cancer. Three-­
quarters of women with endometrial cancer are postmenopausal. The
5 |  CORPUS UTERI
median age at diagnosis is 63 years.1,2
Endometrial carcinoma is classified into two different types
5.1 | Endometrial hyperplasia
(Fig. 7 and Table 1). Type I tumors (Fig. 7A) (about 80%), endome-
Endometrial hyperplasia forms a morphologic continuum of abnormal trioid carcinomas, are often preceded by endometrial hyperplasia
proliferation ranging from focal glandular crowding or simple hyper- or EIN and are associated with estrogenic stimulation. They occur
plasia to well-­differentiated adenocarcinoma. mainly in pre-­ or perimenopausal women and are associated with
obesity, hyperlipidemia, anovulation, infertility, and late meno-
pause. Typically, most endometrioid carcinomas are confined to the
5.1.1 | Pathology
uterus and follow a favorable course. In contrast, type II tumors
The 2014 WHO scheme distinguishes only two categories of endo- (Fig. 7B) (about 10%) are nonendometrioid, largely serous carcino-
metrial hyperplasia: (1) hyperplasia without atypia; and (2) atypical mas, arising occasionally in endometrial polyps or from precancer-
hyperplasia/endometrial intraepithelial neoplasia (EIN).1,2 ous lesions in atrophic endometria (endometrial “intraepithelial”
Prat and Mutch |
      99

Six main molecular alterations have been described in type I endo-


metrioid carcinomas: microsatellite instability (25%–30% of the cases);
PTEN mutations (30%–60%); PIK3CA mutations (26%–39%); ARID1A
(20%); K-RAS mutations (10%–30%); and CTNNB1 (β-­catenin) muta-
tions with nuclear protein accumulation (25%–38%). In contrast, most
type II nonendometrioid carcinomas have p53 mutations, Her-­2/neu
amplification, and loss of heterozygosity on several chromosomes.
Nonendometrioid carcinomas may also derive from endometrioid car-
cinoma with microsatellite instability through tumor progression and
subsequent p53 mutations.10
The Cancer Genome Atlas (TCGA) has conducted the most compre-
hensive genomic analysis of endometrial carcinomas reported to date.11
TCGA has expanded the dualistic classification of endometrial carci-
F I G U R E   7   Carcinoma of the endometrium. (A) Endometrioid
noma (types I and II) to four distinct molecular subgroups: (1) an ultra-
carcinoma. Polypoid tumor with only superficial myometrial
invasion (left). Well-­differentiated (grade 1) adenocarcinoma. The mutated POLE subgroup; (2) a hypermutated microsatellite unstable
neoplastic glands resemble normal endometrial glands (right). (B) subgroup; (3) a copy-­number low/microsatellite stable subgroup; and
Nonendometrioid carcinoma. Large hemorrhagic and necrotic tumor (4) a copy-­number high/serous-­like subgroup. Even if overlapping of
with deep myometrial invasion (left). Nonendometrioid (serous) the molecular genetics findings makes it still difficult to separate signif-
carcinoma exhibiting stratification of anaplastic tumor cells and
icant prognostic categories, POLE mutations predict favorable progno-
abnormal mitoses (severe cytologic atypia) (right).
sis, particularly in high-­grade tumors. On the other hand, patients with
endometrioid tumors that are serous-­like at the molecular level might
carcinoma). Type II tumors are not associated with estrogen stim- benefit from treatments that are typically used for serous carcinomas.11
ulation or hyperplasia, readily invade myometrium and vascular Molecular classification of grade 3 endometrial endometrioid carcino-
spaces, and are highly lethal. 1 mas reveals that these tumors are a mixture of molecular subtypes of
Endometrial cancer is the most common extracolonic cancer in endometrial carcinoma, rather than a homogeneous group. Molecular
women with hereditary nonpolyposis colon cancer syndrome (also markers identify prognostic subgroups, with therapeutic implications.
known as Lynch syndrome)—a defect in DNA mismatch repair that is
also associated with ovarian, urothelial, and breast cancers.10
5.2.2 | Pathology
5.2.2.1 | Endometrioid carcinoma of the endometrium
5.2.1 | Molecular pathogenesis This type of endometrial cancer is composed entirely of glandular cells
A dualistic model of endometrial carcinogenesis has been proposed. and is the most common histologic variant (80%–85%). The FIGO sys-
According to this model, normal endometrial cells transform into tem divides this tumor into three grades on the basis of the ratio of
endometrioid carcinoma through replication errors, so-­called “micro- glandular to solid elements, the latter signifying poorer differentiation.
satellite instability,” and subsequent accumulation of mutations in Less common histologic variants include endometrioid adenocarci-
oncogenes and tumor suppressor genes. For nonendometrioid carci- noma with squamous differentiation and the mucinous and secretory
nomas, alterations of p53 and loss of heterozygosity on several chro- types, both associated with good prognosis.1–3,5
mosomes drive malignant transformation.10
5.2.2.2 | Nonendometrioid endometrial carcinomas
These are aggressive as a group, and histologic grading is not clinically
T A B L E   1   Clinicopathologic features of endometrial carcinoma. useful, all cases being considered high grade.

Type I: Endometrioid Type II: Serous


1. Serous carcinoma histologically resembles, and behaves like,
carcinoma carcinoma
high-grade serous carcinoma of the ovary (Fig. 7B). It often shows
Age Pre-­ and perimenopausal Postmenopausal
transtubal spread to peritoneal surfaces. An intraepithelial form
Unopposed estrogen Present Absent
has been termed “serous endometrial intraepithelial carcinoma”
Hyperplasia precursor Present Absent (serous EIC), not to be confused with EIN, described earlier.
Grade Low High Patients with this type of tumor need to be staged and treated
Myometrial invasion Superficial Deep as if they had ovarian cancer.
Growth behavior Stable Progressive 2. Clear cell carcinoma is a tumor of older women. It contains large
Genetic alterations Microsatellite instability, TP53 mutations, cells with abundant cytoplasmic glycogen (“clear cells”) or cells with
PTEN, PIK3CA, CTNNB1 loss of bulbous nuclei that line glandular lumina (“hobnail cells”). Clear cell
(β-­catenin) heterozygosity
carcinomas have poor prognosis.
|
100       Prat and Mutch

3. Carcinosarcoma (malignant mixed mesodermal tumor): In this Tumors that have penetrated the myometrium or invaded lymphatics
highly malignant tumor, pleomorphic epithelial cells intermingle are more likely to have spread beyond the uterus. Endometrial can-
with areas showing mesenchymal differentiation. These mixed cers involving the cervix have a poorer prognosis. Spread outside the
neoplasms are derived from a common clone thought to be of epi- uterus entails the worst outlook.5,12 As of 1988, this is a surgically
1,2
thelial origin. Overall 5-year survival is 25%. staged disease.

5.2.3 | Clinical features 5.3 | Endometrial sarcomas

Unlike cervical cancer, endometrial cancer may spread directly to Currently, endometrial sarcomas are classified into three categories:

para-­aortic lymph nodes, thereby skipping pelvic nodes. Patients with (1) low-­grade endometrial stromal sarcoma (LG-­ESS); (2) high-­grade

advanced cancers may also develop pulmonary metastases (40% of endometrial stromal sarcoma (HG-­ESS); and (3) undifferentiated endo-

cases with metastases). metrial sarcoma (UES).2 LG-­ESSs represent less than 2% of uterine

Women with well-­differentiated cancers confined to the endo- cancers. They may be polypoid or may diffusely invade the myome-

metrium are usually treated by simple hysterectomy and frequently trium. The tumor cells resemble endometrial stromal cells in the pro-

bilateral salpingo-­oophorectomy. Postoperative radiation is con- liferative phase. Nuclear atypia may be minimal to severe and mitotic

sidered if: (1) the tumor is poorly differentiated or nonendome- activity may be restrained. Expression of CD-­10 and estrogen and

trioid in type; (2) myometrium is deeply invaded (more than 50% progesterone receptors helps confirm the diagnosis. The most com-

of the myometrium); (3) the cervix is involved; or (4) lymph nodes mon cytogenetic abnormality of LG-­ESS is a recurrent translocation

contain metastases. involving chromosomes 7 and 17 [t(7;17) (p15;q21)], which results in

Survival in endometrial carcinoma is related to multiple factors: a fusion between JAZF1 and SUZ12 (formerly designated as JJAZ1).1,2

(1) stage, histologic type, and, for endometrioid tumors, grade; (2) This generally portends a better prognosis.

age; and (3) other risk factors, such as progesterone receptor activity, The recently re-­established HG-­ESS has features intermediate

depth of myometrial invasion (Fig. 8), and extent of lymphovascular between LG-­ESS and undifferentiated sarcomas. It may appear as

invasion.12 Actuarial survival of all patients with endometrial cancer an intracavitary polypoid or a mural mass. Microscopically, it con-

following treatment is 80% after 2 years, decreasing to 65% after sists predominantly of high-­grade round-­cells that are sometimes

10 years. Serous carcinomas have an overall survival of less than 50% associated with a low-­grade spindle cell component usually fibro-

and account for more than half of the mortality from this disease. myxoid. Mitotic activity is very striking and typically more than 10
per 10 high-­power fields (HPFs). Necrosis is usually present. HG-­ESS
typically harbors the YWHAE-FAM22 genetic fusion as a result of
t(10;17) (q22;p13).1,2
Higher-­grade poorly differentiated sarcomas originating in the endo-
metrium are designated as undifferentiated endometrial sarcoma.1,2

5.3.1 | Clinical features
Many years may elapse before LG-­ESSs recur clinically, and metasta-
ses may occur even if the original tumor was confined to the uterus
at initial surgery. Recurrences usually involve the pelvis first, followed
by lung metastases. Prolonged survival and even cure are feasible,
despite metastases. By contrast, UES recur early, generally with wide-
spread metastases. Compared with patients with LG-­ESSs, those with
HG-­ESSs and undifferentiated endometrial sarcoma have earlier and
more frequent recurrences (often <1 year) and are more likely to die
of disease. LG-­ESSs can be successfully treated with surgery followed
by progestin therapy, with an expectation of 90% survival 10 years

F I G U R E   8   Endometrial carcinoma. Measurements of depth to after diagnosis.1,2


which tumor invades. (A) Full thickness of myometrial wall, measured
from where endometrium adjacent to tumor is normal (or hyperplasic).
(B) Component of tumor exophytic and rising above imaginary line 5.4 | Uterine adenosarcoma
drawn between adjacent normal endometrium. (C) Depth of invasion.
Uterine (müllerian) adenosarcoma is a distinctive low-­grade tumor
(D) Tumor-­free zone. (E) Width of tumor. A tumor is generally reported
with benign glandular epithelium and malignant stroma. It should
as measuring n x n x n and ‘C’ cm invasive into a wall ‘A’ cm thick.
Reproduced with permission (copyright Elsevier) from Mutter and Prat,1 be distinguished from carcinosarcoma, which is highly aggressive in
Chapter 35. which both epithelial and stromal elements are malignant. One-­fourth
Prat and Mutch |
      101

of patients with adenosarcoma, particularly cases with myometrial HPFs. Size is important as tumors less than 5 cm in diameter almost
invasion and sarcomatous overgrowth, eventually succumb to local never recur.
recurrence or metastatic spread.1,2 Most leiomyosarcomas are large and are advanced when detected.
They are usually fatal despite combinations of surgery, radiation ther-
apy, and chemotherapy. Five-­year survival is about 25%.1,2 Almost all
5.5 | Leiomyosarcoma
leiomyosarcomas are high-­grade tumors and, usually, their diagnosis
Leiomyosarcoma is a malignancy of smooth muscle origin whose inci- is straightforward; however, a small fraction of uterine smooth mus-
dence is only 1/1000 that of leiomyoma. It accounts for 2% of uterine cle tumors show atypical histologic features that are insufficient for
malignancies. Its pathogenesis is uncertain. Women with leiomyosar- the diagnosis of malignancy or have an unpredictable clinical behav-
comas are on average more than a decade older (age above 50 years) ior. These tumors have been designated as smooth muscle tumors of
than those with leiomyomas, and the malignant tumors are larger uncertain malignant potential (STUMP), but the term atypical smooth
(10–15 cm vs 3–5 cm).1,2 muscle tumors, as introduced by the 2014 WHO classification of
tumors,2 seems preferable in view of their favorable behavior in most
cases. The latter term simply describes the morphologic findings
5.5.1 | Pathology
avoiding the words “uncertain” and “malignant,” which create unnec-
Leiomyosarcoma should be suspected if an apparent leiomyoma is essary concern for the patient.
soft, shows areas of necrosis on gross examination, or has irregu- In two studies of 41 and 16 cases of “STUMP,” only 3 (7%) and
lar borders (invasion of adjacent myometrium). Mitotic activity (10 2 (12%) patients developed recurrences, respectively. Recurrence
or more mitoses per 10 HPFs), nuclear atypia, and geographical occurred, several years after hysterectomy, in the form of “STUMP” in
necrosis are the best diagnostic criteria (Fig. 9A,B). Myxoid and three cases and as leiomyosarcoma in the other two. All five patients
epithelioid leiomyosarcomas may contain only five mitoses per 10 were alive and disease free after prolonged follow-­up.13,14 As indi-
cated previously, when account is taken of mitotic count, myometrial
invasion, nuclear atypia, tumor cell necrosis, size of tumor, and age of
(A)
the patient, tumors can be allocated to benign or malignant categories
with greater certainty and the term “of uncertain malignancy” can be
avoided in most cases.1,2

6 | FALLOPIAN TUBE

Tumors of the fallopian tube are rare. Most primary malignan-


cies are carcinomas, with peak incidence among women aged
50–60 years. Recent observations suggest that some cases of
high-­grade serous carcinoma of the ovary (see below) may arise
from the fimbriated end of the fallopian tube. Tubal carcinomas
(B) behave similarly to ovarian carcinoma and frequently appear as a
solid mass in the wall of a grossly dilated tube, but may sometimes
only be identified upon microscopic examination. The tumor is
bilateral in 25% of cases. Prognosis is poor, as the disease is almost
always detected at advanced stage.1,2 These tumors are treated
like an ovarian cancer.

6.1 | Risk reducing salpingo-­oophorectomy


An increasingly common indication for salpingectomy is prophylac-
tic for patients who have BRCA1/2 mutations, a personal history of
breast cancer, or strong family history of breast and/or tubo-­ovarian
cancer. Typically the specimen is grossly unremarkable, however
these fallopian tubes, along with the corresponding ovaries, should
be submitted entirely for histologic examination.1,2 Prophylactic sal-
F I G U R E   9   Leiomyosarcoma of the uterus. (A) A zone of
pingectomy is becoming standard at the time of hysterectomy and
coagulative tumor necrosis appears demarcated from the viable tumor.
(B) The tumor shows considerable nuclear atypia and abundant emerging data suggest a significant reduction in ovarian cancer risk
mitotic activity. when the fallopian tubes are removed.15
|
102       Prat and Mutch

The protocol for sectioning and extensively examining the fim- absence of stromal invasion: (1) benign; (2) borderline malignancy; and
briated end (SEE-­FIM protocol) (Fig. 10) was developed for process- (3) carcinoma.
ing risk-­reducing salpingo-­oophorectomy specimens.16 The entire Common epithelial neoplasms most commonly affect nulliparous
tube is initially fixed for at least 4 hours to prevent denuding of women and occur least frequently in women in whom ovulation has
the mucosal epithelial cells. Then, the fimbriated end is amputated been suppressed (e.g. by pregnancy or oral contraceptives). Whereas
from the proximal tube and sectioned longitudinally into multiple the lifetime risk of developing ovarian cancer in the general pop-
(at least four) sections and the entire tube is submitted for histo- ulation is 1.6%, women with one-­first-­degree relative with ovarian
logic review.1,2 cancer have a 5% risk. Also, women with a family history of ovarian
carcinoma are at greater risk of breast cancer and vice versa. Defects
in repair genes implicated in hereditary breast cancers, BRCA-1 and
7 |  OVARY
BRCA-2, are incriminated in familial ovarian cancers as well. As for
endometrial carcinoma, women with hereditary nonpolyposis colon
7.1 | Ovarian tumors
cancer (HNPCC) are also at greater risk of ovarian cancer.1,16
There are many types of ovarian tumors including benign, borderline, Epithelial ovarian tumors are primarily classified according
and malignant types. About two-­thirds occur in women of reproduc- to cell type into serous, mucinous, endometrioid, clear cell, tran-
tive age. Approximately 80% of ovarian tumors are benign. Almost sitional, and squamous cell tumors.1,2,16 However, none of these
90% of malignant and borderline tumors are diagnosed after the age cells are found in the normal ovary and their development has
1,16
of 40 years. long been attributed to müllerian “neometaplasia” of the ovarian
Ovarian tumors are classified by the ovarian cell type of origin. surface epithelium (mesothelium). During embryonic life, the celo-
Most are common epithelial tumors (approximately 60%). Other mic cavity is lined by mesothelium which also covers the gonadal
important groups are germ cell tumors (30%), sex cord/stromal tumors ridge. The same mesothelial lining gives rise to müllerian ducts,
(8%), and tumors metastatic to the ovary. Common epithelial tumors from which the fallopian tubes, uterus, and vagina arise (Fig. 11).
account for about 90% of ovarian malignancies, high-­grade serous Thus, the tumor cells would resemble morphologically the epithelia
adenocarcinoma being the most common (70%). Some cases of high-­ of the fallopian tube, endometrium, or endocervix. 1,16 Recently, it
16
grade serous carcinomas may arise in the fallopian tubes. has been hypothesized that cytokeratin7-­positive embryonic/stem
Ovarian cancer is the second most frequent gynecologic malig- cells would give rise to immunophenotypically distinct neoplas-
nancy after endometrial cancer and carries a higher mortality rate than tic progeny,17 which would support the old concept of “mullerian
all other female genital cancers combined. It is difficult to detect early neometaplasia.” Besides the mesothelial origin, there is now com-
in its evolution when it is still curable and, as such, over three-­fourths pelling evidence that a number of what have been thought to be
of patients already have extraovarian tumor spread to the pelvis or primary ovarian cancers actually originate in other pelvic organs
abdomen at the time of diagnosis.1,16 and involve the ovary secondarily. In fact, it has been shown that
some high-­grade serous carcinomas arise from precursor epi-
thelial lesions in the distal fimbriated end of the fallopian tube,
7.2 | Epithelial tumors
whereas endometrioid and clear cell carcinomas originate from
Tumors of common epithelial origin can be broadly classified, accord- ovarian endometriosis.16
ing to cell proliferation, degree of nuclear atypia, and presence or

7.3 | Borderline tumors
Borderline tumors show epithelial proliferation greater than that seen
in their benign counterparts and variable nuclear atypia; however, in
contrast to carcinomas, there is absence of stromal invasion, and their
prognosis is much better than that of carcinomas.
Serous borderline tumors generally occur in women aged
20–50 years (average, 46 years). Serous tumors are more commonly
bilateral (34%) than mucinous ones (6%) or other types. The tumors
vary in size, although mucinous tumors may be gigantic. Serous bor-
derline tumors have one or more cysts lined to varying extents by
papillary projections, ranging from fine and exuberant to grapelike
clusters. These structures show: (1) epithelial stratification; (2) moder-
F I G U R E   1 0   Sectioning and extensively examining the fimbriated
ate nuclear atypia; and (3) mitotic activity. By definition, the presence
end (SEE-­FIM protocol) for cutting in a fallopian tube specimen. Cross-­
of more than focal microinvasion (i.e. discrete nests of epithelial cells
sections cut at approximately 3 mm intervals of the length of the tube,
and the fimbriated end amputated and cut in the longitudinal axis at <3 mm into the ovarian stroma) identifies a tumor as low-­grade serous
1–2 mm intervals. carcinoma (LGSC), rather than a borderline tumor.1,2
Prat and Mutch |
      103

F I G U R E   1 1   Histogenesis of ovarian epithelial tumors.

Despite the lack of ovarian stromal invasion, serous borderline patterns of spread, molecular events during oncogenesis, responses
tumors—particularly those with exophytic growth—can implant on to chemotherapy, and outcomes. With progress toward subtype-­
peritoneal surfaces (Fig. 12A) and, rarely, (about 10% of peritoneal specific management of ovarian cancer, accurate subtype assignment
implants), progress to LGSC and invade the underlying tissues (Fig. 12B). is becoming increasingly important.
Histopathologically, invasive peritoneal implants and LGSC are identical
lesions only distinguished by the timing of the disease and the volume
7.4.1 | Serous carcinomas
of the tumor. Whereas invasive implants are early superficial lesions of
microscopic or small macroscopic size (≤1–2 cm), LGSC frequently pres- 7.4.1.1 | Molecular pathogenesis
1,2,16
ents as bulky disease (peritoneal carcinomatosis). Low-­grade and high-­grade serous carcinomas are fundamen-
Surgical cure is almost always possible if the serous borderline tally different tumors. Whereas low-­grade tumors are frequently
tumor is confined to the ovaries. Even if it has spread to the pelvis associated with serous borderline tumors and have mutations of
or abdomen, 90% of patients are alive after 5 years. Although there KRAS or BRAF oncogenes, high-­grade serous carcinomas lack ovar-
is a significant rate of late recurrence, the tumors rarely recur beyond ian precursor lesions and have a high frequency of mutations in
10 years. Late progression to low-­grade serous carcinoma has been TP53, but not in KRAS or BRAF. Interestingly, carcinomas arising
reported in approximately 7% of cases.1,2,16 After fertility-­sparing sur- in patients with germline BRCA1 or BRCA2 mutations (hereditary
gery, mucinous borderline tumors may “recur” as carcinomas in the ovarian cancers) are almost invariably the high-­grade serous type
contralateral ovary; however, such tumors should be considered inde- and commonly have TP53 mutations. An undetermined number of
pendent primary tumors.18,19 BRCA1- or BRCA2-related tumors arise from the epithelium of the
fimbriated end of the fallopian tube, suggesting that at least some
sporadic high-­grade ovarian and “primary” peritoneal serous carci-
7.4 | Malignant epithelial tumors (carcinomas)
nomas may actually develop from the distal fallopian tube and “spill
Carcinomas of the ovary are most common in women aged over” onto the adjacent tissues (Table 2). 1,16
40–60 years, and are rare under the age of 35 years. Based on light
microscopy and molecular genetics, ovarian carcinomas are classified 7.4.1.2 | Pathology
into five main subtypes, which, in descending order of frequency, High-­grade serous carcinomas are the most common ovarian cancers
are: high-­grade serous carcinomas (>70%), endometrioid carcinomas and most patients present with advanced stage disease (approxi-
(10%), clear cell carcinomas (10%), mucinous carcinomas (3%–4%), mately 80%). Two-­thirds of serous cancers with extraovarian spread
and low-­grade serous carcinomas (<5%)16 (Table 2). These subtypes, are bilateral. They are predominantly solid masses, usually with necro-
which account for 98% of ovarian carcinomas, can be reproducibly sis and hemorrhage and typically show obvious stromal invasion.
diagnosed and are inherently different diseases, as indicated by dif- Most tumors have a high nuclear grade with highly cellular papillae
ferences in epidemiologic and genetic risk factors, precursor lesions, and solid areas (Fig. 13A). The mitotic rate is very high. Psammoma
|
104       Prat and Mutch

(A) 7.4.2.2 | Pathology
Mucinous carcinomas are usually large, unilateral, multilocular cystic
masses containing mucinous fluid. They often exhibit papillary archi-
tecture (Fig. 13C). Since benign and malignant components may coexist
within a single specimen, these tumors should be sampled extensively.
Mucinous tumors are bilateral in only 5% of the cases; thus, finding
bilateral or unilateral mucinous tumors smaller than 10 cm should raise
suspicion of metastases from a mucinous carcinoma elsewhere (e.g.
gastrointestinal tract).
The category of mucinous borderline tumor with intraepithelial
carcinoma is reserved for tumors that lack architectural features of
invasive carcinoma but, focally, show unequivocally malignant cells
lining glandular spaces. Mucinous borderline tumors with intraepi-
(B)
thelial carcinoma have a very low likelihood of recurrence.1,2,16
Mucinous carcinomas showing expansile or confluent glandular
growth appear to have a more favorable prognosis than mucinous car-
cinomas with destructive stromal invasion. The combination of exten-
sive infiltrative stromal invasion, high nuclear grade, and tumor rupture
should be considered a strong predictor of recurrence for Stage I muci-
nous carcinomas.1,2,16
Pseudomyxoma peritonei is a clinical condition of abundant
gelatinous or mucinous ascites in the peritoneum, fibrous adhe-
sions, and frequently mucinous tumors involving the ovaries. The
appendix is also involved by a similar mucinous tumor in 60% of
the cases and appears normal in the remaining 40%. Current data
suggest that in most cases the ovarian tumors are metastases from
F I G U R E   1 2   Peritoneal implants of serous borderline tumor.
the appendiceal lesions.1,2
(A) Noninvasive desmoplastic implant. The implant invaginates
between adjacent lobules of omental fat. A few nests of tumor
cells are present within a loose fibroblastic stroma. (B) Invasive
omental implant. The tumor glands and papillae appear disorderly 7.4.3 | Endometrioid carcinoma
distributed within a dense fibrous stroma and resemble a low-­grade
serous carcinoma. Endometrioid adenocarcinoma histologically resembles its uter-
ine counterpart (Fig. 13D), may have areas of squamous differen-
bodies are often present.1,2,16 Ovarian cancers formerly designated as tiation, and is second only to serous adenocarcinoma in frequency.
transitional cell carcinomas represent histologic variants of high-­grade It accounts for 10% of all ovarian cancers. These tumors occur most
serous carcinoma and carry TP53 mutations. commonly after menopause. Up to half of these cancers are bilateral
Low-­grade serous carcinomas show irregular stromal invasion and, at diagnosis, most tumors are either confined to the ovary or
with small, tight nests of tumor cells within variable desmoplasia. The within the pelvis.1,2
uniformity of the nuclei is the principal criterion for distinguishing
low-­ and high-­grade serous carcinomas (Fig. 13B). Low-­grade serous 7.4.3.1 | Molecular pathogenesis
carcinomas rarely progress to high-­grade tumors.1,2,16 Endometrioid carcinomas are thought to arise by malignant transfor-
mation of endometriosis, and not from ovarian surface epithelium.
The most common genetic abnormalities in sporadic endometri-
7.4.2 | Mucinous carcinoma
oid carcinoma of the ovary are somatic mutations of the ARID1A,
7.4.2.1 | Molecular pathogenesis β-­catenin (CTNNB1), and PTEN genes and microsatellite instability.
Mucinous ovarian tumors are often heterogeneous. Benign, border- Endometrioid borderline tumors also have CTNNB1 (β-­catenin gene)
line, noninvasive, and invasive carcinoma components may coexist mutations (Table 2).16
within the same tumor. Such a morphologic continuum suggests that
tumor progression occurs from cystadenoma and borderline tumor to 7.4.3.2 | Pathology
noninvasive, microinvasive, and invasive carcinomas. This hypothesis Although they may be cystic, most endometrioid carcinomas are
is supported by KRAS mutations in mucinous tumors: 56% of cystade- largely solid with areas of necrosis. These tumors are graded like
nomas and 85% of carcinomas express mutated KRAS, with borderline their uterine counterparts. Between 15% and 20% of patients also
tumors being intermediate (Table 2).1,16 harbor a uterine endometrioid carcinoma. Strong data suggest that
Prat and Mutch |
      105

T A B L E   2   Main types of ovarian carcinoma.

High-­grade serous Low-­grade serous Mucinous Endometrioid Clear cell

Usual stage at diagnosis Advanced Early or advanced Early Early Early


Presumed tissue of Tubal metaplasia in inclusions Serous borderline Adenoma–borderline– Endometriosis, Endometriosis,
origin/precursor lesion of ovarian surface epithelium tumor carcinoma sequence; adenofibroma adenofibroma
or fallopian tube teratoma
Genetic risk BRCA1/2 ? ? HNPCC ?
Significant molecular p53 and BRCA pathways BRAF or K-­RAS K-RAS PTEN, β-­catenin, HNF-­1β
abnormalities HER2 ARID1A ARID1A
PIK3CA PIC3CA
PTEN
K-RAS
MI
Proliferation High Low Intermediate Low Low
Response to primary 80% 26%–28% 15% ? 15%
chemotherapy
Prognosis Poor Favorable Favorable Favorable Intermediate

most of these cases arise independently, although some may be tumor removed as possible. Adjuvant chemotherapy is used to treat
metastases from one or the other. This distinction has important distant occult sites of tumor spread.
prognostic implications.1,2

7.5 | Germ cell tumors


7.4.4 | Clear cell carcinoma
Tumors derived from germ cells make up one-­fourth of ovarian
This enigmatic ovarian cancer is closely related to endometrioid tumors. In adult women, ovarian germ cell tumors are virtually all
adenocarcinoma, and often occurs in association with endometriosis. benign (mature cystic teratoma, dermoid cyst), but in children and
It constitutes 5%–10% of all ovarian cancers usually occurring after young adults, they are largely cancerous. In children, germ cell tumors
menopause. The most common genetic abnormalities are somatic are the most common ovarian cancer (60%); they are extremely rare
mutations of the ARID1A, PTEN, and PIK3CA genes.1,16 after menopause. Rarely, germ cell tumors may arise from pre-­existing
Although patients typically present with Stage I or II disease, clear somatic neoplasms of the female genital tract. In these cases, the tera-
cell carcinomas have a poor prognosis compared with other low-­ toid tumors derive most likely from a pluripotent stem cell population
stage ovarian carcinomas. Clear cell carcinomas of the ovary resem- of somatic neoplasms.1,2
ble their counterparts in the vagina, cervix, and corpus; they show Neoplastic germ cells may differentiate along several lines producing:
sheets or tubules of malignant cells with clear cytoplasm (Fig. 13E).
1. Dysgerminomas are composed of neoplastic germ cells, similar
7.4.4.1 | Clinical features to oogonia of fetal ovaries. After dysgerminomas the differen-
By the time ovarian cancers are diagnosed, many have metastasized tiation is typically extraembryonic or embryonic
to (i.e. implanted on) the surfaces of the pelvis, abdominal organs, or 2. Teratomas differentiate toward somatic (embryonic or adult) tissues.
bladder. Ovarian tumors have a tendency to implant in the peritoneal 3. Yolk sac tumors form extraembryonic endoderm and mesenchyme
cavity on the diaphragm, paracolic gutters, and omentum. Lymphatic and, less frequently, embryonic endodermal derivatives (intestine
spread is preferentially to para-­aortic lymph nodes near the origin of and liver).
the renal arteries and to a lesser extent to external iliac (pelvic) or 4. Choriocarcinomas feature cells similar to those covering the
inguinal lymph nodes.1,2 ­placental villi.
Survival for patients with malignant ovarian tumors is generally
poor. The most important prognostic index is the surgical stage of the Malignant germ cell tumors in women older than 40 years usu-
20
tumor at the time it is detected. Overall, 5-­year survival is only 35%. ally result from transformation of one of the components of a benign
Prognostic indices for epithelial tumors also include histologic type cystic teratoma. Malignant germ cell tumors tend to be highly
(grade) and the size of the residual neoplasm. aggressive; however, with current chemotherapy, survival rates for
Surgery is the mainstay of therapy. It removes the primary tumor, many exceed 80%.1,2
establishes the diagnosis, and determines the extent of spread. The Recent stem cell research has provided several highly diagnos-
peritoneal surfaces, omentum, liver, subdiaphragmatic recesses, and tic pluripotency markers, including transcription factors (SALL4,
all abdominal regions must be visualized, and as much metastatic LIN28, OCT3/4, and SOX2) and cytoplasmic/membranous proteins
|
106       Prat and Mutch

F I G U R E   1 3   Representative examples of the five main types of ovarian carcinoma, which together account for 98% of cases: (A) High-­grade
serous carcinoma; (B) Low-­grade serous carcinoma; (C) Mucinous carcinoma; (D) Endometrioid carcinoma; and (E) Clear cell carcinoma.

(glypican-­3) that are sequentially expressed in malignant germ cell 12p. c-Kit mutations are seen in 25%–50% of tumors, most commonly
1,2
tumors according to their differentiation stage in exon 17, not in the exon 11 location that confers susceptibility to
imatinib therapy.
Dysgerminomas are treated surgically; 5-­year survival for patients
7.5.1 | Dysgerminoma
with Stage I tumor approaches 100%. Because the tumor is highly
Dysgerminoma is the ovarian counterpart of testicular seminoma, responsive to chemotherapy, even for higher-­stage tumors 5-­year sur-
and is composed of primordial germ cells. It accounts for less than 2% vival rates still exceed 90%.
of ovarian cancers in all women. Most patients are between 10 and
30 years. The tumors are bilateral in about 15% of cases.
7.5.2 | Teratoma
7.5.1.1 | Pathology Teratoma is a tumor of germ cell origin that differentiates toward
Dysgerminomas are often large and firm and have a bosselated exter- somatic structures. Most teratomas contain tissues from at least two,
nal surface. The cut surface is soft and fleshy. They contain large nests and usually all three, embryonic layers. Immature teratomas contain
of monotonously uniform tumor cells that have clear glycogen-­filled elements derived from the three germ layers. However, unlike mature
cytoplasm and irregularly flattened central nuclei. Fibrous septa con- cystic teratomas, immature teratomas contain embryonal tissues.
taining lymphocytes traverse the tumor.1,2 These tumors account for 20% of malignant tumors in women under
The tumor cells show diffuse nuclear expression for the stem cell/ the age of 20. Microscopically, they show multiple components such
primitive germ cell nuclear transcription factors OCT3/4, NANOG, as immature neural tissue (neuroepithelial rosettes and glia), glands,
and SALL4. The majority of dysgerminomas show isochromosome and other structures found in mature cystic teratomas. Grading is
Prat and Mutch |
      107

based on the amount of immature tissue present. Survival correlates cells, white zones of stroma, and focal hemorrhages. Random nuclear
1,2
with tumor grade. arrangement about a central degenerative space (Call-­Exner bodies)
gives a characteristic follicular pattern. Tumor cells secrete α-­inhibin,
a protein that suppresses pituitary release of follicle-­stimulating hor-
7.5.3 | Yolk sac tumor
mone (FSH). Besides α-­Inhibin, calretinin, and FOXL2 are the most
Yolk sac tumors are highly malignant neoplasms of women under important positive immunoreactions.1,2
the age of 30 that histologically resemble the endoderm and mes- The most common chromosomal abnormalities are trisomy 12, tri-
enchyme of the primitive yolk sac (extra-­embryonal) and embryonal somy 14, monosomy 16, deletion of 16q, and monosomy 22. Missense
somatic tissues (intestine and liver). They are typically large, with somatic point mutations in the FOXL2 gene (402 C to G) are found in
extensive necrosis and hemorrhage. The most common histotype over 90% of adult granulosa cell tumors.
is the reticular form. Schiller-­Duval bodies are characteristic. They
consist of papillae that protrude into spaces lined by tumor cells,
7.7.2 | Clinical features
resembling the glomerular spaces. The papillae are covered by a
mantle of embryonal cells and contain a fibrovascular core and a Three-­f ourths of granulosa cell tumors secrete estrogens.
central blood vessel. Thus, endometrial hyperplasia is a common presenting sign.
Yolk sac tumor secretes α-­fetoprotein that should be stained for in Endometrial adenocarcinoma may develop if a functioning granu-
all germ cell tumors. Detection of α-­fetoprotein in the blood is useful losa cell tumor remains undetected. At diagnosis, 90% of granu-
for diagnosis and for monitoring the effectiveness of therapy. Once losa cell tumors are within the ovary (Stage I). Over 90% of these
uniformly fatal, 5-­year survival with chemotherapy for Stage I yolk sac patients survive 10 years. Tumors that have extended into the
tumors exceeds 80%.1,2 pelvis and lower abdomen have a poorer prognosis. Late recur-
rence after surgical removal is not uncommon after 5–10 years
and is usually fatal. 1,2
7.5.4 | Choriocarcinoma
Choriocarcinoma of the ovary is a rare tumor that mimics the epithelial
7.8 | Sertoli-­Leydig cell tumors
covering of placental villi, namely, cytotrophoblast and syncytiotropho-
blast. The pregnancy test is positive and the elevated serum level of Ovarian Sertoli-­Leydig cell tumors are rare androgen-­secreting
human chorionic gonadotropin may lead to precocious sexual develop- mesenchymal neoplasms of low malignant potential that resem-
ment in young girls or menstrual abnormalities in older patients. In women ble embryonic testis. Tumor cells typically secrete weak androgens
of reproductive age, however, it may also be a metastasis from an intrau- (dehydroepiandrosterone). Sertoli-­Leydig cell tumors occur at all
terine gestational tumor. The tumor is unilateral, solid, and widely hemor- ages but are most common in young women of childbearing age.
rhagic. Although highly aggressive, it responds well to chemotherapy.1,2 They vary from well to poorly differentiated and some have heterolo-
gous elements (e.g. mucinous glands and, rarely, even skeletal muscle
and cartilage).
7.6 | Sex cord/stromal tumors
Mutations in DICER1, a gene encoding an RNase III endoribo-
These tumors represent 10% of ovarian tumors, vary from benign to nuclease, are found in 60% of Sertoli-­Leydig cell tumors. Germline
low-­grade malignant, and may differentiate toward female (granulosa mutations in this gene are seen in familiar multinodular goiter with
and theca cells) or male (Sertoli and Leydig cells) structures.1,2 Sertoli-­Leydig tumor, and tumor susceptibility includes pleuropul-
monary blastoma in childhood. Sertoli-­Leydig cell tumor has been
reported in association with cervical embryonal rhabdomyosarcoma
7.7 | Granulosa cell tumor
in four patients.21
Granulosa cell tumors are the prototypical functional neoplasms of Nearly half of all patients with Sertoli-­Leydig cell tumors exhibit
the ovary associated with estrogen secretion. They should be con- signs of virilization. Initial signs are often defeminization, manifested
sidered low-­grade malignancies because of their potential for local as breast atrophy, amenorrhea, and loss of hip fat. Once the tumor is
spread and the rare occurrence of distant metastases. removed, these signs disappear or at least lessen. Well-­differentiated
Most granulosa cell tumors occur after menopause (adult form) tumors are virtually always cured by surgical resection, but poorly dif-
and are unusual before puberty. A juvenile form occurs in children and ferentiated ones may metastasize.1,2
young women and has distinct clinical and pathologic features (hyper-
estrinism and precocious puberty).
7.9 | Steroid cell tumor
Steroid cell tumors of the ovary, also called lipid cell tumors, are com-
7.7.1 | Pathology
posed of cells that resemble lutein cells, Leydig cells, and adrenal cor-
Adult-­type granulosa cell tumors are large and focally cystic to solid. The tical cells. Most steroid cell tumors are hormonally active, usually with
cut surface shows yellow areas, due to lipid-­rich luteinized granulosa androgenic manifestations.
|
108       Prat and Mutch

4. Rogers LJ, Cuello MA. Cancer of the vulva. Int J Gynecol Obstet.
7.10 | Tumors metastatic to the ovary 2018;143(Suppl.2):4–13.
5. Mutch DG. The new FIGO staging system for cancers of the vulva, cer-
About 3% of cancers found in the ovaries arise elsewhere, mostly in
vix, endometrium and sarcomas. Gynecol Oncol. 2009;115:325–328.
the large intestine, breast, endometrium, and stomach, in descending 6. Ferlay J, Soerjomataram I, Ervik M, et  al. GLOBOCAN 2012 v1.0,
order. These tumors vary from microscopic lesions to large masses. Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11
Metastatic tumors large enough to cause symptoms originate most [Internet]. Lyon, France: IARC; 2013.
7. Stoler MH. Human papillomaviruses and cervical neoplasia: A model
often in the colon and may be estrogen secreting as they activate the
for carcinogenesis. Int J Gynecol Pathol. 2000;19:16–28.
ovarian stroma. 8. Bhatla N, Aoki D, Sharma DN, Sankaranarayanan R. Cancer of the cer-
Krukenberg tumors are metastases to the ovary, composed of vix uteri. Int J Gynecol Obstet. 2018;143(Suppl.2):22–36.
nests of mucin-­filled “signet-­ring” cells in a cellular stroma derived 9. Trimble CL, Method M, Leitao M, et  al.; Society of Gynecologic
Oncology Clinical Practice Committee. Management of endometrial
from the ovary. The stomach is the primary site in 75% of cases and
precancers. Obstet Gynecol. 2012;120:1160–1175.
most of the rest are from the colon.1,2 10. Matias-Guiu X, Prat J. Molecular pathology of endometrial carcinoma.
Bilateral ovarian involvement and multinodularity suggest a meta- Histopathology. 2013;62:111–123.
static carcinoma, and both ovaries are grossly involved in 75% of cases. 11. The Cancer Genome Atlas Research Network, Kandoth C, Schultz N,
et al. Integrated genomic characterization of endometrial carcinoma.
Nature. 2013;497:67–73.
AUTHOR CONTRI BUTI O N S 12. Prat J. Prognostic parameters of endometrial carcinoma. Hum Pathol.
2004;35:649–662.
JP and DM reviewed and updated the pathology chapter published in 13. Guntupalli SR, Ramirez PT, Anderson ML, Milam MR, Bodurka DC,
the 2015 Cancer Report. Malpica A. Uterine smooth muscle tumor of uncertain malignant
potential: A retrospective analysis. Gynecol Oncol. 2009;113:324–326.
14. Ip PP, Cheung AN, Clement PB. Uterine smooth muscle tumors of
ACKNOWLE DGME N TS uncertain malignant potential (STUMP): A clinicopathologic analysis
of 16 cases. Am J Surg Pathol. 2009;33:992–1005.
This chapter updates the information published in the FIGO Cancer 15. Falconer H, Yin L, Grönberg H, Altman D. Ovarian cancer risk after
Report 2015 (Prat J. Pathology of cancers of the female genital tract. salpingectomy: A nationwide population-­based study. J Natl Cancer
Inst. 2015;107:Pii:dju410.
Int J Gynecol Obstet. 2015;131(Suppl.2):S132-­45), with approval
16. Prat J. D’Angelo E, Espinosa I. Ovarian carcinomas: at least five
granted by the original author.
­different diseases with distinct histological features and molecular
genetics. Hum Pathol. 2018 Jun 23. [Epub ahead of print].
17. Crum CP, Herfs M, Ning G, et  al. Through the glass darkly:
CO NFLI CTS OF I NT E RE S T
Intraepithelial neoplasia, top-­down differentiation, and the road to
ovarian cancer. J Pathol. 2013;231:402–412.
The authors have no conflicts of interest to declare.
18. Uzan C, Nikpayam M, Ribassin-Majed L, et al. Influence of histological
subtypes on the risk of an invasive recurrence in a large series of stage
REFERENCES I borderline ovarian tumor including 191 conservative treatments.
Ann Oncol. 2014;25:1312–1319.
1. Mutter GL, Prat J, eds. Pathology of the Female Reproductive Tract, 3rd 19. Prat J. The results of conservative (fertility-­sparing) treatment in bor-
edn. Edinburgh: Churchill Livingstone (Elsevier); 2014. derline ovarian tumors vary depending on age and histological type.
2. Kurman RJ, Carcangiu ML, Herrington CS, Young RH, eds. WHO Ann Oncol. 2014;25:1255–1258.
Classification of Tumours of Female Reproductive Organs. 4th edn. IARC: 20. Prat J; FIGO Committee on Gynecologic Oncology. Staging classifi-
Lyon; 2014. cation for cancer of the ovary, fallopian tube, and peritoneum. Int J
3. Darragh T, Colgan T, Cox JT, et al. The Lower Anogenital Squamous Gynecol Obstet. 2014;124:1–5.
Terminology Standardization Project for HPV-­associated lesions: 21. McClean GE, Kurian S, Walter N, Kekre A, McCluggage WG. Cervical
Background and consensus recommendations from the College of embryonal rhabdomyosarcoma and ovarian Sertoli-­Leydig cell
American Pathologists and the American Society for Colposcopy and tumour: A more than coincidental association of two rare neoplasms?
Cervical Pathology. J Lower Genit Tract Dis. 2012;16:205–242. J Clin Pathol. 2007;60:326–328.
DOI: 10.1002/ijgo.12609

FIGO CANCER REPORT 2018

Cancer of the vulva

Linda J. Rogers1,2,* | Mauricio A. Cuello3

1
Division of Gynecological Oncology, Groote
Schuur Hospital/University of Cape Town, Abstract
Cape Town, South Africa Vulvar cancer is an uncommon gynecological malignancy primarily affecting
2
South African Medical Research Council/
­postmenopausal women. There is no specific screening and the most effective strategy
University of Cape Town Gynaecological
Cancer Research Centre (SA MRC/UCT to reduce vulvar cancer incidence is the opportune treatment of predisposing and
GCRC), Cape Town, South Africa
­preneoplastic lesions associated with its development. While vulvar cancer may be
3
Division of Obstetrics and
asymptomatic, most women present with vulvar pruritus or pain, or have noticed a
Gynecology, School of Medicine, Pontificia
Universidad Católica de Chile, Santiago, Chile lump or ulcer. Therefore, any suspicious vulvar lesion should be biopsied to exclude
invasion. Once established, the most common subtype is squamous cell carcinoma.
*Correspondence
Linda J. Rogers, Department of Obstetrics and Treatment of vulvar cancer depends primarily on histology and surgical staging.
Gynecology, Groote Schuur Hospital, Cape
Treatment is predominantly surgical, particularly for squamous cell carcinoma, although
Town, South Africa.
Email: linda.rogers@uct.ac.za concurrent chemoradiation is an effective alternative, particularly for advanced tumors.
Management should be individualized, and carried out by a ­multidisciplinary team in a
cancer center experienced in the treatment of these tumors.

KEYWORDS
Cancer staging; Chemotherapy; Diagnostic imaging; FIGO Cancer Report; Radiotherapy; Risk
factors; Surgery; Therapy; Vulvar cancer; Vulvar neoplasms

1 | INTRODUCTION greater (Bartholin glands) vestibular glands.7 Most malignancies are


associated with the skin of the labia. Malignancies arising from the
Vulvar cancer is uncommon, accounting for only 2%–5% of gynecologic clitoris and vestibular glands are extremely rare.
malignancies. Squamous cell carcinoma (SCC) of the vulva, the most com- Lymphatic drainage from the vulva is primarily to the inguinofem-
mon subtype, has traditionally been regarded as a disease of postmeno- oral region, and secondarily to the external and internal iliac region.
pausal women, although the mean age of incidence has fallen in recent This drainage is shared with the inferior third of the vaginal tube and
years owing to the increase in HPV infections worldwide.1,2 Reinforcing the most external portion of the anus (below the anal sphincter).
this epidemiological change, differences in terms of current incidence or Depending on the localization of the primary tumor, its size, and its
age at presentation can be found between countries and regions; some closeness to the midline, lymphatic drainage can be unilateral or bilat-
may be explained by a different local HPV prevalence or other risk fac- eral. Additionally, if the lesion is close to or on the clitoris, drainage can
tors (e.g. ethnic distribution, smoking, atrophy or inflammation, HIV).3–6 be directly to the iliac region.8

3 | PREVENTION
2 | ANATOMY
3.1 | Primary prevention (vaccination)
The external genitalia comprise the vulva and the mons pubis or pubic
area. The vulva is located in the anterior triangle of the perineum. The As for cervical premalignant lesions predisposing to cervical cancer,
elements that make up the vulva include the labia minora and major, persistent HPV infection, particularly by HPV 16 subtype, has been
clitoris, bulb of the vaginal vestibule, and the lesser (Skene glands) and associated with the long-­term development of high-­grade squamous

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

4  |  wileyonlinelibrary.com/journal/ijgo Int J Gynecol Obstet 2018; 143 (Suppl. 2): 4–13
Rogers and Cuello       5|
intraepithelial lesion (HSIL) and SCC of the vulva.9–11 The introduc- T A B L E   1   Vulvar intraepithelial neoplasia (VIN) terminology
tion of HPV vaccination as a primary prevention strategy in cervical changes.
cancer has been shown to also reduce the prevalence of noncervical
LAST (Lower
premalignant lesions among vaccinated women.12 Long-­term trends
Anogenital Squamous
analyses by the Norwegian Cancer Register also show promising esti- ISSVD 1986 ISSVD 2004 Terminology) 2012
mates of reduction in HPV-­associated cases of vulvar cancer in future
VIN 1 Flat condylomata or LSIL
years, among HPV-­vaccinated communities.13 HPV effect
VIN 2 and VIN 3 VIN, usual type: HSIL
1. VIN, warty type
3.2 | Secondary prevention (screening)
2. VIN, basaloid type
3. VIN, mixed
There is no evidence for specific screening for vulvar cancer. Self-­
examination in women with lichen sclerosus, a condition related to vulvar Differentiated VIN, differentiated Differentiated VIN
14 VIN type (dVIN)
cancer development, should be encouraged. In addition, there should
Source: Hoang et al.,20 Bornstein et al.,21 Sideri et al.22
be early evaluation of any patient with signs (e.g. pigmented lesions, irreg-
ular ulcers) or symptoms (e.g. chronic vulvar pruritus) commonly associ-
ated with vulvar disease, who could be a candidate for skin biopsy.15 inhibitors (e.g. tacrolimus) or retinoids and photodynamic therapy
Finally, women who are known to have squamous intraepithelial for selected cases and/or cases resistant to corticosteroid therapy.
lesion (SIL) of the cervix, vagina, or anus should have inspection of the In women, surgery is restricted to scarring processes leading to func-
vulva as part of their follow-­up colposcopy visits.16 tional impairment.23
dVIN represents less than 5% of preneoplastic lesions of the
vulva. However, it is characterized by a higher rate of progression to
3.3 | Tertiary prevention (management of
squamous vulvar carcinoma, shorter time interval to progression, and
premalignant lesions)
higher recurrence rate than HSIL. It is rarely associated with persistent
An effective strategy to reduce vulvar cancer incidence is the oppor- HPV infection (less than 2%). Excision (with 0.5–1 cm margins) consti-
tune treatment of predisposing and preneoplastic lesions associated tutes the treatment of choice, to allow proper evaluation and exclusion
with vulvar cancer development. of occult invasion.24,25
17
There are two main pathological pathways that lead to vulvar SCC : Multiple treatment modalities exist for the management of HSIL,
but simple excision with 5-­mm margins and 4-­mm depth is the most
1. Keratinizing SCC usually occurs in older women and is often common. Excision has the advantage of excluding invasion histologi-
associated with lichen sclerosus and/or differentiated vulvar cally, but the lack of preservation of vulvar skin results in psychosex-
intraepithelial neoplasia (dVIN). ual morbidity, particularly in younger women. An alternative option
2. Warty/basaloid SCC generally occurs in younger women, is to preserve anatomy is the carbon dioxide laser, but this lacks the
caused by persistent infection with oncogenic strains of HPV assessment of occult invasion. A less destructive option is the use of
(particularly HPV 16, 18, 31 and 33), and has SIL as its precur- imiquimod 5% to avoid scarring and sexual dysfunction, particularly
sor lesion.6,18 Lesions are frequently multifocal, and may be in smaller lesions. Moderate quality evidence shows that response
associated with SIL in other parts of the lower genital tract (e.g. rates with imiquimod and cidofovir, another topical treatment, are
cervix, vagina, anus). HIV infection and cigarette smoking are similar at 6 months compared with surgical management or laser
also common predisposing factors. 1,3,9,19
vaporization.26 There is very little evidence of the effectiveness of
topical treatment for HSIL among immunocompromised women.26
As shown in Table 1, the terminology and definitions for prema- Independent of chosen treatment and margin status, there is risk of
lignant or precursor lesions of vulvar cancer have been reviewed and recurrence (up to 30%–40%). Therefore, a close follow-­up is recom-
changed in the last decades. Currently, such lesions arising from the mended for at least 2–3 years.24
vulva and the anus are all included and named as “lower anogenital
squamous intraepithelial lesions.” Under this classification, three sub-
4 | MANAGEMENT OF VULVAR CANCER
types are distinguished for the vulva: low-­grade squamous intraepi-
thelial lesions (LSIL); high-­grade squamous intraepithelial lesions
4.1 | Anatomy of disease spread
(HSIL); and differentiated variant. Such distinction correlates with the
risk of developing cancer over time.20–22
4.1.1 | Primary site
To date, there is no definitive treatment for conditions such as
lichen sclerosus. Cornerstone measures include avoiding exposure Malignant tumors of the vulva should be histologically confirmed, and
to precipitating factors (e.g. trauma by local irritants, occlusive moist are classified as such when the primary site of origin of the tumor is
environment) and the use of potent and ultrapotent topical cortico- the vulva. This includes tumors that involve both the vulva and vagina,
steroids. Alternative options include the use of topical calcineurin but excludes secondary tumors from genital and extragenital sites.27
6       | Rogers and Cuello

4. Adenocarcinoma, not otherwise specified


4.1.2 | Lymph nodes
5. Bartholin gland carcinoma
Inguinal and femoral nodes are the first sites of spread, followed by
the pelvic nodes. Depending on tumor size and its localization (closer
4.4 | Histological grades
to the middle line or to the clitoris), the risk of nodal involvement can
be unilateral or bilateral. 1. GX: Grade cannot be assessed
2. G1: Well differentiated
3. G2: Moderately differentiated
4.1.3 | Metastatic sites
4. G3: Poorly or undifferentiated
Women who have pelvic lymph node metastases or extrapelvic spread
are considered to have Stage IV disease.
4.5 | Treatment
The treatment of vulvar cancer depends primarily on histology and stag-
4.2 | Surgical staging
ing. Other variables influencing management are age, coexistence of
Vulvar cancer has been surgically staged since 1988 and final comorbidities, and performance status of the patient. Treatment is pre-
diagnosis is based on histological evaluation of the vulvar and dominantly surgical, particularly for SCC, although concurrent chemora-
lymph node specimens.19,27,28 The FIGO staging of vulvar car- diation is an effective alternative, particularly for advanced tumors, and
cinoma was revisited and last changed in 2009 by the FIGO those where exenteration would be necessary to achieve adequate sur-
Committee on Gynecologic Oncology (Table 2).16 This system gical margins.29 Management should be individualized, and carried out
is applicable for most of the malignancies rising from the vulva, by a multidisciplinary team in a cancer center experienced in the treat-
except melanoma. ment of these tumors.27,28,30 Other therapies such as chemotherapy and
immunotherapies are usually reserved for metastatic or palliative set-
tings, or for the treatment of rare histologies such as melanoma.28,30–34
4.3 | Histopathological types
Squamous cell carcinomas (SCC) account for the vast majority of vul-
5 | MANAGEMENT OF SQUAMOUS
var cancers (more than 80%), and melanomas are the next most com-
CELL CARCINOMA
mon cancer. Rarer histological types include:

5.1 | Presenting symptoms
1. Basal cell carcinoma
2. Verrucous carcinoma While vulvar cancer may be asymptomatic, most women present with
3. Paget’s disease of the vulva vulvar pruritus or pain, or have noticed a lump or ulcer. They may

T A B L E   2   FIGO staging of carcinoma of the vulva.16

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 >2 cm in size or with stromal invasion >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 1. With 1 lymph node metastasis (≥5 mm), or
2. With 1–2 lymph node metastasis(es) (<5 mm)
IIIB 1. With 2 or more lymph node metastases (≥5 mm), or
2. With 3 or more lymph node metastases (<5 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:
1. upper urethral and/or vaginal mucosa, bladder mucosa, rectal mucosa, or fixed to pelvic bone, or
2. 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.
Rogers and Cuello |
      7

also have abnormal bleeding or discharge, and many will have a his-
6.1 | Microinvasive vulvar cancer (Stage IA)
tory of vulvar symptoms due to underlying lichen sclerosus or HSIL.
Advanced vulvar cancer may present with a lump in the groin due to Stage IA vulvar carcinoma is defined as a lesion measuring 2 cm or less
lymph node metastases.16 in diameter, with a depth of invasion of 1.0 mm or less. The depth of
invasion is measured from the epithelial–stromal junction of the most
adjacent superficial dermal papilla to the deepest point of invasion.27
5.2 | Diagnosis
These lesions should be managed with radical wide local excision, and
Any suspicious vulvar lesion should be biopsied to exclude invasion. groin node dissection is not necessary.35
This can be done under local anesthetic with a 3 or 4 mm Keyes biopsy
instrument, or with an incisional or wedge biopsy. Even if the lesion is
6.2 | Early vulvar cancer
small, it is better not to excise the entire lesion at the time of biopsy,
as this makes the subsequent definitive surgery difficult to plan.16 Early vulvar cancers are those confined to the vulva, and where there
If the diameter of the lesion is 2 cm or less, and the depth of are no suspicious lymph nodes, either on clinical examination, or with
stromal invasion is less than or equal to 1 mm on the initial biopsy, ultrasound or other radiological assessment.19,27
it is usual to do a radical wide local excision of the lesion to assess The gold standard of treatment for early vulvar cancers is radical wide
the maximum depth of invasion. If no part of the lesion has a depth local excision of the tumor. This is as effective as a radical vulvectomy
of invasion greater than 1 mm, then this excision is adequate defin- in preventing local recurrence, but substantially decreases the psycho-
itive treatment. 35,36
sexual morbidity of the treatment.40–42 Associated preinvasive disease
should also be excised to exclude any other areas of invasion, and to
prevent new tumors arising in the so-­called “abnormal field.” While the
5.3 | Investigations
surgeon should aim for surgical margins of 2 cm to achieve pathological
margins of at least 8 mm (allowing for shrinkage of the fixed tissue), it is
1. Cervical cytology, and colposcopy of the cervix and vagina, if now recognized that many “recurrent” vulvar cancers are probably new
applicable, due to the association of HPV-related cancers with tumors that have developed in the surrounding abnormal tissue, rather
other squamous intraepithelial lesions. than recurrences due to inadequate margins.43 The deep margin of the
2. Full blood count, biochemical profile, liver profile, and HIV testing. excision should be the inferior fascia of the urogenital diaphragm and, if
3. Chest X-ray. necessary, the distal 1 cm of the urethra can be removed to achieve an
4. CT or MRI scan of the pelvis and groins may be helpful, especially adequate margin, without compromising urinary continence.27,35
for locally advanced tumors, to detect any enlarged lymph nodes The appropriate management of the groin lymph nodes is the
in the groins or pelvis, erosion into underlying bone, or other most important factor in reducing mortality from early vulvar cancer,
metastases.37 In addition, CT or MRI could be useful in further as recurrences in the groin are associated with poorer survival despite
treatment planning. using multimodal therapies as “rescue” treatments.44 The current stan-
18 18
5. F fluorodeoxyglucose ( F-FDG) positron emission tomography dard involves resection of the primary tumor and lymph nodes through
with computed tomography (PET-CT) can more effectively assess separate incisions.28 This approach allows better healing compared
and detect inguinofemoral lymph node involvement compared with with en bloc resection of the vulva and groins.45 Both inguinal and
CT, influencing the planning of primary surgery and inguinal lymph femoral nodes should be removed, as inguinal node dissection alone is
node dissection to determine the optimum surgical extent without associated with a higher incidence of groin recurrence.46 While some
sentinel lymph node dissection and use of frozen sections.37–39 reviews have suggested that radiation alone can control microscopic
Additionally, PET-CT might be used with larger tumors when meta- groin disease,47,48 a small randomized trial suggested that groin dissec-
28
static disease is suspected or in the recurrence scenario. tion, with postoperative irradiation for patients with positive nodes, is
superior to groin irradiation.49
All women who have Stage IB or Stage II cancers should have an
6 |  SURGICAL MANAGEMENT OF VULVAR inguinofemoral lymphadenectomy.
SQUAMOUS CE LL CARCIMOMA Less than 1% of patients who have small lateral lesions (less than
4 cm and ≥2 cm from the vulvar midline) and negative ipsilateral nodes
Surgical management of vulvar cancer should be individualized, and have metastases in the contralateral groin nodes, and therefore an ipsi-
the most conservative operation that will result in cure of the disease lateral groin dissection is adequate treatment for these patients.28,35
35,36
should be performed. Patients who have tumors closer to (<2 cm) or crossing the mid-
More importantly, when treatment options are considered, the line, especially those involving the anterior labia minora, and those
most appropriate treatment of: (1) the primary lesion; and (2) the women who have very large lateral tumors (>4 cm), or positive ipsilat-
groin lymph nodes, should be considered independently of each eral nodes, should have a bilateral groin node dissection.50
other, to maximize the chance of cure, while minimizing treatment-­ Since the findings of the GROINSS-­V study—a European multi-
related morbidity.27,28,30,32,35,36 center observational study on the sentinel lymph node procedure
|
8       Rogers and Cuello

in vulvar cancer—were published, the sentinel lymph node is being In terms of radiotherapy, radiation fields during external beam
utilized increasingly in the management of women with early vulvar radiotherapy (EBRT) should include the inguinofemoral and external
cancer. The aim of the procedure is to detect nodal metastases in the and internal iliac lymph nodes in most patients. If there are many or
“sentinel” node (which primarily drains the tumor), and then to omit bulky positive inguinal nodes, or if pelvic node metastases are sus-
a full lymphadenectomy in sentinel node negative patients, thereby pected, the upper border of the radiation field might be extended.61
decreasing the morbidity associated with a complete inguinofemoral Sometimes, brachytherapy can be added as a boost to anatomically
node dissection.8,51 amenable primary tumors.
Indications for a sentinel node procedure, as per the GROINSS-­V There is a variety of radiation techniques from which to choose,
study,51 are: depending on the patient’s body size and shape, and the extent of
the disease (e.g. 3D conformal/Anterior–Posterior/Posterior–Anterior
1. Unifocal tumors confined to the vulva [AP/PA] fields, intensity-­modulated radiation therapy [IMRT]). To
2. Tumors less than 4 cm in diameter ensure adequate tumor coverage, clinical examination, imaging find-
3. Stromal invasion more than 1 mm ings (CT or MRI), and nodal size should be considered to properly
4. Clinically negative groin nodes define the target volume during 3D planning.28,61
Combined photon and electron techniques are frequently used
Sentinel lymph nodes are identified using both radio-­labelled tech- to treat the regional nodes, without overdosing the femoral heads.
netium and blue dye.51,52 It is important to adequately include both the superficial and deep
In the GROINSS-­V study, 403 women were included and groin inguinal lymph nodes. Under-­dosage of superficial inguinal nodes by
recurrences occurred in 2.3% of patients, with a median follow-­up of high-­energy photon beams is a risk in thin patients, and care should be
35 months. Disease-­specific survival was 97% after 3 years, and surgi- taken to avoid this. Enough energy must be used to cover the femoral
cal morbidity was substantially reduced.51 nodes, if electron beams are used.27
Of note, when an ipsilateral sentinel lymph node is not detected, a IMRT or other inverse-­planned, computer-­controlled radiation-­
complete ipsilateral inguinofemoral lymphadenectomy must be done. delivery techniques are more modern methods that have been used in
In addition, if an ipsilateral sentinel lymph node is positive, a complete recent years to treat vulvar cancer. The benefits of this are decreased
bilateral inguinofemoral lymphadenectomy is recommended.28,53 acute radiation adverse effects in skin and soft tissue, but as the
A particular scenario in early disease is in the management of treatment planning and delivery of IMRT are complex, and the risk of
patients with positive groin nodes. The Gynecologic Oncology Group under-­dosage of the target is substantial, these techniques are best
protocol #37 showed that patients who were found to have more than utilized by clinicians who have the necessary expertise.27,28
one or grossly positive nodes at inguinal lymph node dissection, had Radiation dose is determined by the initial extent of disease, and
improved outcomes if they had adjuvant pelvic and inguinal radiation any known residual. After a groin lymphadenectomy where micro-
compared with those who had pelvic node dissection.54,55 A more scopic inguinal metastases are found, 50 Gy in 1.8–2.0 Gy fractions is
recent study, AGO-­CaRT-­1, also reported that women with positive usually sufficient. In the case of multiple positive nodes or extracap-
groin nodes who received adjuvant radiotherapy directed at the groins sular spread, radiation doses up to 60 Gy can be given to a reduced
56
had improved survival. volume. Gross residual disease usually requires 60–70 Gy to achieve a
Several studies have demonstrated the prognostic importance of high likelihood of regional disease control.27,28,61
the number and size of groin node metastases, as well as the pres- A 2015 analysis of the National Cancer Data Base (NCDB) sug-
ence of extracapsular spread.57–59 Patients with one small lymph node gested that women with node-­positive vulvar cancer benefitted the
metastasis appear to have a good prognosis after inguinofemoral most from the addition of chemotherapy to radiation.62
lymphadenectomy alone, unless extracapsular spread is present, and
these women do not appear to benefit from adjuvant radiation.58–60
6.3. | Advanced vulvar cancer
Therefore, indications for pelvic and groin irradiation in patients with
positive groin nodes are: Advanced vulvar cancer includes tumors that extend beyond the
vulva, and/or where there are bulky positive groin nodes.27 The man-
1. Presence of extracapsular spread. agement of women with advanced vulvar cancer is complex, and
2. Two or more positive groin nodes.27 should be individualized and carried out by a multidisciplinary team.
When confronted with advanced vulvar cancer, ideally the sta-
All patients who have a positive sentinel lymph node (one or more tus of the groin nodes should be determined before treatment is
positive nodes), besides undergoing a full inguinofemoral lymph node planned.27,28,30,32 Patients with clinically suspicious nodes should have
dissection, should receive radiotherapy to the groins and pelvis if indi- fine needle aspiration (FNA) or biopsy of their nodes, and pelvic CT, MRI,
cated. The sequel to the GROINSS-­V trial, GROINSS-­V II, is investigating or PET-­CT may be helpful in determining the extent of inguinal and pel-
the efficacy of groin radiation without inguinofemoral lymphadenec- vic lymphadenopathies and the presence of distant metastatic disease.63
tomy for patients with a single positive sentinel lymph node 2 mm or If there are no suspicious nodes either clinically or on imaging,
less in diameter.27,28 bilateral inguinofemoral lymphadenectomy may be performed, and if
Rogers and Cuello |
      9

the nodes are negative, radiotherapy to the groins and pelvic nodes more to minimize local disease recurrence. Multiple retrospective
will not be necessary. However, if histology reveals positive nodes, studies have tried to assess the factors that may determine vulvar
then adjuvant radiation to the groin and pelvis should be offered as recurrence, and other clinical determinants besides inadequate exci-
for early stage disease.61 sion margins have been suggested, although it is unclear which combi-
In cases where surgery is thought to be inappropriate for the indi- nation of factors is most significant.43,75
vidual patient, primary chemoradiation may be used to treat the pri- Two types of local recurrences, those at the same site as the origi-
mary tumor as well as the groin and pelvic nodes.28,47,48,61 nal (primary) tumor and those at a different vulvar site, were described
In patients who have clinically positive nodes, enlarged groin and by Rouzier et al.76 An analysis of vulvar cancer patients from the Royal
pelvic nodes should be removed if possible, and the patient given Hospital for Women in Sydney showed that primary site recurrences
64
postoperative groin and pelvic radiation. Full lymphadenectomy occurred with a median disease-­free interval of 21 months, and were
should not be performed because a full groin dissection followed by associated with a histological margin of 8 mm or less, as reported
groin irradiation may result in severe lymphedema. in several other papers.40,43,77 “Recurrences” at remote vulvar sites
Ulcerated or fixed groin lymph nodes should be biopsied to con- occurred later, with a median disease-­free interval of 69 months, and
firm the diagnosis, and then treated with primary radiation, with or were more commonly associated with lichen sclerosus.78,79
without chemotherapy. If there is an incomplete response to radiation, As most vulvar squamous carcinomas arise in a background of
the nodes may then be resected if appropriate.65 atypical skin such as HSIL, lichen sclerosus, and dVIN, and as they
In terms of the management of the primary tumor, surgical excision characteristically recur locally but often at sites remote from the orig-
of the primary tumor with clear surgical margins and without sphinc- inal tumor, it is suggested that many “recurrences” may actually be
ter damage, whenever possible, constitutes the optimum way to treat second primary tumors, which arise in a “field of cancerization”—an
advanced vulvar cancer, as well as to palliate symptoms such as local area of genetically-­altered preneoplastic epithelium that has the pre-
pain and offensive discharge.35 disposition to undergo malignant transformation.43
If adequate excision of the primary tumor can only be achieved by Patients with close (less than 5 mm) surgical margins benefit from
exenteration and the formation of a bowel or urinary stoma, radiother- postoperative radiotherapy, if it is not possible to re-­excise the mar-
apy (with or without concurrent chemotherapy) may be a preferred gins.80 A study of 205 women with vulvar cancer from Boston reported
treatment alternative. Survival is improved if any postradiation resid- that margins of 5 mm or less posed the highest risk of vulvar recur-
66,67
ual tumor is resected. rence, and that patients who received a dose of more than or equal to
Concurrent chemoradiation is a well-­described treatment alter- 56 Gy had a lower risk of relapse than those who received less than or
native for those patients with large tumors in whom primary surgical equal to 50.4 Gy.81
resection would damage central structures (anus, urethra), and long-­ Occasionally, the positive margins may be boosted with
68–72
term complete responses have been reported. The groin nodes brachytherapy, although care must be taken to avoid the risk of necro-
and pelvis may need to be included in the radiation field depending on sis. An alternative is to treat the operative bed with an appositional
the status of the groin nodes, as determined initially.27,28,61 electron field or conformal external beam irradiation.27,61
Neoadjuvant treatment with cisplatin and 5-­fluorouracil, or other
chemotherapy combinations, has been reported to be effective for pres-
7 | RARE VULVAR MALIGNANCIES
ervation of the anal sphincter and/or urethra in patients with advanced
vulvar cancer.73,74 This is the subject of ongoing clinical research.
7.1 | Vulvar melanoma
In relation to radiotherapy planning in advanced vulvar cancer, if
the groin nodes are positive and meet the previously described indi- Vulvar melanoma is the second most common vulvar malignancy.
cations for adjuvant radiation, the radiation treatment fields should Any pigmented vulvar lesion should be biopsied or excised for
include the pelvis, inguinal nodes, and vulva. These should be treated diagnosis, unless it has been present and unchanged for some
to a total dose of at least 50 Gy, with attention to adequate coverage time.24 The majority of vulvar melanomas involve the clitoris or
of the inguinal nodes.27,61 labia minora.27 The Clark or Breslow modifications of the staging
Gross disease or high-­risk areas may be boosted either with appo- system—as included in the American Joint Committee on Cancer
sitional fields of electrons selected to provide an adequate dose to (AJCC) system and based on depth of invasion—should be used for
the surface and at depth, or with conformal external beam therapy. the staging of these lesions rather than the FIGO staging system
Large vulvar tumors probably require 60–70 Gy to achieve local con- since it is the only system prospectively proven to be correlated
trol, although the relationship between dose and local control remains with recurrence and survival.82–84
27,61
the subject of ongoing investigation. Surgery is the treatment of choice for vulvar melanomas. Lesions
should be treated by radical wide local excision, with margins around
the lesion of at least 1 cm.27 The current trend leans toward more
6.4 | Close surgical margins
conservative resection of vulvar melanomas because no survival dif-
Most vulvar cancer recurrences occur on the vulva. It is thought that ference has been found in patients undergoing local excision versus
surgeons should aim for tumor-­free pathological margins of 8 mm or those with radical vulvectomy.85
|
10       Rogers and Cuello

The role of lymph node dissection is also controversial and, to urethra or anus also present a management challenge, and may
date, no survival advantage has been shown for inguinal lymph- require laser therapy.6 Another conservative treatment option is local
84
adenectomy, although the Intergroup Surgical Melanoma Program’s imiquimod.93 A Cochrane meta-­analysis that investigated treatment
prospective, multi-­institutional randomized trial of elective node options concluded that there was no “best” intervention for vulvar
dissection versus observation for intermediate thickness cutane- Paget disease.94
ous melanomas (1–4 mm) revealed that elective node dissection If an underlying adenocarcinoma is present, treatment should be
resulted in a significantly better survival for patients aged 60 years radical wide local excision with margins of at least 1 cm. Inguinofemoral
or younger, patients with tumors 1–2 mm thick, and patients with no lymphadenectomy should be performed, with adjuvant radiation for
tumor ulceration.86 the same indications as for squamous carcinomas.95
Sentinel lymph node evaluation has also been explored for vulvar
melanoma, and although it is feasible, a false-­negative rate of 15%
has been reported87; it has been suggested that the procedure may 8 | PATHOLOGY CONSIDERATIONS
increase the risk of locoregional recurrences,88 and therefore it is not
current standard practice. In relation to specimen analyses, the following should be noted:

1. Orientation: correct orientation of the surgical specimen is


7.2 | Bartholin gland cancer
important.
Bartholin gland carcinomas are rare forms of vulvar malignancy and it 2. Photographs: these should be taken of the whole specimen, as well
is unclear what proportion is associated with high-­risk HPV infection. as the origin of each tissue block.
Cancers of the Bartholin glands may be either transitional cell or SCC 3. Measurements: size of the specimen, dimensions of any visible
that arise from the duct, or adenocarcinomas arising from the glands tumor, macroscopic tumor-free margins, and tumor depth (sections
themselves. There are also adenoid cystic and adenosquamous vari- taken through the tumor). Sections should also be taken from ure-
ants. All SCCs showed diffuse and intense p16 expression consistent thral, anal, and vaginal resection margins.27
89
with the presence of HPV. The diagnosis is often made after resec- 4. Lymph nodes: these should be dissected out, the site from which
tion of a persistent or recurrent Bartholin cyst.27 they are removed recorded, and a full cross-section of each lymph
Bartholin gland carcinomas are effectively treated with a radical node should be embedded.27
hemivulvectomy and bilateral groin dissection; however, due to the
location of these tumors, deep in the ischiorectal fossa, adequate sur- The following histological points should be noted:
gical margins are difficult to achieve and postoperative radiation may
decrease the likelihood of local recurrence.90 1. Tumor type.
Radical wide local excision alone is adequate treatment for ade- 2. Depth of invasion: measured from the epithelial–stromal junction
noid cystic lesions, and adjuvant radiation is recommended for posi- of the adjacent dermal papilla to the deepest point of invasion by
tive margins or perineural invasion.91 the tumor.
3. Tumor grade.
4. Histological measurement of tumor-free margins and statement as
7.3 | Paget disease of the vulva
to whether the tumor is completely excised.
Extramammary Paget disease is rare, and can affect the apocrine 5. Presence or absence of perineural or vascular space invasion.
glands of the vulva. There are two types: the primary form begins as 6. Nature of the adjacent squamous epithelium, e.g. dVIN, lichen scle-
an intraepithelial lesion, but the secondary form is due to invasion rosus, and HPV-associated changes.
from an underlying adenocarcinoma, which may be anorectal, urothe- 7. Sites and number of nodes examined, number of positive nodes,
lial, or genital tract carcinoma (e.g. endocervical or endometrial).92 and presence or absence of extracapsular extension.27
Vulvar Paget disease occurs predominantly in postmenopausal
women who present with vulvar pruritus and pain and, on examina-
AU T HO R CO NT R I B U T I O NS
tion, an eczematoid weeping lesion is often seen.24 Diagnosis is usu-
ally confirmed by biopsy, which will also help to differentiate between LR and MC contributed equally in designing, planning, reviewing lit-
24
an intraepithelial and an invasive lesion. erature, manuscript writing, and updating references.
The treatment of choice for intraepithelial Paget disease is wide
local excision. It is challenging to achieve clear margins as histologi-
AC KNOW L ED G M ENTS
cal changes often extend far beyond what is visible macroscopically;
even with adequate margins, recurrence rates are high. Due to the This chapter reworks and updates the information published in the
high recurrence rate and surgical morbidity, there is a current move FIGO Cancer Report 2015 (Hacker NF, Eifel PJ, van der Velden
to perform less radical resection for intraepithelial lesions, with re-­ J. Cancer of the vulva. Int J Gynecol Obstet. 2015;131(Suppl.2):
excision at a later date should lesions recur.93 Lesions involving the S76–83).
Rogers and Cuello |
      11

CO NFLI CTS OF I NT E RE ST 20. Hoang LN, Park KJ, Soslow RA, Murali R. Squamous precursor
lesions of the vulva: Current classification and diagnostic challenges.
The authors have no conflicts of interest to declare. Pathology. 2016;48:291–302.
21. Bornstein J, Bogliatto F, Haefner HK, et  al. The 2015 International
Society for the Study of Vulvovaginal Disease (ISSVD) terminol-
ogy of vulvar squamous intraepithelial lesions. Obstet Gynecol.
REFERENCES
2016;127:264–268.
1. Barlow EL, Kang YJ, Hacker NF, Canfell K. Changing trends in vul- 22. Sideri M, Jones RW, Wilkinson EJ, et  al. Squamous vulvar intraepi-
var cancer incidence and mortality rates in Australia Since 1982. Int J thelial neoplasia: 2004 modified terminology, ISSVD Vulvar Oncology
Gynecol Cancer. 2015;25:1683–1689. Subcommittee. J Reprod Med. 2005;50:807–810.
2. Kang YJ, Smith M, Barlow E, Coffey K, Hacker N, Canfell K. Vulvar 23. Fistarol SK, Itin PH. Diagnosis and treatment of lichen sclerosus: An
cancer in high-­income countries: Increasing burden of disease. Int J update. Am J Clin Dermatol. 2013;14:27–47.
Cancer. 2017;141:2174–2186. 24. Allbritton JI. Vulvar neoplasms, benign and malignant. Obstet Gynecol
3. Butt JL, Botha MH. Vulvar cancer is not a disease of the elderly: Clin North Am. 2017;44:339–352.
Treatment and outcome at a tertiary referral centre in South Africa. S 25. Eva LJ, Ganesan R, Chan KK, Honest H, Luesley DM. Differentiated-­
Afr Med J. 2017;107:1000–1004. type vulvar intraepithelial neoplasia has a high-­risk association with vul-
4. Muigai J, Jacob L, Dinas K, Kostev K, Kalder M. Potential delay in the var squamous cell carcinoma. Int J Gynecol Cancer. 2009;19:741–744.
diagnosis of vulvar cancer and associated risk factors in women treated 26. Lawrie TA, Nordin A, Chakrabarti M. Medical and surgical treat-
in German gynecological practices. Oncotarget. 2018;9:8725–8730. ments for usual-­type vulvar intraepithelial neoplasia. JAMA Oncol.
5. Xiao X, Meng YB, Bai P, et al. Vulvar cancer in China: Epidemiological 2016;2:1647–1648.
features and risk analysis. J Cancer. 2017;8:2950–2958. 27. Hacker NF, Eifel PJ, van der Velden J. Cancer of the vulva. Int J Gynecol
6. Faber MT, Sand FL, Albieri V, Norrild B, Kjaer SK, Verdoodt F. Obstet. 2015;131(Suppl 2):S76–S83.
Prevalence and type distribution of human papillomavirus in squa- 28. Koh WJ, Greer BE, Abu-Rustum NR, et  al. Vulvar cancer, version
mous cell carcinoma and intraepithelial neoplasia of the vulva. Int J 1.2017, NCCN clinical practice guidelines in oncology. J Natl Compr
Cancer. 2017;141:1161–1169. Canc Netw. 2017;15:92–120.
7. Cobos GA, Pomeranz MK. A general approach to the evaluation and 29. Rao YJ, Chin RI, Hui C, et al. Improved survival with definitive chemo-
the management of vulvar disorders. Obstet Gynecol Clin North Am. radiation compared to definitive radiation alone in squamous cell
2017;44:321–327. carcinoma of the vulva: A review of the National Cancer Database.
8. Coleman RL, Ali S, Levenback CF, et al. Is bilateral lymphadenectomy Gynecol Oncol. 2017;146:572–579.
for midline squamous carcinoma of the vulva always necessary? An 30. Oonk MHM, Planchamp F, Baldwin P, et  al. European Society of
analysis from Gynecologic Oncology Group (GOG) 173. Gynecol Gynaecological Oncology Guidelines for the management of patients
Oncol. 2013;128:155–159. with vulvar cancer. Int J Gynecol Cancer. 2017;27:832–837.
9. Rantshabeng PS, Moyo S, Moraka NO, et al. Prevalence of oncogenic 31. O’Donnell RL, Verleye L, Ratnavelu N, Galaal K, Fisher A, Naik R.
human papillomavirus genotypes in patients diagnosed with anogen- Locally advanced vulva cancer: A single centre review of anovulvec-
ital malignancies in Botswana. BMC Infect Dis. 2017;17:731. tomy and a systematic review of surgical, chemotherapy and radio-
10. Hampl M, Sarajuuri H, Wentzensen N, Bender HG, Kueppers V. therapy alternatives. Is an international collaborative RCT destined
Effect of human papillomavirus vaccines on vulvar, vaginal, and for the “too difficult to do” box? Gynecol Oncol. 2017;144:438–447.
anal intraepithelial lesions and vulvar cancer. Obstet Gynecol. 32. Saito T, Tabata T, Ikushima H, et  al. Japan Society of Gynecologic
2006;108:1361–1368. Oncology guidelines 2015 for the treatment of vulvar cancer and vag-
11. Serrano B, de Sanjose S, Tous S, et al. Human papillomavirus genotype inal cancer. Int J Clin Oncol. 2018;23:201–234.
attribution for HPVs 6, 11, 16, 18, 31, 33, 45, 52 and 58 in female 33. Leitao MM Jr. Management of vulvar and vaginal melanomas:
anogenital lesions. Eur J Cancer. 2015;51:1732–1741. Current and future strategies. Am Soc Clin Oncol Educ Book.
12. Garland SM, Paavonen J, Jaisamrarn U, et al. Prior human papilloma- 2014;34:e277–e281.
virus-­16/18 AS04-­adjuvanted vaccination prevents recurrent high 34. Mahner S, Prieske K, Grimm D, et  al. Systemic treatment of vulvar
grade cervical intraepithelial neoplasia after definitive surgical ther- cancer. Expert Rev Anticancer Ther. 2015;15:629–637.
apy: Post-­hoc analysis from a randomized controlled trial. Int J Cancer. 35. Dellinger TH, Hakim AA, Lee SJ, Wakabayashi MT, Morgan RJ, Han
2016;139:2812–2826. ES. Surgical management of vulvar cancer. J Natl Compr Canc Netw.
13. Hansen BT, Campbell S, Nygard M. Long-­term incidence trends of 2017;15:121–128.
HPV-­related cancers, and cases preventable by HPV vaccination: A 36. Micheletti L, Preti M. Surgery of the vulva in vulvar cancer. Best Pract
registry-­based study in Norway. BMJ Open. 2018;8:e019005. Res Clin Obstet Gynaecol. 2014;28:1074–1087.
14. Halonen P, Jakobsson M, Heikinheimo O, Riska A, Gissler M, Pukkala E. 37. Lin G, Chen CY, Liu FY, et al. Computed tomography, magnetic reso-
Lichen sclerosus and risk of cancer. Int J Cancer. 2017;140:1998–2002. nance imaging and FDG positron emission tomography in the man-
15. Palumbo AR, Fasolino C, Santoro G, et al. Evaluation of symptoms and agement of vulvar malignancies. Eur Radiol. 2015;25:1267–1278.
prevention of cancer in menopause: The value of vulvar exam. Transl 38. Robertson NL, Hricak H, Sonoda Y, et al. The impact of FDG-­PET/CT
Med UniSa. 2016;15:74–79. in the management of patients with vulvar and vaginal cancer. Gynecol
16. Hacker NF. Revised FIGO staging for carcinoma of the vulva. Int J Oncol. 2016;140:420–424.
Gynecol Obstet. 2009;105:105–106. 39. Dolanbay M, Ozcelik B, Abdulrezzak U, Serin IS, Kutuk MS, Uludag
17. Rakislova N, Clavero O, Alemany L, et al. Histological characteristics S. F-­18 fluoro-­D-­glucose (FDG)-­positron emission tomography
of HPV-­associated and -­independent squamous cell carcinomas of (PET)/computed tomography (CT) in planning of surgery and sen-
the vulva: A study of 1,594 cases. Int J Cancer. 2017;141:2517–2527. tinel lymph node screening in vulvar cancers. Arch Gynecol Obstet.
18. Pils S, Gensthaler L, Alemany L, Horvat R, de Sanjose S, Joura EA. 2016;293:1319–1324.
HPV prevalence in vulvar cancer in Austria. Wien Klin Wochenschr. 40. De Hullu JA, Hollema H, Lolkema S, et al. Vulvar carcinoma. The price
2017;129:805–809. of less radical surgery. Cancer. 2002;95:2331–2338.
19. Hacker NF, Barlow EL. Staging for vulvar cancer. Best Pract Res Clin 41. Khanna N, Rauh LA, Lachiewicz MP, Horowitz IR. Margins for cervical
Obstet Gynaecol. 2015;29:802–811. and vulvar cancer. J Surg Oncol. 2016;113:304–309.
|
12       Rogers and Cuello

42. Vitale SG, Valenti G, Biondi A, Rossetti D, Frigerio L. Recent trends in 61. Gaffney DK, King B, Viswanathan AN, et al. Consensus recommenda-
surgical and reconstructive management of vulvar cancer: Review of tions for radiation therapy contouring and treatment of vulvar carci-
literature. Updates Surg. 2015;67:367–371. noma. Int J Radiat Oncol Biol Phys. 2016;95:1191–1200.
43. Yap J, O’Neill D, Nagenthiran S, Dawson CW, Luesley DM. Current 62. Gill BS, Bernard ME, Lin JF, et al. Impact of adjuvant chemotherapy
insights into the aetiology, pathobiology, and management of local with radiation for node-­positive vulvar cancer: A National Cancer
disease recurrence in squamous cell carcinoma of the vulva. BJOG. Data Base (NCDB) analysis. Gynecol Oncol. 2015;137:365–372.
2017;124:946–954. 63. Viswanathan C, Kirschner K, Truong M, Balachandran A, Devine
44. Frey JN, Hampl M, Mueller MD, Gunthert AR. Should groin recurrence C, Bhosale P. Multimodality imaging of vulvar cancer: Staging,
still be considered as a palliative situation in vulvar cancer patients? A therapeutic response, and complications. AJR Am J Roentgenol.
brief report. Int J Gynecol Cancer. 2016;26:575–579. 2013;200:1387–1400.
45. Hacker NF, Leuchter RS, Berek JS, Castaldo TW, Lagasse LD. Radical 64. Hyde SE, Valmadre S, Hacker NF, Schilthuis MS, Grant PT, van der
vulvectomy and bilateral inguinal lymphadenectomy through sepa- Velden J. Squamous cell carcinoma of the vulva with bulky positive
rate groin incisions. Obstet Gynecol. 1981;58:574–579. groin nodes-­nodal debulking versus full groin dissection prior to radi-
46. Stehman FB, Bundy BN, Dvoretsky PM, Creasman WT. Early stage ation therapy. Int J Gynecol Cancer. 2007;17:154–158.
I carcinoma of the vulva treated with ipsilateral superficial inguinal 65. Montana GS, Thomas GM, Moore DH, et  al. Preoperative
lymphadenectomy and modified radical hemivulvectomy: A pro- chemo-­radiation for carcinoma of the vulva with N2/N3 nodes:
spective study of the Gynecologic Oncology Group. Obstet Gynecol. A gynecologic oncology group study. Int J Radiat Oncol Biol Phys.
1992;79:490–497. 2000;48:1007–1013.
47. Katz A, Eifel PJ, Jhingran A, Levenback CF. The role of radiation ther- 66. Boronow RC, Hickman BT, Reagan MT, Smith RA, Steadham RE.
apy in preventing regional recurrences of invasive squamous cell car- Combined therapy as an alternative to exenteration for locally
cinoma of the vulva. Int J Radiat Oncol Biol Phys. 2003;57:409–418. advanced vulvovaginal cancer. II. Results, complications, and dosim-
48. Petereit DG, Mehta MP, Buchler DA, Kinsella TJ. Inguinofemoral radi- etric and surgical considerations. Am J Clin Oncol. 1987;10:171–181.
ation of N0, N1 vulvar cancer may be equivalent to lymphadenec- 67. Hacker NF, Berek JS, Juillard GJ, Lagasse LD. Preoperative
tomy if proper radiation technique is used. Int J Radiat Oncol Biol Phys. radiation therapy for locally advanced vulvar cancer. Cancer.
1993;27:963–967. 1984;54:2056–2061.
49. Stehman FB, Bundy BN, Thomas G, et  al. Groin dissection versus 68. Beriwal S, Coon D, Heron DE, et al. Preoperative intensity-­modulated
groin radiation in carcinoma of the vulva: A Gynecologic Oncology radiotherapy and chemotherapy for locally advanced vulvar carci-
Group study. Int J Radiat Oncol Biol Phys. 1992;24:389–396. noma. Gynecol Oncol. 2008;109:291–295.
50. Stehman FB, Look KY. Carcinoma of the vulva. Obstet Gynecol. 69. Cunningham MJ, Goyer RP, Gibbons SK, Kredentser DC, Malfetano JH,
2006;107:719–733. Keys H. Primary radiation, cisplatin, and 5-­fluorouracil for advanced
51. Van der Zee AG, Oonk MH, De Hullu JA, et al. Sentinel node dissec- squamous carcinoma of the vulva. Gynecol Oncol. 1997;66:258–261.
tion is safe in the treatment of early-­stage vulvar cancer. J Clin Oncol. 70. Landoni F, Maneo A, Zanetta G, et al. Concurrent preoperative che-
2008;26:884–889. motherapy with 5-­fluorouracil and mitomycin C and radiotherapy
52. Levenback CF, Ali S, Coleman RL, et al. Lymphatic mapping and sen- (FUMIR) followed by limited surgery in locally advanced and recurrent
tinel lymph node biopsy in women with squamous cell carcinoma vulvar carcinoma. Gynecol Oncol. 1996;61:321–327.
of the vulva: A gynecologic oncology group study. J Clin Oncol. 71. Lupi G, Raspagliesi F, Zucali R, et al. Combined preoperative chemo-
2012;30:3786–3791. radiotherapy followed by radical surgery in locally advanced vulvar
53. Covens A, Vella ET, Kennedy EB, Reade CJ, Jimenez W, Le T. Sentinel carcinoma. A pilot study. Cancer. 1996;77:1472–1478.
lymph node biopsy in vulvar cancer: Systematic review, meta-­ 72. Moore DH, Ali S, Koh WJ, et al. A phase II trial of radiation therapy and
analysis and guideline recommendations. Gynecol Oncol. 2015;137: weekly cisplatin chemotherapy for the treatment of locally-­advanced
351–361. squamous cell carcinoma of the vulva: A gynecologic oncology group
54. Homesley HD, Bundy BN, Sedlis A, Adcock L. Radiation therapy ver- study. Gynecol Oncol. 2012;124:529–533.
sus pelvic node resection for carcinoma of the vulva with positive 73. Aragona AM, Cuneo N, Soderini AH, et al. Tailoring the treatment of
groin nodes. Obstet Gynecol. 1986;68:733–740. locally advanced squamous cell carcinoma of the vulva: Neoadjuvant
55. Kunos C, Simpkins F, Gibbons H, Tian C, Homesley H. Radiation therapy chemotherapy followed by radical surgery: Results from a multicenter
compared with pelvic node resection for node-­positive vulvar cancer: study. Int J Gynecol Cancer. 2012;22:1258–1263.
A randomized controlled trial. Obstet Gynecol. 2009;114:537–546. 74. Geisler JP, Manahan KJ, Buller RE. Neoadjuvant chemotherapy
56. Mahner S, Jueckstock J, Hilpert F, et  al. Adjuvant therapy in lymph in vulvar cancer: Avoiding primary exenteration. Gynecol Oncol.
node-­positive vulvar cancer: The AGO-­CaRE-­1 study. J Natl Cancer 2006;100:53–57.
Inst. 2015;107:pii:dju426. 75. Te Grootenhuis NC, Pouwer AW, de Bock GH, et al. Prognostic factors
57. Origoni M, Sideri M, Garsia S, Carinelli SG, Ferrari AG. Prognostic for local recurrence of squamous cell carcinoma of the vulva: A sys-
value of pathological patterns of lymph node positivity in squamous tematic review. Gynecol Oncol. 2018;148:622–631.
cell carcinoma of the vulva stage III and IVA FIGO. Gynecol Oncol. 76. Rouzier R, Haddad B, Plantier F, Dubois P, Pelisse M, Paniel
1992;45:313–316. BJ. Local relapse in patients treated for squamous cell vul-
58. Paladini D, Cross P, Lopes A, Monaghan JM. Prognostic significance of var carcinoma: Incidence and prognostic value. Obstet Gynecol.
lymph node variables in squamous cell carcinoma of the vulva. Cancer. 2002;100:1159–1167.
1994;74:2491–2496. 77. Chan JK, Sugiyama V, Pham H, et  al. Margin distance and other
59. van der Velden J, van Lindert AC, Lammes FB, et  al. Extracapsular clinico-­pathologic prognostic factors in vulvar carcinoma: A multivari-
growth of lymph node metastases in squamous cell carcinoma ate analysis. Gynecol Oncol. 2007;104:636–641.
of the vulva. The impact on recurrence and survival. Cancer. 78. Ragupathy K, Grandidge L, Strelley K, Wang H, Tidy J. Early and late
1995;75:2885–2890. vulvar cancer recurrences: Are they different? J Obstet Gynaecol.
60. Fons G, Groenen SM, Oonk MH, et  al. Adjuvant radiotherapy in 2016;36:518–521.
patients with vulvar cancer and one intra capsular lymph node metas- 79. Tantipalakorn C, Robertson G, Marsden DE, Gebski V, Hacker NF.
tasis is not beneficial. Gynecol Oncol. 2009;114:343–345. Outcome and patterns of recurrence for International Federation of
Rogers and Cuello |
      13

Gynecology and Obstetrics (FIGO) stages I and II squamous cell vulvar 87. Dhar KK, Das N, Brinkman DA, Beynon JL, Woolas RP. Utility of senti-
cancer. Obstet Gynecol. 2009;113:895–901. nel node biopsy in vulvar and vaginal melanoma: Report of two cases
80. Faul CM, Mirmow D, Huang Q, Gerszten K, Day R, Jones MW. and review of the literature. Int J Gynecol Cancer. 2007;17:720–723.
Adjuvant radiation for vulvar carcinoma: Improved local control. Int J 88. de Hullu JA, Hollema H, Hoekstra HJ, et al. Vulvar melanoma: Is there
Radiat Oncol Biol Phys. 1997;38:381–389. a role for sentinel lymph node biopsy? Cancer. 2002;94:486–491.
81. Viswanathan AN, Pinto AP, Schultz D, Berkowitz R, Crum CP. 89. Nazeran T, Cheng AS, Karnezis AN, Tinker AV, Gilks CB. Bartholin
Relationship of margin status and radiation dose to recurrence in gland carcinoma: Clinicopathologic features, including p16 expression
post-­operative vulvar carcinoma. Gynecol Oncol. 2013;130:545–549. and clinical outcome. Int J Gynecol Pathol. 2018. Feb 5 [Epub ahead of
82. Iacoponi S, Rubio P, Garcia E, et al. Prognostic factors of recurrence print]
and survival in vulvar melanoma: Subgroup analysis of the VULvar 90. Di Donato V, Casorelli A, Bardhi E, et al. Bartholin gland cancer. Crit
CANcer study. Int J Gynecol Cancer. 2016;26:1307–1312. Rev Oncol Hematol. 2017;117:1–11.
83. Moxley KM, Fader AN, Rose PG, et  al. Malignant melanoma of 91. Yang SY, Lee JW, Kim WS, et  al. Adenoid cystic carcinoma of the
the vulva: An extension of cutaneous melanoma? Gynecol Oncol. Bartholin’s gland: Report of two cases and review of the literature.
2011;122:612–617. Gynecol Oncol. 2006;100:422–425.
84. Phillips GL, Bundy BN, Okagaki T, Kucera PR, Stehman FB. Malignant 92. Parashurama R, Nama V, Hutson R. Paget’s disease of the vulva: A
melanoma of the vulva treated by radical hemivulvectomy. A pro- review of 20 years’ experience. Int J Gynecol Cancer. 2017;27:791–793.
spective study of the Gynecologic Oncology Group. Cancer. 1994;73: 93. Black D, Tornos C, Soslow RA, Awtrey CS, Barakat RR, Chi DS. The out-
2626–2632. comes of patients with positive margins after excision for intraepithe-
85. Nobbenhuis MA, Lalondrelle S, Larkin J, Banerjee S. Management of lial Paget’s disease of the vulva. Gynecol Oncol. 2007;104:547–550.
melanomas of the gynaecological tract. Curr Opin Oncol. 2014;26: 94. Edey KA, Allan E, Murdoch JB, Cooper S, Bryant A. Interventions for
508–513. the treatment of Paget’s disease of the vulva. Cochrane Database Syst
86. Balch CM, Soong SJ, Bartolucci AA, et  al. Efficacy of an elective Rev. 2013;(10):CD009245.
regional lymph node dissection of 1 to 4  mm thick melanomas for 95. Karam A, Dorigo O. Treatment outcomes in a large cohort of
patients 60 years of age and younger. Ann Surg. 1996;224:255–263; patients with invasive Extramammary Paget’s disease. Gynecol Oncol.
discussion 263–256. 2012;125:346–351.
DOI: 10.1002/ijgo.12610

FIGO CANCER REPORT 2018

Cancer of the vagina

Tracey S. Adams1,2,* | Mauricio A. Cuello3

1
Department of Gynecological Oncology,
Groote Schuur Hospital/University of Cape Abstract
Town, Cape Town, South Africa Diagnosis of a primary vaginal cancer is rare because most of these lesions will be
2
South African Medical Research Council/
metastatic from another primary site. Although cancer of the vagina is more common
University of Cape Town Gynaecological
Cancer Research Centre (SA MRC/UCT in postmenopausal women, an increase in young women being diagnosed with primary
GCRC), Cape Town, South Africa
vaginal cancer has been reported, especially in countries with a high HIV prevalence.
3
Division of Obstetrics and Gynecology,
This will be associated with persistence of high-­risk HPV infection. The emphasis
School of Medicine, Pontificia Universidad
Católica de Chile, Santiago, Chile should be on primary prevention with prophylactic HPV vaccination. Once there is a
suspicion of a primary vaginal cancer, this should be confirmed histologically with
*Correspondence
Tracey S. Adams, Department of Obstetrics biopsy. Staging has been done clinically, similar to cervical cancer; however, there is a
and Gynecology, Groote Schuur Hospital/
role for imaging in assisting with staging as this is often a difficult assessment.
University of Cape Town Observatory, Cape
Town, South Africa. Treatment should be individualized and depends on stage as well as histologic ­subtype.
Email: tracey.adams@uct.ac.za
It is prudent to refer cases to centers of excellence with experience in dealing with this
rare gynecological cancer.

KEYWORDS
Adenocarcinoma; FIGO Cancer Report; HPV; Imaging techniques; Individualized treatment;
Sarcoma; Staging; Vaginal cancer

1 | INTRODUCTION 2 | ANATOMY

Primary vaginal cancer is rare, constituting only 1%–2% of all female The vagina is an elastic muscular tube comprising many mucosal folds.
genital tract malignancies and only 10% of all vaginal malignant It extends from the cervix of the uterus to the hymenal ring, posterior
neoplasms.1 It is strictly defined as a cancer found in the vagina to the bladder and anterior to the rectum. Its elasticity and dimen-
without clinical or histologic evidence of cervical or vulvar cancer, sions vary depending on a woman’s age, parity, previous surgeries,
or a prior history of these cancers within five years.2 To support and hormonal status. Such characteristics can make proper examina-
this, most suspicious lesions of malignancy in the vagina will corre- tion (limited by pain or narrowness of the introitus) and identification
spond to metastatic lesions of cervical or vulvar cancer, or others of small malignant lesions difficult.
metastasizing to the vagina (e.g. breast, endometrium, trophoblast,
ovary, lymphoma).
3 | PREVENTION
Historically these cancers are more common in elderly and post-
menopausal women. If vaginal malignancy is found in younger women,
3.1 | Primary prevention (vaccination)
it is etiologically linked to cervical cancer, specifically with regard to the
persistence of high-­risk HPV infections.3 Vaginal cancers, although rare, As with premalignant cervical lesions and carcinoma of the cervix,
are increasingly being seen in younger women owing to the increase in persistent HPV infection—particularly the HPV 16 subtype—has
persistent high-­risk HPV infections, especially in settings with a high been associated with the long-­term development of high-­grade squa-
HIV prevalence. mous intraepithelial lesion (HSIL) and carcinoma of the vagina.4–6 The

This is an open access article under the terms of the Creative Commons AttributionLicense, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

14  |  wileyonlinelibrary.com/journal/ijgo Int J Gynecol Obstet 2018; 143 (Suppl. 2): 14–21
Adams and Cuello |
      15

introduction of HPV vaccination as a primary prevention strategy in depends on the number and location of the lesions, and the degree of
cervical cancer has also been shown to reduce the prevalence of non- suspicion for an invasive cancer, as well as the availability of various
cervical premalignant lesions among vaccinated women.7 Long-­term treatment options, cost of treatment, and skill of the treating doctor.
trends analyses from the Norwegian Cancer Register also show prom-
ising estimates of reduction in HPV-­associated cases of vaginal cancer
3.3.1 | Surgical excision
in future years among HPV-­vaccinated communities.8
The advantages of excising HSIL of the vagina is that it provides a
pathological description of the tissue removed, especially when there
3.2 | Secondary prevention (screening)
is a suspicion of microinvasion on colposcopic evaluation. Most HSIL
There is no evidence to indicate routine screening for vaginal cancer lesions are in the vault or upper third of the vagina and excision is ideal
after hysterectomy for benign disease. If a hysterectomy has been in this situation.17 Morbidity can be associated with the proximity of
performed for persistent HSIL after repeated excisional procedures of structures such as the bladder and rectum, as well as associated dys-
the cervix, vault smears are recommended for long-­term follow-­up.9,10 pareunia as a result of vaginal shortening and stenosis. Women should
In those cases where screening for vaginal cancer is indicated, HPV be extensively counselled preoperatively.
tests adequately correlate with cytological findings.11 Independently
of that, co-­testing seems to offer better accuracy in the diagnosis of
3.3.2 | CO2 laser vaporization
recurrent disease—as observed during follow-­up after treatment of
premalignant cervical lesions.12 Laser vaporization is an alternative option to excision, especially for multi­
focal lesions. Since there is no histologic specimen with this procedure, it
is imperative to ensure that there is no concern regarding cancer.18,19
3.3 | Tertiary prevention (management of
precancerous lesions)
3.3.3 | Topical fluorouracil
As the majority of vaginal cancers are of squamous histology, a com-
mon etiology is shared with cervical cancer. This is the persistence Topical application of 5FU has been used in an effort to avoid the
of high-­risk/oncogenic HPV infections. Co-­factors include immu- mutilating adverse effects of excision and laser ablation. Older studies
nosuppression and cigarette smoking. Smoking in combination with have shown comparable efficacy to laser and excision20,21. However,
high-­risk HPV increases the risk of progression to vaginal HSIL when these are retrospective in nature with small numbers. More recent
compared with nonsmokers.13 studies have shown inferior efficacy.15,22 A recent retrospective study
The terminology for precancerous HPV-­associated vaginal lesions reviewed recurrence rate in 576 women treated with topical 5FU,
has changed over time. In 2014, the WHO classification replaced the excision, or laser ablation. The statistically significant factors that con-
three-­tiered classification used previously (VAIN 1–3) with squamous tributed to recurrence and progression were the presence of high-­risk
intraepithelial lesion (SIL). This is divided into two categories: low HPV positivity and treatment modality. Laser and excision were found
grade (LSIL) and high grade (HSIL). LSIL (VAIN 1) may be associated to be superior to topical management with 5FU in reducing recur-
with either low-­risk or high-­risk HPV and it represents productive rences. It was also found that laser was more effective than excision in
or transient infections that may regress. In contrast, HSIL represents reducing recurrences of multifocal lesions, whereas surgical excision
transforming high-­risk infections (previously VAIN 2–3).14 was preferable with unifocal HSIL.23
Women with HSIL are usually asymptomatic and the majority of
women are aged over 60 years. HSIL can be seen in younger women,
3.3.4 | Topical 5% imiquimod
especially in immunocompromised individuals (HIV and transplant
patients). Colposcopy with acetic acid and/or Lugol iodine is indicated Imiquimod (applied topically either in creams or drug-­embedded
if a woman has an abnormal vaginal cytological smear and no gross tampons) is an immune response modulator that activates the innate
abnormality. Biopsies of colposcopically abnormal areas (acetowhite immune response and subsequently induces proinflammatory fac-
areas with punctation and/or punctation and mosaicism) is essential tors such as interferons. It provides an alternative noninvasive and
for the diagnosis. safe method of treating women with vaginal HSIL, especially young
Risk of progression of HSIL to invasive cancer has been found to women with multifocal lesions or older women who wish to avoid
15,16
range between 2% and 12%. surgical modalities. Studies on imiquimod are few, but a recent rand-
Treatment of precancerous lesions of the vagina must be individu- omized controlled trial (RCT) as well as a systematic review and meta-­
alized. Biopsy proven LSIL lesions can be followed up with observation analysis (including this RCT) found that this is a safe and effective
only (repeat smears and colposcopy), especially if women have non-­ treatment with regression rates similar to laser. HPV clearance rates
oncogenic strains of HPV. were greater than 50%, which was superior to laser.24,25
The various modalities of treatment for HSIL lesions include laser In general, independent of therapy chosen, most patients require
ablation, surgical excision, and topical treatments such as imiquimod and more than one therapy for adequate control. Since recurrence rate
topical chemotherapy with 5-­fluorouracil (5FU). Choice of treatment is high with any single therapy, close and extended follow-­up is
|
16       Adams and Cuello

recommended in all cases. As in treated premalignant cervical lesions, accounts for about 8%–10% of cases. Lymphomas, sarcomas, and
both HPV testing and cytology are recommended. melanomas of the vagina are extremely rare.
All malignancies must be confirmed histologically. Since 80% of
cases are metastatic or secondary tumors, it is important to ensure
4 |  MANAGEMENT OF INVASIVE that there is no other primary site by careful examination and appro-
CANCER OF THE VAGINA priate investigations. Although biopsy can be obtained under local
anesthesia, in many instances examination under sedation or general
Women usually present with bleeding or an odorous discharge. anesthesia should be performed to undertake an adequate exam and
Locally advanced disease may result in pain as well, and symptoms of obtain a biopsy.
disseminated disease depend on the site of metastases. By definition, tumors in the vagina that touch or extend to the
Diagnosis of vaginal cancer is made with directed biopsy of the external os of the cervix should be classified as cervical cancer.
lesion and a clinical assessment that ensures there is no evidence of
tumor on the cervix or the vulva. Biopsy can be performed in an out-
patient setting; however, sometimes examination under anesthesia is 7 | GRADING
required for diagnosis and adequate assessment, particularly in young
girls and elderly women where the exam can become painful or limited 1. GX: Grade cannot be assessed.
by narrowness of the introitus. 2. G1: Well differentiated.
As primary vaginal cancer is rare, treatment is complex, often indi- 3. G2: Moderately differentiated.
vidualized, and much of the management is extrapolated from cervical 4. G3: Poorly or undifferentiated.
cancer owing to its similar etiology and anatomical location. It is thus
recommended that women diagnosed with primary vaginal cancer are
referred to a tertiary unit whenever possible. Accordingly, outcomes 8 | STAGING
are influenced by the experience of the medical team in treating this
type of rare tumor.26 Vaginal carcinoma is primarily clinically staged. This is based on
the results of a physical exam, biopsy, and imaging tests performed
before treatment selection. The 2009 FIGO staging for primary
5 |  PATTERNS OF SPREAD
vaginal cancer is described and compared with other systems
in Table 1.
5.1 | Direct extension
A vaginal tumor may extend to the surrounding pelvic soft tissue
structures, including paravaginal tissue, parametria, urethra, bladder, 9 | ROLE OF IMAGING TECHNIQUES IN
and rectum. Most tumors occur in the upper third of the vagina, espe- DIAGNOSIS, STAGING, AND TREATMENT
cially the posterior wall.
Vaginal cancer is currently staged clinically, as is cervical cancer.
Modern imaging techniques such as computed tomography (CT),
5.2 | Lymphatic spread
magnetic resonance imaging (MRI), and positron emission tomog-
The lymphatic drainage of the vagina is complex. The upper vagina raphy (PET) are encouraged to guide management; however, these
drains by lymphatics to the pelvic lymph nodes. This includes the tests should not be used to change the initial clinical staging.28 In
obturator, internal iliac (hypogastric), and external iliac nodes. Spread this regard, the FIGO Gynecologic Oncology Committee recom-
to the para-­aortic lymph nodes is rare. The lower vagina drains to the mends that wherever available, imaging should be used to better
inguinal and femoral nodes (groin nodes). Cancer in the mid-­vagina define tumor volume and extension of disease. The Committee also
27
may follow both pelvic and groin node routes. encourages registering of imaging findings and their influence on
therapeutic decisions as a complement of assigned clinical FIGO
stage. This will allow further analyses that could change the current
5.3 | Hematogenous spread
staging system.
Hematogenous dissemination to lung, liver, and bone are usually late
manifestations.
9.1 | Magnetic resonance imaging
As extrapolated from cervical cancer, MRI is more sensitive in detect-
6 |  HISTOLOGIC SUBTYPES ing tumor size, as well as paravaginal or parametrial involvement.29,30
In primary vaginal tumors, clinical assessment may be difficult and
The predominant histologic subtype in primary vaginal cancer is squa- MRI may be a useful tool in individual cases owing to its superior soft
mous carcinoma, which comprises 90% of cases. Adenocarcinoma tissue resolution.26
Adams and Cuello |
      17

T A B L E   1   Comparison of staging systems for vaginal cancer.

AJCC stage Stage grouping (TNM) FIGO stage Stage description

IA T1a I The cancer is only in the vagina and is no larger than 2.0 cm (4/5 inch) (T1a)
N0 It has not spread to nearby lymph nodes (N0) or to distant sites (M0)
M0
IB T1b I The cancer is only in the vagina and is larger than 2.0 cm (4/5 inch) (T1b)
N0 It has not spread to nearby lymph nodes (N0) or to distant sites (M0)
M0
IIA T2a II The cancer has grown through the vaginal wall, but not as far as the pelvic wall and is
no larger than 2.0 cm (4/5 inch) (T2a)
N0 It has not spread to nearby lymph nodes (N0) or to distant sites (M0)
M0
IIB T2b II The cancer has grown through the vaginal wall, but not as far as the pelvic wall and is
larger than 2.0 cm (4/5 inch) (T2b)
N0 It has not spread to nearby lymph nodes (N0) or to distant sites (M0)
M0
III T1 to T3 III The cancer can be any size and might be growing into the pelvic wall, and/or growing
into the lower one-­third of the vagina and/or has blocked the flow of urine
(hydronephrosis), which is causing kidney problems (T1 to T3)
N1 It has also spread to nearby lymph nodes in the pelvis or groin (inguinal) area (N1) but
M0 not distant sites (M0)

OR
T3 III The cancer is growing into the pelvic wall, and/or growing into the lower one-­third of
the vagina and/or has blocked the flow of urine (hydronephrosis), which is causing
kidney problems (T3)
N0 It has not spread to nearby lymph nodes (N0) or to distant sites (M0)
M0
IVA T4 IVA The cancer is growing into the bladder or rectum or is growing out of the pelvis (T4)
Any N It might or might not have spread to lymph nodes in the pelvis or groin (inguinal area)
M0 (Any N). It has not spread to distant sites (M0)

IVB Any T IVB The cancer has spread to distant organs such as the lungs or bones (M1). It can be
any size and might or might not have grown into nearby structures or organs (Any T)
Any N It might or might not have spread to nearby lymph nodes (Any N)
M1

9.2 | PET-­CT 10.1 | Surgery
As in cervical cancer, the use of PET-­CT in primary vaginal cancer has The role of surgery is limited in primary vaginal cancer since the pri-
been found superior compared with other imaging modalities for detect- mary tumor is in close proximity to the bladder, urethra, and rectum.
ing nodal disease.31 It is also useful in detecting recurrent disease. In general, primary treatment with surgery is limited to early and small
lesions confined to the vaginal mucosa (less than 2 cm). There are
no RCTs and most of the literature is retrospective in nature. There
10 | TREATMENT may be a role in certain additional situations beyond early disease, as
listed below.
The treatment of carcinoma of the vagina depends primarily on his-
tology, tumor volume, anatomical localization of the lesion, stage of 1. Surgical management of Stage I disease (early disease)
the disease, and age of the patient. Adversely, owing to anatomical a. Upper vaginal disease
localization, both reproductive potential (young women) and sexual i. The tumor is limited to the mucosa in Stage 1 disease.
function (any age) can be affected. Different modalities of treatment ii. If the uterus is in situ, radical hysterectomy, vaginectomy
can be offered to patients affected by this disease including surgery, aiming for 1 cm disease-free margins, and pelvic lymphade-
radiotherapy, chemotherapy or its combination. nopathy should be offered. If the uterus has been removed,
|
18       Adams and Cuello

radical vaginectomy as above with pelvic lymphadenopathy comparison to radiation on its own.39,40 Chemoradiation may be con-
can be performed.32,33 sidered in the treatment of vaginal cancer following a more recent
b. Lower vaginal disease retrospective review suggesting a potential improvement in overall
i. Radical wide local excision with 1 cm margins can be offered, and disease-­free survival.41 Although this was a small review (71
in addition to bilateral groin node dissection patients), it showed a significant difference between both overall sur-
2. Ovarian transposition/surgery pre-radiation vival and disease-­free survival when comparing women who received
In young women with vaginal cancer requiring radiation as primary radiation alone versus chemoradiation as primary treatment (three-­
treatment, ovarian transposition can be offered prior to definitive year overall survival of 56% versus 79% and three-­year disease-­free
radiation treatment in an effort to prevent the adverse effects of survival of 43% versus 73%).41
radiation-induced menopause. In selected cases, laparoscopic or
extraperitoneal removal of bulky lymph nodes can be offered as part
11 | SPECIAL SITUATIONS
of staging and treatment planning.
3. Central recurrence after radiation treatment
11.1 | Adenocarcinoma of the vagina
Pelvic exenteration is a possibility if the recurrence is central and
isolated. These patients require extensive counselling regarding Primary adenocarcinoma of the vagina accounts for 8%–10% of cases.
the risks and morbidity of surgery, as well as the impact on quality In 1971, Herbst et al.42 described the association between diethyl-
of life and body image. stilbestrol (DES) and clear cell adenocarcinoma (cervix/vagina) in
4. Palliative management of recurrent or advanced disease daughters who had intrauterine exposure during the first 16 weeks
In women with advanced (Stage IV disease) or recurrent disease of pregnancy. Most of these cases were diagnosed between the ages
who present with vesicovaginal or rectovaginal fistulae, a palliative of 14 and 22 years. A recent retrospective review of 420 cases found
urinary diversion or colostomy can be offered to improve quality of that the majority of cases were in younger women, but there was a
life before definitive management with radiation treatment. small second peak at 42 years.43 Intrauterine DES has been banned
since the 1970s and it is expected, therefore, that clear cell adeno-
carcinomas attributable to intrauterine DES exposure will disappear in
10.2 | Radiation
next decades. For women in doubt or certain of intrauterine ­exposure
In the majority of cases and especially in advanced stages, radiation to DES, closer follow-­up is recommended including annual ­cytology
constitutes the cornerstone of treatment for this disease. This is a (the so-­called four-­quadrant pap test, with extensive sampling of the
combination of external beam radiation (EBRT) and intracavitary radi- cervix and vagina) and careful visualization of both the cervix and
otherapy or brachytherapy (ICRT). The principal advantage of radia- vagina (including colposcopy and Lugol staining whenever there is
tion is organ preservation. a clinical suspicion).44,45 In addition, the so-­called “DES daughters”
As in other pelvic malignancies, MRI imaging has become an should be screened closely with mammography for the slight increase
essential component for properly defining tumor volume and the spa- in risk of developing breast cancer.46
tial relationship of the tumor with neighboring organs during treat- Non-­DES adenocarcinoma is exceptionally rare and acts as a dif-
ment planning. ferent entity with a different natural history. This may include endo-
EBRT to the pelvis includes the external iliac and obturator nodes metrioid (arising from endometriosis) or mucinous subtypes, which are
as per standard of care. In addition, the inguinal nodes may be included typically diagnosed in postmenopausal women.
if the tumor is in the distal vagina.
The optimal or lower threshold dose is 70 Gy, which has been
11.1.1 | Treatment
shown to improve outcomes.34–36 Doses higher than 70 Gy result in
significant grade 3 and 4 toxicities.37 Most adenocarcinomas are treated in the same manner as
Intensity modulated radiation therapy (IMRT) is an advanced form squamous carcinomas.
of radiation that allows for higher dosages of radiation to be delivered
to the cancer. Although studies in vaginal cancer are limited, this form
11.2 | Vaginal melanoma
of radiation may allow improved dosages to the cancer, with fewer
adverse effects because dose to the adjacent structures is limited.38 Vaginal melanoma is exceptionally rare and is typically diagnosed in
elderly women. The incidence is approximately three women per 10
million women per year.47
10.3 | Concurrent chemotherapy with radiation
Modern management of vaginal cancer often combines concurrent
11.2.1 | Treatment
chemotherapy such as cisplatin or 5FU. This has been extrapo-
lated from the successful outcomes of this treatment with cervical There is no standard of treatment in the literature owing to its rar-
cancer.38 However, most studies using chemoradiation in vaginal ity, but surgical excision either by wide local excision or colpectomy
cancer are limited owing to the small numbers of cases and lack of with or without pelvic exenteration has been described.48,49 Adjuvant
Adams and Cuello |
      19

treatment in the form of radiation or immunotherapy (interferon-­alpha) histology. Additional factors influencing prognosis in squamous sub-
can be considered, although there is limited evidence to support this. type are the tumor volume (>4 cm), its location outside of the upper
Palliative chemotherapy or radiation can be used in advanced disease.50 third of the vagina, HPV status, and MIB-­1 index.54 As with other
malignancies, age, reproductive and sexual functions, and perfor-
mance status can influence the selection of specific therapies and
11.3 | Sarcoma botryoides
potentially affect survival outcomes. However, the non-­negotiable
Rhabdomyosarcomas are the most common soft tissue cancers in premise is to offer the best option in terms of survival. When com-
children and adolescents, accounting for 4%–6% of all malignancies paring histologies, the better outcome is obtained in squamous car-
in this age group. Twenty percent of these occur in the lower genital cinoma of the vagina when diagnosed at earlier stages (I or II). In the
51,52
tract, and more than 50% are of the embryonal histologic subtype. last three decades, incorporation of imaging techniques during diag-
The 2013 WHO classification of tumors of soft tissue and bone nosis and treatment planning has influenced decision making and the
stratified rhabdomyosarcomas into four main histologic subtypes53: selection of treatment for cases with similar clinical stage but with
different tumor volume. As in cervical cancer, the addition of concur-
1. Typical embryonal variant (most common accounting for 58% rent chemotherapy to radiotherapy has also improved survival in the
of cases): this comprises the classic botryoides squamous histology.
2. Spindle cell/sclerosing variant Since vaginal carcinoma is a rare entity, most of the available
3. Alveolar variant data in terms of therapeutic outcomes come from single institutions
4. Pleomorphic variant and include cohorts of patients from different epochs where differ-
ent treatment modalities were used. For example, one of the largest
Most rhabdomyosarcomas in children are in the vagina, whereas studies published by the MD Anderson Cancer Center includes 193
adolescents have predominantly cervical lesions. Sarcoma botryoides patients treated over two decades. This study reported five-­year
often presents in the first few years of life with bleeding and nodular disease-­specific survival rates of 85% for 50 patients with Stage I
lesions filling and possibly protruding from the vagina (grape-­like). More disease, 78% for 97 patients with Stage II, and 58% for 46 patients
advanced stages of disease may present with abdominal pain, an abdom- with Stages III–IVA.55 A more recent retrospective analysis from
inal mass, or symptoms of distant metastases. the Department of Radiation Oncology at Stanford University, also
including a long experience over five decades, reflects the positive
impact of incorporating new imaging technologies (e.g. MRI and
11.3.1 | Treatment
PET-­CT) in defining primary lesion characteristics, unveiling hid-
These tumors are rare and, as a result, treatment is based on the case den metastatic disease, and influencing treatment planning. Adding
reports and series that have been collected over time. There is no level imaging techniques no doubt influences decision making, favoring
1 evidence to support best treatment options. It is recommended that the use of chemoradiation and IMRT, and results in the improvement
a multidisciplinary team be involved in decisions regarding treatment, of locoregional control rate, and distant metastasis-­free and overall
especially when this involves children and adolescents. Ideally these survival. This is particularly useful in the management of large vol-
patients should be referred to centers of excellence where there has ume tumors (particularly ≥4 cm) minimizing grade 3–4 toxicities.37
been appropriate experience in dealing with these cases. Confirming such an improvement, a recent review from Gadducci
Treatment previously involved extensive surgery, but a series et al. 54 summarized the data available and obtained a five-­year over-
of case reports has suggested that less radical surgery can be done. all survival that varies between 35% and 78%, with a severe late
These reports have shown good outcomes in terms of survival as well complication rate between 9.4% and 23.1% among cases of squa-
as quality of life if the patients are selected carefully. Unfavorable mous carcinoma of the vagina when treated with different standards
factors such as the cervix as the primary site, large lesion size, and of radiotherapy.
extent of the disease may require radical surgical intervention or che- In terms of prognoses for other histologies, young women with
motherapy. In the absence of these factors, wide local excision and early stage DES-­related adenocarcinomas treated with either radi-
chemotherapy have been used with good success rates. Neoadjuvant ation, surgery, or a multimodal approach have good outcomes with
chemotherapy is another primary choice that can be used, followed by reported five-­year survival of 80%–87%.56,57 This is in contrast to non-­
surgical resection. Radiotherapy should be avoided if possible, as there DES-­related adenocarcinomas, which have a worse prognosis owing
are long-­term adverse effects. to an increased risk of both local and distant metastases. A review of
26 women with non-­DES-­related vaginal cancer at the MD Anderson
Cancer Center found an overall five year survival of 34%.58
12 | PROGNOSES AND OUTCOMES OF Vaginal melanoma has a very poor prognosis, with a five-­year
VAGINAL CANCER overall survival of 15%.49 In a recent systematic review of 805 cases
reported over the last 20 years (mostly case reports), the mean
The main determinant of prognoses in carcinoma of the vagina is the recurrence-­free survival was short, at 16 months, with a mean overall
stage of disease at the time of diagnosis, independently of subjacent survival of only 22 months.50
|
20       Adams and Cuello

Finally, in relation to sarcoma botryoides, most of the available 7. Garland SM, Paavonen J, Jaisamrarn U, et al. Prior human papilloma-
information in the literature comes from the reports of small series virus-­16/18 AS04-­adjuvanted vaccination prevents recurrent high
grade cervical intraepithelial neoplasia after definitive surgical ther-
from single institutions (many of them combining different variants
apy: Post-­hoc analysis from a randomized controlled trial. Int J Cancer.
including the embryonal or botryiodes subtype and from different ori- 2016;139:2812–2826.
gins in the genital tract). A recent report from Peking Union Medical 8. Hansen BT, Campbell S, Nygard M. Long-­term incidence trends of
College Hospital, including eight rhabdomyosarcoma cases of the HPV-­related cancers, and cases preventable by HPV vaccination: A
registry-­based study in Norway. BMJ Open. 2018;8:e019005.
female genital tract over a 20-­year period, showed that after com-
9. Farghaly H, Bourgeois D, Houser PM, Padmanabhan V, Lage JM, Hoda
bined therapy with local excision and chemotherapy, most patients RS. Routine vaginal pap test is not useful in women status-­post hys-
can achieve a good prognosis. The prognosis is highly correlated with terectomy for benign disease. Diagn Cytopathol. 2006;34:640–643.
tumor site and histologic type.59 Similarly, in a recent survey of 144 10. Videlefsky A, Grossl N, Denniston M, Sehgal R, Lane JM, Goodenough
G. Routine vaginal cuff smear testing in post-­hysterectomy patients
eligible cases of rhabdomyosarcomas of the lower female genital tract
with benign uterine conditions: When is it indicated? J Am Board Fam
in the SEER database (between 1973 and 2013)—75.7% correspond- Pract. 2000;13:233–238.
ing to the embryonal subtype—the estimated five-­year overall survival 11. Bansal M, Austin RM, Zhao C. Correlation of histopathologic fol-
rate was 68.4%. Among the factors associated with better overall low-­up findings with vaginal human papillomavirus and low-­grade
squamous intraepithelial lesion papanicolaou test results. Arch Pathol
survival were younger age, absence of distant metastasis, embryonal
Lab Med. 2011;135:1545–1549.
histology, negative lymph nodes, and the performance of surgery. An
12. Hui Y, Hansen K, Murthy J, Chau D, Sung CJ, Quddus MR. Relevance
important conclusion of this survey was that for prepubescent girls of the pap test: A report of HPV-­DNA test-­negative high-­grade squa-
and adolescents, the use of radical surgery did not confer a survival mous intraepithelial lesions of the female lower genital tract. Acta
benefit compared with local tumor excision.60 Cytol. 2016;60:445–450.
13. Sherman JF, Mount SL, Evans MF, Skelly J, Simmons-Arnold L,
Eltabbakh GH. Smoking increases the risk of high-­grade vaginal
intraepithelial neoplasia in women with oncogenic human papilloma-
AUTHOR CONTRI B UTI O N S
virus. Gynecol Oncol. 2008;110:396–401.
14. Reich O, Regauer S, Marth C, et  al. Precancerous lesions of the
TA and MC shared the concept design, literature review, and writing
cervix, vulva and vagina according to the 2014 WHO classifica-
of the manuscript. tion of tumors of the female genital tract. Geburtshilfe Frauenheilkd.
2015;75:1018–1020.
15. Dodge JA, Eltabbakh GH, Mount SL, Walker RP, Morgan A. Clinical
ACKNOWLE DG ME NTS features and risk of recurrence among patients with vaginal intraepi-
thelial neoplasia. Gynecol Oncol. 2001;83:363–369.
This chapter reworks and updates the information published in the
16. Hodeib M, Cohen JG, Mehta S, et  al. Recurrence and risk of pro-
FIGO Cancer Report 2015 (Hacker NF, Eifel PJ, van der Velden J. gression to lower genital tract malignancy in women with high grade
Cancer of the vagina. Int J Gynecol Obstet. 2015;131(Suppl.2):S84–87). VAIN. Gynecol Oncol. 2016;141:507–510.
17. Boonlikit S, Noinual N. Vaginal intraepithelial neoplasia: A retrospec-
tive analysis of clinical features and colpohistology. J Obstet Gynaecol
CO NFLI CTS OF I NTE RE S T Res. 2010;36:94–100.
18. Campagnutta E, Parin A, De Piero G, Giorda G, Gallo A, Scarabelli C.
The authors have no conflicts of interest to declare. Treatment of vaginal intraepithelial neoplasia (VAIN) with the carbon
dioxide laser. Clin Exp Obstet Gynecol. 1999;26:127–130.
19. Perrotta M, Marchitelli CE, Velazco AF, Tauscher P, Lopez G, Peremateu
MS. Use of CO2 laser vaporization for the treatment of high-­grade
REFERENCES
vaginal intraepithelial neoplasia. J Low Genit Tract Dis. 2013;17:
1. Adhikari P, Vietje P, Mount S. Premalignant and malignant lesions of 23–27.
the vagina. Diagn Histopathology. 2016;23:28–34. 20. Ballon SC, Roberts JA, Lagasse LD. Topical 5-­fluorouracil in the
2. Edge SB, Compton CC The American Joint Committee on Cancer: The treatment of intraepithelial neoplasia of the vagina. Obstet Gynecol.
7th edition of the AJCC cancer staging manual and the future of TNM. 1979;54:163–166.
Ann Surg Oncol. 2010;17:1471–1474. 21. Krebs HB. Treatment of vaginal intraepithelial neoplasia with laser and
3. Hellman K, Silfversward C, Nilsson B, Hellstrom AC, Frankendal B, topical 5-­fluorouracil. Obstet Gynecol. 1989;73:657–660.
Pettersson F. Primary carcinoma of the vagina: Factors influencing the 22. Rome RM, England PG. Management of vaginal intraepithelial neo-
age at diagnosis. The radiumhemmet series 1956–96. Int J Gynecol plasia: A series of 132 cases with long-­term follow-­up. Int J Gynecol
Cancer. 2004;14:491–501. Cancer. 2000;10:382–390.
4. Hampl M, Sarajuuri H, Wentzensen N, Bender HG, Kueppers V. 23. Kim MK, Lee IH, Lee KH. Clinical outcomes and risk of recurrence
Effect of human papillomavirus vaccines on vulvar, vaginal, and among patients with vaginal intraepithelial neoplasia: A comprehen-
anal intraepithelial lesions and vulvar cancer. Obstet Gynecol. sive analysis of 576 cases. J Gynecol Oncol. 2018;29:e6.
2006;108:1361–1368. 24. Tainio K, Jakobsson M, Louvanto K, et al. Randomised trial on treat-
5. Lamos C, Mihaljevic C, Aulmann S, et al. Detection of human papil- ment of vaginal intraepithelial neoplasia-­Imiquimod, laser vaporisa-
lomavirus infection in patients with vaginal intraepithelial neoplasia. tion and expectant management. Int J Cancer. 2016;139:2353–2358.
PLoS ONE. 2016;11:e0167386. 25. Tranoulis A, Laios A, Mitsopoulos V, Lutchman-Singh K, Thomakos N.
6. Serrano B, de Sanjose S, Tous S, et al. Human papillomavirus genotype Efficacy of 5% imiquimod for the treatment of vaginal intraepithelial
attribution for HPVs 6, 11, 16, 18, 31, 33, 45, 52 and 58 in female neoplasia-­a systematic review of the literature and a meta-­analysis.
anogenital lesions. Eur J Cancer. 2015;51:1732–1741. Eur J Obstet Gynecol Reprod Biol. 2017;218:129–136.
Adams and Cuello |
      21

26. Gardner CS, Sunil J, Klopp AH, et al. Primary vaginal cancer: Role of MRI 44. Hanselaar AG, Boss EA, Massuger LF, Bernheim JL. Cytologic exam-
in diagnosis, staging and treatment. Br J Radiol. 2015;88:20150033. ination to detect clear cell adenocarcinoma of the vagina or cervix.
27. Cardenes HR, Schilder JM, Roth LM. Vagina. In: Barakat RR, Markman Gynecol Oncol. 1999;75:338–344.
M, Randall ME, eds. Principles and Practice of Gynecologic Oncology. 45. Saslow D, Solomon D, Lawson HW, et  al. American cancer soci-
Philadelphia: Lippincott Williams & Wilkins; 2009:591–622. ety, American society for colposcopy and cervical pathology, and
28. Rajaram S, Maheshwari A, Srivastava A. Staging for vaginal cancer. American society for clinical pathology screening guidelines for the
Best Pract Res Clin Obstet Gynaecol. 2015;29:822–832. prevention and early detection of cervical cancer. Am J Clin Pathol.
29. Bipat S, Glas AS, van der Velden J, Zwinderman AH, Bossuyt PM, 2012;137:516–542.
Stoker J. Computed tomography and magnetic resonance imaging in 46. Palmer JR, Hatch EE, Rosenberg CL, et  al. Risk of breast cancer in
staging of uterine cervical carcinoma: A systematic review. Gynecol women exposed to diethylstilbestrol in utero: Preliminary results
Oncol. 2003;91:59–66. (United States). Cancer Causes Control. 2002;13:753–758.
30. Hricak H, Gatsonis C, Chi DS, et al. Role of imaging in pretreatment eval- 47. Leitao MM Jr, Cheng X, Hamilton AL, et  al. Gynecologic Cancer
uation of early invasive cervical cancer: Results of the intergroup study InterGroup (GCIG) consensus review for vulvovaginal melanomas. Int
American College of Radiology Imaging Network 6651-­Gynecologic J Gynecol Cancer. 2014;24:S117–S122.
Oncology Group 183. J Clin Oncol. 2005;23:9329–9337. 48. Creasman WT, Phillips JL, Menck HR. The national cancer data base
31. Lamoreaux WT, Grigsby PW, Dehdashti F, et al. FDG-­PET evaluation report on cancer of the vagina. Cancer. 1998;83:1033–1040.
of vaginal carcinoma. Int J Radiat Oncol Biol Phys. 2005;62:733–737. 49. Kirschner AN, Kidd EA, Dewees T, Perkins SM. Treatment
32. Di Donato V, Bellati F, Fischetti M, Plotti F, Perniola G, Panici PB. approach and outcomes of vaginal melanoma. Int J Gynecol Cancer.
Vaginal cancer. Crit Rev Oncol Hematol. 2012;81:286–295. 2013;23:1484–1489.
33. Stock RG, Chen AS, Seski J. A 30-­year experience in the management 50. Rapi V, Dogan A, Schultheis B, Hartmann F, Rezniczek GA, Tempfer
of primary carcinoma of the vagina: Analysis of prognostic factors and CB. Melanoma of the vagina: Case report and systematic review of
treatment modalities. Gynecol Oncol. 1995;56:45–52. the literature. Anticancer Res. 2017;37:6911–6920.
34. Kirkbride P, Fyles A, Rawlings GA, et  al. Carcinoma of the vagina– 51. Villella JA, Bogner PN, Jani-Sait SN, Block AM, Lele S.
experience at the Princess Margaret Hospital (1974–1989). Gynecol Rhabdomyosarcoma of the cervix in sisters with review of the litera-
Oncol. 1995;56:435–443. ture. Gynecol Oncol. 2005;99:742–748.
35. Lian J, Dundas G, Carlone M, Ghosh S, Pearcey R. Twenty-­year 52. Zeisler H, Mayerhofer K, Joura EA, et  al. Embryonal rhabdomyosar-
review of radiotherapy for vaginal cancer: An institutional experience. coma of the uterine cervix: Case report and review of the literature.
Gynecol Oncol. 2008;111:298–306. Gynecol Oncol. 1998;69:78–83.
36. Perez CA, Grigsby PW, Garipagaoglu M, Mutch DG, Lockett MA. 53. Rudzinski ER, Anderson JR, Hawkins DS, Skapek SX, Parham DM, Teot
Factors affecting long-­term outcome of irradiation in carcinoma of LA. The World Health Organization classification of skeletal muscle
the vagina. Int J Radiat Oncol Biol Phys. 1999;44:37–45. tumors in pediatric rhabdomyosarcoma: A report from the children’s
37. Hiniker SM, Roux A, Murphy JD, et al. Primary squamous cell carci- oncology Group. Arch Pathol Lab Med. 2015;139:1281–1287.
noma of the vagina: Prognostic factors, treatment patterns, and out- 54. Gadducci A, Fabrini MG, Lanfredini N, Sergiampietri C. Squamous cell
comes. Gynecol Oncol. 2013;131:380–385. carcinoma of the vagina: Natural history, treatment modalities and
38. Green JA, Kirwan JM, Tierney JF, et al. Survival and recurrence after con- prognostic factors. Crit Rev Oncol Hematol. 2015;93:211–224.
comitant chemotherapy and radiotherapy for cancer of the uterine cer- 55. Frank SJ, Jhingran A, Levenback C, Eifel PJ. Definitive radiation ther-
vix: A systematic review and meta-­analysis. Lancet. 2001;358:781–786. apy for squamous cell carcinoma of the vagina. Int J Radiat Oncol Biol
39. Nashiro T, Yagi C, Hirakawa M, et al. Concurrent chemoradiation for Phys. 2005;62:138–147.
locally advanced squamous cell carcinoma of the vagina: Case series 56. Senekjian EK, Frey KW, Stone C, Herbst AL. An evaluation of stage
and literature review. Int J Clin Oncol. 2008;13:335–339. II vaginal clear cell adenocarcinoma according to substages. Gynecol
40. Samant R, Lau B, E C, Le T, Tam T. Primary vaginal cancer treated with Oncol. 1988;31:56–64.
concurrent chemoradiation using cis-­platinum. Int J Radiat Oncol Biol 57. Senekjian EK, Frey KW, Anderson D, Herbst AL. Local therapy in stage I
Phys. 2007;69:746–750. clear cell adenocarcinoma of the vagina. Cancer. 1987;60:1319–1324.
41. Miyamoto DT, Viswanathan AN. Concurrent chemoradiation for vagi- 58. Frank SJ, Deavers MT, Jhingran A, Bodurka DC, Eifel PJ. Primary ade-
nal cancer. PLoS ONE. 2013;8:e65048. nocarcinoma of the vagina not associated with diethylstilbestrol (DES)
42. Herbst AL, Ulfelder H, Poskanzer DC. Adenocarcinoma of the vagina. exposure. Gynecol Oncol. 2007;105:470–474.
Association if maternal stilbestrol therapy with tumor appearance in 59. Yang J, Yang J, Yu M, Yuan Z, Cao D, Keng S. Clinical study on female
young women. N Engl J Med. 1971;284:878–881. genital tract rhabdomyosarcoma in childhood: Changes during 20
43. Huo D, Anderson D, Palmer JR, Herbst AL. Incidence rates and years in one center. Int J Gynecol Cancer. 2017;27:311–314.
risks of diethylstilbestrol-­related clear-­cell adenocarcinoma of the 60. Nasioudis D, Alevizakos M, Chapman-Davis E, Witkin SS, Holcomb K.
vagina and cervix: Update after 40-­year follow-­up. Gynecol Oncol. Rhabdomyosarcoma of the lower female genital tract: An analysis of
2017;146:566–571. 144 cases. Arch Gynecol Obstet. 2017;296:327–334.
DOI: 10.1002/ijgo.12611

FIGO CANCER REPORT 2018

Cancer of the cervix uteri

Neerja Bhatla1,* | Daisuke Aoki2 | Daya Nand Sharma3 | Rengaswamy Sankaranarayanan4

1
Department of Obstetrics and
Gynecology, All India Institute of Medical Abstract
Sciences, New Delhi, India Since the publication of the last FIGO Cancer Report there have been giant strides in
2
Department of Obstetrics and
the global effort to reduce the burden of cervical cancer, with WHO announcing a call
Gynecology, Keio University School of
Medicine, Tokyo, Japan for elimination. In over 80 countries, including LMICs, HPV vaccination is now included
3
Department of Radiation Oncology, All India in the national program. Screening has also seen major advances with implementation
Institute of Medical Sciences, New Delhi, India
of HPV testing on a larger scale. However, these interventions will take a few years to
4
Early Detection and Prevention
Group, International Agency for Research on show their impact. Meanwhile, over half a million new cases are added each year.
Cancer, Lyon, France Recent developments in imaging and increased use of minimally invasive surgery have

*Correspondence changed the paradigm for management of these cases. The FIGO Gynecologic
Neerja Bhatla, Department of Obstetrics and Oncology Committee has revised the staging system based on these advances. This
Gynecology, All India Institute of Medical
Sciences, New Delhi, India. chapter discusses the management of cervical cancer based on the stage of disease,
Email: nbhatla@aiims.ac.in including attention to palliation and quality of life issues.

KEYWORDS
Cervix; FIGO Cancer Report; Gynecologic cancer; HPV vaccination; Radiation; Screening;
Staging; Surgery

1 | INTRODUCTION the junction with the uterus to the external os which opens into the
vagina and is lined by columnar epithelium. Almost all cases of cervi-
Globally, cervical cancer continues to be one of the most common cal carcinoma originate in the transformation zone from the ecto-­ or
cancers among females, being the fourth most common after breast, endocervical mucosa. The transformation zone is the area of the cer-
colorectal, and lung cancer. In 2012, it was estimated that there were vix between the old and new squamocolumnar junction.
approximately 527 600 new cases of cervical cancer with 265 700 The fact that the cervix can be easily visualized and sampled, and
1
deaths annually. In low-­ and middle-­income countries (LMICs), it is can be treated by freezing and burning with little or no anesthesia, has
more common, being the second most common cancer in incidence contributed to the understanding of the natural history of this can-
among women and the third most common in terms of mortality. cer along with the development of simple outpatient techniques of
The majority of new cases and deaths (approximately 85% and 90%, screening and prevention.
respectively) occur in low-­resource regions or among people from
socioeconomically weaker sections of society.
3 | EARLY DETECTION AND
PREVENTION OF CERVICAL CANCER
2 | ANATOMICAL CONSIDERATIONS
It is now recognized that cervical cancer is a rare long-­term out-
The cervix, which is the lowermost part of the uterus, is a cylindrical-­ come of persistent infection of the lower genital tract by one of
shaped structure composed of stroma and epithelium. The intravaginal about 15 high-­risk HPV types, which is termed the “necessary”
part, the ectocervix, projects into the vagina and is lined by squamous cause of cervical cancer. Of the estimated 530 000 new cervi-
epithelium. The endocervical canal extends from the internal os at cal cancer cases annually, HPV 16 and HPV 18 account for 71%

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

22  |  wileyonlinelibrary.com/journal/ijgo Int J Gynecol Obstet 2018; 143 (Suppl. 2): 22–36
Bhatla ET AL. |
      23

of cases; while HPV types 31, 33, 45, 52, and 58 account for for immunocompromised patients irrespective of age, the recom-
2,3
another 19% of cervical cancer cases It is well documented that mendation is for three doses (0.5 mL at 0, 1, 6 months).5 WHO has
nearly 90% of incident HPV infections are not detectable within reviewed the latest data and concluded that there is no safety con-
a period of 2 years from the acquisition of infection and persist cern regarding HPV vaccines.5
only in a small proportion. It is debatable whether the virus is There is evidence for the effectiveness of vaccination at the
completely cleared or whether it remains latent in basal cells with population level in terms of reduced prevalence of high-­risk HPV
the potential for reactivation in some cases. Persistent HPV infec- types, and reduction in anogenital warts and high-­grade cervical
tion denotes the presence of the same type-­specific HPV DNA on abnormalities caused by the vaccine types among young women;
repeated sampling after 6–12 months. Only one-­tenth of all infec- there is some evidence of cross-­protection from nonvaccine types
tions become persistent, and these women could develop cervical also. There is no evidence of type replacement6–8 Recent observa-
precancerous lesions. tional studies have reported evidence for effectiveness in prevent-
This knowledge has resulted in the development of new initia- ing high-­risk HPV infections following a single dose and further
tives for prevention and early detection. The two major approaches long-­term follow-­up will clarify the role of one dose in preventing
for control of cervical cancer involve: (1) prevention of invasive can- cervical neoplasia.9,10
cer by HPV vaccination; and (2) screening for precancerous lesions.
Prevention and elimination are potential possibilities but the tragedy is
3.2 | Secondary prevention of cervical cancer by
that it is not yet prevented on a large scale in many LMICs due to lack
early detection and treatment of precancerous lesions
of efficient and effective intervention programs. WHO has recently
given a call to action for elimination of cervical cancer. This is foresee- Even with the advent of effective vaccines, screening will remain a
able if implemented in earnest in successful public health programs priority for cervical cancer prevention for several decades. Cervical
achieving high coverage. cancer screening has been successful in preventing cancer by
detection and treatment of precursor lesions, namely, high-­grade
cervical intraepithelial neoplasia (CIN 2 and 3) and adenocarcinoma
3.1 | Primary prevention of cervical cancer with HPV
in-­situ (AIS).
vaccination
Several cervical screening strategies have been found to be
The fact that more than 80% of women followed over time will effective in varied settings. The tests used widely include conven-
acquire at least one high-­risk HPV infection suggests the ubiquitous tional cytology (Pap smear), in recent years liquid-­based cytology
nature of the HPV infection and reflects the ease of transmission. The and HPV testing, and, in LMICs, visual inspection with acetic acid
estimated cross-­sectional HPV prevalence worldwide among healthy (VIA).11 While the Pap smear is still the major workhorse of screen-
women is around 11.7%, with the highest in Sub-­Saharan Africa at ing and is associated with substantial declines in cervical cancer risk
around 24%, and country-­specific prevalence ranging between 2% in high-­income countries, it is a challenging and resource intensive
and 42% globally4 Age-­specific cross-­sectional HPV prevalence peaks technology that is not feasible in low-­resource settings11 where poor
at 25% in women aged less than 25 years, which suggests that the organization, coverage, and lack of quality assurance result in sub-
infection is predominantly transmitted through the sexual route fol- optimal outcomes. In the context of declining HPV infections after
lowing sexual debut. Thus, prophylactic HPV vaccination as a preven- the introduction of HPV vaccines a decade ago, many healthcare sys-
tive strategy should target women before initiation of sexual activity, tems are considering switching to primary HPV screening, which has
focusing on girls aged 10–14 years. higher sensitivity and negative predictive value, and allows extended
Three prophylactic HPV vaccines are currently available in many screening intervals or even a single lifetime screening in low-­
countries for use in females and males from the age of 9 years for resource settings.12,13 VIA involves detection of acetowhite lesions
the prevention of premalignant lesions and cancers affecting the on the cervix 1 minute after application of 3%–5% freshly prepared
cervix, vulva, vagina, and anus caused by high-­risk HPV types: a acetic acid. In view of its feasibility, VIA screening has been widely
bivalent vaccine targeting HPV16 and HPV18; a quadrivalent vac- implemented in opportunistic settings in many low-­income countries
cine targeting HPV6 and HPV11 in addition to HPV16 and HPV18; in Sub-­Saharan Africa. A single-­visit approach (SVA) for screening
and a nonavalent vaccine targeting HPV types 31, 33, 45, 52, and with rapid diagnosis and treatment improves coverage, eliminates
58 in addition to HPV 6, 11, 16, and 18. The last two vaccines tar- follow-­up visits, and makes screening more time and cost-­efficient in
get anogenital warts caused by HPV 6 and 11 in addition to the low-­resource settings.14–16 VIA screening is particularly suitable for
above-­mentioned malignant and premalignant lesions. All the vac- SVA and WHO has issued guidelines for implementing SVA in public
cines are recombinant vaccines composed of virus-­like particles health settings.
(VLPs) and are not infectious since they do not contain viral DNA. A single screening modality will never be universally applicable,
For girls and boys aged 9–14 years, a two-­dose schedule (0.5 mL at but it is possible to adapt cost-­effective means of cervical cancer
0 and 5–13 months) is recommended. If the second vaccine dose screening to each country. The screening strategy chosen must be
is administered earlier than 5 months after the first dose, a third feasible, simple, safe, accurate, acceptable, and easily accessible to
dose is recommended. For those aged 15 years and above, and highest-­risk women. A judicious combination of HPV vaccination and
24       | Bhatla ET AL.

screening has enormous potential to eliminate cervical cancer in the is shown in Table 1 (presented at the FIGO XXII World Congress of
foreseeable future. Gynecology and Obstetrics17).

4.1 | Diagnosis and evaluation of cervical cancer


4 |  FIGO STAGING
4.1.1 | Microinvasive disease
Cervical cancer spreads by direct extension into the parametrium,
vagina, uterus and adjacent organs, i.e., bladder and rectum. It also Diagnosis of Stage IA1 and IA2 is made on microscopic examination
spreads along the lymphatic channels to the regional lymph nodes, of a LEEP (loop electrosurgical excision procedure) or cone biopsy
namely, obturator, external iliac and internal iliac, and thence to the specimen, which includes the entire lesion. It can also be made on
common iliac and para-­aortic nodes. Distant metastasis to lungs, a trachelectomy or hysterectomy specimen. The depth of invasion
liver, and skeleton by the hematogenous route is a late phenomenon. should not be greater than 3 mm or 5 mm, respectively, from the base
Until now, the FIGO staging was based mainly on clinical exam- of the epithelium, either squamous or glandular, from which it origi-
ination with the addition of certain procedures that were allowed by nates. The horizontal dimension is no longer considered in the 2018
FIGO to change the staging. In 2018, this has been revised by the FIGO revision as it is subject to many artefactual errors. Note must be made
Gynecologic Oncology Committee to allow imaging and pathologi- of lymphovascular space involvement, which does not alter the stage,
cal findings, where available, to assign the stage. The revised staging but may affect the treatment plan. Extension to the uterine corpus is

T A B L E   1   FIGO staging of cancer of the cervix uteri (2018).

Stage Description

I The carcinoma is strictly confined to the cervix (extension to the uterine corpus should be disregarded)
IA Invasive carcinoma that can be diagnosed only by microscopy, with maximum depth of invasion <5 mma
IA1 Measured stromal invasion <3 mm in depth
IA2 Measured stromal invasion ≥3 mm and <5 mm in depth
IB Invasive carcinoma with measured deepest invasion ≥5 mm (greater than Stage IA), lesion limited to the cervix uterib
IB1 Invasive carcinoma ≥5 mm depth of stromal invasion, and <2 cm in greatest dimension
IB2 Invasive carcinoma ≥2 cm and <4 cm in greatest dimension
IB3 Invasive carcinoma ≥4 cm in greatest dimension
II The carcinoma invades beyond the uterus, but has not extended onto the lower third of the vagina or to the pelvic wall
IIA Involvement limited to the upper two-­thirds of the vagina without parametrial involvement
IIA1 Invasive carcinoma <4 cm in greatest dimension
IIA2 Invasive carcinoma ≥4 cm in greatest dimension
IIB With parametrial involvement but not up to the pelvic wall
III The carcinoma involves the lower third of the vagina and/or extends to the pelvic wall and/or causes hydronephrosis or nonfunction-
ing kidney and/or involves pelvic and/or para-­aortic lymph nodesc
IIIA The carcinoma involves the lower third of the vagina, with no extension to the pelvic wall
IIIB Extension to the pelvic wall and/or hydronephrosis or nonfunctioning kidney (unless known to be due to another cause)
IIIC Involvement of pelvic and/or para-­aortic lymph nodes, irrespective of tumor size and extent (with r and p notations)c
IIIC1 Pelvic lymph node metastasis only
IIIC2 Para-­aortic lymph node metastasis
IV The carcinoma has extended beyond the true pelvis or has involved (biopsy proven) the mucosa of the bladder or rectum. (A bullous
edema, as such, does not permit a case to be allotted to Stage IV)
IVA Spread to adjacent pelvic organs
IVB Spread to distant organs

When in doubt, the lower staging should be assigned.


a
Imaging and pathology can be used, where available, to supplement clinical findings with respect to tumor size and extent, in all stages.
b
The involvement of vascular/lymphatic spaces does not change the staging. The lateral extent of the lesion is no longer considered.
c
Adding notation of r (imaging) and p (pathology) to indicate the findings that are used to allocate the case to Stage IIIC. Example: If imaging indicates pelvic
lymph node metastasis, the stage allocation would be Stage IIIC1r, and if confirmed by pathologic findings, it would be Stage IIIC1p. The type of imaging
modality or pathology technique used should always be documented.
Source: Bhatla et al.17
Bhatla ET AL. |
      25

also disregarded for staging purposes as it does not in itself alter either FIGO no longer mandates any biochemical investigations or inves-
the prognosis or management. The margins should be reported to be tigative procedures; however, in patients with frank invasive carci-
negative for disease. If the margins of the cone biopsy are positive for noma, a chest X-­ray, and assessment of hydronephrosis (with renal
invasive cancer, the patient is allocated to Stage IB1.18 ultrasound, intravenous pyelography, CT, or MRI) should be done. The
Clinically visible lesions, and those with larger dimensions, are allo- bladder and rectum are evaluated by cystoscopy and sigmoidoscopy
cated to Stage IB, subdivided in the latest staging as IB1, IB2, and IB3 only if the patient is clinically symptomatic. Cystoscopy is also recom-
based on the maximum diameter of the lesion. mended in cases of a barrel-­shaped endocervical growth and in cases
where the growth has extended to the anterior vaginal wall. Suspected
bladder or rectal involvement should be confirmed by biopsy and his-
4.1.2 | Invasive disease
tologic evidence. Bullous edema alone does not warrant a case to be
In the case of visible lesions, a punch biopsy may generally suffice, but allocated to Stage IV.
if not satisfactory a small loop biopsy or cone may be required. Clinical
assessment is the first step in allocation of staging.
4.2 | Pathologic staging
Imaging evaluation may now be used in addition to clinical exam-
ination where resources permit. The revised staging permits the use In case a surgical specimen is available or where image-­guided fine-­
of any of the imaging modalities according to available resources, i.e. needle aspiration cytology has been done, the pathologic report is an
ultrasound, CT, MRI, positron emission tomography (PET), to provide important source for accurate assessment of the extent of disease. As
information on tumor size, nodal status, and local or systemic spread. in the case of imaging, the pathologic methods should also be recorded
The accuracy of various methods depends on the skill of the operator. for future evaluation. The stage is to be allocated after all imaging and
MRI is the best method of radiologic assessment of primary tumors pathology reports are available. It cannot be altered later, for example
greater than 10 mm.19–23 However, ultrasound has also been shown to at recurrence. The 2018 FIGO staging includes involvement of nodes
24
have good diagnostic accuracy in expert hands. The modality used in and thus enables both the selection and evaluation of therapy, as well
assigning staging should be noted for future evaluation. Imaging has the as estimation of the prognosis and calculation of end results.
advantage of the ability to identify additional prognostic factors, which The FIGO and TNM classifications have been virtually identical
can guide the choice of treatment modality. The goal is to identify the in describing the anatomical extent of disease. The TNM nomencla-
most appropriate method and to avoid dual therapy with surgery and ture has hitherto been used for the purpose of documenting nodal
radiation as this has the potential to greatly augment morbidity. and metastatic disease status.38 The revised FIGO classification is now
For detection of nodal metastasis greater than 10 mm, PET-­CT is more closely aligned with the TNM classification in this respect as well.
more accurate than CT and MRI, with false-­negative results in 4%–15% In some cases, hysterectomy is performed in the presence of
20,25–28
of cases. In areas with a high prevalence of tuberculosis and unsuspected invasive cervical carcinoma that is diagnosed later on
inflammation, especially HIV-­endemic areas, large lymph nodes are not histopathology. Such cases cannot be clinically staged or included in
necessarily metastatic. The clinician may make the decision on imaging therapeutic statistics for obvious reasons, but reporting them sepa-
or, when possible, can use fine needle aspiration or biopsy to establish rately is desirable.
27,29,30
or exclude metastases. This is especially true in advanced stages,
where surgical assessment of para-­aortic lymph nodes may be used
4.3 | Histopathology
to tailor treatment according to extent of disease.31–33 They can be
accessed by minimally invasive surgery or laparotomy. Surgical exclu- It is essential that all cancers must be confirmed by microscopic exam-
sion of para-­aortic lymph node involvement has been reported to have ination. Cases are classified as carcinomas of the cervix if the primary
a better prognosis than radiographic exclusion alone.34 growth is in the cervix. All histologic types must be included. The his-
A review of 22 articles that assessed the safety and impact of pre- topathologic types, as described in the World Health Organization’s
treatment para-­aortic lymph node surgical staging (PALNS) found that 2014 Tumours of the Female Reproductive Organs39 are:
18% (range, 8%–42%) of patients with Stage IB–IVA cervical cancer
had para-­aortic lymph node metastases.35 The mean complication rate 1. Squamous cell carcinoma (keratinizing; non-keratinizing; papillary, bas­
of PALNS was 9% (range 4%–24%), with lymphocyst formation being aloid, warty, verrucous, squamotransitional, lymphoepithelioma-like)
the most common. In another study, up to 35% of clinically assessed 2. Adenocarcinoma (endocervical; mucinous, villoglandular, endometrioid)
Stage IIB and 20% of Stage III tumors were reported to have posi- 3. Clear cell adenocarcinoma
tive para-­aortic nodes.36 In the revised staging, all these cases will be 4. Serous carcinoma
assigned to Stage IIIC as lymph node involvement confers a worse 5. Adenosquamous carcinoma
prognosis.37 If only pelvic nodes are positive, it is Stage IIIC1; if para-­ 6. Glassy cell carcinoma
aortic nodes are also involved it is Stage IIIC2. A further notation must 7. Adenoid cystic carcinoma
be added to indicate whether this allocation is based on only imaging 8. Adenoid basal carcinoma
assessment (r) or whether pathological confirmation is available (p). In 9. Small cell carcinoma
due course, the data can be analyzed and reported accordingly. 10. Undifferentiated carcinoma
|
26       Bhatla ET AL.

Grading by any of several methods is encouraged, but it is not a basis be recommended.40 Any route can be chosen, i.e. abdominal,
for modifying the stage groupings in cervical carcinoma. Histopathologic vaginal, or laparoscopic. When LVSI is evident, pelvic lymphad-
grades are as follows: enectomy should be considered, along with modified radical hys-
terectomy.41,42 If fertility is desired, cervical conization with close
1. GX: Grade cannot be assessed follow-­up will be adequate.
2. G1: Well differentiated
3. G2: Moderately differentiated 5.1.1.2 | Stage IA2
4. G3: Poorly or undifferentiated Since there is a small risk of lymph node metastases in these cases,42–45
pelvic lymphadenectomy is performed in addition to type B radical
hysterectomy or more radical surgery.46,47 In low risk cases, simple
5 |  MANAGEMENT OF CERVICAL CANCER hysterectomy or trachelectomy, with either pelvic lymphadenec-
tomy or sentinel lymph node assessment, may be adequate surgi-
Management of cervical cancer is primarily by surgery or radiation cal treatment.48,49 When the patient desires fertility, she may be
therapy, with chemotherapy a valuable adjunct. offered a choice of the following: (1) cervical conization with lapa-
roscopic (or extraperitoneal) pelvic lymphadenectomy; or (2) radi-
cal abdominal, vaginal, or laparoscopic trachelectomy with pelvic
5.1 | Surgical management
lymphadenectomy.50,51
Surgery is suitable for early stages, where cervical conization, total
simple hysterectomy, or radical hysterectomy may be selected accord- 5.1.1.3 | Post-­treatment follow-­up
ing to the stage of disease and extent of spread of cervical cancer. Follow-­up with 3-­monthly Pap smears for 2 years, then 6-­monthly
Table 2 shows the types of radical hysterectomy. In Stage IVA, there for the next 3 years is recommended after treatment of microinvasive
is a place for pelvic exenteration in selected cases. carcinoma. With normal follow-­up at 5 years, the patient can return to
the routine screening schedule according to the national guidelines.40

5.1.1 | Microinvasive cervical carcinoma: FIGO


Stage IA 5.1.2 | Invasive cervical carcinoma: FIGO Stage IB1,
IB2, IIA1
5.1.1.1 | Stage IA1
The treatment is completed with cervical conization unless there Surgical treatment is the preferred modality for the treatment of
is lymphovascular space invasion (LVSI) or tumor cells are present Stage IB1, IB2, and IIA1 lesions. It would usually consist of type C radi-
at the surgical margin. In women who have completed childbear- cal hysterectomy with pelvic lymphadenectomy.52–54 The routes of
ing or elderly women, total extrafascial hysterectomy may also surgery may be open or minimally invasive, i.e. laparoscopic or robotic.

T A B L E   2   Types of radical hysterectomy.

Simple extrafascial hysterectomy Modified radical hysterectomy Radical hysterectomy

Piver and Rutledge Type I Type II Type III


Classification
Querleu and Morrow Type A Type B Type C
classification
Indication Stage IA1 Type IA1 with LVSI. IA2 Stage IB1 and IB2, selected
Stage IIA
Uterus and cervix Removed Removed Removed
Ovaries Optional removal Optional removal Optional removal
Vaginal margin None 1–2 cm Upper one-­quarter to one-­third
Ureters Not mobilized Tunnel through broad ligament Tunnel through broad ligament
Cardinal ligaments Divided at uterine and cervical border Divided where ureter transits broad Divided at pelvic side wall
ligaments
Uterosacral ligaments Divided at cervical border Partially removed Divided near sacral origin
Urinary bladder Mobilized to base of bladder Mobilized to upper vagina Mobilized to middle vagina
Rectum Not mobilized Mobilized below cervix Mobilized below cervix
Surgical approach Laparotomy or laparoscopy or Laparotomy or laparoscopy or Laparotomy or laparoscopy
robotic surgery robotic surgery or robotic surgery
Bhatla ET AL. |
      27

practice. It may have some role in early stage cervical cancer, i.e. FIGO
5.1.2.1 | FIGO Stage IB1
Stage IA, IB1, and IB2.58–60 Dual labeling using blue dye and radiocol-
FIGO Stage IB1 is considered as low risk with the following criteria:
loid increases the accuracy of sentinel lymph nodes can be performed
largest tumor diameter less than 2 cm, cervical stromal invasion less
with.61,62 Indocyanine green dye with near infrared technique has
than 50%, and no suspicious lymph nodes on imaging. The standard
been used in robotic surgery and laparoscopy. Pelvic lymphadenec-
management is a type C radical hysterectomy, but modified radical
tomy needs to be considered if LVSI is present.
hysterectomy may be considered in these cases. Pelvic lymphadenec-
The route of surgery may be laparotomy or minimally invasive
tomy should always be included on account of the high frequency of
surgery, either laparoscopic or robotic. The LACC trial (Laparoscopic
lymph node involvement.46,47
Approach to Cervical Cancer) compared the overall survival with open
A pelvic nerve-­sparing surgical procedure is recommended in
surgery versus laparoscopy or robotic surgery in early stage cervical
patients undergoing radical hysterectomy, in so far as radical curabil-
cancer and showed a decreased overall survival (3 of 312 vs 19 of 319,
ity is maintained, as intrapelvic injuries to the autonomic nerves (i.e.
HR 6.00, 95% CI, 1.48–20.3, P=0.004). Disease-­free survival events
hypogastric nerve, splanchnic nerve, and pelvic plexus) often lead to
showed a three-­fold increase in the minimally invasive surgery group
impairment of urination, defecation, and sexual function, and conse-
(7 of 312 vs 27 of 319, HR 3.74, 95% CI, 1.63–8.58; P=0.002). Rates of
quent deterioration of the postoperative quality of life (QOL).55,56
intraoperative complications did not differ by treatment received (11%
In young women desiring fertility sparing, a radical trachelectomy
in both). They concluded that hysterectomy by a minimally invasive
may be performed, indicated for Stage IA2–IB1 tumors measuring
route was associated with higher rates of recurrence than the open
less than or equal to 2 cm in largest diameter.57 The cervix along with
approach in early-­stage cervical cancer patients.63 Further studies may
the parametrium is removed followed by anastomosis of the uterus
be required to further confirm these findings.
with the vaginal end. Trachelectomy can be done by open abdominal,
vaginal, or by minimally invasive routes. When a vaginal approach is
planned, the pelvic nodes are first removed laparoscopically and sent 5.1.3 | FIGO Stage IB3 and IIA2
for frozen section to confirm node negativity and then proceed with
In Stage IB3 and IIA2, the tumors are larger and the likelihood of
the radical trachelectomy vaginally. Alternatively, the nodes may be
high risk factors such as positive lymph nodes, positive parametria,
first be assessed by conventional pathologic methods and the radical
or positive surgical margins that increase the risk of recurrence and
trachelectomy done as a second surgery after 1 week.
require adjuvant radiation after surgery are high. Other risk factors
that increase the risk of pelvic recurrence even when nodes are not
5.1.2.2 | FIGO Stage IB2 and IIA1 involved include: largest tumor diameter greater than 4 cm, LVSI, and
In FIGO Stage IB2 and IIA1 cervical cancer, surgery or radiotherapy invasion of outer one-­third of the cervical stroma.64,65 In such cases,
can be chosen as the primary treatment depending on other patient adjuvant whole pelvic irradiation reduces the local failure rate and
factors and local resources, as both have similar outcomes. The advan- improves progression-­free survival compared with patients treated
tages of surgical treatment are: (1) that it is feasible to determine the with surgery alone.65 However, the dual modality treatment increases
postoperative stage precisely on the basis of histopathologic findings, the risk of major morbidity to the patient.
thereby enabling individualization of postoperative treatment for The treatment modality must, therefore, be determined based on
each patient; (2) that it is possible to treat cancers that are likely to the availability of resources and tumor-­ and patient-­related factors.
be resistant to radiotherapy; and (3) that it is possible to conserve Concurrent platinum-­based chemoradiation (CCRT) is the preferred
ovarian function. Intraoperative transpositioning of the ovaries high treatment option for Stage IB3 to IIA2 lesions. It has been demon-
in the paracolic gutters away from the radiation field, in case it should strated that the prognosis is more favorable with CCRT, rather than
be required subsequently, is also feasible. The preservation of ovar- radiotherapy alone, as postoperative adjuvant therapy as well in terms
ian and sexual function makes surgery the preferred mode in younger of overall survival, progression-­free survival, and local and distant
women. Type C radical hysterectomy represents a basic procedure for recurrences.52,66,67
the treatment of cervical cancer, consisting of removal of the uterus, In areas where radiotherapy facilities are scarce, neoadjuvant che-
parametrium, upper vagina, and a part of the paracolpium, along with motherapy (NACT) has been used with the goal of: (1) down-­staging of
pelvic lymphadenectomy. As for the adjacent connective tissues, the the tumor to improve the radical curability and safety of surgery; and
anterior vesicouterine ligament (anterior and posterior leaf), lateral (2) inhibition of micrometastasis and distant metastasis. There is no
cardinal ligaments, and posterior sacrouterine and rectovaginal liga- unanimity of view as to whether it improves prognosis compared with
ments are cut from the uterus at sufficient distances from their attach- the standard treatment.68,69
ments to the uterus. Lymphadenectomy constitutes one of the bases The extent of surgery after NACT remains the same, i.e. radical
of this surgical procedure, and the extent of regional lymph node exci- hysterectomy and pelvic lymphadenectomy. The greater difficulty is
sion includes the parametrial nodes, obturator nodes, external, inter- in determining the indications for adjuvant therapy which are often
nal, and common iliac nodes. kept the same as those after primary surgery.66,67 However, it must be
The role of sentinel lymph node (SLN) mapping in cervical cancer remembered that NACT may give a false sense of security by masking
is still experimental and needs more evidence to include into routine the pathologic findings and thus affecting evaluation of indications
|
28       Bhatla ET AL.

for adjuvant radiotherapy/CCRT. NACT surgery is best reserved for arm (28% vs 12%), likely due to contributions from both treatment
research settings or those areas where radiotherapy is unavailable. modalities. An update of the same trial with 20-­year follow-­up data
This is especially true in patients with very large tumors or adenocarci- has shown marginally better results with radiotherapy compared with
noma, which have lower response rates.70 surgery (77% vs 72%, P=0.280).80 Multivariate analysis confirmed
that risk factors for survival are histopathologic type (P=0.020), tumor
diameter (P=0.008), and lymph node status (P<0.001).80
5.1.4 | FIGO Stage IVA or recurrence
Rarely, patients with Stage IVA disease may have only central disease
5.2.2 | Adjuvant radiotherapy
without involvement to the pelvic sidewall or distant spread. Such
cases, or in case of such a recurrence, pelvic exenteration can be con- Following radical hysterectomy, PORT with or without chemotherapy
sidered but usually has a poor prognosis.71–75 is indicated for patients with adverse pathologic factors such as posi-
tive pelvic nodes, parametrial infiltration, positive margins, deep stro-
mal invasion, etc. According to various prognostic factors, patients
5.2 | Radiation management
may be categorized into high-­risk, intermediate-­risk, or low-­risk dis-
In LMICs, the majority of patients present with locally advanced dis- ease. High-­risk disease includes patients with either positive surgical
ease,76 where surgery plays a limited role, and radiotherapy has an margins or lymph node metastases or parametrial spread, and such
important role. Over the last two decades, development of sophisti- patients should be offered PORT with chemotherapy since the GOG
cated planning and delivery techniques, and introduction of computer 109 trial has shown overall survival advantage.67 Intermediate-­risk
technology and imaging have galvanized the practice of radiotherapy, patients with any two of three factors (tumor size more than 4 cm,
resulting in improved clinical outcome and reduced toxicity.77,78 lymphovascular invasion, deep stromal invasion) require PORT64,81
Apart from its curative role, radiotherapy can also be used as adju- and no chemotherapy should be offered to these patients. All other
vant therapy for operated patients to prevent locoregional recurrence, patients following radical hysterectomy are termed as low-­risk disease
although the role of “dual modality” is discouraged, and as palliative patients and do not need any adjuvant therapy.
therapy for alleviating distressing symptoms in patients with advanced Tumor size of more than 4 cm is a well-­known risk factor. Since
incurable disease. 2009 it was incorporated in the FIGO staging system as Stage IB2
and now in the 2018 staging revision as Stage IB3. Recent literature,
especially with the advent of more and more fertility sparing surgery
5.2.1 | Radiation therapy for early stage disease
suggests tumor size more than 2 cm is a risk factor.82–91. In a recent
(FIGO Stage IA, IB1, IB2, and IIA1)
study, Gemer et al.91 evaluated various clinical and pathologic risk fac-
Although surgery is preferred for early stage disease, in cases with tors that may reduce the rate of multimodality treatment of early cer-
contraindications for surgery or anesthesia, radiotherapy provides vical cancer. The authors observed that 89% of patients with tumors
equally good results in terms of local control and survival. Treatment 2 cm or greater and LVSI received radiotherapy and 76% of patients
decision should be made on the basis of clinical, anatomic, and social with tumors 2 cm or greater and depth of invasion greater than 10 mm
factors. Patients with microinvasive disease have been treated by received radiotherapy. They suggest that in patients with early cervical
intracavitary radiation therapy (ICRT) alone with good results if sur- cancer, evaluation of tumor size and LVSI should be undertaken before
gery is contraindicated owing to medical problems. Selected patients performing radical hysterectomy to tailor treatment and to reduce the
with very small Stage IB1 disease (less than 1 cm) may also be treated rate of employing both radical hysterectomy and chemoradiation. In
with ICRT alone, particularly if there are relative contraindications to view of the above-­mentioned emerging literature, tumor size of more
external beam radiation therapy (EBRT).79 A dose of 60–65 Gy equiv- than 2 cm has been taken as the first cut-­off in the 2018 revision of
alent is usually prescribed to Point A. Combination of EBRT and ICRT the FIGO staging system.
is also an option for such patients. PORT consists of whole pelvic EBRT to cover the tumor bed and
Both surgery and radiotherapy remain viable options for early draining lymph node areas. A dose of 45–50 Gy is usually prescribed.
stage disease. Definitive radiotherapy or concurrent chemoradiation Intensity modulated radiation therapy (IMRT), an advanced and refined
(CCRT) is preferred in patients likely to require postoperative radio- technique of irradiation, has been explored in the postoperative set-
therapy to avoid compounding treatment-­related morbidity. There is ting to reduce the toxicity.92,93 A recent Phase III trial93 revealed
a single randomized trial comparing surgery and radiotherapy52 but improved patient reported outcomes at week five with IMRT, with no
none comparing surgery to CCRT, which is the current standard in difference after treatment completion. Therefore, postoperative pelvic
patients treated by definitive radiotherapy. Landoni et al.52 random- IMRT remains investigational until further data are published.
ized patients with IB or IIA cervical cancer to surgery with or with- The role of vaginal brachytherapy boost following EBRT is not
out postoperative radiotherapy (PORT) versus definitive radiotherapy clear; however, it may be considered for patients with close or pos-
alone. PORT was administered to 64% of patients in the surgery arm. itive margins, large or deeply invasive tumors, parametrial or vaginal
The two treatment arms resulted in similar overall survival (83%) and involvement, or extensive LVSI.94 Vaginal cuff brachytherapy is usually
disease-­free survival (74%); severe morbidity was higher in the surgery delivered by ovoids or cylinders to the upper one-­third of the residual
Bhatla ET AL. |
      29

vagina and should include two weekly fractions of high dose rate T A B L E   3   Field design for the pelvic radiotherapy.
(HDR) brachytherapy of 6 Gy each prescribed to 5 mm from the vagi-
Field Border Landmark
nal cylinder/ovoid surface.
AP-­PA Superior L4–5 vertebral interspace
fields Inferior 2 cm below the obturator foramen or 3 cm
5.2.3 | Radiation therapy for FIGO Stage inferior to distal disease, whichever is lower
IB3 and IIA2 Lateral 1.5–2 cm lateral to the pelvic brim

Although feasible, surgery as initial treatment is not encouraged for Lateral Superior Same as AP-­PA field
fields Inferior Same as AP-­PA field
patients with Stage IB3 and IIA2 disease since 80% of them require
52 Anterior Anterior to the pubic symphysis
PORT or CCRT. It is well known that the addition of adjuvant radi-
otherapy to surgery increases morbidity and thus compromises the Posterior 0.5 cm posterior to the anterior border of the
quality of life.95,96 Additionally, combined modality treatment will S2/3 vertebral junction. May include the
entire sacrum to cover the disease extent
unnecessarily overburden the surgical and radiation facilities, which
are already inadequate in low-­resource countries. Therefore, CCRT
is the standard of care for Stage IB3 and IIA2 disease. CCRT includes
higher energy beams resulting in more homogeneous dose delivery to
external radiation and intracavitary brachytherapy.65,66
deep tissues with relative sparing of superficial tissues. Recently, con-
formal radiotherapy techniques like 3D-­CRT and IMRT are increasingly
5.2.4 | Radiation therapy for FIGO Stage IIB–IVA being used with encouraging results in terms of reduced toxicity owing
to relative sparing of normal tissues (Fig. 1).
Concurrent chemoradiation is considered the standard treatment for
Although EBRT plays an important role in the treatment of cervical
patients with locally advanced cervical cancer (LACC). The chemo-
cancer, ICRT is also an extremely important component of curative
therapy regimen is intravenous administration of weekly cisplatin dur-
treatment of cervical cancer since it delivers a high central dose to the
ing the course of EBRT.
Based on the results of five large randomized trials67,97–100 that
tested addition of chemotherapy to pelvic radiation, the National (A)
Cancer Centre issued an alert in 1999 that all patients with locally
advanced cervical cancer should receive CCRT.67 These stud-
ies67,97–100 demonstrated that CCRT had a significant survival
advantage of 10%–15% at 5 years after treatment compared with
radiotherapy alone. A subsequent meta-­analysis showed maximum
benefit of chemoradiation of 6% in Stage IB2 (now termed IB3) to
Stage IIB and only 3% benefit in Stage IIIB patients.101 Concurrent
chemoradiotherapy also reduced local and distant recurrence, and
improved disease-­free survival.
A once-­weekly infusion of cisplatin (40 mg/m2 weekly with appro-
priate hydration) for 5–6 cycles during external beam therapy is a
commonly used concurrent chemotherapy regimen.99,102 For patients
(B)
who are unable to receive platinum chemotherapy, 5–fluorouracil-­
based regimens are an acceptable alternative.102–104 Data on the
toxicity associated with concurrent chemotherapy and extended field
irradiation are limited.105,106
Additional adjuvant chemotherapy after concurrent chemoradio-
therapy is being explored in an international randomized controlled
trial (OUTBACK Trial).107
The combination of EBRT and ICRT maximizes the likelihood of
locoregional control while minimizing the risk of treatment compli-
cations. The primary goal of EBRT is to sterilize local disease and to
shrink the tumor to facilitate subsequent ICRT. Standard EBRT should
deliver a dose of 45–50 Gy to the whole pelvis by 2 or 4 field box
technique (Table 3) encompassing uterus, cervix, adnexal structures,
F I G U R E   1   CT scan images showing radiotherapy planning
parametria, and pelvic lymph nodes. Although EBRT is commonly
using: (A) conventional four-­field box technique; and (B) intensity
delivered by a Cobalt-­60 teletherapy machine in several low-­resource modulated radiation therapy (IMRT) planning. Normal tissues such as
countries, linear accelerators are preferred nowadays as they provide bladder and bowel are relatively spared in IMRT planning.
|
30       Bhatla ET AL.

primary tumor and reduced doses to adjacent normal organs owing to (A)
sharp dose fall-­off.
Standard ICRT is usually performed using a tandem and two
ovoids, or a tandem and ring. Any of the dose rate systems, namely
low-­dose-­rate (LDR), high-­dose-­rate (HDR), or pulsed-­dose-­rate (PDR)
may be practiced as all three yield comparable survival rates.108 The
dose is usually prescribed to Point A or to high-­risk clinical target vol-
ume (HRCTV) if image-­based planning is used.
With an LDR system, a dose of 30–40 Gy is prescribed in one or
two sessions. With HDR, various dose fraction schedules are used,
employing a dose of 5.5–8 Gy by 3–5 weekly fractions. Owing to
resource constraints and long travelling distances in low-­resource
countries, delivering three instead of five fractions is often more
realistic and allows for treatment of a higher number of patients. The
total combined dose with EBRT and ICRT should be in the range of
80–90 Gy. Though PDR is rarely used, the overall treatment time and (B)
dose in PDR remains almost the same as in LDR except that the treat-
ment is given in multiple hourly pulses each lasting for a few minutes.
If ICRT is not feasible either due to distorted anatomy or inade-
quate dosimetry, then interstitial brachytherapy should be considered.
Interstitial brachytherapy consists of insertion of multiple needles/
catheters into the primary tumor and parametria (Fig. 2) through the
perineum with the help of a template. Due to the risk of trauma to
normal structures like bowel and bladder, use of ultrasound imaging
(especially transrectal) is suggested during the implant procedure.109
Completion of the radiotherapy protocol within the stipulated time
is an important goal as it has a direct correlation on the outcome. In
retrospective analyses, patients whose radiotherapy treatment times
exceeded 9–10 weeks had significantly higher rates of pelvic failure
F I G U R E   2   Interstitial brachytherapy implant: (A) clinical image
when compared with women whose treatment was completed in less of a patient showing the perineal template and the steel needles;
than 6–7 weeks.110,111 Currently the recommendation is to complete (B) CT scan image showing the brachytherapy needles inserted into
the entire protocol of EBRT and brachytherapy within 8 weeks. the pelvis.

Patients with an ECOG (Eastern Cooperative Oncology Group)


5.2.5 | FIGO Stage IVB/distant metastases
performance status of 0–2 may be considered for palliative systemic
Presentation with distant metastatic disease is rare, reported in about 2% chemotherapy. Where feasible, these patients could be offered partic-
of cases. A management plan should consider that the median duration ipation in clinical trials, especially when the interval to relapse is less
of survival with distant metastatic disease is approximately 7 months. than 12 months.
Concurrent chemoradiation may have better response than systemic GOG 240 studied the efficacy of antiangiogenic therapy with bev-
chemotherapy with overall and disease-­free survivals of 69% and 57%, acizumab, a humanized anti-­VEGF monoclonal antibody. When incor-
respectively, reported in patients with positive para-­aortic and supraclavic- porated in the treatment of recurrent and metastatic cervical cancer,
112
ular lymph nodes. Currently there is no role for prophylactic extended it showed increased overall survival (17.0 months vs 13.3 months, HR
field radiotherapy (EFRT) in locally advanced cervical cancer. When para-­ for death 0.71, 98% CI 0.54–0.95, P=0.004 in a one-­sided test).115
aortic nodes are involved, EFRT with concurrent chemotherapy should be The treatment is presently expensive and patients and their families
used. IMRT may be used in such patients to reduce the toxicity. need to be counseled. Adverse effects include increased incidence of
Despite limited response rates, cisplatin has been the standard hypertension, thromboembolic events, and gastrointestinal fistulae.
chemotherapy used in the setting of distant metastatic disease.113
Given low response rates to cisplatin alone after concurrent chemo-
5.2.6 | Radiation therapy after inadvertent
radiation, recent evidence supports the use of platinum doublets over
incomplete surgery
cisplatin alone, although with very modest benefits in response rates.
Cisplatin may be combined with taxanes, topotecan, 5-­fluorouracil, Invasive cervical cancer may be found during pathologic evaluation
gemcitabine, or vinorelbine.114 Carboplatin-­paclitaxel combination has of the specimen of a simple hysterectomy for an apparent benign
also been successful in these cases. condition. Inadvertent simple hysterectomy is considered inadequate
Bhatla ET AL. |
      31

surgery for invasive cervical carcinoma and subsequent therapy is thereof. The risk of both pelvic and distant failure increases in propor-
required for all such cases. In such a situation, the extent of the dis- tion to tumor volume.120,121 Most recurrences are seen within 3 years
ease should be assessed by a PET/CT scan if available, or a pelvic and and the prognosis is poor, as most patients die from progressive disease
abdominal CT or MRI scan, and chest imaging. The subsequent treat- with uremia being the most common terminal event.119,122 The treat-
ment plan is formulated based on the histologic and radiologic findings. ment plan depends on the patient’s performance status, site and extent
Although PORT for patients following inadvertent simple hyster- of recurrence and/or metastases, and prior treatment received.123
116,117
ectomy has been shown to be beneficial, the outcome for such If there is extensive local disease or distant metastatic disease,
patients even after PORT remains very poor with 5-­year recurrence-­ the patient is assigned to palliative therapy, with best supportive care
free survival of 49%,33 and therefore CCRT is generally added. In a and symptom control the recommended management. However, if the
116
study from India, Sharma et al. reported the results of 83 patients performance status is good and there is only limited metastatic dis-
treated with PORT following either inadvertent simple hysterec- ease, a trial of platinum doublet chemotherapy is justified, counseling
tomy (33 patients) or radical hysterectomy (50 patients). The 5-­year the patient and her family with respect to the limited benefits with
recurrence-­free survival was found to be significantly inferior in respect to response rate and progression-­free survival.113 Local recur-
patients who underwent PORT after inadvertent simple hysterectomy rence that cannot be salvaged with surgery or radiotherapy is likely to
(49% vs 72%, respectively; P=0.04). PORT, therefore, does not com- have a very poor response to systemic chemotherapy.
pensate for lack of adequate surgery.
In centres where the expertise is available, some of these patients
5.4.1 | Local recurrence
may be found suitable for repeat laparotomy with parametrectomy
and pelvic lymphadenectomy. The procedure is challenging due to The pelvis is the most common site of recurrence and patients who
previous scarring, adhesions, and distortion of anatomy, but does have have only locally recurrent disease after definitive therapy, whether
the potential for curative surgery as well as allow assessment of the surgery or radiotherapy, are in a more favorable situation as the dis-
118
need for adjuvant CCRT. ease is potentially curable. Good prognostic factors are the presence
of an isolated central pelvic recurrence with no involvement of the
pelvic sidewall, a long disease-­free interval from previous therapy, and
5.3 | Post-­treatment follow-­up
the largest diameter of the recurrent tumor is less than 3 cm.74,124
In a systematic review of 17 retrospective studies that followed up When the pelvic relapse follows primary surgery, it may be treated
women treated for cervical cancer, the median time to recurrence by either radical chemoradiation or pelvic exenteration. Confirmation
ranged from 7 to 36 months after primary treatment.119 Therefore, of recurrence with a pathologic specimen obtained by biopsy is essen-
closer clinical follow-­up in the 2–3 years after treatment may be impor- tial prior to proceeding with either therapy. Radical irradiation with or
tant. Routine follow-­up visits are recommended every 3–4 months for without concurrent chemotherapy) may result in 5-­year disease-­free
the first 2–3 years, then 6-­monthly until 5 years, and then annually for survival rates of 45%–74% with isolated pelvic failure after primary
life. At each visit, history taking and clinical examination are carried surgery.125,126 The extent of recurrent disease and involvement of pel-
out to detect treatment complications and psychosexual morbidity, as vic lymph nodes are prognostic factors for survival.127
well as assess for recurrent disease. Concurrent chemotherapy with either cisplatin and/or
Routine imaging is not indicated. Special circumstances, such as 5-­fluorouracil may improve outcome.128 IMRT is reported to be supe-
involved high pelvic lymph nodes, may justify interval imaging of the rior to conventional concurrent chemoradiation yielding better dose
abdomen to assess for potentially curable progression of disease. In sparing of small bowel, rectum, and bladder than chemoradiation with
the systematic review, asymptomatic recurrent disease was detected significantly higher 5-­year overall survival and progression-­free sur-
using physical exam (29%–71%), chest X-­ray (20%–47%), CT (0%– vival rates (35.4% vs 21.4%; 26.1% and 15.1%, respectively).
34%), and vaginal vault cytology (0%–17%). Frequent vaginal vault Pelvic exenteration may be feasible in some patients in whom
cytology does not significantly improve the detection of early disease there is no evidence of intraperitoneal or extrapelvic spread, and there
recurrence. Patients should return to annual population-­based screen- is a clear tumor-­free space between the recurrent disease and the pel-
ing after 5 years of disease-­free survival.119 vic sidewall.71–75 Owing to its high morbidity, it is reserved for those
Women under the age of 50 years who have lost ovarian function with expected curative potential and requires careful patient selec-
should be considered for menopausal hormone therapy. As women tion regarding the associated physical and psychological demands. A
age, the routine exam should include other age-­indicated well-­woman PET/CT scan is the most sensitive noninvasive test to determine any
checks also to ensure quality of life, including assessment of thyroid sites of distant disease, and should be performed prior to exentera-
and renal status. tion, if possible.129–136 Patient assessment and counseling regarding
the implications and ability to manage stoma and ostomy sites must
also be addressed prior to surgery.137 The overall survival is 10% but
5.4 | Recurrent disease
careful selection of patients has been reported to yield a 5-­year sur-
Recurrences may occur locally in the pelvic or para-­aortic, the patient vival with pelvic exenteration in the order of 30%–60%,71,72,74 and an
may develop distant metastases, or there may be a combination operative mortality of less than 10%.138
|
32       Bhatla ET AL.

Broadly, the management of cervical cancer in pregnancy fol-


5.4.2 | Para-­aortic nodal recurrence
lows the same principles as in the nonpregnant state. Before
The second most common site of recurrence is in the para-­aortic 16–20 weeks of pregnancy, patients are treated without delay. The
lymph nodes. Where there is isolated para-­aortic nodal recurrence, mode of therapy can be either surgery or chemoradiation depend-
curative-­intent radiation therapy or chemoradiation, can achieve long-­ ing on the stage of the disease. Radiation often results in sponta-
139
term survival in approximately 30% of cases. Better outcomes are neous abortion of the conceptus. From the late second trimester
seen in asymptomatic patients with low-­volume recurrences occur- onward, surgery and chemotherapy can be used in selected cases
ring more than 24 months from initial treatment. while preserving the pregnancy.143 When the diagnosis is made
after 20 weeks, delaying definitive treatment is a valid option for
Stages IA2 and IB1 and 1B2, which has not been shown to have any
5.5 | Comprehensive palliative care
negative impact on the prognosis compared with nonpregnant con-
Symptom control is the essence of palliative care and plays a major role trols.144–146 Timing of delivery requires a balance between mater-
in maintaining dignity and quality of life. As the disease progresses, nal and fetal health interests. When delivered at a tertiary center
patients may present with a wide range of symptoms that need to be with appropriate neonatal care, delivery by classical cesarean and
managed with individual attention. Common symptoms of advanced radical hysterectomy at the same time is undertaken not later than
cervical cancer include: pain, ureteric obstruction causing renal failure, 34 weeks of pregnancy.
hemorrhage, malodorous vaginal discharge, lymphedema, and fistulae. For more advanced disease, the impact of treatment delay on sur-
Patients require support from the corresponding clinical services as vival is not known. Neoadjuvant chemotherapy may be administered
well as psychosocial care and support for their families and caregiv- to prevent disease progression in women with locally advanced cervi-
ers. Typically a tiered approach to pain is practiced. Access to oral cal cancer when a treatment delay is planned.147,148
morphine is improving within LMICs and is an important aspect of pal-
liative care. The availability of home care teams in many regions and
AU T HO R CO NT R I B U T I O NS
involvement of nongovernmental organizations in this effort can help
minimize the need to transport the patient to hospital and save costs All authors contributed to the manuscript at all stages including
too. In terminal cases, some patients may require the services of a design, planning, data abstraction, and manuscript writing.
hospice facility as well.

AC KNOW L ED G M ENTS
5.5.1 | Palliative radiotherapy
This chapter reworks and updates the information pub-
Common symptoms in patients with advanced incurable disease lished in the FIGO Cancer Report 2015 (Bermudez A, Bhatla
include vaginal bleeding, pelvic pain, malodorous discharge, and symp- N, Leung E. Cancer of the cervix uteri. Int J Gynecol Obstet.
toms related to metastatic disease, which may be distressing to the 2015;131(Suppl.2):S88–95), with approval granted by the original
patient. Short course radiotherapy is very effective in palliation of authors. Dr Jayashree Natarajan’s help in reviewing the literature
such symptoms. Although there is no standard dose fraction schedule, is gratefully acknowledged.
a dose of 20 Gy in five fractions over 1 week or 30 Gy in 10 fractions
over 2 weeks is commonly practiced.140 In patients with severe vaginal
CO NFL I C TS O F I NT ER ES T
bleeding, a short course of EBRT may be tried and, if it fails, ICRT can
be highly effective in controlling the intractable bleeding.141 Control of NB has received research funding through her Institute from MSD,
bleeding is usually achieved after 12–48 hours of radiotherapy. GlaxoSmithKline, and Digene/Qiagen Inc. DA has received hono-
In patients with pain arising from enlarged para-­aortic or supra- raria from AstraZeneca KK, Chugai Pharmaceutical Co. Ltd, Ono
clavicular nodes, skeletal metastases,142 and symptoms associated Pharmaceutical Co. Ltd, and Takeda Pharmaceutical Co. Ltd. DNS and
with cerebral metastases, palliative radiotherapy should be given via RS have no conflicts of interest to declare.
larger fractions over shorter periods of time. Commonly used sched-
ules include large single fractions, 20 Gy in five fractions, and 30 Gy
REFERENCES
in 10 fractions.
1. Ferlay J, Soerjomataram I, Ervik M, et  al. GLOBOCAN 2012 v1.0,
Cancer Incidence and Mortality Worldwide: IARC CancerBase No.
6 |  SPECIAL SITUATIONS 11 [Internet]. Lyon, France: International Agency for Research on
Cancer; 2013.
2. Bosch FX, Lorincz A, Muñoz N, et  al. The causal relation
6.1 | Cervical cancer during pregnancy between human papillomavirus and cervical cancer. J Clin Pathol.
2002;55:244–265.
Adequate management of these patients requires a multidisciplinary
3. IARC Working Group. Human Papillomaviruses: IARC Monographs on
team. The plan must be discussed with the patient and, preferably, her the Evaluation of Carcinogenic Risks to Humans. Lyon: International
partner, as their wishes are to be respected. Agency for Research on Cancer; 2007:90.
Bhatla ET AL. |
      33

4. Bruni L, Diaz M, Castellsague X, et al. Cervical human papillomavirus 23. Kodama J, Mizutani Y, Hongo A, Yoshinouchi M, Kudo T, Okuda
prevalence in 5 continents: Meta-­analysis of 1 million women with H. Optimal surgery and diagnostic approach of stage IA2 squa-
normal cytological findings. J Infect Dis. 2010;202:1789–1799. mous cell carcinoma of the cervix. Eur J Obstet Gynecol Reprod Biol.
5. World Health Organization. Human papillomavirus vaccines: WHO 2002;101:192–195.
position paper, May 2017. Wkly Epidemiol Rec. 2017;92:241–268. 24. Grigsby PW, Siegel BA, Dehdashti F. Lymph node staging by positron
6. Saccucci M, Franco EL, Ding L, et al. Non-­vaccine-­type human pap- emission tomography in patients with carcinoma of the cervix. J Clin
illomavirus prevalence after vaccine introduction: No evidence for Oncol. 2001;19:3745–3749.
type replacement but evidence for cross-­protection. Sex Transm Dis. 25. Fischerova D, Cibula D, Stenhova H, et al. Transrectal ultrasound and
2018;45:260–265. magnetic resonance imaging in staging of early cervical cancer. Int J
7. Drolet M, Bénard É, Boily MC, et  al. Population-­level impact and Gynecol Cancer. 2008;18:766–772.
herd effects following human papillomavirus vaccination pro- 26. Yang WT, Lam WW, Yu MY, Cheung TH, Metreweli C. Comparison
grammes: A systematic review and meta-­analysis. Lancet Infect Dis. of dynamic helical CT and dynamic MR imaging in the evaluation
2015;15:565–580. of pelvic lymph nodes in cervical carcinoma. Am J Roentgenol.
8. Garland SM, Kjaer SK, Muñoz N, et al. Impact and effectiveness of the 2000;175:759–766.
quadrivalent human papillomavirus vaccine: A systematic review of 27. Havrilesky LJ, Kulasingam SL, Matchar DB, Myers ER. FDG-­PET
10 years of real-­world experience. Clin Infect Dis. 2016;63:519–527. for management of cervical and ovarian cancer. Gynecol Oncol.
9. Sankaranarayanan R, Joshi S, Muwonge R, et al. Can a single dose of 2005;97:183–191.
human papillomavirus (HPV) vaccine prevent cervical cancer? Early 28. Rose PG, Adler LP, Rodriguez M, Faulhaber PF, Abdul-Karim FW,
findings from an Indian study Vaccine. 2018;36:4783–4791. Miraldi F. Positron emission tomography for evaluating para-­aortic
10. Kreimer AR, Herrero R, Sampson JN, et  al. Evidence for single-­ nodal metastasis in locally advanced cervical cancer before surgical
dose protection by the bivalent HPV vaccine-­Review of the staging: A surgicopathologic study. J Clin Oncol. 1999;17:41–45.
Costa Rica HPV vaccine trial and future research studies. Vaccine. 29. Sakurai H, Suzuki Y, Nonaka T, et  al. FDG-­PET in the detection
2018;36:4774–4782. of recurrence of uterine cervical carcinoma following radiation
11. Sankaranarayanan R. Screening for cancer in low-­ and middle-­ therapy–tumor volume and FDG uptake value. Gynecol Oncol.
income countries. Ann Glob Health. 2014;80:412–417. 2006;100:601–607.
12. Sankaranarayanan R, Nene BM, Shastri SS, et al. HPV screening for 30. Yen T-C, Ng K-K, Ma S-Y, et  al. Value of dual-­phase
cervical cancer in rural India. N Engl J Med. 2009;360:1385–1394. 2-­fluoro-­2-­deoxy-­d-­glucose positron emission tomography in cervi-
13. Ronco G, Giorgi-Rossi P, Carozzi F, et al. Efficacy of human papilloma- cal cancer. J Clin Oncol. 2003;21:3651–3658.
virus testing for the detection of invasive cervical cancers and cer- 31. Hertel H, Köhler C, Elhawary T, Michels W, Possover M, Schneider
vical intraepithelial neoplasia: A randomised controlled trial. Lancet A. Laparoscopic staging compared with imaging techniques in the
Oncol. 2010;11:249–257. staging of advanced cervical cancer. Gynecol Oncol. 2002;87:46–51.
14. Shiferaw N, Salvador-Davila G, Kassahun K, et  al. The single-­visit 32. Ramirez PT, Jhingran A, Macapinlac HA, et  al. Laparoscopic extra-
approach as a cervical cancer prevention strategy among women peritoneal para-­aortic lymphadenectomy in locally advanced cervical
with HIV in Ethiopia: Successes and lessons learned. Glob Health Sci cancer: A prospective correlation of surgical findings with positron
Pract. 2016;4:87–98. emission tomography/computed tomography findings. Cancer.
15. Msyamboza KP, Phiri T, Sichali W, et  al. Cervical cancer screening 2011;117:1928–1934.
uptake and challenges in Malawi from 2011 to 2015: Retrospective 33. Marnitz S, Köhler C, Roth C, Füller J, Hinkelbein W, Schneider A. Is
cohort study. BMC Public Health. 2016;16:806. there a benefit of pretreatment laparoscopic transperitoneal surgical
16. Parham GP, Mwanahamuntu MH, Kapambwe S, et  al. Population-­ staging in patients with advanced cervical cancer? Gynecol Oncol.
level scale-­up of cervical cancer prevention services in a low-­resource 2005;99:536–544.
setting: Development, implementation, and evaluation of the cervical 34. Gold MA, Tian C, Whitney CW, Rose PG, Lanciano R. Surgical versus
cancer prevention program in Zambia. PLoS ONE. 2015;10:e0122169. radiographic determination of para-­aortic lymph node metastases
17. Bhatla N, Berek J, Cuello M, et al. New revised FIGO staging of cer- before chemoradiation for locally advanced cervical carcinoma: A
vical cancer (2018). Abstract S020.2. Presented at the FIGO XXII Gynecologic Oncology Group Study. Cancer. 2008;112:1954–1963.
World Congress of Gynecology and Obstetrics. Rio de Janeiro, Brazil, 35. Smits RM, Zusterzeel PLM, Bekkers RLM. Pretreatment retroperito-
October 14-19, 2018. Int J Gynecol Obstet 2018;143(Suppl.3):DOI: neal para-­aortic lymph node staging in advanced cervical cancer: A
10.1002/ijgo.12584. review. Int J Gynecol Cancer. 2014;24–6:973–983.
18. Roman LD, Felix JC, Muderspach LI, Agahjanian A, Qian D, Morrow 36. Heller PB, Maletano JH, Bundy BN, Barnhill DR, Okagaki T.
CP. Risk of residual invasive disease in women with microinva- Clinical-­pathologic study of stage IIB, III, and IVA carcinoma of
sive squamous cancer in a conization specimen. Obstet Gynecol. the cervix: Extended diagnostic evaluation for paraaortic node
1997;90:759–764. metastasis–a Gynecologic Oncology Group study. Gynecol Oncol.
19. Hricak H, Gatsonis C, Chi DS, et  al. Role of imaging in pretreat- 1990;38:425–430.
ment evaluation of early invasive cervical cancer: Results of 37. Delgado G, Bundy B. Prospective surgico pathological study of dis-
the intergroup study American College of Radiology Imaging ease free interval in patients with Stage IB squamous cell carcinoma
Network 6651-­Gynecologic Oncology Group 183. J Clin Oncol. of cervix. A gynecological oncology Group study. Gynecol Oncol.
2005;23:9329–9337. 1990;38:352–357.
20. Bipat S, Glas AS, van der Velden J, Zwinderman AH, Bossuyt PM, 38. Edge SB, Byrd DR, Compton CC, Faiz AG, Greene FL, Trotti A, eds.
Stoker J. Computed tomography and magnetic resonance imaging in Gynecologic Sites. In: AJCC Cancer Staging Manual. 7th edn. New
staging of uterine cervical carcinoma: A systematic review. Gynecol York; London: Springer; 2010:395–402.
Oncol. 2003;91:59–66. 39. Kurman RJ, Carcangiu ML, Herrington CS, Young RH. WHO
21. Hricak H, Yu KK. Radiology in invasive cervical cancer. Am J Classification of Tumours of Female Reproductive Organs, 4th edn.
Roentgenol. 1996;167:1101–1108. Lyon: IARC; 2014.
22. Subak LL, Hricak H, Powell CB, Azizi L, Stern JL. Cervical carcinoma: 40. Lee SW, Kim YM, Son WS, et al. The efficacy of conservative man-
Computed tomography and magnetic resonance imaging for preop- agement after conization in patients with stage IA1 microinvasive
erative staging. Obstet Gynecol. 1995;86:43–50. cervical carcinoma. Acta Obstet Gynecol Scand. 2009;88:209–215.
|
34       Bhatla ET AL.

41. Sevin BU, Nadji M, Averette HE, et  al. Microinvasive carcinoma of 61. Levenback C, Coleman RL, Burke TW, et al. Lymphatic mapping and
the cervix. Cancer. 1992;70:2121–2128. sentinel node identification in patients with cervix cancer undergo-
42. Elliott P, Coppleson M, Russell P, et al. Early invasive (FIGO stage IA) ing radical hysterectomy and pelvic lymphadenectomy. J Clin Oncol.
carcinoma of the cervix: A clinico-­pathologic study of 476 cases. Int 2002;20:688–693.
J Gynecol Cancer. 2000;10:42–52. 62. Hauspy J, Beiner M, Harley I, Ehrlich L, Rasty G, Covens A.
43. Webb JC, Key CR, Qualls CR, Smith HO. Population-­based study of Sentinel lymph nodes in early stage cervical cancer. Gynecol Oncol.
microinvasive adenocarcinoma of the uterine cervix. Obstet Gynecol. 2007;105:285–290.
2001;97(5 Pt 1):701–706. 63. Ramireza PT, Frumovitza M, Parejab R, Lopezc A, Vieirad MA,
44. van Meurs H, Visser O, Buist MR, et al. Frequency of pelvic lymph Ribeiroe RA. Phase III randomized trial of laparoscopic or robotic
node metastases and parametrial involvement in stage IA2 cervical versus abdominal radical hysterectomy in patients with early stage
cancer: A population-­based study and literature review. Int J Gynecol cervical cancer: LACC Trial. Abstract presented at the 49th Annual
Cancer. 2009;19:21–26. Meeting of the Society of Gynecologic Oncology, March 24–27,
45. Costa S, Marra E, Martinelli GN, et  al. Outcome of conservatively 2018. New Orleans, LA, USA.
treated microinvasive squamous cell carcinoma of the uterine cervix 64. Rotman M, Sedlis A, Piedmonte MR, et  al. A phase III randomized
during a 10-­year follow-­up. Int J Gynecol Cancer. 2009;19:33–38. trial of postoperative pelvic irradiation in Stage IB cervical carcinoma
46. Bouchard-Fortier G, Reade CJ, Covens A. Non-­radical surgery with poor prognostic features: Follow-­up of a gynecologic oncology
for small early-­stage cervical cancer. Is it time? Gynecol Oncol. group study. Int J Radiat Oncol Biol Phys. 2006;65:169–176.
2014;132:624–627. 65. Sedlis A, Bundy BN, Rotman MZ, Lentz SS, Muderspach LI, Zaino RJ.
47. Kato T, Takashima A, Kasamatsu T, et  al.; Gynecologic Oncology A randomized trial of pelvic radiation therapy versus no further ther-
Study Group of the Japan Clinical Oncology Group. Clinical tumor apy in selected patients with stage IB carcinoma of the cervix after
diameter and prognosis of patients with FIGO stage IB1 cervical can- radical hysterectomy and pelvic lymphadenectomy: A Gynecologic
cer (JCOG0806-­A). Gynecol Oncol. 2015;137:34–39. Oncology Group Study. Gynecol Oncol. 1999;73:177–183.
48. Coutant C, Cordier AG, Guillo E, Ballester M, Rouzier R, Daraï E. 66. Rose PG, Ali S, Watkins E, et  al.; Gynecologic Oncology Group.
Clues pointing to simple hysterectomy to treat early-­stage cervical Long-­term follow-­up of a randomized trial comparing concurrent
cancer. Oncol Rep. 2009;22:927–934. single agent cisplatin, cisplatin-­based combination chemotherapy,
49. Frumovitz M, Sun CC, Schmeler KM, et al. Parametrial involvement or hydroxyurea during pelvic irradiation for locally advanced cer-
in radical hysterectomy specimens for women with early-­stage cer- vical cancer: A Gynecologic Oncology Group Study. J Clin Oncol.
vical cancer. Obstet Gynecol. 2009;114:93–99. 2007;25:2804–2810.
50. Shepherd JH, Spencer C, Herod J, Ind TE. Radical vaginal trachelec- 67. Peters WA III, Liu PY, Barrett RJ II, et al. Concurrent chemotherapy
tomy as a fertility-­sparing procedure in women with early-­stage cer- and pelvic radiation therapy compared with pelvic radiation therapy
vical cancer-­cumulative pregnancy rate in a series of 123 women. alone as adjuvant therapy after radical surgery in high-­risk early-­
BJOG. 2006;113:719–724. stage cancer of the cervix. J Clin Oncol. 2000;18:1606–1613.
51. Quinn MA, Benedet JL, Odicino F, et al. Carcinoma of the cervix uteri. 68. Neoadjuvant Chemotherapy for Locally Advanced Cervical Cancer
FIGO 26th Annual Report on the Results of Treatment in Gynecological Meta-analysis Collaboration. Neoadjuvant chemotherapy for locally
Cancer. Int J Gynecol Obstet. 2006;95(Suppl.1):S43–S103. advanced cervical cancer: A systematic review and meta-­analysis
52. Landoni F, Maneo A, Colombo A, et al. Randomised study of radical of individual patient data from 21 randomised trials. Eur J Cancer.
surgery versus radiotherapy for stage Ib-­IIa cervical cancer. Lancet. 2003;39:2470–2486.
1997;350:535–540. 69. Mossa B, Mossa S, Corosu L, Marziani R. Follow-­up in a long-­term
53. Eifel PJ, Morris M, Wharton JT, Oswald MJ. The influence of tumor randomized trial with neoadjuvant chemotherapy for squamous cell
size and morphology on the outcome of patients with FIGO stage IB cervical carcinoma. Eur J Gynaecol Oncol. 2010;31:497–503.
squamous cell carcinoma of the uterine cervix. Int J Radiat Oncol Biol 70. Dastidar GA, Gupta P, Basu B, Shah JK, Seal SL. Is neo-­adjuvant
Phys. 1994;29:9–16. chemotherapy a better option for management of cervical cancer
54. Landoni F, Maneo A, Cormio G, et al. Class II versus class III radical patients of rural India? Indian J Cancer. 2016;53–1:56–59.
hysterectomy in stage IB-­IIA cervical cancer: A prospective random- 71. Shingleton HM, Soong SJ, Gelder MS, Hatch KD, Baker VV, Austin
ized study. Gynecol Oncol. 2001;80:3–12. JM Jr. Clinical and histopathologic factors predicting recurrence and
55. Fujii S, Takakura K, Matsumura N, et al. Anatomic identification and survival after pelvic exenteration for cancer of the cervix. Obstet
functional outcomes of the nerve sparing Okabayashi radical hyster- Gynecol. 1989;73:1027–1034.
ectomy. Gynecol Oncol. 2007;107:4–13. 72. Rutledge FN, Smith JP, Wharton JT, O’Quinn AG. Pelvic exenteration:
56. Roh JW, Lee DO, Suh DH, et  al. Efficacy and oncologic safety of Analysis of 296 patients. Am J Obstet Gynecol. 1977;129:881–892.
nerve-­sparing radical hysterectomy for cervical cancer: A random- 73. Morley GW, Hopkins MP, Lindenauer SM, Roberts JA. Pelvic exen-
ized controlled trial. J Gynecol Oncol. 2015;26:90–99. teration, University of Michigan: 100 patients at 5 years. Obstet
57. Abu-Rustum NR, Sonoda Y, Black D, et  al. Fertility-­sparing radical Gynecol. 1989;74:934–943.
abdominal trachelectomy for cervical carcinoma: Technique and 74. Estape R, Angioli R. Surgical management of advanced and recurrent
review of the literature. Gynecol Oncol. 2006;103:807–813. cervical cancer. Semin Surg Oncol. 1999;16:236–241.
58. Martínez-Palones JM, Gil-Moreno A, Pérez-Benavente MA, Roca I, 75. Benn T, Brooks RA, Zhang Q, et al. Pelvic exenteration in gynecologic
Xercavins J. Intraoperative sentinel node identification in early stage oncology: A single institution study over 20 years. Gynecol Oncol.
cervical cancer using a combination of radiolabeled albumin injection 2011;122:14–18.
and isosulfan blue dye injection. Gynecol Oncol. 2004;92:845–850. 76. Sankaranarayanan R. Overview of cervical cancer in the developing
59. van de Lande J, Torrenga B, Raijmakers PG, et  al. Sentinel lymph world. FIGO 26th annual report on the results of treatment in gyne-
node detection in early stage uterine cervix carcinoma: A systematic cological cancer. Int J Gynecol Obstet. 2006;95:S205–S210.
review. Gynecol Oncol. 2007;106:604–613. 77. Dutta S, Nguyen NP, Vock J, et  al. Image-­guided radiotherapy and
60. Gortzak-Uzan L, Jimenez W, Nofech-Mozes S, et al. Sentinel lymph brachytherapy for cervical cancer. Front Oncol. 2015;5:64.
node biopsy vs. pelvic lymphadenectomy in early stage cervi- 78. Harkenrider MM, Alite F, Silva SR, Small W Jr. Image-­based
cal cancer: Is it time to change the gold standard? Gynecol Oncol. brachytherapy for the treatment of cervical cancer. Int J Radiat Oncol
2010;116:28–32. Biol Phys. 2015;92:921–934.
Bhatla ET AL. |
      35

79. Grigsby PW, Perez CA. Radiotherapy alone for medically inoperable negative para-­aortic lymph nodes: A Gynecologic Oncology Group
carcinoma of the cervix: Stage IA and carcinoma in situ. Int J Radiat and Southwest Oncology Group study. J Clin Oncol. 1999;17:
Oncol Biol Phys. 1991;21:375. 1339–1348.
80. Landoni F, Colombo A, Milani R, Placa F, Zanagnolo V, Mangioni C. 98. Morris M, Eifel PJ, Lu J, et al. Pelvic radiation with concurrent chemo-
Randomized study between radical surgery and radiotherapy for the therapy compared with pelvic and para-­aortic radiation for high-­risk
treatment of stage IB-­IIA cervical cancer: 20-­year update. J Gynecol cervical cancer. N Engl J Med. 1999;340:1137–1143.
Oncol. 2017;28:e34. 99. Rose PG, Bundy BN, Watkins EB, et al. Concurrent cisplatin-­based
81. Fuller AF Jr, Elliott N, Kosloff C, et al. Determinants of increased risk radiotherapy and chemotherapy for locally advanced cervical cancer.
for recurrence in patients undergoing radical hysterectomy for stage N Engl J Med. 1999;340:1144–1153.
IB and IIA carcinoma of the cervix. Gynecol Oncol. 1989;33:34–39. 100. Keys HM, Bundy BN, Stehman FB, et  al. Cisplatin, radiation, and
82. Póka R, Molnár S, Daragó P, et al. Intention-­to-­treat analysis of radical adjuvant hysterectomy compared with radiation and adjuvant
trachelectomy for early-­stage cervical cancer with special reference hysterectomy for bulky stage IB cervical carcinoma. N Engl J Med.
to oncologic failures. Int J Gynecol Cancer. 2017;27:1438–1445. 1999;340:1154–1161.
83. Bentivegna E, Maulard A, Pautier P, Chargari C, Gouy S, Morice P. 101. National Cancer Institute. Concurrent chemoradiation for cervical
Fertility results and pregnancy outcomes after conservative treat- cancer. Clinical announcement. NCI; 1999.
ment of cervical cancer: A systematic review of the literature. Fertil 102. Vale C, Tierney JF, Stewart LA, et al. Reducing uncertainties about
Steril. 2016;106:1195–1211. the effects of chemoradiotherapy for cervical cancer: A systematic
84. Hauerberg L, Høgdall C, Loft A, et al. Vaginal radical trachelectomy review and meta-­analysis of individual patient data from 18 random-
for early stage cervical cancer. Results of the Danish National Single ized trials. J Clin Oncol. 2008;26:5802–5812.
Center Strategy. Gynecol Oncol. 2015;138:304–310. 103. Kim YS, Shin SS, Nam JH, et  al. Prospective randomized com-
85. Park JY, Joo WD, Chang SJ, et al. Long-­term outcomes after fertility-­ parison of monthly fluorouracil and cisplatin versus weekly cis-
sparing laparoscopic radical trachelectomy in young women with platin concurrent with pelvic radiotherapy and high-­dose rate
early-­stage cervical cancer: An Asan Gynecologic Cancer Group brachytherapy for locally advanced cervical cancer. Gynecol Oncol.
(AGCG) study. J Surg Oncol. 2014;110:252–257. 2008;108:195–200.
86. Plante M, Gregoire J, Renaud MC, Roy M. The vaginal radical tra- 104. Lanciano R, Calkins A, Bundy BN, et al. Randomized comparison of
chelectomy: An update of a series of 125 cases and 106 pregnancies. weekly cisplatin or protracted venous infusion of fluorouracil in com-
Gynecol Oncol. 2011;121:290–297. bination with pelvic radiation in advanced cervix cancer: A gyneco-
87. Plante M, Renaud MC, Sebastianelli A, Gregoire J. Simple vaginal logic oncology group study. J Clin Oncol. 2005;23:8289–8295.
trachelectomy: A valuable fertility-­preserving option in early-­stage 105. Varia MA, Bundy BN, Deppe G, et  al. Cervical carcinoma met-
cervical cancer. Int J Gynecol Cancer. 2017;27:1021–1027. astatic to para-­aortic nodes: Extended field radiation therapy
88. Ramirez PT, Pareja R, Rendón GJ, Millan C, Frumovitz M, Schmeler with concomitant 5-­fluorouracil and cisplatin chemotherapy: A
KM. Management of low-­risk early-­stage cervical cancer: Should Gynecologic Oncology Group study. Int J Radiat Oncol Biol Phys.
conization, simple trachelectomy, or simple hysterectomy replace 1998;42:1015–1023.
radical surgery as the new standard of care? Gynecol Oncol. 106. Grigsby PW, Lu JD, Mutch DG, Kim RY, Eifel PJ. Twice-­daily fraction-
2014;132:254–259. ation of external irradiation with brachytherapy and chemotherapy
89. Schmeler KM, Frumovitz M, Ramirez PT. Conservative management in carcinoma of the cervix with positive para-­aortic lymph nodes:
of early stage cervical cancer: Is there a role for less radical surgery? Phase II study of the Radiation Therapy Oncology Group 92-­10. Int J
Gynecol Oncol. 2011;120:321–325. Radiat Oncol Biol Phys. 1998;41:817–822.
90. Zhang Q, Li W, Kanis MJ, et al. Oncologic and obstetrical outcomes 107. National Cancer Institute. Clinical Trials Home Page. http://www.
with fertility-­sparing treatment of cervical cancer: A systematic cancer.gov/clinicaltrials. Accessed February 8, 2018.
review and meta-­analysis. Oncotarget. 2017;8:46580–46592. 108. Sharma DN, Rath GK, Gandhi AK, et al. Low-­dose-­rate versus high-­
91. Gemer O, Lavie O, Gdalevich M, et  al. Evaluation of clinical and dose-­rate versus pulsed-­dose-­rate intracavitary brachytherapy in
pathologic risk factors may reduce the rate of multimodality treat- cervical carcinoma: A mono-­institutional comparative study. Int J
ment of early cervical cancer. Am J Clin Oncol. 2016;39:37–42. Radiat Oncol Biol Phys. 2015;93(3 Suppl):e278.
92. Small W, Mell LK, Anderson P, et al. Consensus guidelines for delin- 109. Sharma DN, Rath GK, Thulkar S, et al. Use of transrectal ultrasound
eation of clinical target volume for intensity-­modulated pelvic radio- for high dose rate interstitial brachytherapy for patients of carci-
therapy in postoperative treatment of endometrial and cervical noma of uterine cervix. J Gynecol Oncol. 2010;21:12–17.
cancer. Int J Radiat Oncol Biol Phys. 2008;71:428–434. 110. Perez CA, Grigsby PW, Castro-Vita H, Lockett MA. Carcinoma of the
93. Klopp A, Yeung A, Deshmukh S, et  al. A phase III randomized trial uterine cervix. I. Impact of prolongation of overall treatment time
comparing patient-­reported toxicity and quality of life (QOL) during and timing of brachytherapy on outcome of radiation therapy. Int J
pelvic intensity modulated radiation therapy as compared to conven- Radiat Oncol Biol Phys. 1995;32:1275–1288.
tional radiation therapy. Int J Radiat Oncol Biol Phys. 2016;96:S3. 111. Lanciano RM, Pajak TF, Martz K, Hanks GE. The influence of treat-
94. Small W Jr, Beriwal S, Demanes DJ, et al. American Brachytherapy ment time on outcome for squamous cell cancer of the uterine cervix
Society consensus guidelines for adjuvant vaginal cuff brachytherapy treated with radiation: A patterns-­of-­care study. Int J Radiat Oncol
after hysterectomy. Brachytherapy. 2012;11:58–67. Biol Phys. 1993;25:391–397.
95. Yeo RM, Chia YN, Namuduri RP, et al. Tailoring adjuvant radiotherapy 112. Lim KC, Howells RE, Evans AS. The role of clinical follow up in early
for stage IB-­IIA node negative cervical carcinoma after radical hys- stage cervical cancer in South Wales. BJOG. 2004;111:1444–1448.
terectomy and pelvic lymph node dissection using the GOG score. 113. Moore DH, Blessing JA, McQuellon RP, et al. Phase III study of cis-
Gynecol Oncol. 2011;123:225–229. platin with or without paclitaxel in stage IVB, recurrent, or persistent
96. Minig L, Patrono MG, Romero N, et al. Different strategies of treat- squamous cell carcinoma of the cervix: A gynecologic oncology
ment for uterine cervical carcinoma stage IB2-­IIB. World J Clin Oncol. group study. J Clin Oncol. 2004;22:3113–3119.
2014;5:86–92. 114. Monk BJ, Sill MW, McMeekin DS, et  al. Phase III trial of four
97. Whitney CW, Sause W, Bundy BN, et  al. Randomized compari- cisplatin-­containing doublet combinations in stage IVB, recurrent, or
son of fluorouracil plus cisplatin versus hydroxyurea as an adjunct persistent cervical carcinoma: A Gynecologic Oncology Group study.
to radiation therapy in stage llB-­IVA carcinoma of the cervix with J Clin Oncol. 2009;27:4649–4655.
|
36       Bhatla ET AL.

115. Tewari KS, Sill MW, Penson RT, et  al. Bevacizumab for advanced 132. Havrilesky LJ, Wong TZ, Secord AA, Berchuck A, Clarke-Pearson DL,
cervical cancer: Final overall survival and adverse event analysis Jones EL. The role of PET scanning in the detection of recurrent cer-
of a randomised, controlled, open-­label, phase 3 trial (Gynecologic vical cancer. Gynecol Oncol. 2003;90:186–190.
Oncology Group 240). Lancet. 2017;390:1654–1663. 133. Chung HH, Jo H, Kang WJ, et al. Clinical impact of integrated PET/
116. Sharma DN, Rath GK, Bhatla N, et al. Postoperative radiotherapy fol- CT on the management of suspected cervical cancer recurrence.
lowing inadvertent simple hysterectomy versus radical hysterectomy Gynecol Oncol. 2007;104:529–534.
for cervical carcinoma. Asian Pac J Cancer Prev. 2011;12:1537–1541. 134. Pallardy A, Bodet-Milin C, Oudoux A, et  al. Clinical and survival
117. Saibishkumar EP, Patel FD, Ghoshal S, et  al. Results of salvage impact of FDG PET in patients with suspicion of recurrent cervical
radiotherapy after inadequate surgery in invasive cervical carci- carcinoma. Eur J Nucl Med Mol Imaging. 2010;37:1270–1278.
noma patients: A retrospective analysis. Int J Radiat Oncol Biol Phys. 135. Mittra E, El-Maghraby T, Rodriguez CA, et  al. Efficacy of 18F-­FDG
2005;63:828–833. PET/CT in the evaluation of patients with recurrent cervical carci-
118. Kinney WK, Egorshin EV, Ballard DJ, Podratz KC. Long-­term survival noma. Eur J Nucl Med Mol Imaging. 2009;36:1952–1959.
and sequelae after surgical management of invasive cervical carci- 136. Kitajima K, Murakami K, Yamasaki E, Domeki Y, Kaji Y, Sugimura K.
noma diagnosed at the time of simple hysterectomy. Gynecol Oncol. Performance of FDG-­PET/CT for diagnosis of recurrent uterine cer-
1992;44:24–27. vical cancer. Eur Radiol. 2008;18:2040–2047.
119. Elit L, Fyles AW, Devries MC, Oliver TK, Fung-Kee-Fung M. Follow-­up 137. Ruth-Sahd LA, Zulkosky KD. Cervical cancer: Caring for patients
for women after treatment for cervical cancer: A systematic review. undergoing total pelvic exenteration. Crit Care Nurse. 1999;19:46–57.
Gynecol Oncol. 2009;114:528–535. 138. Höckel M, Dornhöfer N. Pelvic exenteration for gynaecological
120. Eifel PJ, Jhingran A, Brown J, Levenback C, Thames H. Time course tumours: Achievements and unanswered questions. Lancet Oncol.
and outcome of central recurrence after radiation therapy for carci- 2006;7:837–847.
noma of the cervix. Int J Gynecol Cancer. 2006;16:1106–1111. 139. Niibe Y, Kenjo M, Kazumoto T, et  al. Multi-­institutional study of
121. Fagundes H, Perez CA, Grigsby PW, Lockett MA. Distant metastases radiation therapy for isolated para-­aortic lymph node recurrence in
after irradiation alone in carcinoma of the uterine cervix. Int J Radiat uterine cervical carcinoma: 84 subjects of a population of more than
Oncol Biol Phys. 1992;24:197–204. 5,000. Int J Radiat Oncol Biol Phys. 2006;66:1366–1369.
122. van Nagell JR, Rayburn W, Donaldson ES, et  al. Therapeutic impli- 140. Sharma DN, Gandhi AK, Adhikari N. Definitive radiation therapy
cations of patterns of recurrence in cancer of the uterine cervix. of locally advanced cervical cancer initially treated with palliative
Cancer. 1979;44:2354–2361. hypofractionated radiation therapy. Int J Radiat Oncol Biol Phys.
123. Eralp Y, Saip P, Sakar B, et  al. Prognostic factors and survival in 2016;96(2 Suppl):e306.
patients with metastatic or recurrent carcinoma of the uterine cer- 141. Biswal BM, Lal P, Rath GK, Mohanti BK. Hemostatic radiother-
vix. Int J Gynecol Cancer. 2003;13:497–504. apy in carcinoma of the uterine cervix. Int J Gynaecol Obstet.
124. Friedlander M, Grogan M; U.S. Preventative Services Task Force. 1995;50:281–285.
Guidelines for the treatment of recurrent and metastatic cervical 142. Chow E, Zeng L, Salvo N, Dennis K, Tsao M, Lutz S. Update on the
cancer. Oncologist. 2002;7:342–347. systematic review of palliative radiotherapy trials for bone metasta-
125. Grigsby PW. Radiotherapy for pelvic recurrence after radical hyster- ses. Clin Oncol (R Coll Radiol). 2012;24:112–124.
ectomy for cervical cancer. Radiat Med. 2005;23:327–330. 143. Amant F, Brepoels L, Halaska MJ, Gziri MM, Calsteren KV.
126. Haasbeek CJ, Uitterhoeve AL, van der Velden J, González DG, Gynaecologic cancer complicating pregnancy: An overview. Best
Stalpers LJ. Long-­term results of salvage radiotherapy for the treat- Pract Res Clin Obstet Gynaecol. 2010;24:61–79.
ment of recurrent cervical carcinoma after prior surgery. Radiother 144. Duggan B, Muderspach LI, Roman LD, Curtin JP, d’Ablaing G 3rd,
Oncol. 2008;89:197–204. Morrow CP. Cervical cancer in pregnancy: Reporting on planned
127. Piura B, Rabinovich A, Friger M. Recurrent cervical carcinoma after delay in therapy. Obstet Gynecol. 1993;82(4 Pt 1):598–602.
radical hysterectomy and pelvic lymph node dissection: A study of 145. Nevin J, Soeters R, Dehaeck K, Bloch B, Van Wyk L. Advanced cer-
32 cases. Eur J Gynaecol Oncol. 2008;29:31–36. vical carcinoma associated with pregnancy. Int J Gynecol Cancer.
128. Lee YS, Kim YS, Kim JH, et al. Feasibility and outcome of concurrent 1993;3:57–63.
chemoradiotherapy for recurrent cervical carcinoma after initial sur- 146. Hunter MI, Tewari K, Monk BJ. Cervical neoplasia in pregnancy.
gery. Tumori. 2010;96:553–559. Part 2: Current treatment of invasive disease. Am J Obstet Gynecol.
129. Sun SS, Chen TC, Yen RF, Shen YY, Changlai SP, Kao A. Value of 2008;199:10–18.
whole body 18F-­fluoro-­2-­deoxyglucose positron emission tomog- 147. Tewari K, Cappuccini F, Gambino A, Kohler MF, Pecorelli S,
raphy in the evaluation of recurrent cervical cancer. Anticancer Res. DiSaia PJ. Neoadjuvant chemotherapy in the treatment of locally
2001;21(4B):2957–2961. advanced cervical carcinoma in pregnancy: A report of two cases
130. Husain A, Akhurst T, Larson S, Alektiar K, Barakat RR, Chi DS. A pro- and review of issues specific to the management of cervical car-
spective study of the accuracy of 18Fluorodeoxyglucose positron cinoma in pregnancy including planned delay of therapy. Cancer.
emission tomography (18FDG PET) in identifying sites of metastasis 1998;82:1529–1534.
prior to pelvic exenteration. Gynecol Oncol. 2007;106:177–180. 148. Boyd A, Cowie V, Gourley C. The use of cisplatin to treat advanced-­
131. Unger JB, Ivy JJ, Connor P, et al. Detection of recurrent cervical can- stage cervical cancer during pregnancy allows fetal development and
cer by whole-­body FDG PET scan in asymptomatic and symptomatic prevents cancer progression: Report of a case and review of the liter-
women. Gynecol Oncol. 2004;94:212–216. ature. Int J Gynecol Cancer. 2009;19:273–276.
DOI: 10.1002/ijgo.12612

FIGO CANCER REPORT 2018

Cancer of the corpus uteri

Frédéric Amant1,2,3,* | Mansoor Raza Mirza4 | Martin Koskas5 | Carien L. Creutzberg6

1
Division of Gynecologic Oncology, University
Hospitals Gasthuisberg, Leuven, Belgium Abstract
2
Center for Gynecologic Oncology Endometrial cancer is the most common gynecological malignancy in high-income
Amsterdam, Netherlands Cancer Institute,
countries. Although the overall prognosis is relatively good, high-­grade endometrial cancers
Amsterdam, Netherlands
3 have a tendency to recur. Recurrence needs to be prevented since the prognosis for
Center for Gynecologic Oncology
Amsterdam, Amsterdam University Medical recurrent endometrial cancer is dismal. Treatment tailored to tumor biology is the optimal
Centers, Amsterdam, Netherlands
strategy to balance treatment efficacy against toxicity. Standard treatment consists of
4
Department of Oncology,
Rigshospitalet, Copenhagen University hysterectomy and bilateral salpingo-­oophorectomy. Lymphadenectomy (with ongoing
Hospital, Copenhagen, Denmark studies of sentinel node biopsy) enables identification of lymph node positive patients who
5
Division of Gynecologic Oncology, Bichat need adjuvant treatment, including radiotherapy and chemotherapy. Adjuvant radiotherapy
University Hospital, Paris, France
6 is used for Stage I–II patients with high-­risk factors and Stage III lymph node negative
Department of Radiation Oncology, Leiden
University Medical Center, Leiden, patients. In advanced disease, a combination of surgery to no residual disease and
Netherlands
chemotherapy results in the best outcome. Surgery for recurrent disease is only advocated
*Correspondence in patients with a good performance status with a relatively long disease-­free interval.
Frédéric Amant, Center for Gynecologic
Oncology Amsterdam, Amsterdam University
KEYWORDS
Medical Centers, Amsterdam, Netherlands.
Email: frederic.amant@uzleuven.be Chemotherapy; Corpus uteri; Endometrial cancer; FIGO Cancer Report; Gynecologic cancer;
Radiotherapy; Surgery

1 |  STAGING 1.1.2 | Nodal stations


The lymphatic system of the corpus uteri is formed by three main lym-
1.1 | Anatomy phatic trunks: utero-­ovarian (infundibulopelvic), parametrial, and presa-
cral. They collectively drain into the hypogastric (also known as internal
1.1.1 | Primary site iliac), external iliac, common iliac, presacral, and para-­aortic nodes. Direct
metastases to the para-­aortic lymph nodes are uncommon. This is sur-
The upper two-­thirds of the uterus located above the internal orifice
prising given that a direct route of lymphatic spread from the corpus
of the uterus is termed the corpus. The fallopian tubes enter at the
uteri to the para-­aortic nodes through the infundibulopelvic ligament
upper lateral corners of an inverse pear-­shaped body. The portion of
has been suggested from anatomical and sentinel lymph node studies.
the muscular organ that is above a line joining the tubouterine orifices
is referred to as the fundus.
Cancer of the corpus uteri is usually referred to as endometrial 1.1.3 | Metastatic sites
cancer, which arises from the epithelial lining of the uterine cavity. The vagina, ovaries, and lungs are the most common metastatic sites.
Its first local extension concerns the myometrium. Cancers arising in
the stromal and muscle tissues of the myometrium are called uter-
1.2 | Rules for classification
ine sarcomas and are not discussed in this overview (readers are
directed to the chapter on uterine sarcomas in this Supplement by Surgical staging of endometrial cancer replaced clinical staging by the
Mbatani et al.1). FIGO Committee on Gynecologic Oncology in 1988 and again revised

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

Int J Gynecol Obstet 2018; 143 (Suppl. 2): 37–50 wileyonlinelibrary.com/journal/ijgo  |  37
|
38       Amant ET AL.

in 2009. Rules for classification include histologic verification of grad- mismatch repair deficiency, TP53 mutation, and a copy number low group
ing and extent of the tumor. without a specific driver mutation, each with a distinct prognosis.3,4

1.3 | Histopathology 1.3.2 | Histopathologic grades (G)

1.3.1 | Histopathologic types (according to WHO/


1. GX: Grade cannot be assessed.
International Society of Gynecological Pathology
2. G1: Well differentiated.
classification)
3. G2: Moderately differentiated.
All tumors are to be microscopically verified. 4. G3: Poorly or undifferentiated.
The histopathologic types of endometrial carcinomas are2:
Degree of differentiation of the adenocarcinoma is another basis for
1. Endometrioid carcinoma: adenocarcinoma; adenocarcino- classification carcinoma of the corpus, which are grouped as follows:
ma-variants (with squamous differentiation; secretory variant;
villoglandular variant; and ciliated cell variant). 1. G1: less than 5% of a nonsquamous or nonmorular solid growth
2. Mucinous adenocarcinoma. pattern.
3. Serous adenocarcinoma. 2. G2: 6%–50% of a nonsquamous or nonmorular solid growth pattern.
4. Clear cell adenocarcinoma. 3. G3: greater than 50% of a nonsquamous or nonmorular solid
5. Undifferentiated carcinoma. growth pattern.
6. Neuroendocrine tumors.
7. Mixed carcinoma (carcinoma composed of more than one
1.3.3 | Pathologic grading notes
type, with at least 10% of each component).
Notable nuclear atypia (pleomorphism and prominent nucleoli), inap-
Apart from the classification of endometrial carcinoma, carcinoma of propriate for the architectural grade, raises the grade of a grade 1 or
the endometrium comprises mixed epithelial and mesenchymal tumors grade 2 tumor by 1. However, this should not be done too easily as
including: grade 2 will then lose its discriminative power.5
Most authors consider serous and clear cell carcinomas high grade
1. Adenomyoma by definition.
2. Atypical polypoid adenomyoma Grading of adenocarcinomas with squamous differentiation is allo-
3. Adenofibroma cated according to the nuclear grade of the glandular component.
4. Adenosarcoma
5. Carcinosarcoma: currently carcinosarcomas, in which both epithe-
1.4 | FIGO staging classification
lial and mesenchymal components are malignant and aggressive
tumors, are considered metaplastic carcinomas, and are treated as Table 1 shows the current FIGO staging classification for cancer of
aggressive carcinomas. the corpus uteri. Comparison of the stage groupings with the TNM
classification is represented in Table 2.
Endometrial cancers have traditionally been classified in one of the
following two categories:
1.4.1 | Regional lymph nodes (N)
1. Types 1 (grade 1 and 2 endometrioid carcinoma) are the most
common endometrial cancers. They may arise from complex 1. NX: Regional lymph nodes cannot be assessed.
atypical hyperplasia and are linked to excess of estrogen stim- 2. N0: No regional lymph node metastasis.
ulation. As they are usually diagnosed at early stages, they 3. N1: Regional lymph node metastasis to pelvic lymph nodes.
present a relatively good prognosis. 4. N2: Regional lymph node metastasis to para-aortic lymph nodes,
2. Types 2 are the least common endometrial tumors. They include with or without positive pelvic lymph nodes.
grade 3 endometrioid tumors as well as tumors of nonendometri-
oid histology, and develop from atrophic endometrium. Type 2
1.4.2 | Distant metastasis (M)
tumors are less hormone sensitive. Since they are diagnosed in later
stages, they are generally more aggressive and have a poorer prog-
nosis than Type 1 endometrial cancer. 1. MX: Distant metastasis cannot be assessed.
2. M0: No distant metastasis.
However, the Cancer Genome Atlas studies have identified four 3. M1: Distant metastasis (includes metastasis to inguinal lymph
molecular subgroups characterized, respectively, by POLE mutation, nodes or intraperitoneal disease).
Amant ET AL. |
      39

T A B L E   1   Cancer of the corpus uteri. extensive LVSI is found.6 The distinction made using LVSI status could
be more relevant than the distinction between Stages IA and IB for
FIGO Stage
predicting survival in Stage I endometrial cancer.7
Ia Tumor confined to the corpus uteri
As a minimum, any enlarged or suspicious lymph nodes should
a
IA No or less than half myometrial invasion be removed in all patients. For high-­risk patients (grade 3, deep myo-
IBa Invasion equal to or more than half of the metrial invasion, cervical extension, serous or clear cell histology),
myometrium
complete pelvic lymphadenectomy and resection of any enlarged
IIa Tumor invades cervical stroma, but does not extend para-­aortic nodes is recommended.
beyond the uterusb
Clinical staging, as designated by FIGO in 1971, applies to a small
IIIa Local and/or regional spread of the tumor
percentage of corpus cancers that are primarily treated with radiation
IIIAa Tumor invades the serosa of the corpus uteri and/or therapy. In those instances, the designation of that staging system
adnexaec
should be noted.
IIIBa Vaginal involvement and/or parametrial involvementc
IIICa Metastases to pelvic and/or para-­aortic lymph nodesc
a
IIIC1 Positive pelvic nodes 2 | INTRODUCTION
a
IIIC2 Positive para-­aortic nodes with or without positive
pelvic lymph nodes
IVa Tumor invades bladder and/or bowel mucosa, and/or 2.1 | Incidence
distant metastases
Endometrial cancer represents the sixth most common malignant
IVAa Tumor invasion of bladder and/or bowel mucosa
disorder worldwide. An estimated 320 000 new cases are diagnosed
IVBa Distant metastasis, including intra-­abdominal
with this malignancy annually. High-­income countries have a greater
metastases and/or inguinal nodes)
incidence of endometrial cancer (5.9%) compared with low-­resource
a
Either G1, G2, or G3. countries (4.0%), although specific mortality is higher in the latter.
b
Endocervical glandular involvement only should be considered as Stage I
The cumulative risk of endometrial cancer up to the age of 75 years
and no longer as Stage II.
c
Positive cytology has to be reported separately without changing the has been estimated as 1.6% for high-­income regions and 0.7% for
stage. low-­income countries.8 This might be attributable to high rates
of obesity and physical inactivity—two major risk factors in high-­
T A B L E   2   Cancer of the corpus uteri: FIGO staging compared with income countries. Specifically, elevated estrogen levels are known to
the TNM classification.a be the most likely cause of the increased risk of endometrial can-

Union for International Cancer Control (UICC)


cer for postmenopausal obese women.9 Conversely, physical activ-
ity and long-­term use of continuous combined estrogen–progestin
FIGO Stage T (tumor) N (lymph nodes) M (metastasis)
therapy are associated with a reduced risk of endometrial cancer.10,11
I T1 N0 M0 Interestingly, obesity is associated with earlier age at diagnosis, and
IA T1a N0 M0 with endometrioid-­type endometrial cancers. Similar associations
IB T1b N0 M0 were not observed with nonendometrioid cancers, consistent with
II T2 N0 M0 different pathways of tumorigenesis.12

III T3 N0–N1 M0 North America and Europe have the highest incidence of endome-
trial cancer, where it is the most frequent cancer of the female genital
IIIA T3a N0 M0
tract and the fourth most common site in women after breast, lung,
IIIB T3b N0 M0
and colorectal cancer.13
IIIC1 T1–T3 N1 M0
In Europe, it represents the eighth most common cancer death
IIIC2 T1–T3 N1 M0
in women, with a reported 23 700 women dying in 2012.7 In North
IVA T4 Any N M0
America, it is the sixth most frequent cause of death, with approxi-
IVB Any T Any N M1 mately 55 000 new cases and 11 000 estimated new deaths each year.3
a
Carcinosarcomas should be staged as carcinoma. The two major factors that contribute to an increase in the inci-
dence of endometrial cancer in high-­income countries are increased
prevalence of obesity and extended life expectancy. Other deter-
1.4.3 | Rules related to staging
minants—such as the widespread decrease in use of estrogen plus
During staging, distance from tumor to serosa should be measured. progestin menopausal hormone therapy—have also been proposed
Other features should also be reported in the pathologic report of the as the cause of the increased incidence rates for endometrial cancer
hysterectomy specimen. For instance, the presence of lymphovas- in North America.14
cular space invasion (LVSI) should also be indicated, as patients with Mortality rates for endometrial cancer showed a decrease in most
LVSI-­positive tumors have a significantly worse prognosis, especially if European Union member states among women born before 1940.
|
40       Amant ET AL.

Improved cancer treatment and access to health care have been sug- diagnosis is higher than for dilatation and curettage; however, it is not
8
gested as contributing to this decrease in cancer mortality. significantly higher than for office endometrial biopsy.21
Full biochemistry (renal and liver function tests), and blood count
also represent routine tests in the diagnosis of corpus uterine cancers.
2.2 | Pathophysiology
A chest X-­ray is often performed as it is a universally available, low-­
Endometrial cancer research has gained some momentum in recent cost examination and the consequences of detecting lung metastases,
years and insights obtained from those studies have significant although rare in early stage disease, are significant. Serum CA125 may
implications in the clinic. Endometrioid adenocarcinoma progresses be of value in advanced disease for follow-­up. Evaluation for metasta-
through a premalignant phase of intraepithelial endometrial neoplasia sis is useful particularly in patients with abnormal liver function tests,
15
in a large proportion of cases. Other histologic types such as serous and clinical findings such as parametrial or vaginal tumor extension. In
and clear cell carcinoma arise as a result of a sequence of genetic high-­risk patients, CT-­based imaging of the chest, abdomen, and pelvis
mutations. Mutations in the tumor suppressor p53 have been shown or PET-­CT may help determine the surgical approach. Cystoscopy and/
to play a pivotal role in serous endometrial cancer.16 or proctoscopy may be helpful if direct extension to the bladder or
rectum is suspected.

2.3 | Diagnosis
The utility of population screening for endometrial cancer remains to 3 | PROGNOSTIC TUMOR
be fully substantiated. 17
Transvaginal ultrasound (TVS) is a possible CHARACTERISTICS FOR HIGH-­R ISK DISEASE
screening test, as it is reasonably sensitive and specific. Screening is
only recommended for high-­risk groups, such as those with Lynch Its early presentation following postmenopausal bleeding results in
type 2 syndrome with a wish for fertility preservation, before the deci- a generally good prognosis, but it should be treated using evidence-­
18
sion for prophylactic hysterectomy is made at a later age. In these based protocols, and where appropriate, by expert multidisciplinary
cases, endometrial surveillance is performed by aspiration biopsy teams. Four main histopathologic criteria are recommended to deter-
and transvaginal ultrasonography starting from the age of 35 years mine high-­risk disease:
(annually until hysterectomy). Prophylactic surgery (hysterectomy and
bilateral salpingo-­oophorectomy), preferably using a minimally inva- 1. Tumor grade 3 (poorly differentiated).
sive approach, should be discussed at the age of 40 as an option for 2. Lymphovascular space invasion.
Lynch type 2 syndrome mutation carriers to prevent endometrial and 3. Nonendometrioid histology (serous, clear cell, undifferentiated,
ovarian cancer.19 small cell, anaplastic, etc.).
After physical examination, endometrial cancer is usually sus- 4. Cervical stromal involvement.
pected with ultrasound—an effective first test with a high negative
predictive value when the endometrial thickness is less than 5 mm.20 MRI scanning and intraoperative frozen section represent the most
Specifically, combination of transvaginal ultrasound with endometrial accurate means of assessing both the depth of myometrial invasion and
biopsies obtained by curettage has been shown to have a negative pre- cervical involvement.22–24 Although CT and MRI are equivalent in terms
20
dictive value of 96%. When a biopsy is required, this can be obtained of evaluating nodal metastases, neither is suitable to replace surgical
usually as an office procedure using a number of disposable instru- lymph node assessment, which provides histological confirmation.25,26
ments developed for this purpose. In patients with diagnostic uncer- PET-­CT is the best imaging method to evaluate lymph node and distant
tainty, hysteroscopy may be performed, and with flexible instruments metastases, and could be considered in high-­risk or advanced stage dis-
can also be done without recourse to general anesthesia. However, ease. The role of PET-­MRI is currently being investigated.
the prognostic role of cells that are transtubally flushed during hys- Nonsurgical staging for endometrial cancer, where extrauterine
teroscopy remains uncertain. Anesthesia might be necessary in cases disease exists, is inherently inaccurate. This is particularly the case for
of cervical stenosis or if patient tolerance does not permit an office the detection of small nodal involvement, intraperitoneal implants,
procedure. Individuals whose pelvic examination is unsatisfactory may and adnexal metastasis.
also be evaluated with transvaginal or abdominal ultrasound to rule
out concomitant adnexal pathology.
After a histopathologic diagnosis of endometrial adenocarcinoma, 4 | SURGICAL STAGING PROCEDURE FOR
other factors need to be assessed. These include the local extent of ENDOMETRIAL CANCER
the tumor, evidence of metastatic disease, as well as perioperative risk.
The pathology report from endometrial sampling should indicate Staging of endometrial cancer was changed from clinical to surgical
at least the tumor type and grade of the lesion. Overall there is only in 1988, by the FIGO Gynecologic Oncology Committee. This recom-
moderate agreement on tumor grade between preoperative endome- mendation has led to considerable debate and effort to define surgical
trial sampling and final diagnosis, with the lowest agreement for grade staging procedures that can be implemented internationally. A gener-
2 carcinomas. Agreement between hysteroscopic biopsy and final ally recommended protocol includes opening of the abdomen with a
Amant ET AL. |
      41

vertical midline incision and peritoneal washings taken immediately by removal of any enlarged or suspicious nodes. Documentation of
from the pelvis and abdomen, followed by careful exploration of the positive nodes identifies a high-­risk population and helps to tailor
intra-­abdominal contents. The omentum, liver, peritoneal cul-­de-­sac, adjuvant treatment. Nodal resection also allows identification of
and adnexal surfaces should be examined and palpated for any pos- node negative patients, potentially reducing the need for external
sible metastases. These procedures should be followed by careful pal- beam radiotherapy.19
pation for suspicious or enlarged nodes in the aortic and pelvic areas. Several parameters advocate for aortic node sampling. These
However, laparoscopic procedures have increasingly been introduced include suspicious aortic or common iliac nodes, grossly positive
as standard, especially for early stage disease, as these have been adnexae, grossly positive pelvic nodes, and high-­grade tumors show-
proven safe and reduce acute treatment-­related complications.27,28 ing full thickness myometrial invasion. Patients with clear cell, papillary
The recommended standard surgical procedure is an extra-­fascial serous, or carcinosarcoma histologic subtypes are also candidates for
total hysterectomy with bilateral salpingo-­oophorectomy. Adnexal aortic node sampling.
removal is recommended even if the tubes and ovaries appear normal,
as they may contain micrometastases. In premenopausal women with
low-­grade early stage disease, ovarian preservation could be consid- 5 | WHO SHOULD PERFORM THE
ered.29,30 Vaginal cuff removal is not advised, nor is there any benefit SURGERY?
from excising parametrial tissue in the usual case. Where obvious cer-
vical stromal involvement is demonstrated preoperatively, a modified Full surgical staging is not required for low-­risk tumors, defined as
radical hysterectomy has been historically performed. However, there well-­differentiated tumors with less than 50% myometrial invasion,
is consensus (ESMO-­ESGO-­ESTRO) that simple hysterectomy with with positive nodes in less than 5% of cases. Women with these
free margins together with pelvic and para-­aortic lymphadenectomy tumors can be safely operated on by a general gynecologist. Patients
may be sufficient.31 at greater risk of extrauterine disease who may require lymphadenec-
The safety of endoscopic surgery for the treatment of endome- tomy should, in contrast, be operated on by gynecological oncolo-
trial cancer has also been the subject of considerable debate. Recent gists. Care provided by gynecologic oncologists has been associated
studies have demonstrated that laparoscopic removal of the uterus with better survival in high-­risk cancers40 and results in efficient use
and adnexae appears to be safe. For instance, not many differences of healthcare resources and minimization of the potential morbidity
have been reported in terms of major complications between abdom- associated with adjuvant radiation.41
inal hysterectomy and laparoscopically assisted vaginal hysterectomy A thorough preoperative assessment, with particular attention to
(LAVH) or total laparoscopic hysterectomy (TLH). Additionally, lapa- the pathology and to radiological features has been defined as the
roscopic interventions are associated with significant decreased risk most effective strategy for the triaging of these patients. Triaging
of major surgical adverse events, shorter hospital stays, less pain, for lymphadenectomy is also possible during surgery. Intraoperative
and faster recoveries.32–34 Owing to the demonstrated oncological assessment mainly involves assessment of myometrial invasion.22,24,39
28,35
safety of the laparoscopic approach, hysterectomy and bilat- Grading on frozen section is possible, though suboptimal compared
eral salpingo-­oophorectomy by this route is recommended in those with preoperative grading.24
patients with no contraindications to laparoscopy (e.g. large-­volume Concerning sentinel lymph node biopsy, several key surgical points
uterus). The endoscopic route also appears safe in high-­risk endome- should be respected42:
trial cancer.36 This approach can be accompanied by a laparoscopic
lymphadenectomy, if surgical staging is to be undertaken. Robotic 1. Expertise of the surgeon and attention to technical detail.
surgery for morbidly obese patients represents a valuable option for 2. Superficial and deep cervical injection of dye.
experienced surgeons. In these instances, surgical management using 3. Complete evaluation of the peritoneal cavity (sentinel lymph node
robotics is safe and presents fewer perioperative complications com- mapping is for clinical Stage I, apparent uterine-confined disease).
pared with open surgery.37 Furthermore, retrospective studies have 4. Sentinel lymph node dissection begins with evaluation of the retro-
suggested equivalent oncologic outcomes compared with traditional peritoneal spaces and identification of the sentinel drainage path-
laparoscopic surgery.38,39 ways that emanate from the parametria, followed by excision of the
The utility of lymphadenectomy of the pelvic and para-­aortic most proximal lymph nodes in the sentinel pathway.
areas is disputed, albeit it is currently mandated through the stag- 5. Any suspicious lymph nodes should be removed regardless of sen-
ing system. Currently, it is advised that complete lymphadenectomy tinel lymph node mapping and frozen section analysis may influ-
is reserved for cases with high-­risk features. In contrast, selective ence the decision to perform para-aortic lymphadenectomy in
node sampling has been deemed dubious as a routine approach. some cases.
Since many individuals with endometrial cancer are obese or elderly, 6. Performance of hemipelvic side-specific lymphadenectomy for
with concomitant medical problems, clinical judgment is required mapping failure has been shown to reduce false-negative staging.
to determine if additional surgery is warranted. Any deeply invasive 7. Enhanced pathology evaluation of sentinel lymph nodes with serial
tumor or radiological suggestion of positive nodes is an indication sectioning and immunohistochemistry stains increases the detec-
for retroperitoneal lymph node evaluation, which might be followed tion of low-volume metastasis.
|
42       Amant ET AL.

prognostic information from lymph node status. A recent meta-­


6 |  WHEN SHOULD SURGERY BE
analysis reported overall detection rates higher than 80%, with 50%
PERFORMED?
bilateral pelvic node detection rate and 17% para-­aortic detection
rate.53 Use of indocyanine green increases the bilateral detection rate
The effect of waiting time for surgical staging on survival outcome
compared with blue dye. Additionally, cervical injection increases the
for endometrial cancer is controversial. It has been suggested that a
bilateral sentinel lymph node detection rate but decreases the para-­
longer waiting time for surgical staging was associated with worse
aortic detection rate compared with alternative injection techniques.
survival outcomes in uterine cancer43 and the delay between diagno-
The sensitivity of sentinel lymph node mapping to detect metasta-
sis and surgery should not exceed 6 weeks.44 However, when focus-
ses is higher than 90%, reaching almost 100% in a meta-­analysis.53
ing on type 1 endometrial cancer only, the waiting time for surgical
Randomized studies have suggested that sentinel lymph node map-
staging was not associated with decreased survival outcome, presum-
ping can safely replace lymphadenectomy in the staging of endome-
ably owing to its indolent growth and resulting excellent prognosis.45
trial cancer.54,55
Apart from the historical distinction between type 1 and 2 endo-
metrial cancer, various approaches (genomic and immunochemistry)
7 |  IS LYMPHADENECTOMY have been conducted to better predict prognosis and subsequently
THERAPEUTIC? adapt therapy. The Cancer Genome Atlas Research Network identified
four groups of endometrial carcinomas based on genomic features.4
Lymphadenectomy is required for accurate staging, yet its therapeu- Similarly, many immunohistochemical markers have been studied to
tic benefits remain controversial. Historically, one case–control study differentiate between low-­ and high-­risk endometrial carcinomas.
suggested that lymphadenectomy may be beneficial therapeutically46 Several studies have tried to develop more applicable variants of the
and another showed it improved prognosis even in node-­positive TCGA classification by using immunohistochemical markers and DNA
women.47 Another retrospective study suggested that complete lym- sequencing techniques that can be done on formalin-­fixed, paraffin
phadenectomy increases survival in patients with grade 3 tumors.48 embedded tissues.56,57 L1 cell adhesion molecule (L1CAM) was intro-
In contrast, two major trials of large-­scale cohorts have shown that duced as a promising biomarker for identification of patients with
pelvic lymphadenectomy offers no therapeutic benefits compared poor outcome, which has been confirmed in subsequent studies.58–60
49,50
with no lymphadenectomy. These studies, however, have been Markers of the p53 pathway,16 hormone receptor expression,61 and
criticized for several limiting factors. First, limited effort with respect microsatellite instability,62 are several of the other relevant biomarkers
to the extent of dissection and lymph node evaluation was made. to predict prognosis of endometrial cancer. Various approaches com-
Second, a high proportion of low-­risk patients in these studies might bining genomic characterization and biomarkers expression provide
have skewed the results. Finally, no direct decision on adjuvant ther- promising results to tailor adjuvant therapy.63–65
apy based on lymphadenectomy was designed as part of the proto-
cols. At present, lymphadenectomy is primarily used for staging and
should be considered in women with high-­risk factors.51 An interna- 8 | ADJUVANT TREATMENT
tional trial of the role of lymphadenectomy to direct adjuvant therapy
for high-­risk endometrial cancer (STATEC) has recently started. The At present, the indication for adjuvant radiation therapy is based on
ongoing ENGOT-­EN2-­DGCG trial (NCT01244789) aims to shed light the presence of risk factors. Low-­risk disease (Stage I, grade 1 or 2
into this issue by comparing survival in patients with Stage I grade 3 with no or superficial myometrial invasion) does not require adjuvant
endometrioid endometrial cancer, Stage I and II type 2 endometrial radiation therapy. This was demonstrated in a Danish cohort study
cancer, or Stage II endometrioid endometrial cancer and without met- of low-­risk women, in which surgery alone resulted in a 96% 5-­year
astatic node after randomization for adjuvant chemotherapy. survival.66 A seminal Norwegian trial,67 which included 621 women
In a retrospective study, para-­aortic lymphadenectomy resulted treated after surgery with vaginal brachytherapy, indicated that over-
in an improved outcome in intermediate and high-­risk patients when all survival was not improved by additional external beam radiation
compared with pelvic lymphadenectomy alone.52 A limiting factor of therapy (EBRT). This study, however, showed that adjuvant radio-
this study was that adjuvant therapy was not comparable in the two therapy reduced the risk of pelvic recurrence. Three other large ran-
groups. In patients who underwent both pelvic and para-­aortic lymph- domized trials (PORTEC-­1 trial,68 the US GOG#99 trial,69 and the UK
adenectomy, 77% received chemotherapy as opposed to only 45% in MRC ASTEC trial70) studying the benefits of pelvic radiation therapy
the pelvic lymphadenectomy group. This uncertainty is the reason why as adjuvant therapy to surgery have supported its indication only for
addition of para-­aortic lymphadenectomy is recommended if pelvic high-­risk patients. The main finding from these trials is the significant
lymphadenectomy is being done, and explains different approaches reduction in the rates of vaginal and pelvic recurrence after EBRT, but
among different centers. without added survival benefit. In contrast, EBRT added to the risk
Sentinel lymph node mapping has been introduced into the sur- of long-­term morbidity. The patients without lymphadenectomy ana-
gical staging of endometrial cancer with the goal to reduce morbid- lyzed in the PORTEC and ASTEC trials presented similar recurrence
ity associated with comprehensive lymphadenectomy and to obtain and survival rates to those with documented node-­negative disease in
Amant ET AL. |
      43

the GOG#99 trial. Additionally, PORTEC-­1 illustrated that most pelvic In the PORTEC-­3 trial, patients with high-­risk Stage I–II (32%
relapses were located in the vaginal vault (75%), and that salvage rates grade 3 and 29% serous or clear cell cancer) or with Stage III (45%)
were high in women who had not had previous radiation therapy.71 endometrial cancer were randomly allocated to pelvic EBRT alone
Other trials have investigated the value of radiation as an adjuvant or EBRT with two concurrent cycles of cisplatin in weeks 1 and 4
therapy in high-­risk patients. The PORTEC-­2 trial compared the adju- of EBRT, followed by four cycles of carboplatin and paclitaxel. At a
vant value of two radiation approaches, EBRT and vaginal brachyther- median follow-­up of 60.2 months, there was no significant difference
apy, in 427 women with high/intermediate risk factors. The patients in overall survival between the arms, but a significant difference in
were randomized to EBRT or vaginal brachytherapy.72 This trial failure-­free survival, with women in the combined chemoradio-
showed that vaginal brachytherapy had excellent vaginal control rates therapy arm having 7% higher failure-­free survival (76% vs 69%;
(<2% at 5 years for both EBRT and vaginal brachytherapy groups), with P=0.022).79 Women with Stage III disease had the highest absolute
minimal adverse effects and significantly better quality of life. Quality benefit of chemoradiotherapy, with 5-­year failure-­free survival of
of life of patients in the brachytherapy group remained the same as 69% versus 58% for radiotherapy alone (P=0.03). The large majority
those of an age-­matched normal population.73 Since this seminal trial, of recurrences were at distant sites (22% vs 28%) and pelvic recur-
vaginal brachytherapy has replaced EBRT as standard adjuvant treat- rence was rare. In view of the toxicity of chemoradiotherapy with
ment for patients with high/intermediate risk factors. significantly more grade 3–4 adverse events during and after treat-
However, in low-­risk patients, adjuvant radiation therapy does not ment and a persisting higher rate of grade 2 sensory neuropathy at
lead to a better survival. In a Danish study, omission of any EBRT or vag- longer term, it can be concluded that the combined schedule cannot
inal brachytherapy for high/intermediate risk disease led to an increase be recommended as a new standard of care for Stage I–II disease, but
in recurrence rates (22% for intermediate risk disease, of which 15% women with Stage III endometrial cancer should be counseled about
locoregional) without affecting survival rates.74 A patient preference the failure-­free survival benefit.
study showed that patient’s preferences are biased toward a treatment In the randomized GOG-­258 trial for Stage III and Stage IV (resid-
75
preventing relapse. However, even treating all women with high/ ual disease <2 cm allowed), 813 patients were randomized to receive
intermediate risk factors (grade 1–2 with deep invasion) is still over- either chemoradiotherapy as used in PORTEC-­3 or six cycles of car-
treatment. The currently ongoing PORTEC-­4a trial investigates the use boplatin and paclitaxel without radiotherapy.80 Addition of radiation
of combined clinicopathologic, immunohistochemical, and molecular therapy to chemotherapy did not improve overall or progression-­
markers to determine the use of adjuvant vaginal brachytherapy or free survival, but the rate of pelvic (7% vs 3%; HR 0.36) and para-­
observation, keeping EBRT only for those with high-­risk factors. aortic nodal relapse (21% vs 10%; HR 0.43) was significantly higher
Since adjuvant radiotherapy alone and adjuvant chemotherapy in the chemotherapy alone arm. In the ongoing ENGOT-­EN2-­DGCG-­
alone have shown similar impact on overall or relapse-­free survival in trial, patients with node-­negative endometrial cancer with high-­risk
76,77
patients operated on for endometrial cancer, several studies have features are randomized to adjuvant chemotherapy (six cycles of
investigated the effect of sequential combination of chemotherapy and carboplatin-­paclitaxel) or observation, with or without brachytherapy
radiotherapy. A meta-­analysis pooling the results of two randomized in both arms. This trial could provide some answers to the questions
trials (NSGO-­EC-­9501/EORTC-­55991 and MaNGO ILIADE-­III) inves- regarding optimal use and optimal schedules of adjuvant therapy for
tigating the therapeutic value of combining adjuvant platinum-­based women with high-­risk endometrial cancer.
chemotherapy with EBRT in patients with risk factors (grade 3 or deep In summary, adjuvant radiation therapy is discouraged in low-­risk
invasion or adverse histologies) found a significant 9% improvement in patients and indicated in high-­risk patients. Specifically, patients with
progression-­free survival (69% vs 78% at 5 years; Hazard Ratio [HR] grade 1–2 tumors and no more than 50% myometrial invasion, or for
0.63) with the addition of chemotherapy to EBRT, and a trend for a 7% those with only a single risk factor, adjuvant radiotherapy is not rec-
improvement in 5-­year overall survival (75% vs 82%; HR 0.69, P=0.07). ommended. For patients with high/intermediate risk factors (at least
Three other large randomized trials (GOG#249, GOG#258, two of the factors: age >60 years, deep myometrial invasion, grade 3,
PORTEC-­3) are currently underway to support and expand on serous or clear cell histology, LVSI), vaginal brachytherapy alone is
those findings. The randomized GOG-­249 trial, which recruited 601 preferable to EBRT, providing excellent vaginal control without impact-
patients with Stage I–II endometrial cancer with high/intermediate or ing on quality of life. In patients with higher-­risk Stage I–II disease
high-­risk factors, compared vaginal brachytherapy plus three cycles (grade 3 and deep invasion and/or LVSI, unfavorable histologies, unfa-
of carboplatin-­paclitaxel chemotherapy with pelvic EBRT alone; vorable molecular factors), pelvic EBRT remains the standard of care.
results showed no differences in relapse-­free survival between the Overall, the need for EBRT decreases when surgical staging identifies
arms, while there was better pelvic control in the pelvic EBRT arm node-­negative disease.19 Surgical staging also allows clinicians to iden-
and more acute toxicity in the chemotherapy arm. From this trial, the tify node-­positive (Stage III) disease that benefits from adjuvant ther-
authors concluded that for Stage I–II endometrial cancer with (high) apy. For women with Stage III endometrial cancer, the combination of
risk features, pelvic EBRT is still the standard of care.78 About 50% adjuvant chemotherapy and radiation therapy seems most effective
of the trial population had grade 1–2 disease with a baseline 5-­year to maximize recurrence-­free survival. Ongoing and new studies with
survival of 86%–91% and for these patients, vaginal brachytherapy more individual assessment of molecular features will investigate their
alone might be preferable. role in directing adjuvant treatment.
|
44       Amant ET AL.

As primary tumors of both the ovary and the endometrium may


9 |  PROGESTOGEN THERAPY
be present in patients with presumed Stage III disease with adnexal
involvement, full surgical staging and expert pathologic examination
Although the use of progesterone therapy has been widely
of the specimen is recommended in these cases.
recognized in the past, a meta-­a nalysis of six randomized tri-
Adjuvant treatment is indicated for women with Stage III disease
als totalizing 3339 women has shown no survival benefit for
as detailed in Section 8 above.
adjuvant progestogen therapy in endometrial cancer. 81 A subse-
Patients with clinical Stage III endometrial carcinoma in which
quently published randomized trial of 1012 women also failed to
surgical resection is not possible are treated primarily by pelvic irra-
demonstrate any survival benefit. 82 However, hormonal therapy
diation, with or without chemotherapy.89 Once therapy has been
can provide prolonged remission of metastatic disease in women
completed, exploratory laparotomy should be considered for those
with grade 1 and/or ER/PR receptor-­p ositive disease. Where
patients whose disease now appears to be resectable.
possible, ER/PR should be determined on a biopsy of the recur-
rent tumor because the hormone receptor status may change
over time. 83
12 | STAGE IV

10 | STAGE II Optimal management in women with Stage IV endometrial cancer


includes cytoreductive surgery, which is associated with superior
10.1 | Occult Stage II disease overall survival outcome.90 In advanced disease, neoadjuvant chem-

Therapeutic management of patients with clinically occult Stage II dis- otherapy is also an option, particularly if postoperative morbidity is

ease is similar to that of patients with Stage I disease. considered likely and/or ascites is present.91 After surgery, platinum-­
based chemotherapy should be considered, based on the trials cited
above. Patients with evidence of extra-­abdominal metastases are usu-
10.2 | Clinical overt Stage II disease ally managed with systemic platinum-­based chemotherapy, or hormo-
In these cases, radical hysterectomy, bilateral salpingo-­ nal therapy if grade 1 and/or receptor positive.
oophorectomy, bilateral pelvic lymphadenectomy, and selec- As neoadjuvant chemotherapy is the treatment of choice in
tive aortic node dissection have been historically used as primary advanced-­stage disease, as well as in relapsed disease, several stud-
treatment. However, it is important to note that this strategy has ies have investigated the optimal combinations of chemotherapeu-
been poorly supported by the medical literature. Results of one of tic agents that represent the most effective neoadjuvant therapy for
the few retrospective studies could not find any survival benefit Stage IV endometrial cancer patients. As the combinations of doxorubi-
from radical hysterectomy for patients with suspected gross cer- cin, cisplatin, and paclitaxel (TAP)92 and carboplatin and paclitaxel have
vical involvement in comparison with simple or modified radical been shown to be most effective, these have been the most studied.
hysterectomy. 31,84
Surgical treatment in patients with suspected The former, however, is much more toxic and resulted in treatment-­
gross cervical involvement is currently under evaluation, as radical related deaths. A comparative trial of the GOG, randomizing to either
hysterectomy increases the risk of adverse events. Preoperative TAP or carboplatin-­paclitaxel chemotherapy has shown both schedules
MRI scanning is advisable to exclude bladder involvement and to have similar efficacy, while carboplatin-­paclitaxel was preferred for
ensure local resectability. Studies indicate excellent results for this lower morbidity; full results have not yet been published.93
approach, with no benefit from the addition of radiation for patients Two randomized trials have compared doxorubicin monotherapy
with negative nodes. 85,86
Adjuvant radiotherapy is usually reserved versus doxorubicin–cisplatin doublet.94,95 Superiority of the combina-
for patients with involved nodes or other adverse factors and/or tion chemotherapy in terms of progression-­free and overall survival,
close or involved surgical margins. with manageable toxicity, was confirmed in both studies. Doxorubicin–
However, neoadjuvant therapy followed by a less extensive simple cisplatin doublet versus doxorubicin–cisplatin–paclitaxel triplet was
hysterectomy can represent an alternative. If surgery is not considered tested in a phase III randomized trial.92 The triplet regimen resulted
feasible because of tumor extension and/or in medically inoperable in a significantly superior progression-­free survival, although this regi-
patients, full pelvic radiotherapy and intracavitary brachytherapy, as in men proved to be too toxic, with treatment-­related deaths despite the
cervical cancer, may be employed either preoperatively or definitively use of growth factors.
with high disease control and survival rates. 87,88 The carboplatin–paclitaxel doublet has been tested in several
phase II studies in advanced-­stage or relapsed disease, demonstrating
a response rate of 65%–75% and progression-­free survival of about
11 | STAGE III 14 months.96–98 The interim results of the GOG-­0209 trial, a nonin-
feriority trial comparing the combination of doxorubicin, cisplatin, and
Most patients with Stage III endometrial cancer are managed by com- paclitaxel (TAP) and G-­CSF versus carboplatin and paclitaxel, show
plete surgical resection of all pelvic and/or nodal disease, followed by that the carboplatin and paclitaxel doublet is not inferior to TAP.98
postoperative EBRT and/or chemotherapy. The better tolerability profile of carboplatin–paclitaxel has led to the
Amant ET AL. |
      45

recommendation of the use of carboplatin and paclitaxel as the stan- medroxyprogesterone acetate (400–600 mg/d) may be appropri-
dard for adjuvant treatment in Stage III and IV disease. ate in these situations. The safety of such an approach has been
Pelvic radiotherapy in Stage IV disease is sometimes considered reported in several studies, for grade 1 endometrial adenocarci-
to provide local tumor control. Similarly, it has also been suggested noma and atypical hyperplasia.100 Few studies reported the safety
that patients with vaginal bleeding or pain from a local tumor mass, of fertility-­sparing management of grade 2 endometrial can-
or with leg edema due to lymph node involvement, should be treated cer.101 However, a recent large retrospective analysis reported an
with pelvic radiotherapy. Palliation of brain or bone metastases can be increased risk associated with uterine preservation in patients with
effectively obtained with short courses (1–5 fractions) of radiotherapy. grade 2 and 3 endometrial adenocarcinoma and suggested such
management should be limited in time.29 Equivocal lesions should
be examined by an experienced pathologist. In cases of complete
13 |  SPECIAL CONSIDERATIONS
response, conception must be encouraged and referral to a fertility
clinic is recommended. Although the literature describes successful
13.1 | Diagnosis post hysterectomy
outcomes, fatal recurrences of endometrial cancer after a conserv-
Several therapeutic management problems have been reported to ative approach have been reported; as such, the patient must be
arise from post hysterectomy diagnosis. This is particularly true in informed about the nonstandard treatment. Hysterectomy should
cases where the adnexae have not been removed, which most often be recommended once childbearing is complete.
arises following vaginal hysterectomy for pelvic organ prolapse. Ovarian preservation, in patients with grade 1 intramucosal endo-
Recommendations for further postoperative therapy are based on metrial adenocarcinoma, might represent a beneficial therapeutic
known risk factors for extrauterine disease related to the histologic option, as this management was not associated with an increase in
grade and depth of myometrial invasion. Individuals with grade 3 cancer-­related mortality in the largest sample available.30
lesions, deep myometrial invasion, or LVSI may be candidates for addi-
tional surgery to remove the adnexae, or adjuvant EBRT. Patients with
a grade 1 or 2 lesion with minimal myometrial invasion and no LVSI 14 | FOLLOW-­U P
involvement generally require no further therapy.
The objectives of follow-­up care for treated endometrial cancer
patients are to provide reassurance, diagnose early recurrence, and
13.2 | Medically inoperable patients
collect data. The clinical and cost-­effectiveness of follow-­up imple-
The most common reasons for endometrial carcinoma to be deemed mentation has been addressed internationally in one prospective102
medically inoperable are morbid obesity and severe cardiopulmonary dis- and several retrospective studies.103–105 Overall, these studies found
ease. In such cases, uterine brachytherapy is advised and has been shown that about 75% of recurrences in endometrial cancer patients are
to achieve cure rates in excess of 70%. In the presence of prognostic symptomatic and 25% asymptomatic. Neither recurrence-­free nor
factors suggesting a high risk of involved nodes it can be combined with overall survival was improved in asymptomatic cases compared with
EBRT.88 Primary radiation therapy for medically inoperable patients with those detected at clinical presentation. Most (65%–85%) recurrences
clinical Stage I and II endometrial adenocarcinoma provides disease con- were diagnosed within 3 years of primary treatment, and 40% of recur-
trol, with fewer than 16% of surviving patients experiencing recurrence.99 rences were local. Another important finding of those studies was that
For patients with a well-­differentiated lesion, contraindications the use of routine follow-­up Pap smears and chest X-­rays is not cost-­
to general anesthesia, and who are unsuitable for radiotherapy, effective. Given the high salvage rate following radiotherapy, it has
high-­dose progestins may be used. Trials using intrauterine hormone been suggested that nonirradiated patients are a group that would
releasing devices instead of oral progestins are underway. In patients benefit from regular follow-­up to detect early vaginal recurrence.106
with contraindications to high-­dose progestins, the uterine hormone Two systematic reviews107,108 documented evidence for the util-
releasing device can be considered. ity of follow-­up examinations, and concluded that follow-­up should
be practical and directed by symptoms and pelvic examination. These
studies also recommend reduction in the frequency of follow-­up visits
13.3 | Diagnosis in young women
for low-­risk patients. Given the low risk of recurrence, vaginal cytology
Since endometrial carcinoma is uncommon in women the age of can be omitted, resulting in reduced healthcare costs.109 It appears
35 years, diagnosis during the reproductive years should be made that visual inspection is sufficient, since positive cytology is merely
with caution, and grade 1 endometrial carcinoma may be confused diagnosed in cases of symptomatic recurrence.104,110,111
with severe atypical hyperplasia. In these women, consideration More recently, studies of minimal follow-­up (nurse led, telephone
should be given to an estrogen-­related underlying condition such based) after the first year have been done and results are awaited.112,113
as a granulosa cell tumor, polycystic ovaries, or obesity. Fertility First results suggest good patient acceptability once prompt access to
preservation is only recommended in grade 1 endometrioid endo- evaluation in case of symptoms is ensured.
metrial cancer not invading the myometrium (as determined by Follow-­up care should also include patient counseling as these
MRI).19 Progestins such as megestrol acetate (160–320 mg/d) or patients are at risk of second cancers following their primary
|
46       Amant ET AL.

endometrial cancer. For instance, the estimated incidence rate of and help to differentiate tumors at low- and high-risk of lymph
Lynch syndrome in an unselected endometrial cancer population is node metastasis. Imaging might be used to determine depth
3%–6%.114 Routine pathologic screening of mismatch repair deficien- of myometrial invasion, cervical involvement, and lymph node
cies in the endometrial cancer specimen, similar to colorectal cancer, enlargement. Level of Evidence C
has been advocated and is increasingly being introduced in practice.115   2. Although lymphadenectomy in clinical Stage I endometrial can-
However, in most women with mismatch repair deficiency this is cer decreases recurrence, it has no impact on overall or relapse-
caused by MLH1 promoter hypermethylation and a test of this before free survival. Level of Evidence A. In the clinic, lymphadenectomy
referring a patient to a clinical geneticist is recommended. Survivors of should be performed for staging only in high-risk cases. There is
endometrial cancer have a three-­fold increased risk of second cancer little evidence to support a therapeutic benefit, but it may be
when compared with a matched population. This risk increase seems used to select women with positive nodes for adjuvant therapy
mainly related to lifestyle factors and genetic susceptibility.116 These and reduce the need for EBRT in node-negative patients. Level
women should be counseled on exercise and weight loss programs. of Evidence C
  3. In patients with Stage I endometrial cancer with low-, intermedi-
ate-, or high/intermediate risk features, adjuvant radiotherapy
15 | RECURRENCE has no impact on survival, but significantly reduces the rate of
pelvic recurrence. Level of Evidence A. In high-risk patients, vagi-
The therapeutic management for localized recurrences includes sur- nal brachytherapy effectively reduces the risk of vaginal relapse.
gery, radiation therapy, or a combination of both. The choice of these Level of Evidence A. EBRT should be considered in patients with
strategies depends on the primary therapy. Screening for distant presumed Stage I–II disease with strong adverse factors, positive
metastases should be performed before deciding on curative treat- nodes, or advanced stage disease to ensure pelvic control. Level
ment. If primary therapy consisted of surgery alone, radiotherapy of Evidence A
represents an effective salvage strategy in cases of vaginal or central   4. The addition of adjuvant chemotherapy to radiotherapy in patients
pelvic recurrence. In these cases, a combination of EBRT and brachy- with high-risk disease improves progression-free survival, but an
therapy, preferably image guided, is usually required. Large recur- overall survival benefit is unproven. Level of Evidence A
rences should be evaluated for excision, followed by radiotherapy.   5. Adjuvant chemotherapy for patients with early stage, high-risk
Alternatively, chemotherapy may be considered to decrease the vol- disease should only be considered for those with serous cancers
ume of the recurrence and hence improve the chances of complete and after individual patient counseling (no proven benefit in over-
surgical resection. Additional chemotherapy with radiotherapy is being all survival), and preferably be done within clinical trials.
evaluated in an ongoing GOG trial. Extended surgery may be justified,   6. Chemotherapy is a more effective strategy compared with whole
especially in patients who have had prior radiation therapy. However, abdominal radiation in patients with Stage IV disease and abdomi-
radical surgery within irradiated fields (especially in the case of side- nal disease with residual nodules less than 2 cm diameter. Level
wall recurrence) frequently results in significant morbidity, such as of Evidence A
treatment-­resistant pain and fistula formation. The results of pelvic   7. Targeted therapy in endometrial cancer should be further devel-
exenteration in properly selected cases (central recurrences without oped and only considered within clinical trials.
signs of distant spread) are similar to those obtained in cervical cancer.   8. The use of adjuvant hormonal therapy (progestogen) has not been
Overall, survival rates in well-­selected patients are in the order of 50%. properly substantiated. Level of Evidence A
Nonlocalized recurrent tumors are usually treated with proges-   9. High-risk and advanced stage endometrial cancer patients should
tin therapy: medroxyprogesterone acetate 50–100 mg three times a be managed where possible by a gynecological oncologist, work-
day or megestrol acetate 80 mg 2–3 times a day. Treatment is contin- ing within a multidisciplinary team. Level of Evidence A
ued as long as the disease is stable or in remission. Maximum clinical 10. Patients with endometrial cancer are frequently old and frail, and
response may only be observed three or more months after therapy this should be taken into consideration when prescribing adjuvant
initiation. Platinum-­based chemotherapy (cisplatin and doxorubicin, or therapy. Professional consensus
carboplatin and paclitaxel) has been recommended for patients with
advanced or recurrent disease, not amenable to cure by surgery and/
or radiotherapy.96,117 Several ongoing trials are currently investigating
AU T HO R CO NT R I B U T I O NS
the clinical applicability of targeted therapies in patients with nonlo-
calized recurrent tumors All authors contributed equally to this manuscript.

AC KNOW L ED G M ENTS
16 | RECOMMENDATIONS FOR PRACTICE
This chapter updates the information published in the FIGO Cancer
 1. A definitive tissue diagnosis must be obtained preoperatively. Report 2015 (Amant F, Mirza MR, Koskas M, Creutzberg CL. Cancer
This will result in better selection of the surgical approach, of the corpus uteri. Int J Gynecol Obstet. 2015;131(Suppl.2):S96–104).
Amant ET AL. |
      47

CO NFLI CTS OF I NTE RE ST bleeding–a Nordic multicenter study. Am J Obstet Gynecol.


1995;172:1488–1494.
The authors have no conflicts of interest to declare. 21. Visser NCM, Reijnen C, Massuger L, Nagtegaal ID, Bulten J,
Pijnenborg JMA. Accuracy of endometrial sampling in endometrial
carcinoma: A systematic review and meta-­analysis. Obstet Gynecol.
2017;130:803–813.
REFERENCES
22. Ugaki H, Kimura T, Miyatake T, et al. Intraoperative frozen section
1. Mbatani N, Olawaiye A, Prat J. Uterine sarcomas. Int J Gynecol assessment of myometrial invasion and histology of endometrial
Obstet. 2018;143(Suppl.2):51–58. cancer using the revised FIGO staging system. Int J Gynecol Cancer.
2. Kurman RJ, Carcangiu ML, Herrington S, Young RH. Tumours of the 2011;21:1180–1184.
Female Reproductive Organs. WHO classification of tumours. Lyon: 23. Cade TJ, Quinn MA, McNally OM, Neesham D, Pyman J, Dobrotwir
IARC Press; 2014. A. Predictive value of magnetic resonance imaging in assessing
3. Piulats JM, Guerra E, Gil-Martin M, et  al. Molecular approaches myometrial invasion in endometrial cancer: Is radiological staging
for classifying endometrial carcinoma. Gynecol Oncol. sufficient for planning conservative treatment? Int J Gynecol Cancer.
2017;145:200–207. 2010;20:1166–1169.
4. Cancer Genome Atlas Research N, Kandoth C, Schultz N, et  al. 24. Ozturk E, Dikensoy E, Balat O, Ugur MG, Aydin A. Intraoperative fro-
Integrated genomic characterization of endometrial carcinoma. zen section is essential for assessment of myometrial invasion but
Nature. 2013;497:67–73. not for histologic grade confirmation in endometrial cancer: A ten-­
5. Scholten AN, Smit VT, Beerman H, van Putten WL, Creutzberg year experience. Arch Gynecol Obstet. 2012;285:1415–1419.
CL. Prognostic significance and interobserver variability of his- 25. DelMaschio A, Vanzulli A, Sironi S, et  al. Estimating the depth of
tologic grading systems for endometrial carcinoma. Cancer. myometrial involvement by endometrial carcinoma: Efficacy of
2004;100:764–772. transvaginal sonography vs MR imaging. AJR Am J Roentgenol.
6. Winer I, Ahmed QF, Mert I, et  al. Significance of lymphovascular 1993;160:533–538.
space invasion in uterine serous carcinoma: What matters more; 26. Epstein E, Blomqvist L. Imaging in endometrial cancer. Best Pract Res
extent or presence? Int J Gynecol Pathol. 2015;34:47–56. Clin Obstet Gynaecol. 2014;28:721–739.
7. Aristizabal P, Graesslin O, Barranger E, et  al. A suggested mod- 27. Janda M, Gebski V, Davies LC, et al. Effect of total laparoscopic hys-
ification to FIGO stage I endometrial cancer. Gynecol Oncol. terectomy vs total abdominal hysterectomy on disease-­free survival
2014;133:192–196. among women with Stage I endometrial cancer: A randomized clini-
8. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cal trial. JAMA. 2017;317:1224–1233.
cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108. 28. Walker JL, Piedmonte MR, Spirtos NM, et al. Recurrence and survival
9. Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-­mass after random assignment to laparoscopy versus laparotomy for com-
index and incidence of cancer: A systematic review and meta-­analysis prehensive surgical staging of uterine cancer: Gynecologic Oncology
of prospective observational studies. Lancet. 2008;371:569–578. Group LAP2 Study. J Clin Oncol. 2012;30:695–700.
10. Friedenreich CM, Neilson HK, Lynch BM. State of the epidemiologi- 29. Gonthier C, Trefoux-Bourdet A, Koskas M. Impact of conservative
cal evidence on physical activity and cancer prevention. Eur J Cancer. managements in young women with Grade 2 or 3 endometrial ade-
2010;46:2593–2604. nocarcinoma confined to the endometrium. Int J Gynecol Cancer.
11. Cust AE. Physical activity and gynecologic cancer prevention. Recent 2017;27:493–499.
Results Cancer Res. 2011;186:159–185. 30. Koskas M, Bendifallah S, Luton D, Darai E, Rouzier R. Safety of
12. Nevadunsky NS, Van Arsdale A, Strickler HD, et al. Obesity and age uterine and/or ovarian preservation in young women with grade 1
at diagnosis of endometrial cancer. Obstet Gynecol. 2014;124(2 Pt intramucous endometrial adenocarcinoma: A comparison of survival
1):300–306. according to the extent of surgery. Fertil Steril. 2012;98:1229–1235.
13. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J 31. Phelippeau J, Koskas M. Impact of radical hysterectomy on survival
Clin. 2018;68:7–30. in patients with Stage 2 Type1 endometrial carcinoma: A matched
14. Wartko P, Sherman ME, Yang HP, Felix AS, Brinton LA, Trabert B. cohort study. Ann Surg Oncol. 2016;23:4361–4367.
Recent changes in endometrial cancer trends among menopausal-­ 32. Walker JL, Piedmonte MR, Spirtos NM, et  al. Laparoscopy com-
age U.S. women. Cancer Epidemiol. 2013;37:374–377. pared with laparotomy for comprehensive surgical staging of uter-
15. Kurman RJ, Kaminski PF, Norris HJ. The behavior of endometrial ine cancer: Gynecologic Oncology Group Study LAP2. J Clin Oncol.
hyperplasia. A long-­term study of “untreated” hyperplasia in 170 2009;27:5331–5336.
patients. Cancer. 1985;56:403–412. 33. Mourits MJ, Bijen CB, Arts HJ, et  al. Safety of laparoscopy versus
16. Edmondson RJ, Crosbie EJ, Nickkho-Amiry M, et  al. Markers of laparotomy in early-­stage endometrial cancer: A randomised trial.
the p53 pathway further refine molecular profiling in high-­risk Lancet Oncol. 2010;11:763–771.
endometrial cancer: A TransPORTEC initiative. Gynecol Oncol. 34. Obermair A, Janda M, Baker J, et al. Improved surgical safety after
2017;146:327–333. laparoscopic compared to open surgery for apparent early stage
17. Jacobs I, Gentry-Maharaj A, Burnell M, et al. Sensitivity of transvag- endometrial cancer: Results from a randomised controlled trial. Eur J
inal ultrasound screening for endometrial cancer in postmenopausal Cancer. 2012;48:1147–1153.
women: A case-­control study within the UKCTOCS cohort. Lancet 35. Galaal K, Bryant A, Fisher AD, Al-Khaduri M, Kew F, Lopes AD.
Oncol. 2011;12:38–48. Laparoscopy versus laparotomy for the management of early stage
18. Smith RA, Cokkinides V, Brawley OW. Cancer screening in the United endometrial cancer. Cochrane Database Syst Rev. 2012;(9):CD006655.
States, 2009: A review of current American Cancer Society guidelines 36. Koskas M, Jozwiak M, Fournier M, et  al. Long-­term oncological
and issues in cancer screening. CA Cancer J Clin. 2009;59:27–41. safety of minimally invasive surgery in high-­risk endometrial cancer.
19. Colombo N, Creutzberg C, Amant F, et  al. ESMO-­ESGO-­ESTRO Eur J Cancer. 2016;65:185–191.
Consensus Conference on Endometrial Cancer: Diagnosis, treatment 37. Bernardini MQ, Gien LT, Tipping H, Murphy J, Rosen BP. Surgical
and follow-­up. Ann Oncol. 2016;27:16–41. outcome of robotic surgery in morbidly obese patient with endo-
20. Karlsson B, Granberg S, Wikland M, et  al. Transvaginal ultraso- metrial cancer compared to laparotomy. Int J Gynecol Cancer.
nography of the endometrium in women with postmenopausal 2012;22:76–81.
|
48       Amant ET AL.

38. Cardenas-Goicoechea J, Shepherd A, Momeni M, et al. Survival anal- 58. Zeimet AG, Reimer D, Huszar M, et al. L1CAM in early-­stage type I
ysis of robotic versus traditional laparoscopic surgical staging for endometrial cancer: Results of a large multicenter evaluation. J Natl
endometrial cancer. Am J Obstet Gynecol. 2014;210:160.e161–160. Cancer Inst. 2013;105:1142–1150.
e111. 59. van der Putten LJ, Visser NC, van de Vijver K, et al. L1CAM expres-
39. Wright JD, Burke WM, Wilde ET, et al. Comparative effectiveness of sion in endometrial carcinomas: An ENITEC collaboration study. Br J
robotic versus laparoscopic hysterectomy for endometrial cancer. J Cancer. 2016;115:716–724.
Clin Oncol. 2012;30:783–791. 60. Bosse T, Nout RA, Stelloo E, et  al. L1 cell adhesion molecule is a
40. Chan JK, Sherman AE, Kapp DS, et  al. Influence of gynecologic strong predictor for distant recurrence and overall survival in early
oncologists on the survival of patients with endometrial cancer. J stage endometrial cancer: Pooled PORTEC trial results. Eur J Cancer.
Clin Oncol. 2011;29:832–838. 2014;50:2602–2610.
41. Roland PY, Kelly FJ, Kulwicki CY, Blitzer P, Curcio M, Orr JW Jr. The 61. Trovik J, Wik E, Werner HM, et al. Hormone receptor loss in endo-
benefits of a gynecologic oncologist: A pattern of care study for metrial carcinoma curettage predicts lymph node metastasis
endometrial cancer treatment. Gynecol Oncol. 2004;93:125–130. and poor outcome in prospective multicentre trial. Eur J Cancer.
42. Holloway RW, Abu-Rustum NR, Backes FJ, et  al. Sentinel lymph 2013;49:3431–3441.
node mapping and staging in endometrial cancer: A Society of 62. Zighelboim I, Goodfellow PJ, Gao F, et  al. Microsatellite instabil-
Gynecologic Oncology literature review with consensus recommen- ity and epigenetic inactivation of MLH1 and outcome of patients
dations. Gynecol Oncol. 2017;146:405–415. with endometrial carcinomas of the endometrioid type. J Clin Oncol.
43. Elit LM, O’Leary EM, Pond GR, Seow HY. Impact of wait times on 2007;25:2042–2048.
survival for women with uterine cancer. J Clin Oncol. 2014;32:27–33. 63. Stelloo E, Bosse T, Nout RA, et  al. Refining prognosis and iden-
44. Strohl AE, Feinglass JM, Shahabi S, Simon MA. Surgical wait time: tifying targetable pathways for high-­risk endometrial cancer; a
A new health indicator in women with endometrial cancer. Gynecol TransPORTEC initiative. Mod Pathol. 2015;28:836–844.
Oncol. 2016;141:511–515. 64. van der Putten LJM, Visser NCM, van de Vijver K, et al. Added value
45. Matsuo K, Opper NR, Ciccone MA, et  al. Time interval between of estrogen receptor, progesterone receptor, and L1 cell adhesion
endometrial biopsy and surgical staging for type I endometrial can- molecule expression to histology-­based endometrial carcinoma
cer: Association between tumor characteristics and survival out- recurrence prediction models: An ENITEC collaboration study. Int J
come. Obstet Gynecol. 2015;125:424–433. Gynecol Cancer. 2018;28:514–523.
46. Kilgore LC, Partridge EE, Alvarez RD, et al. Adenocarcinoma of the 65. Kommoss S, McConechy MK, Kommoss F, et  al. Final validation
endometrium: Survival comparisons of patients with and without of the ProMisE molecular classifier for endometrial carcinoma
pelvic node sampling. Gynecol Oncol. 1995;56:29–33. in a large population-­based case series. Ann Oncol. 2018;29:
47. Larson DM, Broste SK, Krawisz BR. Surgery without radiother- 1180–1188.
apy for primary treatment of endometrial cancer. Obstet Gynecol. 66. Poulsen HK, Jacobsen M, Bertelsen K, et al. Adjuvant radiation ther-
1998;91:355–359. apy is not necessary in the management of endometrial carcinoma
48. Cragun JM, Havrilesky LJ, Calingaert B, et al. Retrospective analysis stage I, low risk cases. Int J Gynecol Cancer. 1996;6:38–43.
of selective lymphadenectomy in apparent early-­stage endometrial 67. Aalders J, Abeler V, Kolstad P, Onsrud M. Postoperative external irra-
cancer. J Clin Oncol. 2005;23:3668–3675. diation and prognostic parameters in stage I endometrial carcinoma:
49. Kitchener H, Swart AM, Qian Q, Amos C, Parmar MK. Efficacy of Clinical and histopathologic study of 540 patients. Obstet Gynecol.
systematic pelvic lymphadenectomy in endometrial cancer (MRC 1980;56:419–427.
ASTEC trial): A randomised study. Lancet. 2009;373:125–136. 68. Creutzberg CL, van Putten WL, Koper PC, et al. Surgery and postop-
50. Benedetti Panici P, Basile S, Maneschi F, et  al. Systematic pelvic erative radiotherapy versus surgery alone for patients with stage-­1
lymphadenectomy vs. no lymphadenectomy in early-­stage endo- endometrial carcinoma: Multicentre randomised trial. PORTEC
metrial carcinoma: Randomized clinical trial. J Natl Cancer Inst. Study Group. Post operative radiation therapy in endometrial carci-
2008;100:1707–1716. noma. Lancet. 2000;355:1404–1411.
51. Aalders JG, Thomas G. Endometrial cancer–revisiting the impor- 69. Keys HM, Roberts JA, Brunetto VL, et al. A phase III trial of surgery
tance of pelvic and para aortic lymph nodes. Gynecol Oncol. with or without adjunctive external pelvic radiation therapy in inter-
2007;104:222–231. mediate risk endometrial adenocarcinoma: A Gynecologic Oncology
52. Todo Y, Kato H, Kaneuchi M, Watari H, Takeda M, Sakuragi N. Group study. Gynecol Oncol. 2004;92:744–751.
Survival effect of para-­aortic lymphadenectomy in endometrial 70. Blake P, Swart AM, Orton J, et al. Adjuvant external beam radiother-
cancer (SEPAL study): A retrospective cohort analysis. Lancet. apy in the treatment of endometrial cancer (MRC ASTEC and NCIC
2010;375:1165–1172. CTG EN.5 randomised trials): Pooled trial results, systematic review,
53. Bodurtha Smith AJ, Fader AN, Tanner EJ. Sentinel lymph node and meta-­analysis. Lancet. 2009;373:137–146.
assessment in endometrial cancer: A systematic review and meta-­ 71. Creutzberg CL, van Putten WL, Koper PC, et al. Survival after relapse
analysis. Am J Obstet Gynecol. 2017;216:459–476.e410. in patients with endometrial cancer: Results from a randomized trial.
54. Rossi EC, Kowalski LD, Scalici J, et al. A comparison of sentinel lymph Gynecol Oncol. 2003;89:201–209.
node biopsy to lymphadenectomy for endometrial cancer staging 72. Nout RA, Smit VT, Putter H, et al. Vaginal brachytherapy versus pelvic
(FIRES trial): A multicentre, prospective, cohort study. Lancet Oncol. external beam radiotherapy for patients with endometrial cancer of
2017;18:384–392. high-­intermediate risk (PORTEC-­2): An open-­label, non-­inferiority,
55. Soliman PT, Westin SN, Dioun S, et al. A prospective validation study randomised trial. Lancet. 2010;375:816–823.
of sentinel lymph node mapping for high-­risk endometrial cancer. 73. Nout RA, Putter H, Jurgenliemk-Schulz IM, et al. Quality of life after
Gynecol Oncol. 2017;146:234–239. pelvic radiotherapy or vaginal brachytherapy for endometrial can-
56. Talhouk A, McConechy MK, Leung S, et  al. A clinically applicable cer: First results of the randomized PORTEC-­2 trial. J Clin Oncol.
molecular-­based classification for endometrial cancers. Br J Cancer. 2009;27:3547–3556.
2015;113:299–310. 74. Ortoft G, Hansen ES, Bertelsen K. Omitting adjuvant radiotherapy
57. Talhouk A, McConechy MK, Leung S, et al. Confirmation of ProMisE: in endometrial cancer increases the rate of locoregional recurrences
A simple, genomics-­based clinical classifier for endometrial cancer. but has no effect on long-­term survival: The Danish Endometrial
Cancer. 2017;123:802–813. Cancer Study. Int J Gynecol Cancer. 2013;23:1429–1437.
Amant ET AL. |
      49

75. Kunneman M, Pieterse AH, Stiggelbout AM, et al. Treatment prefer- 92. Fleming GF, Brunetto VL, Cella D, et al. Phase III trial of doxorubicin
ences and involvement in treatment decision making of patients with plus cisplatin with or without paclitaxel plus filgrastim in advanced
endometrial cancer and clinicians. Br J Cancer. 2014;111:674–679. endometrial carcinoma: A Gynecologic Oncology Group Study. J Clin
76. Susumu N, Sagae S, Udagawa Y, et al. Randomized phase III trial of Oncol. 2004;22:2159–2166.
pelvic radiotherapy versus cisplatin-­based combined chemotherapy 93. Miller D, Filiaci V, Fleming G, et  al. Late-­breaking Abstract 1:
in patients with intermediate-­ and high-­risk endometrial cancer: Randomized phase III noninferiority trial of first line chemotherapy
A Japanese Gynecologic Oncology Group study. Gynecol Oncol. for metastatic or recurrent endometrial carcinoma: A Gynecologic
2008;108:226–233. Oncology Group study. Gynecol Oncol. 2012;125:771.
77. Maggi R, Lissoni A, Spina F, et al. Adjuvant chemotherapy vs radio- 94. van Wijk FH, Aapro MS, Bolis G, et al. Doxorubicin versus doxoru-
therapy in high-­risk endometrial carcinoma: Results of a randomised bicin and cisplatin in endometrial carcinoma: Definitive results of
trial. Br J Cancer. 2006;95:266–271. a randomised study (55872) by the EORTC Gynaecological Cancer
78. Randall M, Filiaci V, McMeekin D, et  al. A Phase 3 trial of pelvic Group. Ann Oncol. 2003;14:441–448.
radiation therapy versus vaginal cuff brachytherapy followed by 95. Thigpen JT, Brady MF, Homesley HD, et al. Phase III trial of doxoru-
paclitaxel/carboplatin chemotherapy in patients with high-­risk, bicin with or without cisplatin in advanced endometrial carcinoma: A
early-­stage endometrial cancer: A Gynecology Oncology Group gynecologic oncology group study. J Clin Oncol. 2004;22:3902–3908.
Study. Int J Rad Oncol Biol Phys. 2017;99:1313. 96. Akram T, Maseelall P, Fanning J. Carboplatin and paclitaxel for the
79. de Boer SM, Powell ME, Mileshkin L, et al. Adjuvant chemoradiother- treatment of advanced or recurrent endometrial cancer. Am J Obstet
apy versus radiotherapy alone for women with high-­risk endometrial Gynecol. 2005;192:1365–1367.
cancer (PORTEC-­3): Final results of an international, open-­label, 97. Hoskins PJ, Swenerton KD, Pike JA, et al. Paclitaxel and carboplatin,
multicentre, randomised, phase 3 trial. Lancet Oncol. 2018;19: alone or with irradiation, in advanced or recurrent endometrial can-
295–309. cer: A phase II study. J Clin Oncol. 2001;19:4048–4053.
80. Matei D, Filiaci VL, Randall M, Steinhoff M, DiSilvestro P, Moxley KM. 98. Sorbe B, Andersson H, Boman K, Rosenberg P, Kalling M. Treatment
A randomized phase III trial of cisplatin and tumor volume directed of primary advanced and recurrent endometrial carcinoma with a
irradiation followed by carboplatin and paclitaxel vs. carboplatin and combination of carboplatin and paclitaxel-­long-­term follow-­up. Int J
paclitaxel for optimally debulked, advanced endometrial carcinoma. Gynecol Cancer. 2008;18:803–808.
J Clin Oncol. 2017;35:5505. 99. Podzielinski I, Randall ME, Breheny PJ, et al. Primary radiation ther-
81. Martin-Hirsch PL, Lilford RJ, Jarvis GJ. Adjuvant progestagen ther- apy for medically inoperable patients with clinical stage I and II endo-
apy for the treatment of endometrial cancer: Review and meta-­ metrial carcinoma. Gynecol Oncol. 2012;124:36–41.
analyses of published randomised controlled trials. Eur J Obstet 100. Koskas M, Uzan J, Luton D, Rouzier R, Darai E. Prognostic factors
Gynecol Reprod Biol. 1996;65:201–207. of oncologic and reproductive outcomes in fertility-­sparing man-
82. COSA‐NZ‐UK Endometrial Cancer Study Groups. Adjuvant medroxy- agement of endometrial atypical hyperplasia and adenocarcinoma:
progesterone acetate in high-­risk endometrial cancer. Int J Gynecol Systematic review and meta-­analysis. Fertil Steril. 2014;101:785–
Cancer. 1998;8: 387–391. 794.e783.
83. Vandenput I, Trovik J, Leunen K, et al. Evolution in endometrial can- 101. Hwang JY, Kim DH, Bae HS, et al. Combined oral medroxyproges-
cer: Evidence from an immunohistochemical study. Int J Gynecol terone/levonorgestrel-­intrauterine system treatment for women
Cancer. 2011;21:316–322. with Grade 2 Stage IA endometrial cancer. Int J Gynecol Cancer.
84. Takano M, Ochi H, Takei Y, et  al. Surgery for endometrial cancers 2017;27:738–742.
with suspected cervical involvement: Is radical hysterectomy needed 102. Allsop JR, Preston J, Crocker S. Is there any value in the long-­term
(a GOTIC study)? Br J Cancer. 2013;109:1760–1765. follow up of women treated for endometrial cancer? Br J Obstet
85. Sartori E, Gadducci A, Landoni F, et  al. Clinical behavior of 203 Gynaecol. 1997;104:122.
stage II endometrial cancer cases: The impact of primary surgical 103. Salvesen HB, Akslen LA, Iversen T, Iversen OE. Recurrence of endo-
approach and of adjuvant radiation therapy. Int J Gynecol Cancer. metrial carcinoma and the value of routine follow up. Br J Obstet
2001;11:430–437. Gynaecol. 1997;104:1302–1307.
86. Mariani A, Webb MJ, Keeney GL, Calori G, Podratz KC. Role of 104. Agboola OO, Grunfeld E, Coyle D, Perry GA. Costs and benefits of
wide/radical hysterectomy and pelvic lymph node dissection in routine follow-­up after curative treatment for endometrial cancer.
endometrial cancer with cervical involvement. Gynecol Oncol. CMAJ. 1997;157:879–886.
2001;83:72–80. 105. Shumsky AG, Stuart GC, Brasher PM, Nation JG, Robertson DI,
87. Lee MH, Aquino-Parsons C, Hoskins PJ, Lim P, Kwon JS. Preoperative Sangkarat S. An evaluation of routine follow-­up of patients treated
radiotherapy for inoperable stage II endometrial cancer: Insights for endometrial carcinoma. Gynecol Oncol. 1994;55:229–233.
into improving treatment and outcomes. J Obstet Gynaecol Can. 106. Ackerman I, Malone S, Thomas G, Franssen E, Balogh J, Dembo A.
2013;35:635–639. Endometrial carcinoma–relative effectiveness of adjuvant irradiation
88. van der Steen-Banasik E, Christiaens M, Shash E, et  al. Systemic vs therapy reserved for relapse. Gynecol Oncol. 1996;60:177–183.
review: Radiation therapy alone in medical non-­operable endome- 107. Fung-Kee-Fung M, Dodge J, Elit L, Lukka H, Chambers A, Oliver T.
trial carcinoma. Eur J Cancer. 2016;65:172–181. Follow-­up after primary therapy for endometrial cancer: A system-
89. Vargo JA, Boisen MM, Comerci JT, et al. Neoadjuvant radiotherapy atic review. Gynecol Oncol. 2006;101:520–529.
with or without chemotherapy followed by extrafascial hysterec- 108. Kew FM, Roberts AP, Cruickshank DJ. The role of routine fol-
tomy for locally advanced endometrial cancer clinically extending to low-­up after gynecological malignancy. Int J Gynecol Cancer.
the cervix or parametria. Gynecol Oncol. 2014;135:190–195. 2005;15:413–419.
90. Barlin JN, Puri I, Bristow RE. Cytoreductive surgery for advanced 109. Salani R, Nagel CI, Drennen E, Bristow RE. Recurrence patterns and
or recurrent endometrial cancer: A meta-­analysis. Gynecol Oncol. surveillance for patients with early stage endometrial cancer. Gynecol
2010;118:14–18. Oncol. 2011;123:205–207.
91. Vandenput I, Van Calster B, Capoen A, et  al. Neoadjuvant chemo- 110. Sartori E, Pasinetti B, Carrara L, Gambino A, Odicino F, Pecorelli
therapy followed by interval debulking surgery in patients with S. Pattern of failure and value of follow-­up procedures in endo-
serous endometrial cancer with transperitoneal spread (stage IV): A metrial and cervical cancer patients. Gynecol Oncol. 2007;107(1
new preferred treatment? Br J Cancer. 2009;101:244–249. Suppl.1):S241–S247.
|
50       Amant ET AL.

111. Berchuck A, Anspach C, Evans AC, et al. Postsurgical surveillance of 115. Moline J, Eng C. Equality in lynch syndrome screening: Why should
patients with FIGO stage I/II endometrial adenocarcinoma. Gynecol we hold patients with endometrial cancer to a different standard? J
Oncol. 1995;59:20–24. Clin Oncol. 2014;32:2277.
112. Nordin AJ. Mode of detection of recurrent gynecological malig- 116. Wiltink LM, Nout RA, Fiocco M, et al. No increased risk of second
nancy: Does routine follow-­up delay diagnosis and treatment? Int J cancer after radiotherapy in patients treated for rectal or endome-
Gynecol Cancer. 2006;16:1746–1748. trial cancer in the randomized TME, PORTEC-­1, and PORTEC-­2 tri-
113. Leeson SC, Beaver K, Ezendam NPM, et al. The future for follow-­up of als. J Clin Oncol. 2015;33:1640–1646.
gynaecological cancer in Europe. Summary of available data and overview 117. Randall ME, Filiaci VL, Muss H, et al. Randomized phase III trial of
of ongoing trials. Eur J Obstet Gynecol Reprod Biol. 2017;210:376–380. whole-­abdominal irradiation versus doxorubicin and cisplatin che-
114. Hampel H, Frankel W, Panescu J, et al. Screening for Lynch syndrome motherapy in advanced endometrial carcinoma: A Gynecologic
(hereditary nonpolyposis colorectal cancer) among endometrial can- Oncology Group Study. J Clin Oncol. 2006;24:36–44.
cer patients. Can Res. 2006;66:7810–7817.
DOI: 10.1002/ijgo.12613

FIGO CANCER REPORT 2018

Uterine sarcomas

Nomonde Mbatani1,2 | Alexander B. Olawaiye3 | Jaime Prat4,*

1
Department of Obstetrics and
Gynecology, Groote Schuur Hospital/ Abstract
University of Cape Town, Cape Town, Uterine sarcomas account for approximately 3%–7% of all uterine cancers. Since
South Africa
2
carcinosarcomas are currently classified as metaplastic carcinomas, leiomyosarcomas
South African Medical Research Council/
University of Cape Town Gynaecological remain the most common subtype. Exclusion of several histologic variants of
Cancer Research Centre (SA MRC/UCT
leiomyoma, as well as atypical smooth muscle tumors (so-­called “smooth muscle
GCRC), Cape Town, South Africa
3 tumors of uncertain malignant potential”), has highlighted that the vast majority
Department of Obstetrics, Gynecology
and Reproductive Sciences, University of of leiomyosarcomas are high-­grade tumors associated with poor prognosis even
Pittsburgh School of Medicine, Pittsburgh,
when apparently confined to the uterus. Low-­grade endometrial stromal sarcomas
PA, USA
4
Department of Pathology, Hospital de la are indolent tumors associated with long-­term survival. High-­grade endometrial
Santa Creu i Sant Pau, Autonomous University stromal sarcomas and undifferentiated endometrial sarcomas behave more
of Barcelona, Barcelona, Spain
aggressively than tumors showing nuclear uniformity. Adenosarcomas have a
*Correspondence favorable prognosis except for tumors showing myometrial invasion or sarcomatous
Jaime Prat, Autonomous University of
Barcelona, Medical School - UD Sant Pau, overgrowth. The prognosis for carcinosarcomas (which are considered here
Barcelona, Spain. in a postscript fashion) is usually worse than that for grade 3 endometrial
Email: jpratdl@gmail.com
carcinomas. Tumor stage is the single most important prognostic factor for
uterine sarcomas.

KEYWORDS
Adenosarcomas; Carcinosarcomas; Endometrial stromal sarcomas; FIGO Cancer Report;
Leiomyosarcomas; Undifferentiated endometrial sarcomas; Uterine sarcomas

1 |  INTRODUCTION uterine sarcomas, as well as in the separate section of “mixed epithelial
and mesenchymal tumors” of the 2014 WHO classification.3
Uterine sarcomas account for approximately 1% of all female genital Tumor stage is the single most important prognostic factor. In the
tract malignancies and 3%–7% of all uterine cancers.1 Their rarity and past, uterine sarcomas were staged using a staging system proposed
histopathological diversity have contributed to the lack of consensus in 1988 for endometrial carcinoma. This has not proven satisfactory
2
on risk factors for poor outcome and optimal treatment. and, in 2009, a new FIGO staging system was developed for uterine
Histologically, uterine sarcomas were classified initially into carci- sarcomas (Table 1).4 The new staging system has two divisions, one for
nosarcomas (malignant mesodermal mixed tumors), accounting for 50% leiomyosarcoma and endometrial stromal sarcoma (ESS), and one for
of cases, leiomyosarcomas (30%), endometrial stromal sarcomas (15%), adenosarcoma. Carcinosarcoma is now staged using the endometrial
and undifferentiated sarcomas (5%). Subsequently, carcinosarcoma has carcinoma staging system.4
been reclassified, largely based on its spreading pattern, as a dediffer- Prolonged use of tamoxifen, a uterine estrogen receptor agonist,
entiated or metaplastic form of endometrial carcinoma. However, as it is associated with a three times risk of sarcoma development.5 There
behaves more aggressively than the usual type of endometrial carci- have been reported cases of radiation-­induced sarcomas occurring
noma, carcinosarcoma is still included in most retrospective studies of long after treatment for other cancers.6

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

Int J Gynecol Obstet 2018; 143 (Suppl. 2): 51–58 wileyonlinelibrary.com/journal/ijgo  |  51
|
52       Mbatani ET AL.

Neither preoperative imaging with ultrasonography nor PET scans T A B L E   1   FIGO staging for uterine sarcomas.
is capable of differentiating between benign or malignant smooth
Stage Definition
muscle masses. The use of diffusion-­weighted magnetic resonance
imaging (DWI) for tumor location and characterization has been sug- Leiomyosarcomas and endometrial stromal sarcomas

gested, but is yet to be validated. I Tumor limited to uterus

Patients with carcinosarcomas and adenosarcomas tend to be IA Less than 5 cm


much older than patients with other sarcomas. IB More than 5 cm
II Tumor extends beyond the uterus,
within the pelvis
2 |  LEIOMYOSARCOMAS IIA Adnexal involvement
IIB Involvement of other pelvic tissues
III Tumor invades abdominal tissues (not
2.1 | Clinical features
just protruding into the abdomen)
After excluding carcinosarcoma, leiomyosarcoma has become the IIIA One site
most common subtype of uterine sarcoma even though it accounts for IIIB More than one site
only 1%–2% of uterine malignancies.2 Approximately 1 in every 800 IIIC Metastasis to pelvic and/or para-­aortic
smooth muscle tumors of the uterus is a leiomyosarcoma.2 It occurs lymph nodes
in women over 40 years of age who usually present with abnormal IV
vaginal bleeding (56%), a palpable pelvic mass (54%), and/or pelvic IVA Tumor invades bladder and/or rectum
pain (22%).2 Signs and symptoms resemble those of the far more com- IVB Distant metastasis
mon leiomyoma and preoperative distinction between the two tumors Adenosarcomas
may be difficult. Malignancy should be suspected by the presence of
I Tumor limited to uterus
tumor growth in postmenopausal women who are not using hormonal
IA Tumor limited to endometrium/
replacement therapy, although it is rare for a leiomyosarcoma to pre- endocervix with no myometrial invasion
sent as a rapidly growing tumor.
IB Less than or equal to half myometrial
invasion
IC More than half myometrial invasion
2.2 | Pathological features
II Tumor extends to the pelvis
Leiomyosarcomas are either single masses or, when associated with
IIA Adnexal involvement
leiomyomas, the largest mass. They are typically voluminous tumors
IIB Tumor extends to extrauterine pelvic
with a mean diameter of 10 cm (only 25% of cases measure less than tissue
5 cm). The cut surface is typically soft, bulging, fleshy, necrotic, hemor-
III Tumor invades abdominal tissues (not
rhagic, and lacks the prominent whorled appearance of leiomyomas. just protruding into the abdomen)
The histopathologic diagnosis of leiomyosarcoma is usually straightfor- IIIA One site
ward as most clinically malignant smooth muscle tumors of the uterus IIIB More than one site
exhibit the constellation of hypercellularity, severe nuclear atypia, and
IIIC Metastasis to pelvic and/or para-­aortic
high mitotic rate generally exceeding 15 mitotic figures per 10 high-­ lymph nodes
power-­fields (MF/10 HPF) (Fig. 1).3 Moreover, one or more supportive IV
clinicopathologic features such as peri-­ or postmenopausal age, extrau-
IVA Tumor invades bladder and/or rectum
terine extension, large size (over 10 cm), infiltrating border, necrosis,
IVB Distant metastasis
and atypical mitotic figures are frequently present. However, epithe-
Carcinosarcomas
lioid and myxoid leiomyosarcomas are two rare variants that may be
Carcinosarcomas should be staged as carcinomas of the
difficult to recognize microscopically as their pathologic features differ
endometrium
from those of ordinary spindle cell leiomyosarcomas. In both tumor
Simultaneous tumors of the uterine corpus and ovary/pelvis in association
types nuclear atypia is usually mild and the mitotic rate often less than
with ovarian/pelvic endometriosis should be classified as independent
3 MF/10 HPF.3 Necrosis may be absent in epithelioid leiomyosarcomas ­primary tumors.
and myxoid leiomyosarcomas are often hypocellular. In the absence of
severe cytologic atypia and high mitotic activity, both tumors are diag- also atypical smooth muscle tumors (so-­called smooth muscle tumors
nosed as sarcomas based on their infiltrative borders. of uncertain malignant potential [STUMPs]) (Box 3). Application
The minimal pathological criteria for the diagnosis of leiomyosar- of the WHO diagnostic criteria3 has allowed distinguishing these
coma are more problematic and, in such cases, the differential diagno- unusual histologic variants of leiomyoma frequently misdiagnosed as
sis includes, not only benign smooth muscle tumors that exhibit variant “well-­differentiated” or “low-­grade” leiomyosarcomas in the past. In
histologic features and unusual growth patterns (Boxes 1 and 2), but a population-­based study of uterine sarcomas from Norway,6 of 356
Mbatani ET AL. |
      53

2.3 | Immunohistochemistry and molecular biology


Leiomyosarcomas usually express smooth muscle markers such as
desmin, h-­caldesmon, smooth muscle actin, and histone deacetylase
8 (HDCA8). However, epithelioid and myxoid leiomyosarcomas may
show lesser degrees of immunoreaction for these markers.3 Also, leio-
myosarcomas are often immunoreactive for CD10 (mainly considered
a marker of endometrial stromal differentiation) and epithelial markers
including keratin and EMA (the latter being more frequently positive
in the epithelioid variant).3 Conventional leiomyosarcomas express
estrogen receptors, progesterone receptors, and androgen receptors
in 30%–40% of cases. Whereas a variable proportion of uterine leio-
myosarcomas has been reported as being immunoreactive for c-­KIT,
no c-KIT mutations have been identified.3
The levels of Ki67 are higher in uterine leiomyosarcomas com-
F I G U R E   1   Leiomyosarcoma. pared with benign smooth muscle tumors. Overexpression of p16 has
been described in uterine leiomyosarcomas and may prove to be a
useful adjunct immunomarker for distinguishing between benign and
malignant uterine smooth muscle tumors.7
Box 1 Leiomyoma variants that may mimic malignancy. The vast majority of uterine leiomyosarcomas are sporadic. Patients
• Mitotically active leiomyoma with germline mutations in fumarate hydratase are believed to be at
• Cellular leiomyoma increased risk for developing uterine leiomyosarcomas as well as uterine
• Hemorrhagic leiomyoma and hormone-induced changes leiomyomas.8 The oncogenic mechanisms underlying the development of
• Leiomyoma with bizarre nuclei (atypical leiomyoma) uterine leiomyosarcomas remain elusive. Overall, uterine leiomyosarcoma
• Myxoid leiomyoma is a genetically unstable tumor that demonstrates complex structural
• Epithelioid leiomyoma chromosomal abnormalities and highly disturbed gene regulation, which
• Leiomyoma with massive lymphoid infiltration likely reflects the end-­state of accumulation of multiple genetic defects.

2.4 | Prognosis
Box 2 Smooth muscle proliferations with unusual growth Leiomyosarcomas diagnosed according to the WHO criteria 3
are
patterns. associated with poor prognosis even when confined to the uterus at
• Disseminated peritoneal leiomyomatosis the time of diagnosis.6,9 Recurrence rate ranges from 53% to 71%.10,11
• Benign metastasizing leiomyoma First recurrences occur in the lungs in 40% of patients and in the pel-
• Intravenous leiomyomatosis vis in only 13%.12 Overall 5-­year survival rate ranges from 15% to
• Lymphangioleiomyomatosis 25% with a median survival of only 10 months in one study.13 In the
Norwegian series, 148 patients with leiomyosarcomas limited to the
uterus had a 5-­year survival of 51% at Stage I and 25% at Stage II (by
the 1988 FIGO staging classification). All patients with tumor spread
Box 3 Atypical smooth muscle tumors (so-­called smooth outside the pelvis died within 5 years.6
muscle tumors of uncertain malignant potential There has been no consistency among various studies regarding
[STUMP]). correlation between survival and patient age, clinical stage, tumor
• Tumor cell necrosis in a typical leiomyoma size, type of border (pushing vs infiltrative), presence or absence of
• Necrosis of uncertain type with ≥10 MF/10 HPFs, or marked necrosis, mitotic rate, degree of nuclear pleomorphism, and vascu-
diffuse atypia lar invasion.3 However, one study,14 found tumor size to be a major
• Marked diffuse or focal atypia with borderline mitotic counts prognostic parameter: five of eight patients with tumors less than
• Necrosis difficult to classify 5 cm in diameter survived, whereas all patients with tumors greater
than 5 cm in diameter died. In a series of 208 uterine leiomyosarco-
mas,2 the only other parameters predictive of prognosis were tumor
tumors classified initially as leiomyosarcomas, the diagnosis was con- grade and stage. In the report from Norway,6 including 245 leiomyo-
firmed in only 259 (73%) cases, whereas 97 (27%) were excluded on sarcomas confined to the uterus, tumor size and mitotic index were
review and reclassified, according to WHO criteria, as leiomyomas or significant prognostic factors and allowed for separation of patients
leiomyoma variants. into three risk groups with marked differences in prognosis.
|
54       Mbatani ET AL.

Ancillary parameters including p53, p16, Ki 67, and Bcl-­2 have or exclusively intramural neoplasms and are divided into benign and
9
been used in leiomyosarcomas to try to predict outcome. It is not malignant based on the type of tumor margin: well-­circumscribed
clear whether they act independently of stage. However, a recent tumors are benign stromal nodules, whereas those exhibiting myo-
study revealed that the combination of tumor size, mitotic index, Ki67, metrial invasion and permeation of myometrial lymphovascular
and Bcl-­2 protein expression allows two groups of leiomyosarcomas to spaces are sarcomas.3 Endometrial sarcomas are further classified
be distinguished, with different survival: tumors greater than or equal by the latest WHO classification, based on how closely the tumor
to 10 cm in diameter, with greater than or equal to 20 MF/10 HPF, resembles proliferative-­type endometrial stroma, into the following
greater than or equal to 10% immunoreactive nuclei for Ki67, and three main categories: (1) low-­grade endometrial stromal sarcoma,
negative for Bcl-­2 had worse prognosis than smaller leiomyosarcomas (2) high-­grade endometrial stromal sarcoma, and (3) undifferentiated
with less than or equal to 20 MF/10 HPF, less than or equal to 10% endometrial sarcoma.3
14
immunoreactive nuclei for Ki67, and positive or negative for Bcl-­2.

4.1 | Low-­grade endometrial stromal sarcoma


2.5 | Treatment
Low-­grade endometrial stromal sarcomas frequently occur in
Treatment of leiomyosarcomas includes total abdominal hysterectomy women between 40 and 55 years of age and more than 50% of
and debulking of the tumor if present outside the uterus. Removal of the patients are premenopausal.22 Some cases have been reported
ovaries and lymph node dissection remain controversial as metastases in women with ovarian polycystic disease, and after estrogen use
to these organs occur in only a small percentage of cases and are fre- or tamoxifen therapy.22 Patients commonly present with abnor-
2
quently associated with intra-­abdominal disease. Ovarian preservation mal uterine bleeding, pelvic pain, and dysmenorrhea, but as many
may be considered in premenopausal patients with early-­stage leiomyo- as 25% are asymptomatic.14 At presentation, extrauterine pelvic
2
sarcomas. Lymph node metastases have been identified in 6.6% and extension, most commonly involving the ovary, is found in up to
11% in two series of patients with leiomyosarcoma who underwent lym- one-­third of patients.22,23
2,15
phadenectomy. In the first series, the 5-­year disease-­specific survival Microscopically, endometrial stromal sarcomas consist of well-­
rate was 26% in patients who had positive lymph nodes compared with differentiated endometrial stromal cells exhibiting only mild nuclear
64.2% in patients who had negative lymph nodes (P<0.001).16 In com- atypia and characteristically invade the lymphovascular spaces of the
pletely resected organ-­confined disease (Stages I and II), the influence myometrium (Fig. 2). Tumor cell necrosis is rarely seen.
of adjuvant systemic therapy or radiotherapy on survival is uncertain. The tumor cells are strongly immunoreactive for CD10, usually
Docetaxel/gemcitabine, doxorubicin, and ifosfamide are all reasonable positive for smooth-­muscle actin and less frequently for desmin (30%),
options for advanced or recurrent disease with response rates ranging but they are negative for h-­caldesmon and HDAC8. Estrogen recep-
16–18
from 17% to 36%. Some tumors may respond to hormonal treat- tors (only alpha isoform), progesterone receptors, androgen receptors,
ment.19 Targeted therapies such as trabectedin and olaratumab have and WT-­1 are typically positive. Nuclear beta-­catenin expression has
been investigated as treatment in advanced stage or metastatic leio- been shown in up to 40% of low-­grade endometrial stromal sarcomas.
myosarcoma with some appreciable disease control.20,21 The most common cytogenetic abnormality of low-­grade endome-
trial stromal sarcomas is a recurrent translocation involving chromo-
somes 7 and 17 t(7;17) (p15;q21)], which results in a fusion between
3 |  ATYPICAL SMOOTH MUSCLE TUMORS JAZF1 and SUZ12 (formerly designated as JJAZ1).24 The fusion can be
(STUMP)

Uterine smooth muscle tumors that show some worrisome histologic


features (i.e. necrosis, nuclear atypia, or mitoses), but do not meet all
diagnostic criteria for leiomyosarcoma, fall into the category of atypi-
cal smooth muscle tumors (STUMP) (Box 2).3 This diagnosis, however,
should be used sparingly and every effort should be made to classify
a smooth muscle tumor into a specific category when possible.3 Most
tumors classified as STUMP have been associated with favorable prog-
nosis and, in these cases, only follow-­up of the patients is recommended.

4 |  ENDOMETRIAL STROMAL TUMORS

Endometrial stromal tumors account for less than 1% of all uterine


tumors1; nevertheless, they represent the second most common
category of mesenchymal uterine tumors. They are predominantly F I G U R E   2   Low-­grade endometrial stromal sarcoma.
Mbatani ET AL. |
      55

detected by fluorescence in situ hybridization as well as by reverse


4.3 | Undifferentiated endometrial sarcoma
transcriptase–polymerase chain reaction.
Low-­grade endometrial stromal sarcomas are indolent tumors This tumor is rare. Patients are typically postmenopausal (mean age
with a favorable prognosis.22 Tumor behavior is characterized by late is 60 years) and have postmenopausal bleeding or signs/symptoms
recurrences even in patients with Stage I disease; thus, long-­term fol- secondary to extrauterine spread.29 Approximately 60% of patients
low-­up is required. About one-­third of patients develop recurrences, present with high-­stage disease (Stage III/IV). The diagnosis of undif-
most commonly in the pelvis and abdomen, and less frequently in ferentiated endometrial sarcoma is applied to tumors that exhibit
the lungs and vagina.22 Stage of the tumor is the most significant myometrial invasion, severe nuclear pleomorphism, high mitotic activ-
prognostic factor. Surgical stage higher than Stage I is a univariate ity and/or tumor cell necrosis, and lack smooth muscle or endometrial
predictor of unfavorable outcome. Five-­year survival for patients stromal differentiation.3 The histological appearance of this tumor is
with Stages I and II tumors is 90% compared with 50% for Stages more like the mesenchymal elements of a carcinosarcoma than a typi-
III and IV.25 cal endometrial stromal tumor. It is variably CD10 positive and typi-
Treatment of low-­grade endometrial stromal sarcomas is cally estrogen receptor and progesterone weakly positive or negative.
largely surgical in the form of hysterectomy and bilateral salpingo-­ Undifferentiated endometrial sarcomas are highly aggressive tumors
oophorectomy. The tumors are often sensitive to hormones and it has that are associated with a very poor prognosis (less than 2 years’
been shown that patients retaining their ovaries have a much higher survival).29 Patients should be treated by hysterectomy and bilateral
26
risk of recurrence (up to 100%). Lymph node dissection does not salpingo-­oophorectomy and adjuvant radiation and/or chemotherapy.
seem to have a role in the treatment of these tumors. Patients may
also receive adjuvant radiation or hormonal treatment with progesta-
tional agents or aromatase inhibitors. Post-­treatment hormone (estro- 5 | ADENOSARCOMA
gen) replacement therapy is discouraged
Müllerian adenosarcoma is a mixed tumor of low malignant potential that
shows an intimate admixture of benign glandular epithelium and low-­
4.2 | High-­grade endometrial stromal sarcoma
grade sarcoma, usually of endometrial stromal type. It represents between
These rare tumors have features that are intermediate between 5% and 10% of all uterine sarcomas. The tumor occurs mainly in the uterus
low-­grade endometrial stromal sarcomas and undifferentiated sarco- of postmenopausal women (average 58 years) but also in adolescents and
mas.27 Patients range in age from 28 to 67 years (mean 50 years) and young adults (30%).30 Most adenosarcomas arise from the endometrium,
usually present with abnormal vaginal bleeding, an enlarged uterus, including the lower uterine segment, but rare tumors develop in the
or a pelvic mass.28 endocervix (5%–10% of cases) and in extrauterine locations.31
The tumors may appear as intracavitary polypoid or mural masses. Adenosarcomas are polypoid tumors of approximately 5–6 cm in
They range in size up to 9 cm (median 7.5 cm) and often show extra- maximum diameter (range, 1–20 cm) that typically fill and distend the
uterine extension at the time of diagnosis. The cut surface is fleshy uterine cavity. Adenosarcomas with sarcomatous overgrowth tend to
with extensive areas of necrosis and hemorrhage. Microscopically, be larger with a fleshy, hemorrhagic, and necrotic cut surface. They
they consist predominantly of high-­grade round-­cells that are some- invade the myometrium more often than conventional adenosarcomas.
times associated with a low-­grade spindle cell component that is Microscopically, the stroma typically concentrates around the glands
most commonly fibromyxoid.28 Mitotic activity is striking and typi- forming periglandular cuffs (Fig. 3). Well-­differentiated tumors may
cally greater than 10 per 10 HPF. Necrosis is usually present. Rarely, exhibit only mild nuclear atypia and very few or no mitoses in the stro-
areas of conventional low-­grade endometrial stromal sarcoma are mal component. However, the presence of hypercellular periglandular
seen. High-­grade endometrial stromal sarcomas are CD10, estrogen cuffs helps to distinguish adenosarcoma from its rarer benign counter-
receptor, and progesterone receptor negative but show strong diffuse part, the adenofibroma.31 Heterologous mesenchymal elements, usually
cyclin D1 immunoreactivity (>70% nuclei). They are also typically c-­Kit rhabdomyosarcoma, are found in 10%–15% of cases. Vaginal or pelvic
positive but DOG1 negative. High-­grade endometrial stromal sarcoma recurrence occurs in approximately 25%–30% of cases at 5 years and
typically harbors the YWHAE-FAM22 genetic fusion as a result of is associated almost exclusively with myometrial invasion and sarcoma-
t(10;17) (q22;p13). tous overgrowth.30,31 Myometrial invasion is found in approximately 15%
These tumors appear to have a prognosis that is intermediate, of cases, but deep invasion in only 5%.30,31 Sarcomatous overgrowth,
between low-­grade endometrial stromal sarcomas and undifferenti- defined as the presence of pure sarcoma, usually of high-­grade and with-
ated uterine sarcomas.28 Compared with low-­grade endometrial stro- out a glandular component, occupying at least 25% of the tumor, has
mal sarcomas, patients with high-­grade endometrial stromal sarcomas been reported in 8%–54% of uterine adenosarcomas.30,31
have earlier and more frequent recurrences (often <1 year) and are Whereas immunoreactions for cell proliferation markers (Ki-­67
more likely to die of disease. Advanced or recurrent tumors should be and P53) are stronger in adenosarcomas with sarcomatous over-
treated aggressively with a combination of radiation and chemother- growth than in typical adenosarcomas, the expression of markers of
apy as they do not respond to conventional treatment for low-­grade cell differentiation (CD10 and PR) is higher in typical adenosarcomas
endometrial stromal sarcomas.28 than in adenosarcomas with sarcomatous overgrowth.31
|
56       Mbatani ET AL.

cancers with vaginal bleeding. Another typical presentation of carci-


nosarcoma is in the form of a polypoid mass that protrudes through
the cervical os.
The epithelial component is serous, or high-­grade carcinoma not
otherwise specified, in about two-­thirds of cases, and endometrioid
carcinoma in approximately one-­third.32 In a recent study, 10% of the
carcinomatous components were FIGO grade 1, 10% grade 2, and 80%
grade 3.32 The homologous components of carcinosarcoma are usually
spindle cell sarcoma without obvious differentiation; many resemble
fibrosarcomas or pleomorphic sarcomas. Almost all are high-­grade sar-
comas. The most common heterologous elements are malignant car-
tilage or skeletal muscle constituting something that resembles either
pleomorphic rhabdomyosarcoma or embryonal rhabdomyosarcoma.1
Carcinosarcomas are highly aggressive tumors—far more aggres-
sive than usual endometrial carcinomas. The overall 5-­year survival
F I G U R E   3   Adenosarcoma. for patients with carcinosarcoma is around 30% and for those with
Stage I disease (confined to the uterus) it is approximately 50%.1,33–35
This is in contrast with other high-­grade endometrial cancers for which
Except when associated with myometrial invasion or sarcomatous
5-­year survival in Stage I disease is approximately 80% or higher.36,37
overgrowth, the prognosis of adenosarcoma is far more favorable than
As a result of its aggressive behavior, adjuvant systemic therapy con-
that of carcinosarcoma; however, about 25% of patients with ade-
sisting of ifosfamide, taxol, and platinum agents is routinely given,
nosarcoma ultimately die of their disease.30 Recurrences are usually
even when the disease is in its early stage.38 Adjuvant radiotherapy is
composed exclusively of mesenchymal elements. Distant metastases,
also commonly utilized.
which occur in 5% of cases, are almost always composed of pure sar-
In carcinosarcomas, there is general agreement that surgical stage
coma (70%). The treatment of choice is total abdominal hysterectomy
is the most important prognostic indicator regardless of how the
with bilateral salpingo-­oophorectomy.
patient was staged. One study found that the presence of heterolo-
gous elements is a poor prognostic factor in patients with FIGO Stage
I tumors.32 Other factors proposed include the histologic grade of the
6 |  CARCINOSARCOMA
carcinomatous and sarcomatous elements, the percentage of tumor
with sarcomatous differentiation, depth of myometrial invasion, and
Carcinosarcoma, also referred to as “malignant müllerian mixed
presence of lymphovascular invasion.1,33–35
tumor,” is a biphasic neoplasm composed of distinctive and separate,
but admixed, malignant-­appearing epithelial and mesenchymal ele-
ments (Fig. 4). The mean age of patients with carcinosarcoma is in the 6.1 | Treatment of carcinosarcomas
seventh decade, but the age range spans from the fourth through the
Primary surgery for early disease includes a hysterectomy, bilateral
ninth decades.32 The disease usually presents like other endometrial
salpingo-­oophorectomy, and pelvic node dissection as the tumor
spread pattern is similar to high-­grade endometrial carcinomas.
Omentectomy is also advocated by some. Complete cytoreduction
should be the aim of surgery, as this may be associated with an overall
survival benefit.
Combination chemotherapy seems to result in fewer recurrences
than whole body irradiation.39 Patients with carcinosarcomas, how-
ever, tend to be elderly with comorbidities. The ideal agents still need
to be established. The results of the Gynecologic Oncology Group 261
study, which aims to compare ifosfamide/paclitaxel versus carbopla-
tin/paclitaxel combinations in patients with advanced stage or recur-
rent carcinosarcoma, are awaited. Radiotherapy is only able to control
pelvic disease.40

6.2 | Follow-­up of sarcomas
Follow-­up should be determined by risk of recurrence. As metasta-
F I G U R E   4   Carcinosarcoma. sis to the lungs is common, efforts must be made to rule these out
Mbatani ET AL. |
      57

remembering that early lesions tend to be asymptomatic but resect- 12. Jones MW, Norris HJ. Clinicopathologic study of 28 uterine leiomyo-
able. Low-­grade sarcoma patients may be followed for local relapse sarcomas with metastasis. Int J Gynecol Pathol. 1995;14:243–249.
13. Kapp DS, Shin JY, Chan JK. Prognostic factors and survival in 1396
every 4–6 months for the first 3–5 years, then yearly. High-­grade
patients with uterine leiomyosarcomas: Emphasis on impact of
tumors can be followed-­up every 3–4 months for the first 2–3 years, lymphadenectomy and oophorectomy. Cancer. 2008;112:820–830.
twice a year for the next 2–3 years, and then annually. 14. D’Angelo E, Espinosa I, Ali R, et al. Uterine leiomyosarcomas: Tumor
size, mitotic index, and biomarkers Ki67, and Bcl-­2 identify two
groups with different prognosis. Gynecol Oncol. 2011;121:328–333.
AUT HOR CONTRI B UTI O N S 15. Hensley ML, Ishill N, Soslow R, et  al. Adjuvant gemcitabine plus
docetaxel for completely resected stages I-­IV high grade uterine
JP wrote the manuscript and prepared the tables and figures. NB leiomyosarcoma: Results of a prospective study. Gynecol Oncol.
updated the treatment of uterine sarcomas and included references. 2009;112:563–567.
ABO updated the treatment of leiomyosarcomas. 16. Sutton GP, Blessing JA, Barrett RJ, et  al. Phase II trial of ifosfamide
and mesna in leiomyosarcoma of the uterus: A Gynecologic Oncology
Group study. Am J Obstet Gynecol. 1992;166:556–559.
ACKNOWLE DG ME NTS 17. Hensley ML, Blessing JA, Mannel R, et al. Fixed-­dose rate gemcitabine
plus docetaxel as first-­line therapy for metastatic uterine leiomyosar-
This chapter updates the information published in the FIGO Cancer coma: A Gynecologic Oncology Group phase II trial. Gynecol Oncol.
Report 2015 (Prat J, Mbatani N. Uterine sarcomas. Int J Gynecol 2008;109:329–334.
18. Seddon B, Strauss SJ, Whelan J, et al. Gemcitabine and docetaxel versus
Obstet. 2015;131(Suppl.2):S105–110), with approval granted by the
doxorubicin as first-­line treatment in previously untreated advanced
original authors. unresectable or metastatic soft-­tissue sarcomas (GeDDiS): A ran-
domised controlled phase 3 trial. Lancet Oncol. 2017;18:1397–1410.
19. Hardman MP, Roman JJ, Burnett AF, et  al. Metastatic uterine leio-
CO NFLI CTS OF I NTE RE S T myosarcoma regression using an aromatase inhibitor. Obstet Gynecol.
2007;110:518–520.
The authors have no conflicts of interest to declare.
20. Monk BJ, Blessing JA, Street DG, et al. A phase II evaluation of trabec-
tedin in the treatment of advanced, persistent, or recurrent uterine
leiomyosarcoma: A gynecologic oncology group study. Gynecol Oncol.
REFERENCES
2012;124:48–52.
1. Major FJ, Blessing JA, Silverberg SG, et al. Prognostic factors in early-­ 21. Tap WD, Jones RL, Van Tine BA, et  al. Olaratumab and doxorubi-
stage uterine sarcoma. A Gynecologic Oncology Group study. Cancer. cin versus doxorubicin alone for treatment of soft-­tissue sarcoma:
1993;71:1702–1709. An open-­label phase 1b and randomised phase 2 trial. Lancet.
2. Giuntoli RL 2nd, Metzinger DS, DiMarco CS, et  al. Retrospective 2016;388:488–497.
review of 208 patients with leiomyosarcoma of the uterus: Prognostic 22. Chang KL, Crabtree GS, Lim-Tan SK, et al. Primary uterine endometrial
indicators, surgical management, and adjuvant therapy. Gynecol Oncol. stromal neoplasms. A clinicopathologic study of 117 cases. Am J Surg
2003;89:460–469. Pathol. 1990;14:415–438.
3. Oliva E. Cellular mesenchymal tumors of the uterus: A review empha- 23. Baker P, Oliva E. Endometrial stromal tumours of the uterus: A prac-
sizing recent observations. Int J Gynecol Pathol. 2014;33:374–384. tical approach using conventional morphology and ancillary tech-
4. Prat J. FIGO staging for uterine sarcomas. Int J Gynaecol Obstet. niques. J Clin Pathol. 2007;60:235–243.
2009;104:177–178. 24. Nucci MR, Harburger D, Koontz J, et  al. Molecular analysis of the
5. Lavie O, Barnett-Griness O, Narod SA, et  al. The risk of devel- JAZF1-­JJAZ1 gene fusion by RT-­PCR and fluorescence in situ
oping uterine sarcoma after tamoxifen use. Int J Gynecol Cancer. hybridization in endometrial stromal neoplasms. Am J Surg Pathol.
2008;18:352–356. 2007;31:65–70.
6. Abeler VM, Royne O, Thoresen S, et  al. Uterine sarcomas in 25. Chan JK, Kawar NM, Shin JY, et al. Endometrial stromal sarcoma: A
Norway. A histopathological and prognostic survey of a total pop- population-­based analysis. Br J Cancer. 2008;99:1210–1215.
ulation from1970 to 2000 including 419 patients. Histopathology. 26. Spano JP, Soria JC, Kambouchner M, et  al. Long-­term survival of
2009;54:355–364. patients given hormonal therapy for metastatic endometrial stromal
7. Chen L, Yang B. Immunohistochemical analysis of p16, p53, and Ki-­ sarcoma. Med Oncol. 2003;20:87–93.
67 expression in uterine smooth muscle tumors. Int J Gynecol Pathol. 27. Kurihara S, Oda Y, Ohishi Y, et al. Endometrial stromal sarcomas and
2008;27:326–332. related high-­grade sarcomas: Immunohistochemical and molecular
8. Ylisaukko-oja SK, Kiuru M, Lehtonen HJ, et al. Analysis of fumarate genetic study of 31 cases. Am J Surg Pathol. 2008;32:1228–1238.
hydratase mutations in a population-­based series of early onset uter- 28. Lee CH, Marino-Enriquez A, Ou W, et al. The clinicopathologic fea-
ine leiomyosarcoma patients. Int J Cancer. 2006;119:283–287. tures of YWHAE-­FAM22 endometrial stromal sarcomas: A histolog-
9. D’Angelo E, Spagnoli LG, Prat J. Comparative clinicopathologic and ically high-­grade and clinically aggressive tumor. Am J Surg Pathol.
immunohistochemical analysis of uterine sarcomas diagnosed using 2012;36:641–653.
the World Health Organization classification system. Hum Pathol. 29. Tanner EJ, Garg K, Leitao MM Jr, et  al. High grade undifferentiated
2009;40:1571–1585. uterine sarcoma: Surgery, treatment, and survival outcomes. Gynecol
10. Koivisto-Korander R, Butzow R, Koivisto AM, et al. Clinical outcome Oncol. 2012;127:27–31.
and prognostic factors in 100 cases of uterine sarcoma: Experience 30. Clement PB, Scully RE. Mullerian adenosarcoma of the uterus: A clin-
in Helsinki University Central Hospital 1990–2001. Gynecol Oncol. icopathologic analysis of 100 cases with a review of the literature.
2008;111:74–81. Hum Pathol. 1990;21:363–381.
11. Mayerhofer K, Obermair A, Windbichler G, et al. Leiomyosarcoma of 31. Gallardo A, Prat J. Mullerian adenosarcoma: A clinicopathologic and
the uterus: A clinicopathologic multicenter study of 71 cases. Gynecol immunohistochemical study of 55 cases challenging the existence of
Oncol. 1999;74:196–201. adenofibroma. Am J Surg Pathol. 2009;33:278–288.
|
58       Mbatani ET AL.

32. Ferguson SE, Tornos C, Hummer A, et  al. Prognostic features subtypes: Similar outcomes belie distinctive biologic differences. Am J
of surgical stage I uterine carcinosarcoma. Am J Surg Pathol. Surg Pathol. 2007;31:979–987.
2007;31:1653–1661. 37. Alektiar KM, McKee A, Lin O, et al. Is there a difference in outcome
33. Silverberg SG, Major FJ, Blessing JA, et  al. Carcinosarcoma (malig- between stage I-­II endometrial cancer of papillary serous/clear cell
nant mixed mesodermal tumor) of the uterus. A Gynecologic and endometrioid FIGO Grade 3 cancer? Int J Radiat Oncol Biol Phys.
Oncology Group pathologic study of 203 cases. Int J Gynecol Pathol. 2002;54:79–85.
1990;9:1–19. 38. Homesley HD, Filiaci V, Markman M, et al. Phase III trial of ifosfamide
34. Yamada SD, Burger RA, Brewster WR, et al. Pathologic variables and with or without paclitaxel in advanced uterine carcinosarcoma: A
adjuvant therapy as predictors of recurrence and survival for patients Gynecologic Oncology Group study. J Clin Oncol. 2007;25:526–531.
with surgically evaluated carcinosarcoma of the uterus. Cancer. 39. Callister M, Ramondetta LM, Jhingran A, et al. Malignant mixed Mullerian
2000;88:2782–2786. tumors of the uterus: Analysis of patterns of failure, prognostic factors,
35. George E, Lillemoe TJ, Twiggs LB, et  al. Malignant mixed mullerian and treatment outcome. Int J Radiat Oncol Biol Phys. 2004;58:786–796.
tumor versus high-­grade endometrial carcinoma and aggressive vari- 40. Reed NS, Mangioni C, Malmstrom H, et al. Phase III randomised study
ants of endometrial carcinoma: A comparative analysis of survival. Int to evaluate the role of adjuvant pelvic radiotherapy in the treatment
J Gynecol Pathol. 1995;14:39–44. of uterine sarcomas stages I and II: An European Organisation for
36. Soslow RA, Bissonnette JP, Wilton A, et  al. Clinicopathologic analy- Research and Treatment of Cancer Gynaecological Cancer Group
sis of 187 high-­grade endometrial carcinomas of different histologic Study (protocol 55874). Eur J Cancer. 2008;44:808–818.
DOI: 10.1002/ijgo.12614

FIGO CANCER REPORT 2018

Cancer of the ovary, fallopian tube, and peritoneum

Jonathan S. Berek1,* | Sean T. Kehoe2 | Lalit Kumar3 | Michael Friedlander4,5

1
Stanford Women’s Cancer Center, Stanford
Cancer Institute, Stanford University School of Abstract
Medicine, Stanford, CA, USA The Gynecologic Oncology Committee of FIGO in 2014 revised the staging of ovarian
2
Institute of Cancer and Genomics, University
cancer, incorporating ovarian, fallopian tube, and peritoneal cancer into the same
of Birmingham, Birmingham, UK
3 ­system. Most of these malignancies are high-­grade serous carcinomas (HGSC). Stage IC
Department of Medical Oncology, All India
Institute of Medical Sciences, New Delhi, India is now divided into three categories: IC1 (surgical spill); IC2 (capsule ruptured before
4
Royal Hospital for Women, Randwick, surgery or tumor on ovarian or fallopian tube surface); and IC3 (malignant cells in the
Sydney, NSW, Australia
5
ascites or peritoneal washings). The updated staging includes a revision of Stage IIIC
University of New South Wales Clinical
School, Sydney, NSW, Australia based on spread to the retroperitoneal lymph nodes alone without intraperitoneal
­dissemination. This category is now subdivided into IIIA1(i) (metastasis ≤10 mm in
*Correspondence
Jonathan S. Berek, Stanford Women’s Cancer greatest dimension), and IIIA1(ii) (metastasis >10 mm in greatest dimension). Stage IIIA2
Center, Stanford Cancer Institute, Stanford is now “microscopic extrapelvic peritoneal involvement with or without positive
University School of Medicine, Stanford,
CA, USA. ­retroperitoneal lymph node” metastasis. This review summarizes the genetics, surgical
Email: jberek@stanford.edu management, chemotherapy, and targeted therapies for epithelial cancers, and the
treatment of ovarian germ cell and stromal malignancies.

KEYWORDS
Cancer staging; Chemotherapy; Fallopian tube; FIGO Cancer Report; Ovarian; Ovary; Peritoneum

end of the fallopian tube.3–8 Therefore, the incidence of fallopian tube


1 |  INTRODUCTION
cancers may have been substantially underestimated. These new data
support the view that high-­grade serous ovarian, fallopian tube, and
1.1 | Primary sites: ovarian, fallopian tube, and
peritoneal cancers should be considered collectively, and that the con-
peritoneal cancer
vention of designating malignancies as having an ovarian origin should
In 2014, the Gynecologic Oncology Committee of FIGO revised the no longer be used, unless that is clearly the origination site. It has
staging to incorporate ovarian, fallopian tube, and peritoneal cancer been suggested that extrauterine tumors of serous histology arising
in the same system. Changing the staging system required extensive in the ovary, fallopian tube, or peritoneum might be described col-
international consultation. The primary site (i.e. ovary, fallopian tube, lectively as “Müllerian carcinomas”1,2 or “pelvic serous carcinomas”.9
or peritoneum) is designated, where possible. When it is not pos- The latter tumor designation is controversial because some peritoneal
sible to clearly delineate the primary site, these should be listed as tumors might arise in extrapelvic peritoneum. Therefore, the simple
“undesignated”.1,2 term “serous carcinoma” is preferred, and most of these are high-­grade
It has been presumed that fallopian tube malignancies were rare.2 serous carcinomas (HGSC).
However, histologic, molecular, and genetic evidence shows that as Although there has been no formal staging for peritoneal cancers,
many as 80% of tumors that were classified as high-­grade serous carci- the FIGO staging system is used with the understanding that it is not
nomas of the ovary or peritoneum may have originated in the fimbrial possible to have a Stage I peritoneal cancer.

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

Int J Gynecol Obstet 2018; 143 (Suppl. 2): 59–78 wileyonlinelibrary.com/journal/ijgo  |  59
|
60       Berek ET AL.

be useful in determining response to chemotherapy, but they do not


1.1.1 | Primary site
contribute to staging.
Ovarian epithelial tumors may arise within endometriosis or cortical
inclusions of Müllerian epithelium, likely a form of endosalpingiosis.
1.2.1 | Fallopian tube involvement
These include low-­grade endometrioid carcinomas, clear cell carcino-
mas, borderline and low-­grade serous carcinomas, and mucinous car- Fallopian tube involvement can be divided into three categories. In the
cinomas. These tumors are thought to evolve slowly from lower-­grade first, an obvious intraluminal and grossly apparent fallopian tube mass
precursor conditions (endometriotic cysts, cystadenomas, etc.) and is seen with tubal intraepithelial carcinoma (carcinoma in situ) that
are classified as type I tumors.5 Fallopian tube carcinomas arise in the is presumed to have arisen in the fallopian tube. These cases should
distal fallopian tube and the majority of these are high-­grade serous be staged surgically with a histologic confirmation of disease. Tumor
carcinomas. These are thought to evolve rapidly from more obscure extension into the submucosa or muscularis and to and beyond the
precursors and are designated as type II tumors.5,6 This latter group serosa can therefore be defined. These features, together with the
encompasses high-­grade endometrioid carcinomas and carcinosarco- laterality and the presence or absence of ascites, should all be taken
mas. All of these high-­grade carcinomas are nearly always associated into consideration.1,3,6,7
5
with mutations in the TP53 gene. In the second scenario, a widespread serous carcinoma is asso-
ciated with a tubal intraepithelial carcinoma. A visible mass in the
endosalpinx may not be seen but the histologic findings should be
1.1.2 | Lymphatic and lymph node drainage
noted in the pathology report since they may indicate a fallopian tube
The lymphatic drainage of the ovaries and fallopian tubes is via the primary. Tumors obliterating both fallopian tube and ovary may belong
utero-­ovarian, infundibulopelvic, and round ligament pathways and an to this group but whether a presumptive assignment of a tubal origin
external iliac accessory route into the following regional lymph nodes: can be made in such cases is controversial given that tubal intraepithe-
external iliac, common iliac, hypogastric, lateral sacral, para-­aortic lial carcinoma cannot be confirmed.
lymph nodes and, occasionally, to the inguinal nodes.1,10–12 The peri- In the third scenario— risk-­reducing salpingo-­oophorectomy—
toneal surfaces can drain through the diaphragmatic lymphatics and tubal intraepithelial carcinoma may be the only finding. It should be
hence to the major venous vessels above the diaphragm. reported as originating in the fallopian tube and managed accordingly.
The majority of early serous cancers detected are found in the fallo-
pian tube, irrespective of genetic risk.15,16
1.1.3 | Other metastatic sites
The peritoneum, including the omentum and pelvic and abdominal vis-
1.2.2 | FIGO staging
cera, is the most common site for dissemination of ovarian and fallo-
pian tube cancers. This includes the diaphragmatic and liver surfaces. The updated, revised FIGO staging system combines the classifica-
Pleural involvement is also seen. Other extraperitoneal or extrapleural tion for ovarian, fallopian tube, and peritoneum cancer. It is based on
sites are relatively uncommon, but can occur.1,10–12 After system- findings made mainly through surgical exploration (as outlined above).
atic pathologic analysis has excluded a tubal or ovarian site of origin, Table 1 presents the 2014 FIGO staging classification for cancer of
malignancies that appear to arise primarily on the peritoneum have an the ovary, fallopian tube, and peritoneum. The equivalents within the
identical spread pattern, and frequently may involve the ovaries and Union for International Cancer Control (UICC) TNM classification are
fallopian tubes secondarily. These “peritoneal” tumors are thought to presented in Table 2.
arise in endosalpingiosis. In addition to these changes, several other modifications of the
former staging system have been made to better prospectively capture
the data. Stage IC is now divided into three categories: IC1 (surgical
1.2 | Classification rules
spill); IC2 (capsule ruptured before surgery or tumor on ovarian or fallo-
Although CT scans can delineate the intra-­abdominal spread of dis- pian tube surface); and IC3 (malignant cells in the ascites or peritoneal
ease to a certain extent, ovarian, fallopian tube, and peritoneal can- washings). Stage IIC has been eliminated. The updated staging includes
cers should be staged surgically. Operative findings determine the a revision of the Stage IIIC based on spread to the retroperitoneal
precise histologic diagnosis, stage, and therefore the prognosis, of the lymph nodes alone without intraperitoneal dissemination, because an
patient.1,9,10,12–14 analysis of these patients indicates that their survival is significantly
In selected patients with advanced-­stage disease, it may be appro- better than those who have intraperitoneal dissemination.17 This cat-
priate to initiate chemotherapy prior to surgical intervention, and in egory is now subdivided into IIIA1(i) (metastasis ≤10 mm in greatest
these cases, there should be histologic or cytologic confirmation of the dimension), and IIIA1(ii) (metastasis >10 mm in greatest dimension).
diagnosis prior to starting neoadjuvant chemotherapy (see 5.2.2. below). Stage IIIA2 is now “microscopic extrapelvic peritoneal involvement
Chest radiograms may serve as a screen for pleural effusions. As with or without positive retroperitoneal lymph node” metastasis. The
distant metastases are infrequent, there is no requirement for other wording of Stage IIIB has been modified to reflect the lymph node sta-
radiological evaluation unless symptomatic. Serum CA125 levels may tus. Stage IVB now includes metastases to the inguinal lymph nodes.
Berek ET AL. |
      61

T A B L E   1   FIGO staging classification for cancer of the ovary, fallopian tube, and peritoneum.

Stage I: Tumor confined to ovaries or fallopian tube(s)


T1-­N0-­M0
IA: Tumor limited to 1 ovary (capsule intact) or fallopian tube; no tumor on ovarian or fallopian tube surface; no malignant
cells in the ascites or peritoneal washings
T1a-­N0-­M0
IB: Tumor limited to both ovaries (capsules intact) or fallopian tubes; no tumor on ovarian or fallopian tube surface; no
malignant cells in the ascites or peritoneal washings
T1b-­N0-­M0
IC: Tumor limited to 1 or both ovaries or fallopian tubes, with any of the following:
IC1: Surgical spill
T1c1-­N0-­M0
IC2: Capsule ruptured before surgery or tumor on ovarian or fallopian tube surface
T1c2-­N0-­M0
IC3: Malignant cells in the ascites or peritoneal washings
T1c3-­N0-­M0
Stage II: Tumor involves 1 or both ovaries or fallopian tubes with pelvic extension (below pelvic brim) or peritoneal cancer
T2-­N0-­M0
IIA: Extension and/or implants on uterus and/or fallopian tubes and/or ovaries
T2a-­N0-­M0
IIB: Extension to other pelvic intraperitoneal tissues
T2b-­N0-­M0
Stage III: Tumor involves 1 or both ovaries or fallopian tubes, or peritoneal cancer, with cytologically or histologically
confirmed spread to the peritoneum outside the pelvis and/or metastasis to the retroperitoneal lymph nodes
T1/T2-­N1-­M0
IIIA1: Positive retroperitoneal lymph nodes only (cytologically or histologically proven):
IIIA1(i) Metastasis up to 10 mm in greatest dimension
IIIA1(ii) Metastasis more than 10 mm in greatest dimension
IIIA2: Microscopic extrapelvic (above the pelvic brim) peritoneal involvement with or without positive retroperitoneal
lymph nodes
T3a2-­N0/N1-­M0
IIIB: Macroscopic peritoneal metastasis beyond the pelvis up to 2 cm in greatest dimension, with or without metastasis to
the retroperitoneal lymph nodes
T3b-­N0/N1-­M0
IIIC: Macroscopic peritoneal metastasis beyond the pelvis more than 2 cm in greatest dimension, with or without metastasis
to the retroperitoneal lymph nodes (includes extension of tumor to capsule of liver and spleen without parenchymal
involvement of either organ)
T3c-­N0/N1-­M0
Stage IV: Distant metastasis excluding peritoneal metastases
Stage IVA: Pleural effusion with positive cytology
Stage IVB: Parenchymal metastases and metastases to extra-­abdominal organs (including inguinal lymph nodes and
lymph nodes outside of the abdominal cavity)
Any T, any N, M1

1.2.2.1 | Regional lymph nodes (N) 1.2.2.2 | Distant metastasis (M)

1. NX: Regional lymph nodes cannot be assessed. 1. MX: Distant metastasis cannot be assessed.
2. N0: No regional lymph node metastasis. 2. M0: No distant metastasis.
3. N1: Regional lymph node metastasis. 3. M1: Distant metastasis (excluding peritoneal metastasis).
|
62       Berek ET AL.

T A B L E   2   Cancer of the ovary, fallopian tube and peritoneum: 3. Endometrioid tumors.


FIGO staging (2014) compared with TNM classification.a 4. Clear cell tumors.
5. Brenner tumors.
UICC
FIGO (designate primary: 6. Undifferentiated carcinomas (this group of malignant tumors is of
Tov, Tft, Tp, or Tx) T N M epithelial structure, but they are too poorly differentiated to be
Stage placed in any other group).
IA T1a N0 M0 7. Mixed epithelial tumors (these tumors are composed of two or
IB T1b N0 M0 more of the five major cell types of common epithelial tumors. The
IC T1c N0 M0 types are usually specified).

IIA T2a N0 M0
8. Cases with high-grade serous carcinoma in which the ovaries and
fallopian tubes appear to be incidentally involved and not the pri-
IIB T2b N0 M0
mary origin can be labeled as peritoneal carcinoma or serous carci-
IIIA T3a N0 M0
noma of undesignated site, at the discretion of the pathologist.
T3a N1 M0
IIIB T3b N0 M0
Epithelial tumors of the ovary and fallopian tube are further sub-
T3b N1 M0
classified by histologic grading, which can be correlated with progno-
IIIC T3c N0–1 M0 sis. This grading system does not apply to nonepithelial tumors.19 Two
T3c N1 M0 grading systems are applied. For non-­serous carcinomas (most endo-
IV Any T Any N M1 metrioid and mucinous), grading is identical to that used in the uterus,
Regional nodes (N) based on architecture with a one-­step upgrade if there is prominent
Nx Regional lymph nodes cannot be nuclear atypia, as follows:
assessed
N0 No regional lymph node metastasis 1. GX: Grade cannot be assessed.
N1 Regional lymph node metastasis 2. G1: Well differentiated.
Distant metastasis (M) 3. G2: Moderately differentiated.
Mx Distant metastasis cannot be assessed 4. G3: Poorly differentiated.

M0 No distant metastasis
Serous carcinomas are the most common in both the ovary and
M1 Distant metastasis (excluding peritoneal
metastasis) tube. More than 90% of fallopian tube carcinomas are serous or high-­
grade endometrioid adenocarcinoma. Other cell types have been
Notes: 1. The primary site—that is, ovary, fallopian tube, or peritoneum—
should be designated where possible. In some cases, it may not be possible to reported, but are rare.1,2,20 Serous carcinomas are graded in a two-­
clearly delineate the primary site, and these should be listed as “undesignated.” grade system befitting their biology. High-­grade serous carcinomas,
2. The histologic type should be recorded. 3. The staging includes a revision of including both classic appearing and those with SET features (solid,
the Stage III patients and allotment to Stage IIIA1 is based on spread to the
endometrioid-­like, and transitional) carry a high frequency of muta-
retroperitoneal lymph nodes without intraperitoneal dissemination, because
tions in TP53.21–23 Low-­grade serous carcinomas are often associated
an analysis of these patients indicates that their survival is significantly better
than those who have intraperitoneal dissemination. 4. Involvement of retrop- with borderline or atypical proliferative serous tumors, often contain
eritoneal lymph nodes must be proven cytologically or histologically. 5. mutations in BRAF and KRAS and contain wild-­type TP53. Most “mod-
Extension of tumor from omentum to spleen or liver (Stage IIIC) should be dif- erately differentiated” serous carcinomas carry mutations in TP53 and
ferentiated from isolated parenchymal splenic or liver metastases (Stage IVB).
a should be combined with the high-­grade tumors.19,22–24
Source: Prat J; FIGO Committee on Gynecologic Oncology. Staging classi-
fication for cancer of the ovary, fallopian tube, and peritoneum. Int J Nonepithelial cancers, although uncommon, are extremely import-
Gynecol Obstet. 2014;124:1–5. ant. These include granulosa cell tumors, germ cell tumors, sarcomas, and
lymphomas. They are discussed below as separate entities. Metastatic
neoplasms to the ovary, such as tumors arising in the breast, lower
1.3 | Histopathologic classification
reproductive tract sites (cervix or uterine carcinomas) and gastrointes-
The majority of cases of ovarian cancer are of epithelial origin. FIGO tinal tract (signet ring cell [Krukenberg] carcinomas, low grade appen-
endorses the WHO histologic typing of epithelial ovarian tumors. It is diceal or pancreaticobiliary mucinous tumors and other neoplasms) are
recommended that all ovarian epithelial tumors be subdivided accord- graded and staged in accordance with their respective sites of origin.1,2
ing to the classification given below.18
The histologic classification of ovarian, fallopian tube, and perito-
neal neoplasia is as follows: 2 | EPIDEMIOLOGY

1. Serous tumors. Malignant tumors of the ovaries occur at all ages with variation in his-
2. Mucinous tumors. tologic subtype by age. For example, in women younger than 20 years
Berek ET AL. |
      63

of age, germ cell tumors predominate, while borderline tumors typi- cancer. A recent study of next generation sequencing of constitu-
cally occur in women in their 30s and 40s—10 or more years younger tional DNA samples from 1915 women with ovarian cancer was
than in women with invasive epithelial ovarian cancers, which mostly carried out to identify germline mutations using a panel of 20 genes
occur after the age of 50 years. including BRCA1 and BRCA2, DNA mismatch repair genes, double
The lifetime risk of a woman in the USA developing ovarian cancer stranded DNA break repair genes such as CHEK2 and ATM, as well
is approximately 1 in 70. Approximately 23% of gynecologic cancers as the BRCA1-associated complex or the BRCA2/Fanconi Anemia
are ovarian in origin, but 47% of all deaths from cancer of the female pathway genes (including BRIP1, BARD1, PALB2, RAD50, RAD51C,
genital tract occur in women with ovarian cancer. Overall, epithelial and RAD51D, among others). About 80% of mutations were in
ovarian cancer accounts for 4% of all new cancer diagnoses in women BRCA1 or BRCA2. About 3% of patients carried mutations in the
1,2,25
and 5% of all cancer-­related deaths. Fanconi Anemia pathway genes, while only 0.4% had mutations in
The overall incidence of epithelial tumors varies from 9 to 17 per mismatch repair genes.33 In an earlier similar study that included
100 000 and is highest in high-­income countries, with the exception of 360 patients, 24% carried germline loss-of-function mutations
Japan.26 However, this incidence rate increases proportionately with including 18% in BRCA1 or BRCA2 and 6% in BARD1, BRIP1, CHEK2,
age. The largest number of patients with epithelial ovarian cancer is MRE11A, MSH6, NBN, PALB2, RAD50, RAD51C, or TP53.34,35
found in the 60–64 years age group. The median age is about a decade 4. Inherited mutations in the mismatch repair genes associated with
earlier in low-­income countries. Lynch syndrome type II. Women carrying these mutations have an
Established risk factors for epithelial ovarian tumors include repro- increased risk of a number of cancers including colon, endometrial,
ductive risk factors. Women who have never had children are twice and ovarian cancer. Typically, the ovarian cancers that occur are
as likely to develop this disease. First pregnancy at an early age, early endometrioid or clear cell histologically and are usually Stage I.35
menopause, and the use of oral contraceptives have been associated
with lower risks of ovarian cancer.27 The relationship of these variables Women with a strong family history of epithelial ovarian, fallopian
to fallopian tube cancer is unclear. tube, or peritoneal cancers, particularly if there is a documented germline
As noted above, it has been previously presumed that fallopian BRCA mutation, are advised to have a risk-­reducing bilateral salpingo-­
tube malignancies were rare; however, this has been challenged by oophorectomy after appropriate counseling and at the completion of
evidence to show that many tumors that were classified as serous car- childbearing. All women who are suspected of carrying a BRCA germ-
cinomas of the ovary or peritoneal cancers appear to have their origin line mutation, based on family history or young age of diagnosis and a
in the fallopian tube.3–7 When the origin is uncertain, the convention high-­grade serous or high-­grade endometrioid cancer, should be offered
of designating all serous cancers, as originating in the ovary should no genetic testing. BRCA mutations may also occur in women without a
longer be used and the term “undesignated origin” may be applied at family history of breast/ovarian cancer, and genetic testing should be
18
the discretion of the pathologist. considered in patients from ethnic groups where there is a high inci-
dence of founder mutations (e.g. Ashkenazi Jewish ancestry), and in
women with high-­grade serous cancers under the age of 70 years.26–30
2.1 | Genetics
Australian guidelines advise that all women with invasive epithelial
Hereditary factors are implicated in approximately 20% of ovarian, fal- ovarian cancer apart from mucinous cancers diagnosed under the age
lopian tube, and peritoneal cancers28–32: of 70 should be offered BRCA mutation testing independent of family
history and histologic subtype.36 In contrast, the Society of Gynecologic
1. Most hereditary ovarian cancers are due to pathogenic mutations Oncology (SGO) and National Comprehensive Cancer Network (NCCN)
in either the BRCA1 or BRCA2 genes. At least 15% of women guidelines recommend that all women diagnosed with ovarian, fallopian
with high-grade nonmucinous ovarian cancers have germline tube, or peritoneal carcinoma, regardless of age or family history, should
mutations in BRCA1/2 and, importantly, almost 40% of these receive genetic counseling and be offered genetic testing.37 Women
women do not have a family history of breast/ovarian cancer. whose family history suggests Lynch syndrome type II should undergo
All women with high-grade nonmucinous invasive ovarian cancers appropriate genetic counseling and testing.
should be offered genetic testing even if they do not have a
family history of breast/ovarian cancer.
2. Inherited deleterious mutations in BRCA1 and BRCA2 are the 3 | SCREENING
major genetic risk factors. Women who carry germline mutations
in BRCA1 and BRCA2 have a substantially increased risk of ovarian, To date, there are no documented effective screening methods that
tubal, and peritoneal cancer—about 20%–50% with BRCA1 and reduce the mortality of ovarian, fallopian tube, or peritoneal cancers.
10%–20% with BRCA2.29–32 Typically, these cancers occur at an Studies using CA125, ultrasonography of the pelvis, and pelvic exami-
earlier age than sporadic cancers, particularly in BRCA1 mutation nation do not have an acceptable level of sensitivity and specificity,
carriers, with a median age of diagnosis in the mid-40s. based on trials carried out in women in the general population and
3. There are a number of other low- to moderate-penetrance genes those in the high-­risk population. The US Preventive Services Task
that can also predispose to ovarian, fallopian tube, or peritoneal Force recommends against screening asymptomatic women for
|
64       Berek ET AL.

ovarian cancer with pelvic examination, pelvic ultrasound, or serum Then a complete physical examination, including general, breast, pel-
38
tumor marker measurements. The low prevalence of disease and vic, and rectal examination, must be performed.1
lack of high-­quality screening methods make it more likely to obtain Prior to surgery a chest radiograph should be taken to screen for
false-­positive results leading to unnecessary interventions. A recent a pleural effusion and a CT scan of the abdomen and pelvis should
study of multimodal screening using CA125 based on a risk of ovarian be performed to delineate the extent of intra-­abdominal disease.
cancer algorithm (ROCA) every 4 months and transvaginal ultrasound However, in the absence of extra-­abdominopelvic disease, radiologi-
annually or earlier where indicated by the ROCA in women at high cal scanning does not replace surgical staging with laparotomy. Tumor
risk of ovarian cancer reported that screening was associated with markers including CA125, and carcinoembryonic antigen (CEA) should
a low rate of high-­volume disease at primary surgery and very high be considered.1 With a high CA125 level, the most common diagnosis
rates of no residual disease after surgery.38 Given that the majority would be epithelial ovarian, fallopian tube, or peritoneal cancer.
of women with advanced stage ovarian cancer, even with complete A gastric or colonic primary with metastases to the ovaries may
resection, will relapse after chemotherapy, this does not seem to be a mimic ovarian cancer, and if the CEA is elevated, this should be con-
good alternative to risk-­reducing surgery. The authors of the screen- sidered. A ratio of more than 25:1 (CA-­125 and CEA) favors an ovarian
ing study concluded that risk-­reducing salpingectomy-­oophorectomy primary though it does not completely rule out a primary in the gas-
remains the treatment choice for women at high risk of ovarian/fal- trointestinal tract.47
lopian tube cancer.38 A current mammogram should be considered as patients are fre-
Women at increased genetic risk should be encouraged to con- quently in the age group where breast cancer is prevalent. A colonos-
sider risk-­reducing bilateral salpingo-­oophorectomy, as this is the most copy is indicated when symptoms suggest possible bowel cancer.1
39,40
effective way to reduce mortality in this population of women. An The following factors point to the presence of a malignancy, and
ACOG bulletin has recommended that opportunistic (at the time of a are useful in the clinical assessment of masses:
clinically indicated hysterectomy) bilateral salpingectomy be considered
in women not at genetic risk who wish to retain their ovaries as a way 1. Age of the patient (young for germ cell, older for epithelial
to reduce their risk of later developing high-­grade serous carcinomas.41 malignancies).
2. Bilaterality.
3. Tumor fixation clinically.
4 | DIAGNOSIS 4. Ascites.
5. Ultrasonographically complex, especially if solid areas.
Patients with epithelial ovarian cancers confined to the ovary or fal- 6. CT finding of metastatic nodules.
lopian tube at initial diagnosis have a very good prognosis.42–45 The 7. Elevated tumor markers.
symptoms are often very insidious and the duration of symptoms not
very different between patients with early stage or advanced stage
disease.13,14 This may reflect the different biological behavior of the 5 | PRIMARY SURGERY
various histologic subtypes; for example, grade 1 serous, clear cell,
mucinous, and endometrioid cancers are commonly early stage at In general, the prognosis of epithelial ovarian, fallopian, and peritoneal
presentation, whereas high-­grade serous cancers are most often Stage malignancies is independently affected by the following1,48,49:
III because of early dissemination by a more aggressive cancer. Tumor
markers such as human gonadotropin (hCG) and alpha-­fetoprotein 1. Stage of the cancer at diagnosis.
(AFP) are mandatory to exclude germ cell tumors in younger patients 2. Histologic type and grade.
with a pelvic mass or suspicious enlargement of an ovary. 3. Maximum diameter of residual disease after cytoreductive surgery.
Approximately two-­thirds of all epithelial “ovarian” cancers are
Stage III or Stage IV at diagnosis. Presenting symptoms include vague
5.1 | Staging laparotomy
abdominal pain or discomfort, menstrual irregularities, dyspepsia,
and other mild digestive disturbances, which may have been pres- A thorough staging laparotomy is an important part of early manage-
ent for only a few weeks.13,14,46 As the disease progresses, abdom- ment. If the preoperative suspicion is malignancy, a laparotomy should
inal distention and discomfort from ascites generally worsen, and be performed. If there is no visible or palpable evidence of metastasis,
may be associated with respiratory symptoms from increased intra-­ the following should be performed for adequate staging1,10,11,13,14:
abdominal pressure or from the transudation of fluid into the pleural
cavities. Abnormal vaginal bleeding is an uncommon symptom. 1. Careful evaluation of all peritoneal surfaces.
Serous fallopian tube and peritoneal cancers present the same as 2. Retrieval of any peritoneal fluid or ascites. If there is none, wash-
ovarian cancer. Past analyses have been biased because many fallo- ings of the peritoneal cavity should be performed.
pian tube cancers have been presumed to arise in the ovaries. 3. Infracolic omentectomy.
A detailed medical history must be taken to ascertain possible risk 4. Selective lymphadenectomy of the pelvic and para-aortic lymph
factors, history of other cancers, and history of cancer in the family. nodes, at least ipsilateral if the malignancy is unilateral.
Berek ET AL. |
      65

5. Biopsy or resection of any suspicious lesions, masses, or adhesions. organs. Systematic pelvic and para-­aortic lymphadenectomy of non-­
6. Random peritoneal biopsies of normal surfaces, including from the enlarged nodes does not improve overall survival, when compared with
undersurface of the right hemidiaphragm, bladder reflection, cul- removal of bulky nodes only, although there is a modest improvement in
de-sac, right and left paracolic recesses, and both pelvic sidewalls. progression-­free survival.51 Level of Evidence A
7. Total abdominal hysterectomy and bilateral salpingo-oophorec-
tomy in most cases.
5.2.2 | Interval debulking
8. Appendectomy for mucinous tumors.
In selected patients with cytologically proven Stage IIIC and IV disease
Upon opening the abdominopelvic cavity, the peritoneal fluid should who may not be good surgical candidates, 3–4 cycles of neoadjuvant
be sent for cytology. In the absence of ascites, irrigation should be per- chemotherapy (NACT) may be given initially, followed by interval
formed and washings sent for cytology. debulking surgery (IDS) and additional chemotherapy as demon-
The laparotomy should proceed with a detailed examination of the strated in the EROTC and CHORUS Trials.52,53 These two randomized
contents, including all of the peritoneal surfaces. In addition to the prospective trials showed that in selected patients, interval debulking
suspicious sites, biopsies from the peritoneal reflection of the bladder, surgery after neoadjuvant chemotherapy showed equivalent survival
the posterior cul-­de-­sac, both paracolic gutters, subdiaphragmatic sur- with less morbidity compared with primary cytoreductive surgery.
faces, and both pelvic sidewalls should be taken. The primary tumor, if NACT followed by IDS may be particularly useful in patients with a
limited to the ovary, should be examined to look for capsular rupture. poor performance status, significant medical co-­morbidities, visceral
All obvious sites of tumor must be removed wherever possible in addi- metastases, and those who have large pleural effusions and/or gross
tion to total hysterectomy and bilateral salpingo-­oophorectomy. The ascites.54 In selected patients whose primary cytoreduction is consid-
omentum, pelvic, and para-­aortic lymph nodes should be removed for ered suboptimal, particularly if a gynecologic oncologist did not per-
histologic examination. form the initial operation, interval debulking may be considered after
In younger women, fertility may be an issue. In these patients, con- 2–3 cycles of systemic chemotherapy.1,52,53,55 Pathologic assessment
servative surgery, with preservation of the uterus and contralateral for residual tumor following neoadjuvant therapy will enable an esti-
ovary, should be considered after informed consent.43 mate of residual disease and pathological response.56 There are recent
Clinical judgment is important in the approach to a pelvic data to indicate that patients who have a good pathological response
mass in the young, reproductive-­aged woman. If the suspicion have a better outcome. A histopathologic scoring system for measur-
is strong for malignancy, open laparotomy is generally indicated. ing response to neoadjuvant chemotherapy has been developed and
Laparoscopy may be more appropriate if the suspicion is more for validated by Bohm et al.57 who reported criteria for defining a chemo-
benign disease, where tumor markers (including hCG and AFP) are therapy response score (CRS) based on a three-­tier system. A CRS
normal. A biopsy of any suspicious lesion can be performed and 3 (complete or near complete pathological response) was associated
frozen section obtained in order to proceed expeditiously with with a better prognosis. Recently, these results have been validated in
definitive surgery. an independent West Australian cohort.58
Ovaries and fallopian tubes should be evaluated as thoroughly as
possible to establish the site of origin. If visible, the entire tube, partic-
6 | CHEMOTHERAPY
ularly the distal portion, should be submitted for pathology and exam-
ined using the SEE-­FIM protocol.32 Ovaries should be scrutinized for
6.1 | Chemotherapy for early stage cancer
coexisting endometriotic cysts, adenofibromas, or other benign condi-
tions that could serve as a nidus of tumor development. The prognosis of patients with adequately staged tumors with Stage
IA and Stage IB grade 1–2 epithelial cancers of the ovary is very good;
adjuvant chemotherapy does not provide additional benefits and is
5.2 | Cytoreductive (debulking) surgery for advanced
not indicated. For higher-­grade tumors and for patients with Stage
stage disease
IC disease, adjuvant platinum-­based chemotherapy is given to most
patients, although there has been debate about the absolute survival
5.2.1 | Primary debulking
benefit in women with Stage IA and IB cancers who have had thorough
At least two-­thirds of patients with ovarian cancer present with Stage III surgical staging.42 All patients with Stage II disease should receive
or IV disease. This may affect the performance status and fitness for sur- adjuvant chemotherapy. The optimal number of cycles in patients
gery. However, the most important prognostic indicator in patients with with Stage I disease has not been definitively established, but typically
advanced stage ovarian cancer is the volume of residual disease after between 3 and 6 cycles are administered. The Gynecologic Oncology
surgical debulking. Therefore, patients whose medical condition per- Group (GOG) 157 study suggested that 3 cycles of carboplatin and
mits should generally undergo a primary laparotomy with total abdomi- paclitaxel was equivalent to 6 cycles, but in subgroup analysis, 6 cycles
nal hysterectomy, bilateral salpingo-­oophorectomy, omentectomy, and appeared superior in patients with high-­grade serous cancers.50
maximal attempt at optimal cytoreduction.1,48–50 This may necessitate There is no evidence to support adjuvant therapy for carcinoma in situ
bowel resection, and occasionally, partial or complete resection of other of the fallopian tube and it is not recommended.1,2,44 Level of Evidence A
|
66       Berek ET AL.

still debated.66–71 The GOG 172 trial compared intravenous paclitaxel


6.2 | Chemotherapy for advanced stage
plus cisplatin with intravenous paclitaxel plus intraperitoneal cispla-
ovarian cancer
tin and paclitaxel in patients with Stage III ovarian or peritoneal car-
Patients who have had primary cytoreduction should receive chemo- cinoma, with no residual disease greater than 1 cm in diameter.68 Only
1,59
therapy following surgery (Table 3). The accepted standard is 6 42% of patients in the intraperitoneal group completed 6 cycles of
cycles of platinum-­based combination chemotherapy, with a platinum the assigned therapy, but the intraperitoneal group had an improve-
(carboplatin or cisplatin) and a taxane (paclitaxel or docetaxel).60–64 ment in progression-­free survival of 5.5 months (23.8 vs 18.3 months;
Docetaxel is an option in patients who have had a significant aller- P=0.05) and an improvement in overall survival of 15.9 months (65.6
gic reaction to paclitaxel or who develop early sensory neuropathy as vs 49.7 months; P=0.03). Level of Evidence A
it has less neurotoxicity, but it is more myelosuppressive than pacli- More recently, the GOG 252 trial reported a median progression-­
taxel.60 The SCOT-­ROC (Scottish Gynecological Cancer Trials Group) free survival of approximately 27–29 months in over 1500 patients
study randomly assigned 1077 women with Stages IC–IV epithelial with optimal Stage II–III disease treated with regimens consisting of
ovarian cancer to carboplatin paclitaxel or docetaxel.60 The efficacy different combinations of intravenous and intraperitoneal cisplatin, car-
of docetaxel was similar to paclitaxel. The median progression-­free boplatin, and paclitaxel, in combination with bevacizumab.69 The treat-
survival was 15.1 versus 15.4 months. The MITO 2 trial randomized ment arms included intravenous carboplatin AUC 6/intravenous weekly
over 800 patients to receive either carboplatin and liposomal doxoru- paclitaxel at 80 mg/m2; intraperitoneal carboplatin AUC 6/intravenous
bicin (PLD) or carboplatin and paclitaxel. The median progression-­free weekly paclitaxel at 80 mg/m; and intravenous paclitaxel at 135 mg/
survival was 19.0 and 16.8 months with carboplatin/PLD and carbo- m2 on day one/intraperitoneal cisplatin at 75 mg/m2 on day two/intra-
65
platin/paclitaxel, respectively. The median overall survival times peritoneal paclitaxel at 60 mg/m2 on day eight. In addition, each arm
were 61.6 and 53.2 months with carboplatin/PLD and carboplatin/ received intravenous bevacizumab at 15 mg/kg with cycles 2 through
paclitaxel, respectively (hazard ratio [HR] 0.89; 95% CI 0.72–1.12; 6 of chemotherapy and then alone for cycles 7 through 22. The median
P=0.32). Carboplatin/PLD produced a similar response rate but dif- progression-­free survival by intent-­to-­treat analysis was 24.9 (intrave-
ferent toxicity (less neurotoxicity and alopecia but more hematologic nous carboplatin), 27.3 (intraperitoneal carboplatin), and 26.0 months
adverse effects) and could also be considered as an option in patients (intraperitoneal cisplatin). An analysis limited to patients with opti-
where paclitaxel cannot be used. mal Stage III tumors and no gross residual disease found a median
Although intraperitoneal chemotherapy has been shown to be progression-­free survival of 31–34 months in all three arms. By compar-
associated with improved progression-­free survival and overall survival ison, the GOG 172 trial comparing intraperitoneal and intravenous che-
in selected patients with optimally debulked Stage III ovarian cancer, motherapy regimens in ovarian cancer had a median progression-­free
it is not widely used outside the USA because of concerns regarding survival of 23.8 months with intraperitoneal cisplatin (vs 18.3 months
increased toxicity and catheter-­related problems, and the benefits are with intravenous) with an improvement in overall survival in favor of
intraperitoneal injection.68 In addition, the median progression-­free sur-

T A B L E   3   Chemotherapy for advanced epithelial ovarian cancer: vival was 60 months in the patients with no residual disease in GOG
recommended regimens.a 172. Differences in the cisplatin arm from the GOG 172 study include a
dose reduction from 100 mg to 75 mg and a shorter infusion time from
Drugs
24 hours to 3 hours.68 If intraperitoneal treatment is used it would be
Standard Administration No. of
regimens Dose (h) Interval treatments appropriate to follow the GOG 172 protocol rather than the modified
protocol with a lower dose of cisplatin accepting the increased toxicity.
Carboplatin AUC=5–6 3 Every 3 wk 6–8 cycles
Combination chemotherapy with either intravenous carboplatin and
Paclitaxel 175 mg/m2
paclitaxel or intraperitoneal cisplatin and paclitaxel (using the GOG 172
Carboplatin AUC=5–6 3 Every 3 wk 6 cycles
protocol) are the standard treatment options for patients with advanced
Paclitaxel 80 mg/m2 Every 18 wk
disease, with evidence to support the addition of bevacizumab in
week
selected patients. The advantages and disadvantages of the intravenous
Carboplatin AUC=5 3 Every 6 cycles
versus intraperitoneal routes of administration of these drugs should be
week
discussed with the patient. Intraperitoneal chemotherapy is applicable
Docetaxel 75 mg/m2 Every 3 wk
only to patients with advanced disease who have had optimal debulking
Cisplatin 75 mg/m2 3 Every 3 wk 6 cycles
and have less than 1 cm residual disease. It should be used only in cen-
Paclitaxel 135 mg/m2
ters that have experience with intraperitoneal chemotherapy.
Carboplatin AUC=5 3 Every 3 wk 6 cycles,
The recommended doses and schedule for intravenous chemother-
(single as
apy are: carboplatin (starting dose AUC 5–6), and paclitaxel (175 mg/
agent)b tolerated
m2), every 3 weeks for 6 cycles,51 or the dose-­dense regimen of carbo-
Abbreviation: AUC, area under the curve dose by the methods of Calvert
platin AUC 6 every 3 weeks for 6 cycles and weekly paclitaxel 80 mg/
et al. 75 and Nagao et al.76
a
Reproduced with permission from Berek et al.,1 p.510. m2.70 The Japanese GOG (JGOG) reported the findings of the latter
b
In patients who are elderly, frail, or poor performance status. regimen and showed improved progression-­free survival and overall
Berek ET AL. |
      67

survival.71 An Italian trial (MITO-­7) investigated a different schedule Unfortunately, the study did not have an arm with intraperitoneal
of weekly carboplatin (AUC 2 mg/mL per min) plus weekly paclitaxel cisplatin alone without HIPEC, therefore it is not possible to know
(60 mg/m2) compared with carboplatin (AUC 6 mg/mL per min, admin- whether the improved survival was due to the addition of intraperito-
istered every 3 weeks) and paclitaxel (175 mg/m2).72 The weekly regi- neal cisplatin alone or HIPEC.
men did not significantly improve progression-­free survival compared In patients who may not tolerate combination chemotherapy
with the conventional regimen (18.8 vs 16.5 months; P=0.18), but because of medical comorbidities or advanced age, single-­agent, intra-
was associated with better quality of life and fewer toxic effects. The venously administered carboplatin (AUC 5–6) can be given.
results of the ICON 8 trial investigating dose-­dense paclitaxel in a non-­ For patients who have a significant hypersensitivity reaction to
Japanese population have been recently presented.73 Over 1500 pre- paclitaxel, an alternative active drug can be substituted (e.g. docetaxel,
dominantly European patients were randomized to receive one of three nanoparticle paclitaxel, or liposomal doxorubicin). Carboplatin hyper-
regimens. Arm 1: carboplatin AUC 5/6 and paclitaxel 175 mg/m2 every sensitivity is very uncommon in the first-­line setting, but is seen in
3 weeks; Arm 2: carboplatin AUC 5/6 every 3 weeks and paclitaxel 10%–20% of patients with recurrent disease who have multiple lines
80 mg/m2 weekly; and Arm 3: carboplatin AUC 2 and paclitaxel 80 mg/ of platinum-­based chemotherapy.80
m2 weekly. All patients had received neoadjuvant chemotherapy with In patients with carboplatin hypersensitivity, desensitization could
planned interval debulking or received chemotherapy after initial pri- be attempted, depending on the severity of the reaction, or alterna-
mary cytoreductive surgery. There was no benefit found for the dose-­ tively cisplatin (50–75 mg/m2) may be an option, but there still may be
dense regimens. The progression-­free survival was 24.4 months with a risk of a severe allergic reaction.
every 3-­week dosing, compared with 24.9 and 25.3 months in arms 2 The treatment of all patients with advanced stage disease is
and 3, respectively.73 These results are very different to the JGOG trial approached in a similar manner, with dose modifications based on the
and it seems that the likely explanation is due to pharmacogenomic toxicity of therapy. Care should be taken when considering combina-
differences between these two ethnic groups.74 tion chemotherapy in patients with a very poor performance status or
The recommended doses and schedule for intraperitoneal chemo- with compromised renal function.
therapy are paclitaxel 135 mg/m2 intravenously on day one, followed
by cisplatin 100 mg/m2 intraperitoneally on day two, followed by
6.3 | Maintenance chemotherapy
paclitaxel 60 mg/m2 intraperitoneally on day eight, every 3 weeks for
6 cycles, as tolerated.68,69 Many centers modify the dose of cispla- Almost 80% of women with advanced-­stage disease who respond to
tin to 75 mg/m2 rather than 100 mg/m2 that was used in GOG 172 first-­line chemotherapy relapse. There have been several trials con-
to reduce toxicity, but this could be questioned based on GOG 262 ducted to determine if there is a benefit of maintenance therapy in
results discussed above.69 Others substitute carboplatin (AUC 5–6) these patients immediately following their primary treatment in an
for cisplatin in the regimen and the same caveats regarding lack of effort to decrease the relapse rate. These were all negative and there
evidence apply.69 The role of intraperitoneal carboplatin is being eval- is no evidence to support maintenance chemotherapy after comple-
uated in JGOG and the results should be available in the near future. tion of first-­line therapy.
Bevacizumab 7.5–15 mg/kg every 3 weeks may be added to these
regimens.77,78 Two studies have reported a modest, but statistically
6.4 | PARP inhibitors
significant increase in progression-­free survival in patients receiving
maintenance bevacizumab following carboplatin, paclitaxel, and con- There is good evidence to support the role of PARP inhibitors as main-
current bevacizumab.77,78 There is no evidence as yet to demonstrate tenance therapy following response to chemotherapy in patients with
an overall survival benefit, but a subgroup analysis of the International platinum-­sensitive recurrent ovarian cancer, as well as monotherapy
Collaboration on Ovarian Neoplasms 7 (ICON7) trial reported an in selected patients with recurrent ovarian cancer.81–85 Patients with
improved median survival (30.3 vs 39.4 months) in patients with sub- BRCA mutations (both germline and somatic) have the greatest ben-
optimal Stage III and Stage IV.77 The role, optimal dose (7.5 mg/kg vs efit, but a subset of patients with tumors with homologous recombi-
15 mg/kg), timing (primary vs recurrent disease), and duration of treat- nation deficiency (HRD) also derive benefit from treatment with PARP
ment of bevacizumab are still debatable. inhibitors; the ongoing challenge is how best to identify these patients.
van Driel et al.79 recently reported results of a randomized trial in The results of these trials are summarized in Table 4.83–85 Readers are
which 245 patients with Stage III epithelial ovarian cancer who had directed to the chapter on targeted therapy in this Supplement by
received 3 cycles of neoadjuvant chemotherapy underwent interval Basu et al.86 for further discussion of PARP inhibitors.
debulking surgery. These patients were then randomized to receive
either 3 more cycles of paclitaxel plus carboplatin with or without
7 | SECONDARY SURGERY
hyperthermic intraperitoneal chemotherapy (HIPEC). The addi-
tion of HIPEC to interval cytoreductive surgery resulted in longer
7.1 | Second-­look laparotomy
recurrence-­free survival (14.2 vs 10.7 months) and overall survival
(45.7 vs 33.9 months) and did not result in higher rates of adverse A second-­look laparotomy (or laparoscopy) was previously performed
effects. These findings are provocative and raise important questions. in patients who have no clinical evidence of disease after completion
|
68       Berek ET AL.

T A B L E   4   Progression-­free survival endpoint in the three phase recurrent disease improves overall survival or quality of life. In asymp-
trials of maintenance PARP inhibitors. tomatic patients with CA125 progression and small volume disease or
no radiological evidence of recurrence, it is appropriate to delay start-
PARP inhibitor Placebo
progression-­ progression-­ ing chemotherapy. However, there may be a subset of patients who
free survival free survival Hazard are suitable for secondary debulking surgery at the time of recurrence.
Study (months) (months) ratio The objectives of follow-­up include:
SOLO 283 19.1 5.5 0.3
NOVA84 1. Early recognition and prompt management of treatment-related
gBRCA 21 5.5 0.27 complications, including provision of psychological support.
Non-­BRCA 9.3 3.9 0.45 2. Early detection of symptoms or signs of recurrent disease.

Non-­BRCA HRD+ 12.9 3.8 0.38


3. Collection of data regarding the efficacy of any treatment and the
85 complications associated with those treatments in patients treated
ARIEL 3
in clinical trials.
gBRCA 16.6 5.4 0.23
4. Promotion of healthy behavior, including screening for breast can-
HRD+ (includes WT/ 13.6 5.4 0.32
cer in patients with early stage disease, and screening for cervical
gBRCA)
cancer in patients having conservative surgery.

of first-­line chemotherapy to determine response to treatment. There are no evidence-­based guidelines regarding the appropriate
Although of prognostic value, it has not been shown to influence sur- follow-­up schedule. During the first year following treatment, patients
vival, and is no longer recommended as part of the standard of care.87 are seen every 3 months with a gradual increase in intervals to every
Level of Evidence C 4–6 months after 2 years and then annually after the fifth year. At
each follow-­up, the patient should have her history retaken, including
any change in family history of cancers and attention to any symptoms
7.2 | Secondary cytoreduction
that could suggest recurrence; a physical and pelvic examination should
Secondary cytoreduction may be defined as an attempt at cytoreduc- be performed. This is an opportunity to refer appropriate patients for
tive surgery at some stage following completion of first-­line chemo- genetic testing if it was not done at diagnosis or during treatment. The
therapy. Retrospective studies suggest that patients benefit if all CA125 has traditionally been checked at regular intervals, but there
macroscopic disease can be removed, which usually means patients has been debate regarding the clinical benefit of using CA125 progres-
with a solitary recurrence. Patients with a disease-­free interval longer sion alone as a trigger for initiating second-­line chemotherapy. A large
than 12–24 months and those with only 1–2 sites of disease appear to MRC OV05-­EORTC 55955 study showed that treating asymptomatic
derive most benefit.88,89 The role of secondary cytoreductive surgery patients with recurrent ovarian cancer with chemotherapy on the basis
is being evaluated in randomized clinical trials. The role of secondary of CA125 progression alone did not improve survival and early treatment
debulking surgery has been addressed in the DESKTOP III trial and in asymptomatic patients had a negative impact on quality of life.91 This
the results recently presented by Dubois on behalf of the AGO.90 This study has generated considerable debate regarding the use of CA125
study included patients with a progression-­free survival of greater than for follow-­up, but most agree that it is reasonable not to immediately
6 months after first-­line chemotherapy and who were considered to initiate treatment unless there is a clear clinical indication to do so. The
be good candidates for surgery based on a positive AGO Study Group timing of treatment should be based on symptoms as well as clinical and
score, defined as an ECOG performance status score of zero, ascites of radiological findings. Imaging tests such as ultrasonography of the pelvis,
500 mL or less, and complete resection at initial surgery. Du Bois et al.90 CT, MRI, and/or positron emission tomography (PET) scans should be
reported that the median progression-­free survival in 204 women who performed only when the clinical findings or the tumor markers suggest
met this criteria and who were randomized to undergo surgery fol- possible recurrence.
lowed by chemotherapy was 19.6 months, compared with 14 months There appears to be no benefit to initiating chemotherapy in an
in 203 women who were randomized to receive only second-­line chem- asymptomatic patient with recurrent disease based only on rising
otherapy. The primary endpoint of the study is overall survival, which CA125 levels in the absence of clinical symptoms or radiological evi-
will only be available in a few years. Level of Evidence C dence of recurrence. In asymptomatic patients with small volume dis-
ease and no radiological evidence of recurrence, close observation is a
reasonable option, as well as entry into an appropriate clinical trial or
8 |  FOLLOW-­U P FOR MALIGNANT possibly a trial of tamoxifen may be considered.
EPITHELIAL TUMORS A Cochrane database systematic review of tamoxifen in unse-
lected women with recurrent ovarian cancer reported a 10% objective
There is no evidence to show that intensive clinical monitoring dur- response and a 32% disease stabilization rate.92 The patients treated
ing follow-­up after completion of primary surgery and chemotherapy were heterogeneous and included asymptomatic patients with rising
with early initiation of chemotherapy in asymptomatic women with CA125 levels, and symptomatic patients with chemotherapy-­resistant
Berek ET AL. |
      69

disease who had been heavily pretreated and had a poor performance progression-­free survival for a combination of carboplatin and pacli-
status. GOG 198 compared tamoxifen and thalidomide in women with taxel versus single-­agent carboplatin.96 Level of Evidence A
recurrent FIGO Stage III or IV epithelial ovarian, tubal, or peritoneal can- For patients with neurotoxicity, gemcitabine97 or liposomal
cer who had completed first-­line chemotherapy, and who subsequently doxorubicin98 may be substituted for paclitaxel. A large GCIG study
had Gynecologic Cancer InterGroup (GCIG) documented CA125 pro- (CALYPSO) compared carboplatin and liposomal doxorubicin (CD) with
gression. The study reported that women who received thalidomide carboplatin and paclitaxel (CP) in 976 patients.99 The CD arm had sta-
had a 31% increased risk of disease progression (HR 1.31), compared tistically superior progression-­free survival compared with the CP arm,
with those who were given tamoxifen.93 The median progression-­free with a median progression-­free survival of 11.3 versus 9.4 months,
survival was 3.2 months in the thalidomide group versus 4.5 months respectively. There was no significant difference in the overall survival
in the tamoxifen group. This suggests that tamoxifen may have a role between the treatment groups. Median overall survival was 33 ver-
in selected patients with a rising CA125 level, and the relationship sus 30.7 months for the CP and CD arms, respectively. The CD arm
between estrogen receptor positivity and benefit of tamoxifen in this was better tolerated with less severe toxicities, and this combination
patient population is being evaluated in current studies. is now widely used. Level of Evidence A
There is evidence that the addition of bevacizumab to the regimen
of carboplatin and gemcitabine improves progression-­free survival com-
9 | CHEMOTHERAPY FOR RECURREN T pared with carboplatin and gemcitabine in platinum-­sensitive disease.
EPITHELIAL MALIGNANCIES In the OCEANS study,100 484 patients with platinum-­sensitive disease
were randomly assigned to carboplatin (AUC 4 on day 1) and gemcitabine
The majority of patients who present with advanced epithelial cancers 1000 mg/m2 on days 1 and 8) with or without bevacizumab (15 mg/kg
of the ovary/fallopian tube/peritoneum will relapse with a median on day 1) with every 21 days cycles. Bevacizumab could be given con-
time to recurrence of 16 months. Patients with recurrent ovarian can- currently with chemotherapy for a maximum of 10 cycles followed by
cer constitute a heterogeneous group with a variable prognosis, and a bevacizumab alone until progression of disease or toxicity. The addition
variable response to further treatment. The most widely used clinical of bevacizumab to carboplatin and gemcitabine resulted in an improve-
surrogate for predicting response to subsequent chemotherapy and ment in progression-­free survival (12 vs 8 months; HR 0.48; 95% CI
prognosis has been the progression-­free interval or the “platinum-­ 0.39–0.61); however, there was no difference in overall survival between
free interval,” which is defined as the time from cessation of primary the two arms. Treatment with bevacizumab was associated with higher
platinum-­based chemotherapy to disease recurrence or progres- rates of serious hypertension (17% vs <1%), proteinuria grade 3 or higher
sion.94,95 This has been useful to define specific patient populations, (9% vs 1%), and noncentral nervous system bleeding (6% vs 1%).100
but it has a number of limitations and depends on how patients are For patients with definite platinum-­resistant disease, enrollment on
followed. In particular, it depends on how recurrence is detected and available clinical trials or treatment with nonplatinum chemotherapy
defined. Patients with a treatment-­free interval of less than 6 months should be considered. There are a number of chemotherapy options
are classified as platinum resistant and generally treated with including liposomal doxorubicin,101 topotecan,101 etoposide,102,103 and
nonplatinum-­based chemotherapy, while those with a treatment-­ gemcitabine.104,105 The reported response rates are low, about 10%,
free interval of more than 6 months are considered to be platinum with a median time to progression of 3–4 months and a median sur-
sensitive and commonly treated with platinum-­based chemotherapy. vival of 9–12 months. Over the last 5 years there have been a number
Patients who progress while on treatment or within 4 weeks of stop- of trials carried out with new agents in patients with platinum-­resistant
ping chemotherapy are classified as platinum refractory.94,95 ovarian cancer, including epothilones, trabectedin106 and perme-
There have been modifications to these definitions, and time trexed107 with no significant increase in response rates or progression-­
to progression or recurrence rather than treatment-­free interval or free survival. No new cytotoxic agent has been approved to treat
platinum-­free interval has been used to define specific patient pop- recurrent ovarian cancer for many years. The role of angiogenesis
ulations. There has been significant change in practice over the last inhibitors in platinum-­resistant ovarian cancer is ­discussed below.
20 years and patients have been routinely followed with regular CA125 The optimal management of a patient with platinum-­resistant or
testing after completion of chemotherapy. For example, the “platinum-­ refractory disease is complex and requires a careful assessment of
resistant” subgroup may include asymptomatic patients with CA125 the patient’s performance status, symptoms, and extent of disease.
progression alone at 3 months post chemotherapy or radiological Attention to symptom control and good palliative care is an essential
evidence of recurrence as well as those who are symptomatic with component of management.
clinical recurrence. The Fourth Ovarian Cancer Consensus Conference With very few exceptions, recurrent disease is not curable and the
reached agreement that distinct patient populations should be based aim of treatment is to maintain quality of life and palliate symptoms
on the interval from last platinum therapy and the time to progression. particularly in patients with platinum-­resistant ovarian cancer.108 There
The progression-­free interval is defined from the last date of platinum are many potential treatment options, including chemotherapy, angio-
dose until progressive disease is documented.94,95 genesis inhibitors, radiation therapy, or surgery in selected patients
For patients whose disease is considered platinum-­sensitive, the and inclusion in clinical trials.89 There is a subset of patients who
ICON 4 study showed advantage in terms of overall survival and may benefit from secondary surgical debulking, but they constitute a
|
70       Berek ET AL.

minority. The role of secondary surgical debulking is being addressed in suboptimal debulking and were evaluable for response evaluation.118
prospective randomized clinical trials. Level of Evidence C The response rate was higher than other studies at 23.1% in this small
subset of patients with LGSOC compared with 90.1% in patients with
HGSOC. The majority of patients with LGSOC will relapse despite
9.1 | PARP inhibitors as monotherapy in patients
treatment and have a relatively long survival (median overall survival
with recurrent ovarian cancer
of 82 months). These patients are often treated with multiple agents
Olaparib is FDA approved for the treatment of patients with gBRCA-­ over many years for recurrent disease with variable degrees of benefit
mutated recurrent ovarian cancer who have received three or more and the impact of treatment on survival is unclear.118
109,110
prior lines of chemotherapy. The FDA granted approval on the
basis of the response rate in a single-­arm study of olaparib in patients
10.1 | Management of low malignant potential
with BRCA mutations and with a wide range of different cancers. The
(borderline) tumors
response rate was 34% in heavily pretreated BRCA-­positive patients
with platinum-­resistant recurrent ovarian cancer and the median Compared with invasive epithelial cancers, borderline tumors tend
110
progression-­free survival was 7.9 months. to affect a younger population and constitute 15% of all epithelial
Rucaparib is also approved for treatment of BRCA-­mutation-­ tumors of the ovary.119 Nearly 75% of these are Stage I at the time of
associated advanced ovarian cancer after completion of treatment with diagnosis. The following can be said for these tumors120:
two or more chemotherapy regimens regardless of whether patients
are platinum-­sensitive or resistant.111 Rucaparib’s approval was based 1. The diagnosis must be based on the pathology of the
primarily on efficacy data from 106 patients with BRCA-­associated primary tumor.
recurrent ovarian cancer who had prior treatment with two or more 2. Extensive sectioning of the tumor is necessary to rule out invasive
chemotherapy regimens and safety data from 377 patients with ovarian cancer.
cancer treated with rucaparib 600 mg orally twice daily on two open-­ 3. The prognosis of these tumors is extremely good, with a 10-year
label, single-­arm trials.112 Investigator-­assessed objective response rate survival of about 95%.
was 54% and the median duration of response was 9.2 months.112 4. Invasive cancers that arise in borderline tumors are often indolent and
generally have a low response to platinum-based chemotherapy.
5. Spontaneous regression of peritoneal implants has been observed.
10 |  MANAGEMENT OF EPITHELIAL 6. Early stage, serous histology, and younger age at diagnosis are
TUMORS OF LOW-­G RAD E associated with a more favorable prognosis.
SEROUS CANCERS 7. Although gross residual disease after primary laparotomy is associ-
ated with poorer prognosis, mortality from the disease remains low.
Low-­grade serous cancers (LGSCs) comprise 5% to 10% of serous 8. Those patients who have invasive implants in the omentum or
ovarian cancers and up to 8% of all ovarian cancers.113 They are other distant sites are more likely to recur earlier. The role of cyto-
typically diagnosed at a younger age than in women with high-­grade toxic chemotherapy is questionable as the response rates are low.
serous ovarian cancer (HGSOC), with a median age of 47–54 years at
diagnosis, and are characterized by a relatively indolent behavior and The causes of death include complications of disease (e.g. small
resistance to cytotoxic chemotherapy.114 In contrast to HGSOC they bowel obstruction) or complications of therapy, and only rarely malig-
do not have TP53 mutations, but may have KRAS or BRAF mutations, nant transformation. The mainstay of treatment is primary surgical
114–116
and activation of the Ras-­Raf-­MEK-­ERK signaling pathway. staging and cytoreduction. For patients with Stage I disease who want
Most patients with low-­grade serous ovarian cancer (LGSOC) have to preserve fertility, conservative surgery with unilateral salpingo-­
advanced-­stage disease at initial diagnosis and the surgical manage- oophorectomy can be considered after intraoperative inspection of the
ment is similar to patients with high-­grade cancers, with attempts at contralateral ovary to exclude involvement.121 For patients with only one
total resection of tumor—with the exception of fertility-­sparing surgery ovary, or bilateral cystic ovaries, a partial oophorectomy or cystectomy
in younger women with tumors confined to the ovary. Neoadjuvant can be considered for fertility preservation. For all other patients, total
platinum-­based chemotherapy for advanced-­stage LGSOC or perito- hysterectomy and bilateral salpingo-­oophorectomy are recommended,
neum was associated with a radiological response rate of 4%, which with maximal cytoreduction if the disease is metastatic.
is much lower than response rates of up to 80% in patients with Patients with borderline tumors in all stages of disease should
HGSOC.117 Similarly, the response rates to chemotherapy have been be treated with surgery. A small percentage of patients with invasive
reported to be low in a number of studies and the rate was only 3.7 implants may respond to chemotherapy but the response to che-
(4.9% in patients with platinum-­sensitive disease and 2.1% in those motherapy is low. Uncommonly, some patients recur early and have
with platinum-­resistant disease) in a report of patients with recurrent higher-­grade invasive cancers and may benefit from chemotherapy.122
114
LGSC. A recent retrospective, exploratory, case-­control analysis In patients with late recurrence of the disease, secondary cytore-
of over 5000 patients receiving adjuvant chemotherapy in clinical duction should be considered, and chemotherapy given only if invasive
trials included 145 patients (2.8%) with LGSOC, of whom 37 had disease is present histologically.
Berek ET AL. |
      71

Hormonal therapy has been reported to be associated with clinical and/or AMH appear to be reliable markers during follow-­up for early
benefit in recurrent and metastatic borderline ovarian tumors as well detection of residual or recurrent disease.132
as LGSC. Hormonal therapy was reported to have a response rate of There is no evidence-­based preference for inhibin B or AMH as a
9% in a retrospective analysis of 64 patients with recurrent LGSC.123 tumor marker.133 Serum inhibin is a useful tumor marker in postmeno-
In 26 patients with LGSC of the ovary or peritoneum, adjuvant hor- pausal women. Level of Evidence C
mone therapy following debulking surgery was associated with a
median progression-­free survival of 22 months and recurrence rate of
14.8%.124 In this small study, survival of the patients treated with adju- 12 | MANAGEMENT OF GERM
vant hormonal therapy was not significantly different to an age-­ and CELL MALIGNANCIES
stage-­matched control group of patients with LGSC treated with sur-
gery and adjuvant chemotherapy. A recent retrospective analysis was This group of ovarian tumors consists of a variety of histologically
reported of 203 patients with LGSC of the ovary or peritoneum who different subtypes that are all derived from the primitive germ cells
received either maintenance/adjuvant hormonal treatment or obser- of the embryonic gonad. Malignant germ cell tumors represent a
vation, based on physician discretion, following primary cytoreductive relatively small proportion of all ovarian tumors. Prior to advances in
surgery and platinum-­based chemotherapy.125 Patients who received chemotherapy, the prognosis for these aggressive tumors was poor.
adjuvant hormonal therapy had significantly longer median progression-­ The use of platinum-­based chemotherapeutic regimes has made germ
free survival (64.9 vs 26.4 months) compared with the patients in the cell malignancies among the most highly curable cancers.127
observation group, without significant prolongation of overall survival
(115.7 vs 102.7 months). The role of maintenance/adjuvant hormonal
12.1 | Presentation
therapy in patients with LGSC will soon be tested in a large NRG trial.
Follow-­up of patients with no evidence of disease is the same as for These are most common ovarian tumors in the second and third decades
those with malignant epithelial carcinomas, but at less frequent inter- of life. They are frequently diagnosed by finding a palpable abdominal
vals. If the contralateral ovary has been retained, it should be followed mass in a young woman who complains of abdominal pain. The following
by transvaginal ultrasonography, at least on an annual basis.1,120,126 are the symptoms of germ cell tumors in order of frequency127:
Level of Evidence C
1. Acute abdominal pain.
2. Chronic abdominal pain.
11 |  MANAGEMENT OF GRANULOSA 3. Asymptomatic abdominal mass.
CELL TUMORS 4. Abnormal vaginal bleeding.
5. Abdominal distention.
Granulosa cell tumors account for about 70% of sex-­cord stromal
tumors and 3%–5% of all ovarian neoplasms.2 There are two types
of granulosa cell tumors: the juvenile and the adult types. Because of
12.2 | Histologic classification
the high estrogen production, the juvenile type typically presents with
sexual precocity, while the adult type may present with postmeno- The classification of germ cell tumors of the ovary is important to
pausal bleeding. The majority of patients are diagnosed with Stage I determine prognosis and for treatment with chemotherapy. Germ cell
tumors. The peak incidence is in the first postmenopausal decade.2,127 tumors are classified as follows2,127:
Granulosa cell tumors are generally indolent (i.e. with a tendency
to late recurrence). Stage at diagnosis is the most important prognostic 1. Dysgerminoma.
factor. Other prognostic factors include age at diagnosis, tumor size, 2. Embryonal carcinoma.
and histologic features. If metastatic, adequate cytoreduction is the 3. Polyembryoma.
mainstay of treatment. If the patient is young and the disease is con- 4. Teratoma (immature; mature; mature with carcinoma [squamous
fined to one ovary, conservative surgery should be performed.128,129 cell, carcinoid, neuroectodermal, malignant struma, etc.]).
The infrequency of the disease, and its protracted course, has 5. Extraembryonal differentiation (choriocarcinoma; endodermal
resulted in a lack of prospective studies. There is no evidence that adju- sinus tumor [yolk sac tumor]).
vant chemotherapy or radiotherapy improves the results of surgery alone
for Stage I disease. The value of postoperative adjuvant chemotherapy
for higher-­risk Stage I disease (tumor size >10 cm, capsule rupture, high
12.3 | Diagnosis, staging, and surgical management
mitotic count) is uncertain, and has not been tested in randomized stud-
ies. Platinum-­based chemotherapy is used for patients with advanced Ovarian germ cell tumors are staged similarly to epithelial carcinomas,
or recurrent disease, with an overall response rate of 63%–80%.129–131 although the staging system used for male germ cell tumors is prob-
Follow-­up is clinical. For patients with elevated levels of inhibin ably more useful. The approach to treatment is based on the principles
B and/or AMH at initial diagnosis of granulosa cell tumors, inhibin B of management of metastatic germ cell tumors of the testis (i.e. low,
|
72       Berek ET AL.

intermediate, and poor risk). Dysgerminoma is the equivalent of semi- T A B L E   5   Follow-­up regime for Stage I germ cell malignancies.a
noma in testicular cancer.134 It is exquisitely sensitive to platinum-­
Regimen Description
based chemotherapy and is radiosensitive. The cure rate is high
Surveillance Baseline CT chest, abdomen, and pelvis, if
irrespective of the stage. The other histologic subtypes are equivalent
not performed preoperatively
to nonseminomatous testicular cancer. The aggressiveness of the dis-
Repeat CT or MRI, abdomen and pelvis at
ease is dependent on the type, the most aggressive being endodermal
3 months after surgery
sinus and choriocarcinoma, but with combination chemotherapy, they
Repeat CT or MRI abdomen plus pelvis at
are highly curable.135–139 12 months
As chemotherapy can cure the majority of patients, even with
Pelvic ultrasound alternate visits (not
advanced disease, conservative surgery is standard in all stages of all when having CT scan) for 2 years if
germ cell tumors. Conservative surgery means laparotomy with careful non-­dysgerminoma and for 3 years if
examination and biopsy of all suspicious areas, with limited cytore- dysgerminoma

duction, thereby avoiding major morbidity. The uterus and the con- Chest X-­ray at alternate visits
tralateral ovary should be left intact. Wedge biopsy of a normal ovary Clinical examination
is not recommended as it defeats the purpose of conservative ther- 1 year Monthly
apy by potentially causing infertility. Patients with advanced disease 2nd year 2 monthly
may benefit from 3 to 4 cycles of neoadjuvant chemotherapy using 3rd year 3 monthly
BEP (bleomycin, etoposide, cisplatin [platinum]) regimen with preser- 4th year 4 monthly
vation of fertility.140 Patients who receive conservative surgery with
Years 5–10 6 monthly
the preservation of one ovary retain acceptable fertility rates despite
Tumor marker followup Samples: serum AFP and hCG, LDH and
adjuvant treatment with chemotherapy. There has been no report of CA 125 (regardless of initial value)
higher adverse obstetric outcome or long-­term unfavorable sequelae
0–6 mo 2 weekly
in the offspring.141–144
7–12 mo 4 weekly
Secondary surgery is of no proven benefit, except in those patients
12–24 mo 8 weekly
whose tumor was not completely resected at the initial operation and
24–36 mo 12 weekly
who had teratomatous elements in their primary tumor. Surgical resec-
36–48 mo 16 weekly
tion of residual masses may be beneficial in such patients, as there may
be mature teratomatous nodules that can continue to increase in size 48+ mo 6 monthly until year 10

(growing teratoma syndrome), and more rarely can undergo malignant Abbreviations: AFP, alpha-­fetoprotein; hCG, human chorionic gonadotro-
transformation over time to an incurable malignancy, e.g. squamous pin; LDH, lactate dehydrogenase.
a
Adapted from Patterson et al.146
cell carcinoma.145

1. Etoposide (E) 100 mg/m2 IV per day for 5 days every 3 weeks


12.4 | Postoperative management and follow-­up of
for 3 cycles.
dysgerminoma
2. Cisplatin (P) 20 mg/m2 IV per day for 5 days every 3 weeks for 3
Patients with Stage IA disease may be observed after surgery. A small cycles.
proportion of patients may recur, but they can be treated success- 3. Bleomycin (B) 30 000 IU IV/IM on days 1/8/15 for 12 weeks
fully at the time of recurrence with a high rate of cure. Patients with (Optional) (Note: bleomycin is dosed in International Units). If bleo-
disease beyond the ovary should receive adjuvant chemotherapy. mycin is omitted, then 4 cycles of EP are commonly used. Note that
Although radiation therapy is effective, ovarian failure makes it unde- various schedules of bleomycin have been used.
sirable for patients with an intact ovary.
A follow-­up surveillance regime for patients with Stage 1A dys- When there is bulky residual disease, it is common to give 3–4
germinoma is outlined in Table 5. This schedule is based on the expe- courses of BEP chemotherapy.148 Level of Evidence B
rience managing seminomas in males and the reports by Patterson The optimal follow-­up schedule has not been clinically investigated
et al.146 and Dark et al.147 This pragmatic follow-­up schedule and has in ovarian germ cancers and the frequency of visits and investigations
not been tested in randomized trials. is controversial. Patients who have Stage I tumors and are offered
surveillance need to be seen regularly and one option is to utilize the
follow-­up regimen presented above.147 Patients who have had chemo-
12.4.1 | Chemotherapy for dysgerminoma
therapy have a lower risk of recurrence and the frequency of CT scans
Dysgerminoma is extremely sensitive to chemotherapy, and treat- can be reduced, which is similar to the approach for testicular germ
ment with chemotherapy cures the majority of patients, even with cell tumors.146 Each follow-­up visit should involve taking a medical his-
advanced disease.127,148 The recommended chemotherapy regimen is tory, physical examination, and tumor marker determination. Although
as follows: tumor markers are important, radiological imaging is also pertinent,
Berek ET AL. |
      73

especially for patients whose tumor markers were not raised at diag- The sarcomatous component is derived from the carcinoma or from a
147
nosis. CT or MRI scans should be performed as clinically indicated. stem cell that undergoes divergent differentiation. Thus, ovarian carci-
Patients who have not received chemotherapy should be followed nosarcomas are best regarded as metaplastic carcinomas.
closely. Ninety percent of relapses in these patients occur within the Pure sarcomas are very rare and should be treated according to
first 2 years. At relapse, with few exceptions, these patients can be the specific histologic subtype. These rare sarcomas include fibrosar-
successfully treated.147 Level of Evidence D comas, leiomyosarcomas, neurofibrosarcomas, rhabdomyosarcomas,
chondrosarcomas, angiosarcomas, and liposarcomas. Their manage-
ment is not discussed here.
12.5 | Postoperative management and follow-­
Patients with early stage MMMTs have a better outcome than
up of nondysgerminoma germ cell malignancies
those with advanced stage disease, but the overall prognosis is poor.
These tumors are highly curable with chemotherapy, even with They should be managed similarly to high-­grade pelvic serous cancers.
advanced disease. Patients with Stage IA grade 1–2 immature teratoma Their rarity prohibits any prospective randomized trials.
have a very good prognosis and should be only observed after primary The principles of surgical management of ovarian MMMTS are the
conservative surgery. Adjuvant chemotherapy does not appear to add same as for high-­grade pelvic serous cancers.127 Following surgery,
any survival benefit in this subgroup of patients. All other patients patients should receive platinum-­based chemotherapy.127,147,148 The
with nondysgerminomas, and higher-­stage and higher-­grade immature follow-­up schedule is as recommended for epithelial malignancies.
teratomas, should receive postoperative adjuvant chemotherapy.127 Level of Evidence C
The recommended chemotherapy regimen is etoposide 100 mg/
m2 per day for 5 days with cisplatin 20 mg/m2 per day for 5 days,
AU T HO R CO NT R I B U T I O NS
and bleomycin at 30 000 IU IM/IV on days 1, 8, and 15 for a total
of 12 weeks of treatment. For patients with good prognosis disease, JB, SK, LK, and MF reviewed and updated the chapter on cancer
3 cycles of BEP are recommended, while patients with intermediate/ of the ovary, fallopian tube, and peritoneum published in the 2015
poor risk disease should receive 4 cycles of BEP.127 Cancer Report.
Patients who relapse after BEP may still attain a durable remission
and cure with second-­line chemotherapy regimens such as paclitaxel–
ifosfamide–cisplatin (TIP).137 High-­dose chemotherapy and autolo- AC KNOW L ED G M ENTS

gous marrow rescue may be considered in selected patients. These This chapter updates the information published in the FIGO Cancer
patients should be managed in specialized units. Report 2015 (Berek JS, Crum C, Friedlander M. Cancer of the ovary,
After chemotherapy, patients with metastatic immature teratomas fallopian tube, and peritoneum. Int J Gynecol Obstet 2015;131:S111–
can sometimes have residual masses, which are composed entirely S122), with approval granted by the original authors.
of mature elements. These masses can grow, and should be resected
after the completion of chemotherapy.149 Level of Evidence B
All patients should have lactate dehydrogenase (LDH), alpha-­ CO NFL I C TS O F I NT ER ES T
fetoprotein (AFP), and human gonadotropin (beta hCG) to monitor
The authors have no conflicts of interest to declare.
response to treatment. All patients treated with chemotherapy should
be followed-­up with medical history, physical examination, and appro-
priate tumor markers in the same way as dysgerminomas. CT or MRI REFERENCES
scans should be performed as clinically indicated.122
1. Berek JS, Friedlander M, Hacker NF. Epithelial ovarian, fallopian
Relapses in patients usually occur within the first 2 years after tube, and peritoneal cancer. In: Berek JS, Hacker NF, eds. Berek and
diagnosis127,137 Level of Evidence D Hacker’s Gynecologic Oncology, 6th edn. Philadelphia: Lippincott
Williams and Wilkins; 2015:464–529.
2. Scully RE, Young RH, Clements PB. Tumors of the Ovary, Maldeveloped
Gonads, Fallopian Tube, and Broad Ligaments. Atlas of Tumor Pathology.
13 |  SARCOMA OF THE OVARY Third series. Washington, DC: Armed Forces Institute of Pathology;
1998.
Ovarian sarcomas are rare and occur primarily in postmenopausal 3. Kindelberger DW, Lee Y, Miron A, Hirsch MS, Feltmate C, Medeiros
patients.127,150 Nevertheless, accurate diagnosis and differentiation F, et  al. Intraepithelial carcinoma of the fimbria and pelvic serous
carcinoma: Evidence for a causal relationship. Am J Surg Pathol.
from other types of primary ovarian cancer are important, as the prog-
2007;31:161–169.
nosis is generally poor. 4. Callahan MJ, Crum CP, Medeiros F, Kindelberger DW, Elvin JA,
There are two types of sarcoma. Malignant mixed Müllerian Garber JE, et  al. Primary fallopian tube malignancies in BRCA-­
tumors (MMMTs), the more common of the two, are biphasic tumors positive women undergoing surgery for ovarian cancer risk reduc-
tion. J Clin Oncol. 2007;25:3985–3990.
composed of both carcinomatous and sarcomatous elements.150,151
5. Kurman RJ, Shih IM. Pathogenesis of ovarian cancer: Lessons from
Most authors agree that most MMMTs are monoclonal in origin and morphology and molecular biology and their clinical implications. Int
should be thought of and managed as a high-­grade epithelial cancer. J Gynecol Pathol. 2008;27:151–160.
|
74       Berek ET AL.

6. Crum CP, Drapkin R, Miron A, Ince TA, Muto M, Kindelberger DW, 27. Negri E, Franceschi S, Tzonou A, Booth M, La Vecchia C, Parazzini F,
et al. The distal fallopian tube: A new model for pelvic serous car- et al. Pooled analysis of 3 European case-­control studies of epithelial
cinogenesis. Curr Opin Obstet Gynecol. 2007;19:3–9. ovarian cancer: I. Reproductive factors and risk of epithelial ovarian
7. Carlson JW, Miron A, Jarboe EA, Parast MM, Hirsch MS, Lee Y, et al. cancer. Int J Cancer. 1991;49:50–56.
Serous tubal intraepithelial carcinoma: Its potential role in primary 28. Lynch HT, Watson P, Lynch JF, Conway TA, Fili M. Hereditary
peritoneal serous carcinoma and serous cancer prevention. J Clin ovarian cancer. Heterogeneity in age at onset. Cancer. 1993;71(2
Oncol. 2008;26:4160–4165. Suppl):573–581.
8. Aziz S, Kuperstein G, Rosen B, Cole D, Nedelcu R, McLaughlin J, et al. 29. Struewing JP, Hartge P, Wacholder S, Baker SM, Berlin M, McAdams
A genetic epidemiological study of carcinoma of the fallopian tube. M, et  al. The risk of cancer associated with specific mutations
Gynecol Oncol. 2001;80:341–345. of BRCA1 and BRCA2 among Ashkenazi Jews. N Engl J Med.
9. Levanon K, Crum C, Drapkin R. New insights into the pathogene- 1997;336:1401–1408.
sis of serous ovarian cancer and its clinical impact. J Clin Oncol. 30. Risch HA, McLaughlin JR, Cole DE, Rosen B, Bradley L, Fan I, et al.
2008;26:5284–5293. Population BRCA1 and BRCA2 mutation frequencies and cancer
10. Deffieux X, Morice P, Thoury A, Camatte S, Duvillard P, Castaigne penetrances: A kin-­cohort study in Ontario, Canada. J Natl Cancer
D. Anatomy of pelvic and para-­aortic nodal spread in patients with Inst. 2006;98:1694–1706.
primary fallopian tube carcinoma. J Am Coll Surg. 2005;200:45–48. 31. Chetrit A, Hirsh-Yechezkel G, Ben-David Y, Lubin F, Friedman E,
11. Baekelandt M, Jorunn Nesbakken A, Kristensen GB, Trope CG, Abeler Sadetzki S, et al. Effect of BRCA1/2 mutations on long-­term survival
VM. Carcinoma of the fallopian tube. Cancer. 2000;89:2076–2084. of patients with invasive ovarian cancer: The national Israeli study of
12. Burghardt E, Girardi F, Lahousen M, Tamussino K, Stettner H. ovarian cancer. J Clin Oncol. 2008;26:20–25.
Patterns of pelvic and paraaortic lymph node involvement in ovarian 32. Medeiros F, Muto MG, Lee Y, Elvin JA, Callahan MJ, Feltmate C,
cancer. Gynecol Oncol. 1991;40:103–106. et al. The tubal fimbria is a preferred site for early adenocarcinoma
13. Bankhead CR, Kehoe ST, Austoker J. Symptoms associated in women with familial ovarian cancer syndrome. Am J Surg Pathol.
with diagnosis of ovarian cancer: A systematic review. BJOG. 2006;30:230–236.
2005;112:857–865. 33. Norquist BM, Harrell MI, Brady MF, et  al. Inherited mutations in
14. Lataifeh I, Marsden DE, Robertson G, Gebski V, Hacker NF. women with ovarian carcinoma. JAMA Oncol. 2016;2:482–490.
Presenting symptoms of epithelial ovarian cancer. Aust N Z J Obstet 34. Walsh T, Casadei S, Lee MK, et  al. Mutations in 12 genes for
Gynecol. 2005;45:211–214. inherited ovarian, fallopian tube, and peritoneal carcinoma iden-
15. Gilbert L, Basso O, Sampalis J, Karp I, Martins C, Feng J, et  al. tified by massively parallel sequencing. Proc Natl Acad Sci USA.
Assessment of symptomatic women for early diagnosis of ovarian 2011;108:18032–18037.
cancer: Results from the prospective DOvE pilot project. Lancet 35. Ryan NAJ, Bolton J, McVey RJ, Evans DG, Crosbie EJ. BRCA and
Oncol. 2012;13:285–291. lynch syndrome-­associated ovarian cancers behave differently.
16. Gilks CB, Irving J, Köbel M, Lee C, Singh N, Wilkinson N, et  al. Gynecol Oncol Rep. 2017;22:108–109.
Incidental nonuterine high-­grade serous carcinomas arise in 36. eviQ. Cancer treatments online [website]. https://www.eviq.org.au.
the fallopian tube in most cases: Further evidence for the Accessed April 3, 2018.
tubal origin of high–grade serous carcinomas. Am J Surg Pathol. 37. Society of Gynecologic Oncology. SGO Clinical Practice Statement:
2015;39:357–364. Genetic testing for ovarian cancer. https://www.sgo.org/clin-
17. Berek JS. Lymph-­node positive stage IIIC ovarian cancer: A separate ical-practice/guidelines/genetic-testing-for-ovarian-cancer/.
entity? Int J Gynecol Cancer. 2009;19(Suppl.2):S18–S20. Accessed April 3, 2018.
18. Kurman RJ, Carcangiu ML, Herrington CS, Young RH, eds. WHO 38. US Preventive Services Task Force, Grossman DC, Curry SJ, et  al.
Classification of Tumours of Female Reproductive Organs. Lyon, France: Screening for ovarian cancer: US Preventive Services Task Force rec-
IACR; 2014:11–40. ommendation statement. JAMA. 2018;319:588–594.
19. Bodurka DC, Deavers MT, Tian C, Sun CC, Malpica A, Coleman RL, 39. Menon U, Skates SJ, Lewis S, Rosenthal AN, Rufford B, Sibley K,
et al. Reclassification of serous ovarian carcinoma by a 2-­tier system: A et al. Prospective study using the risk of ovarian cancer algorithm to
Gynecologic Oncology Group Study. Cancer. 2012;118:3087–3094. screen for ovarian cancer. J Clin Oncol. 2005;23:7919–7926.
20. Edge SB, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A, eds. 40. Buys SS, Partridge E, Black A, Johnson CC, Lamerato L, Isaacs C, et al.
AJCC Cancer Staging Manual. Ovary. 7th edn. New York: Springer; Effect of screening on ovarian cancer mortality: The Prostate, Lung,
2010. Colorectal, and Ovarian (PLCO) cancer screening randomized con-
21. Soslow RA, Han G, Park KJ, Garg K, Olvera N, Spriggs DR, et  al. trolled trial. JAMA. 2011;305:2295–2303.
Morphologic patterns associated with BRCA1 and BRCA2 genotype 41. Committee on Gynecologic Practice. Committee opinion no.620:
in ovarian carcinoma. Mod Pathol. 2012;25:625–636. Salpingectomy for ovarian cancer prevention. Obstet Gynecol.
22. Kalloger SE, Köbel M, Leung S, Mehl E, Gao D, Marcon KM, et  al. 2015;125:279–281.
Calculator for ovarian carcinoma subtype prediction. Mod Pathol. 42. Trimbos JB, Vergote I, Bolis G, Vermorken JB, Mangioni C, Madronal
2011;24:512–521. C, et  al. Impact of adjuvant chemotherapy and surgical staging in
23. Roh MH, Yassin Y, Miron A, Mehra KK, Mehrad M, Monte NM, et al. early stage ovarian carcinoma: European Organisation for Research
High-­grade fimbrial-­ovarian carcinomas are unified by altered p53, and Treatment of Cancer-­Adjuvant Chemotherapy in Ovarian
PTEN and PAX2 expression. Mod Pathol. 2010;23:1316–1324. Neoplasm Trial. J Natl Cancer Inst. 2003;95:113–125.
24. Ayhan A, Kurman RJ, Yemelyanova A, Vang R, Logani S, Seidman JD, 43. Zanetta G, Chiari S, Rota S, Bratina G, Maneo A, Torri V, et  al.
et al. Defining the cut point between low-­grade and high-­grade ovar- Conservative surgery for stage I ovarian carcinoma in women of
ian serous carcinomas: A clinicopathologic and molecular genetic childbearing age. Br J Obstet Gynaecol. 1997;104:1030–1035.
analysis. Am J Surg Pathol. 2009;33:1220–1224. 44. Young RC, Walton LA, Ellenberg SS, Homesley HD, Wilbanks GD,
25. Siegel R, Ward E, Brawley O, Jemal A. Cancer statistics, 2011: The Decker DG, et al. Adjuvant therapy in stage I and stage II epithelial
impact of eliminating socioeconomic and racial disparities on prema- ovarian cancer. Results of two prospective randomized trials. N Engl
ture cancer deaths. CA Cancer J Clin. 2011;61:212–236. J Med. 1990;322:1021–1027.
26. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global can- 45. Bell J, Brady M, Lage J, Look KY, Spirtos N, Walker J, et  al. A
cer statistics. CA Cancer J Clin. 2011;61:69–90. Randomized phase III trial of three versus six cycles of carboplatin
Berek ET AL. |
      75

and aclitaxel as adjuvant treatment in early stage ovarian epithelial and cisplatin in patients with stage III and stage IV ovarian cancer. N
carcinoma: A Gynecologic Oncology Group study. Gynecol Oncol. Engl J Med. 1996;334:1–6.
2006;102:432–439. 62. Ozols RF, Bundy BN, Greer B, Greer BE, Fowler JM, Clarke-Pearson
46. Nagle CM, Francis JE, Nelson AE, Zorbas H, Luxford K, de Fazio A, et al. D, et  al. Phase III trial of carboplatin and paclitaxel compared cis-
Reducing time to diagnosis does not improve outcomes for women platin and paclitaxel in patients with optimally resected stage III
with symptomatic ovarian cancer: A report from the Australian ovarian cancer: A Gynecologic Oncology Group study. J Clin Oncol.
Ovarian Cancer Study Group. J Clin Oncol. 2011;29:2253–2258. 2003;21:3194–3200.
47. Sørensen SS, Mosgaard BJ. Combination of cancer antigen 125 and 63. Gemignani M, Hensley M, Cohen R, Venkatraman E, Saigo PE,
carcinoembryonic antigen can improve ovarian cancer diagnosis. Barakat RR. Paclitaxel-­based chemotherapy in carcinoma of the fal-
Dan Med Bull. 2011;58:A4331. lopian tube. Gynecol Oncol. 2001;80:16–20.
48. Hacker NF, Berek JS, Lagasse LD, Nieberg RK, Elashoff RM. Primary 64. Bookman MA, Brady MF, McGuire WP, Harper PG, Alberts DS,
cytoreductive surgery for epithelial ovarian cancer. Obstet Gynecol. Friedlander M, et  al. Evaluation of new platinum-­based treatment
1983;61:413–420. regimens in advanced-­stage ovarian cancer: A Phase III Trial of the
49. Bristow RE, Tomacruz RS, Armstrong DK, Trimble EL, Montz FJ. Gynecologic Cancer InterGroup. J Clin Oncol. 2009;27:1419–1425.
Survival effect of maximal cytoreductive surgery for advanced ovar- 65. Pignata S, Scambia G, Ferrandina G, Savarese A, Sorio R, Breda E,
ian carcinoma during the platinum era: A meta-­analysis. J Clin Oncol. et  al. Carboplatin plus paclitaxel versus carboplatin plus pegylated
2002;20:1248–1259. liposomal doxorubicin as first-­line treatment for patients with ovar-
50. Chan JK, Tian C, Fleming GF, Monk BJ, Herzog TJ, Kapp DS, et al. ian cancer: The MITO-­2 randomized phase III trial. J Clin Oncol.
The potential benefit of 6 vs. 3 cycles of chemotherapy in sub- 2011;29:3628–3635.
sets of women with early-­stage high-­risk epithelial ovarian cancer: 66. Alberts DS, Liu PY, Hannigan EV, O’Toole R, Williams SD, Young JA,
An exploratory analysis of a Gynecologic Oncology Group study. et  al. Intraperitoneal cisplatin plus intravenous cyclophosphamide
Gynecol Oncol. 2010;116:301–306. versus intravenous cisplatin plus intravenous cyclophosphamide for
51. Benedetti Panici P, Maggioni A, Hacker NF, Landoni F, Ackermann stage III ovarian cancer. N Engl J Med. 1996;335:1950–1955.
S, Campagnutta E, et al. Systematic aortic and pelvic lymphadenec- 67. Markman M, Bundy BN, Alberts DS, Fowler JM, Clark-Pearson
tomy versus resection of bulky nodes only in optimally debulked DL, Carson LF, et  al. Phase III trial of standard-­dose intravenous
advanced ovarian cancer. J Natl Cancer Inst. 2005;97:560–566. cisplatin plus paclitaxel versus moderately high-­dose intravenous
52. Vergote I, Tropé CG, Amant F, Kristensen GB, Ehlen T, Johnson N, paclitaxel and intraperitoneal cisplatin in small-­volume stage III
et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or ovarian cancer: An intergroup study of the Gynecologic Oncology
IV ovarian cancer. N Engl J Med. 2010;363:943–953. Group, Southwestern Oncology Group, and the Eastern Cooperative
53. Kehoe S, Hook J, Nankivell M, Jayson GC, Kitchener H, Lopes T, et al. Oncology Group. J Clin Oncol. 2001;19:1001–1007.
Primary chemotherapy versus primary surgery for newly diagnosed 68. Armstrong DK, Bundy B, Wenzel L, Huang HQ, Baergen R, Lele S,
advanced ovarian cancer (CHORUS): An open-­label, randomised, et al. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl
controlled, non-­inferiority trial. Lancet. 2015;386:249–257. J Med. 2006;354:34–43.
54. Kumar L, Pramanik R, Kumar S, Bhatla N, Malik S. Neoadjuvant che- 69. Chan JK, Brady MF, Penson RT, Huang H, Birrer MJ, Walker JL, et al.
motherapy in gynaecological cancers – Implications for staging. Best Weekly vs. every-­3-­week paclitaxel and carboplatin for ovarian can-
Pract Res Clin Obstet Gynaecol. 2015;29:790–801. cer. N Engl J Med. 2016;374:738–748.
55. van der Burg ME, van Lent M, Buyse M, Kobierska A, Colombo N, Favalli 70. Jaaback K, Johnson N, Lawrie TA. Intraperitoneal chemotherapy for
G, et al. The effect of debulking surgery after induction chemotherapy the initial management of primary epithelial ovarian cancer. Cochrane
on the prognosis in advanced epithelial ovarian cancer. Gynecological Database Syst Rev 2011;(11):CD005340.
Cancer Cooperative Group of the European Organization for Research 71. Katsumata N, Yasuda M, Isonishi S, Takahashi F, Michimae H, Kimura
and Treatment of Cancer. N Engl J Med. 1995;332:629–634. E, et  al. Long-­term results of dose-­dense paclitaxel and carbopla-
56. Ferron JG, Uzan C, Rey A, Gouy S, Pautier P, Lhommé C, et  al. tin versus conventional paclitaxel and carboplatin for treatment of
Histological response is not a prognostic factor after neoadjuvant advanced epithelial ovarian, fallopian tube, or primary peritoneal
chemotherapy in advanced-­stage ovarian cancer with no residual cancer (JGOG 3016): A randomised, controlled, open-­label trial.
disease. Eur J Obstet Gynecol Reprod Biol. 2009;147:101–105. Lancet Oncol. 2013;14:1020–1026.
57. Bohm S, Faruqi A, Said I, Lockley M, Brockbank E, Jeyarajah A, et al. 72. Pignata S, Scambia G, Katsaros D, Gallo C, Pujade-Lauraine E, De
Chemotherapy response score: Development and validation of a Placido S, et  al. Carboplatin plus paclitaxel once a week versus
system to quantify histopathologic response to neoadjuvant chemo- every 3 weeks in patients with advanced ovarian cancer (MITO-­7):
therapy in tubo-­ovarian high-­grade serous carcinoma. J Clin Oncol. A randomised, multicentre, open-­label, phase 3 trial. Lancet Oncol.
2015;33:2457–2463. 2014;15:396–405.
58. Coghlan E, Meniawy TM, Munro A, Bulsara M, Stewart CJ, Tan A, 73. Clamp AR, McNeish I, Dean A, et al. ICON8: A GCIG phase III ran-
et  al. Prognostic role of histological tumor regression in patients domised trial evaluating weekly dose-­ dense chemotherapy integra-
receiving neoadjuvant chemotherapy for high-­grade serous tubo-­ tion in first-­line epithelial ovarian/fallopian tube/primary peritoneal
ovarian carcinoma. Int J Gynecol Cancer. 2017;27:708–713. carcinoma (EOC) treatment: Results of primary progression-­ free
59. Aabo K, Adams M, Adnitt P, Alberts DS, Athanazziou A, Barley V, survival (PFS) analysis. Annal Oncol. 2017;28(Suppl.5):1229–1235.
et  al. Chemotherapy in advanced ovarian cancer: Four system- 74. Fuh KC, Shin JY, Kapp DS, Brooks RA, Ueda S, Urban RR, et  al.
atic meta-­analyses of individual patient data from 37 random- Survival differences of Asian and Caucasian epithelial ovarian cancer
ized trials. Advanced Ovarian Cancer Trialists’ Group. Br J Cancer. patients in the United States. Gynecol Oncol. 2015;136:491–497.
1998;78:1479–1487. 75. Calvert AH, Newell DR, Gumbrell LA, O’Reilly S, Burnell M, Boxall FE,
60. Vasey PA, Paul J, Birt A, Junor EJ, Reed NS, Symonds RP, et  al. et al. Carboplatin dosage: Prospective evaluation of a simple formula
Docetaxel and cisplatin in combination as first-­line chemotherapy based on renal function. J Clin Oncol. 1989;7:1748–1756.
for advanced epithelial ovarian cancer. Scottish Gynaecological 76. Nagao S, Fujiwara K, Imafuku N, Kagawa R, Kozuka Y, Oda T, et al.
Cancer Trials Group. J Clin Oncol. 1999;17:2069–2080. Difference of carboplatin clearance estimated by the Cockroft-­Gault,
61. McGuire WP, Hoskins WJ, Brady MF, Kucera PR, Partridge EE, Look Jelliffe, Modified-­Jelliffe. Wright or Chatelut formula. Gynecol Oncol.
KY, et al. Cyclophosphamide and cisplatin compared with paclitaxel 2005;99:327–333.
|
76       Berek ET AL.

77. Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, 94. Markman M, Rothman R, Hakes T, Reichman B, Hoskins W, Rubin
Kristensen G, et al. A phase 3 trial of bevacizumab in ovarian cancer. S, et  al. Second-­line platinum therapy in patients with ovarian
N Engl J Med. 2011;365:2484–2496. cancer  previously treated with cisplatin. J Clin Oncol. 1991;9:
78. Burger RA, Brady MF, Bookman MA, Fleming GF, Monk BJ, Huang 389–393.
H, et al. Incorporation of bevacizumab in the primary treatment of 95. Trimble E, Tinker A, Alberts D, Avall-Lundqvist E, Brady M, Harter P,
ovarian cancer. N Engl J Med. 2011;365:2473–2483. et al. Clinical trials in recurrent ovarian cancer. Int J Gynecol Cancer.
79. van Driel WJ, Koole SN, Sikorska K, Schagen van Leeuwen JH, 2011;21:771–775.
Schreuder HW, Hermans RH, et  al. Hyperthermic intraperitoneal 96. Parmar MK, Ledermann JA, Colombo N, du Bois A, Delaloye JF,
chemotherapy in ovarian cancer. N Engl J Med. 2018;378:230–240. Kristensen GB, et  al. Paclitaxel plus platinum-­based chemotherapy
80. Markman M1, Kennedy A, Webster K, Elson P, Peterson G, Kulp B, versus conventional platinum-­based chemotherapy in women with
et al. Clinical features of hypersensitivity reactions to carboplatin. J relapsed ovarian cancer: The ICON4/AGO-­OVAR-­2.2 trial. Lancet.
Clin Oncol. 1999;17:1141. 2003;361:2099–2106.
81. Ledermann J, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, 97. Pfisterer J, Vergote I, du Bois A, Eisenhauer E; AGO-OVAR; NCIC
et  al. Olaparib maintenance therapy in platinum-­sensitive relapsed CTG; EORTC GCG. Combination therapy with gemcitabine and
ovarian cancer. N Engl J Med. 2012;366:1382–1392. carboplatin in recurrent ovarian cancer. Int J Gynecol Cancer.
82. Ledermann J, Harter P, Gourley C, Friedlander M, Vergote I, Rustin 2005;15(Suppl.1):36–41.
G, et  al. Olaparib maintenance therapy in patients with platinum-­ 98. Pfisterer J, Ledermann JA. Management of platinum-­sensitive recur-
sensitive relapsed serous ovarian cancer: A preplanned retrospective rent ovarian cancer. Semin Oncol. 2006;33(2 Suppl.6):S12–S16.
analysis of outcomes by BRCA status in a randomised phase 2 trial. 99. Wagner U, Marth C, Largillier R, Kaern J, Brown C, Heywood M,
Lancet Oncol. 2014 July;15:852–861. et al. Final overall survival results of phase III GCIG CALYPSO trial
83. Pujade-Lauraine E, Ledermann JA, Selle F, Gebski V, Penson RT, Oza of pegylated liposomal doxorubicin and carboplatin vs paclitaxel
AM, et al. Olaparib tablets as maintenance therapy in patients with and carboplatin in platinum-­sensitive ovarian cancer patients. Br J
platinum-­sensitive, relapsed ovarian cancer and a BRCA1/2 muta- Cancer. 2012;107:588–591.
tion (SOLO2/ENGOT-­Ov-­21): A double-­blind, randomised, placebo-­ 100. Aghajanian C, Blank SV, Goff BA, Judson PL, Teneriello MG,
controlled, phase 3 trial. Lancet Oncol. 2017;18:1274–1284. Husain A, et  al. OCEANS: A randomized, double-­blind, placebo-­
84. Mirza MR, Monk BJ, Herrstedt J, Oza AM, Mahner S, Redondo A, controlled phase III trial of chemotherapy with or without beva-
et al. Niraparib maintenance therapy in platinum-­sensitive, recurrent cizumab in patients with platinum-­sensitive recurrent epithelial
ovarian cancer. N Engl J Med. 2016;375:2154–2164. ovarian, primary peritoneal, or fallopian tube cancer. J Clin Oncol.
85. Coleman RL, Oza AM, Lorusso D, Aghajanian C, Oaknin A, Dean 2012;30:2039–2045.
A, et  al. Rucaparib maintenance treatment for recurrent ovarian 101. Gordon AN, Fleagle JT, Guthrie D, Parkin DE, Gore ME, Lacave AJ.
carcinoma after response to platinum therapy (ARIEL3): A ran- Recurrent epithelial ovarian carcinoma: A randomized phase III study
domised, double-­blind, placebo-­controlled, phase 3 trial. Lancet. of pegylated liposomal doxorubicin versus topotecan. J Clin Oncol.
2017;390:1949–1961. 2001;19:3312–3322.
86. Basu P, Mukhopadhyay A, Konishi I. Targeted therapy for gyneco- 102. Hoskins PJ, Swenerton KD. Oral etoposide is active against platinum-­
logic cancers: Toward the era of precision medicine. Int J Gynecol. resistant epithelial ovarian cancer. J Clin Oncol. 1994;12:60–63.
2018;143(Suppl.2):131–136. 103. Rose PG, Blessing JA, Mayer AR, Homesley HD. Prolonged oral
87. Dowdy SC, Constaninou CL, Hartman LC, Keeney GL, Suman etoposide as second-­line therapy for platinum-­resistant and
VJ, Hillman DW, et  al. Long term follow-­up of women with ovar- platinum-­sensitive ovarian carcinoma: A Gynecologic Oncology
ian cancer after positive second-­look laparotomy. Gynecol Oncol. Group study. J Clin Oncol. 1998;16:405–410.
2003;91:563–568. 104. Friedlander M, Millward MJ, Bell D, Bugat R, Harnett P, Moreno
88. Tay EH, Grant PT, Gebski V, Hacker NF. Secondary cytoreductive JA, et  al. A phase II study of gemcitabine in platinum pre-­treated
surgery for recurrent epithelial ovarian cancer. Obstet Gynecol. patients with advanced epithelial ovarian cancer. Ann Oncol.
2002;99:1008–1013. 1998;9:1343–1345.
89. Chi DS, McCaughty K, Diaz JP, Huh J, Schwabenbauer S, Hummer 105. Shapiro JD, Millward MJ, Rischin D, Michael M, Walcher V, Francis
AJ, et al. Guidelines and selection criteria for secondary cytoreduc- PA, et al. Activity of gemcitabine in patients with advanced ovarian
tive surgery in patients with recurrent, platinum-­sensitive epithelial cancer: Responses seen following platinum and paclitaxel. Gynecol
ovarian carcinoma. Cancer. 2006;106:1933–1939. Oncol. 1996;63:89–93.
90. Du Bois A, Vergote I, Ferron G, Reuss A, Meier W, Greggi S, et  al. 106. Monk BJ, Herzog TJ, Kaye SB, Krasner CN, Vermorken JB, Muggia
Randomized controlled phase III study evaluating the impact of FM, et al. Trabectedin plus pegylated liposomal doxorubicin in recur-
secondary cytoreductive surgery in recurrent ovarian cancer: AGO rent ovarian cancer. J Clin Oncol. 2010;28:3107–3114.
DESKTOP III/ENGOT ov20. J Clin Oncol. 2017;15(Suppl):5501. 107. Colombo N, Kutarska E, Dimopoulos M, Bae D-S, Rzepka-Gorska
91. Rustin GJ, van der Burg ME, Griffin CL, Guthrie D, Lamont A, I, Bidzinski M, et  al. Randomized, open-­label, phase III study com-
Jayson GC, et  al. Early versus delayed treatment of relapsed ovar- paring patupilone (EPO906) with pegylated liposomal doxorubicin
ian cancer (MRC OV05/EORTC 55955): A randomised trial. Lancet. in platinum-­refractory or -­resistant patients with recurrent epithelial
2010;376:1155–1163. ovarian, primary fallopian tube, or primary peritoneal cancer. J Clin
92. Williams C, Simera I, Bryant A. Tamoxifen for relapse of ovarian can- Oncol. 2012;30:3841–3847.
cer. Cochrane Database Syst Rev. 2010;(3):CD001034. 108. Butow P, Stockler M, Gainford C, Martyn J, Oza A, Donovan HS,
93. Hurteau JA, Brady MF, Darcy KM, McGuire WP, Edmonds P, Pearl et  al. Symptom control in patients with recurrent ovarian cancer:
ML, et  al. Randomized phase III trial of tamoxifen versus tha- Measuring the benefit of palliative chemotherapy in women with
lidomide in women with biochemical-­recurrent-­only epithelial platinum refractory/resistant ovarian cancer. Int J Gynecol Cancer.
ovarian, fallopian tube or primary peritoneal carcinoma after a 2009;19(Suppl.2):S44–S48.
complete response to first-­line platinum/taxane chemotherapy 109. Oncology Nurse Advisor. FDA approves Lynparza to treat advanced
with an evaluation of serum vascular endothelial growth factor ovarian cancer. 2014. https://www.oncologynurseadvisor.com/dai-
(VEGF): A Gynecologic Oncology Group Study. Gynecol Oncol. ly-oncology-news/fda-approves-lynparza-to-treat-advanced-ovari-
2010;119:444–450. an-cancer/article/389417/. Accessed June 14, 2018.
Berek ET AL. |
      77

110. Kaufman B, Shapira-Frommer R, Schmutzler RK, Audeh MW, 127. Berek JS, Friedlander M, Hacker NF. Germ cell and nonepithelial
Friedlander M, Balmaña J, et  al. Olaparib monotherapy in patients ovarian cancer. In: Berek JS, Hacker NF, eds. Berek and Hacker’s
with advanced cancer and a germline BRCA 1/2 mutation. J Clin Gynecologic Oncology, 6th edn. Philadelphia: Lippincott Williams and
Oncol. 2015;33:244–250. Wilkins; 2015:530–559.
111. US Food and Drug Administration. FDA grants accelerated 128. Colombo N, Parma G, Zanagnolo V, Insinga A. Management of ovar-
approval to new treatment for advanced ovarian cancer. FDA ian stromal cell tumors. J Clin Oncol. 2007;25:2944–2951.
News release. 2016. www.fda.gov/NewsEvents/Newsroom/Press 129. Schumer ST, Cannistra SA. Granulosa cell tumors of the ovary. J Clin
Announcements/ucm533873.htm. Accessed April 3, 2018. Oncol. 2003;21:1180–1189.
112. Swisher EM, Lin KK, Oza AM, Scott CL, Giordano H, Sun J, et  al. 130. Pautier P, Gutierrez-Bonnaire M, Rey A, Sillet-Bach I, Chevreau C,
Rucaparib in relapsed, platinum-­sensitive high-­grade ovarian car- Kerbrat P, et al. Combination of bleomycin, etoposide, and cisplatin
cinoma (ARIEL2 Part 1): An international, multicentre, open-­label, for the treatment of advanced ovarian granulosa cell tumors. Int J
phase 2 trial. Lancet Oncol. 2017;18:75–87. Gynecol Cancer. 2008;18:446–452.
113. Gershenson DM, Sun CC, Lu KH, Coleman RL, Sood AK, Malpica A, 131. Brown J, Shvartsman HS, Deavers MT, Ramondetta LM, Burke TW,
et al. Clinical behavior of Stage II-­IV low-­grade serous carcinoma of Munsell MF, et al. The activity of taxanes compared with bleomycin,
the ovary. Obstet Gynecol. 2006;108:361–368. etoposide, and cisplatin in the treatment of sex cord-­stromal ovarian
114. Gershenson DM, Sun CC, Bodurka D, Coleman RL, Lu KH, Sood AK, tumors. Gynecol Oncol. 2005;97:489–496.
et al. Recurrent low-­grade serous ovarian carcinoma is relatively che- 132. Lappöhn RE, Burger HG, Bouma J, Bangah M, Krans M. Inhibin as
moresistant. Gynecol Oncol. 2009;114:48–52. a marker for granulosa cell tumor. Acta Obstet Gynecol Scand Suppl.
115. Wong K-K, Tsang YT, Deavers MT, Mok SC, Zu Z, Sun C, et al. BRAF 1992;155:61–65.
mutation is rare in advanced-­stage low-­grade ovarian serous carci- 133. Geerts I, Vergote I, Neven P, Billen J. The role of inhibins B and
nomas. Am J Pathol. 2010;177:1611–1617. antimüllerian hormone for diagnosis and follow-­up of granulosa cell
116. Hsu C-Y, Bristow R, Cha MS, Wang BG, Ho C-L, Kurman RJ, et  al. tumors. Int J Gynecol Cancer. 2009;19:847–855.
Characterization of active mitogen-­activated protein kinase in ovar- 134. Winter C, Albers P. Testicular germ cell tumors: Pathogenesis, diag-
ian serous carcinomas. Clin Cancer Res. 2004;10:6432–6436. nosis and treatment. Nat Rev Endocrinol. 2011;7:43–53.
117. Schmeler KM, Sun CC, Bodurka DC, Deavers MT, Malpica A, Coleman 135. Kondagunta GV, Motzer RJ. Chemotherapy for advanced germ cell
RL, et al. Neoadjuvant chemotherapy for low-­grade serous carcinoma tumors. J Clin Oncol. 2006;24:5493–5502.
of the ovary or peritoneum. Gynecol Oncol. 2008;108:510–514. 136. Williams S, Blessing JA, Liao SY, Ball H, Hanjani P. Adjuvant ther-
118. Grabowski JP, Harter P, Heitz F, Pujade-Lauraine E, Reuss A, apy of ovarian germ cell tumors with cisplatin, etoposide, and bleo-
Kristensen G, et al. Operability and chemotherapy responsiveness in mycin: A trial of the Gynecologic Oncology Group. J Clin Oncol.
advanced low-­grade serous ovarian cancer. An analysis of the AGO 1994;12:701–706.
Study Group metadatabase. Gynecol Oncol. 2016;140:457–462. 137. Williams SD, Blessing JA, Hatch KD, Homesley HD. Chemotherapy
119. Lalwani N, Shanbhogue AK, Vikram R, Nagar A, Jagirdar J, Prasad of advanced dysgerminoma: Trials of the Gynecologic Oncology
SR. Current update on borderline ovarian neoplasms. AJR Am J Group. J Clin Oncol. 1991;9:1950–1955.
Roentgenol. 2010;194:330–336. 138. Gershenson DM, Morris M, Cangir A, Kavanagh JJ, Stringer CA,
120. Kennedy AW, Hart WR. Ovarian papillary serous tumors of low Edwards CL. Treatment of malignant germ cell tumors of the
malignant potential (serous borderline tumors). A long-­term fol- ovary with bleomycin, etoposide, and cisplatin. J Clin Oncol.
low-­up study, including patients with microinvasion, lymph node 1990;8:715–720.
metastasis, and transformation to invasive serous carcinoma. Cancer. 139. Williams SD, Blessing JA, DiSaia PJ, Major FJ, Ball HG 3rd, Liao SY.
1996;78:278–286. Second-­look laparotomy in ovarian germ cell tumors: The gyneco-
121. Morice P, Denschlag D, Rodolakis A, Reed N, Schneider A, Kesic logic oncology group experience. Gynecol Oncol. 1994;52:287–291.
V, et  al. Recommendations of the Fertility Task Force of the 140. Talukdar S, Kumar S, Bhatla N, Mathur S, Thulkar S, Kumar L. Neo-­
European Society of Gynecologic Oncology about the conserva- adjuvant chemotherapy in the treatment of advanced malignant
tive management of ovarian malignant tumors. Int J Gynecol Cancer. germ cell tumors of ovary. Gynecol Oncol. 2014;132:28–32.
2011;21:951–963. 141. Wu PC, Huang RL, Lang JH, Huang HF, Lian LJ, Tang MY. Treatment
122. Shih KK, Zhou QC, Aghajanian C, Huh J, Soslow RA, Morgan JC, of malignant ovarian germ cell tumors with preservation of fertility:
et  al. Patterns of recurrence and role of adjuvant chemotherapy A report of 28 cases. Gynecol Oncol. 1991;40:2–6.
in stage II-­IV serous ovarian borderline tumors. Gynecol Oncol. 142. Zanetta G, Bonazzi C, Cantu M, Binidagger S, Locatelli A, Bratina G,
2010;119:270–273. et al. Survival and reproductive function after treatment of malignant
123. Gershenson DM, Sun CC, Iyer RB, Malpica AL, Kavanagh JJ, Bodurka germ cell ovarian tumors. J Clin Oncol. 2001;19:1015–1020.
DC, et  al. Hormonal therapy for recurrent low-­grade serous carci- 143. Casey AC, Bhodauria S, Shapter A, Nieberg R, Berek JS, Farias-Eisner
noma of the ovary or peritoneum. Gynecol Oncol. 2012;125:661–666. R. Dysgerminoma: The role of conservative surgery. Gynecol Oncol.
124. Fader AN, Bergstrom J, Jernigan A, Tanner EJ 3rd, Roche KL, Stone 1996;63:352–357.
RL, et  al. Primary cytoreductive surgery and adjuvant hormonal 144. Low JJ, Perrin LC, Crandon AJ, Hacker NF. Conservative surgery
monotherapy in women with advanced low-­grade serous ovarian to preserve ovarian function in patients with malignant germ cell
carcinoma: Reducing overtreatment without compromising survival? tumors. A review of 74 cases. Cancer. 2000;89:391–398.
Gynecol Oncol. 2017;147:85–91. 145. Mathew GK, Singh SS, Swaminathan RG, Tenali SG. Laparotomy for
125. Gershenson DM, Bodurka DC, Coleman RL, Lu KH, Malpica A, post chemotherapy residue in ovarian germ cell tumors. J Postgrad
Sun CC. Hormonal maintenance therapy for women with low-­ Med. 2006;52:262–265.
grade serous cancer of the ovary or peritoneum. J Clin Oncol. 146. Patterson DM, Murugaesu N, Holden L, Seckl MJ, Rustin GJ. A review
2017;35:1101–1111. of the close surveillance policy for stage I female germ cell tumors of
126. Zanetta G, Rota S, Chiari S, Bonazzi C, Bratina G, Mangioni C. the ovary and other sites. Int J Gynecol Cancer. 2008;18:43–50.
Behavior of borderline tumors with particular interest to persistence, 147. Dark GG, Bower M, Newlands ES, Paradinas F, Rustin GJ.
recurrence, and progression to invasive carcinoma: A prospective Surveillance policy for stage I ovarian germ cell tumors. J Clin Oncol.
study. J Clin Oncol. 2001;19:2658–2664. 1997;15:620–624.
|
78       Berek ET AL.

148. Huddart RA, Purkalne G; ESMO Guidelines Task Force. ESMO 150. Le T, Krepart GV, Lotocki RJ, Heywood MS. Malignant mixed meso-
Minimum Clinical Recommendations for diagnosis, treatment and dermal ovarian tumor treatment and prognosis: A 20-­year experi-
follow-­up of mixed or non-­seminomatous germ cell tumors (NSGCT). ence. Gynecol Oncol. 1997;65:237–240.
Ann Oncol. 2005;16(Suppl.1):i37–i39. 151. Sood AK, Sorosky JI, Gelder MS, Buller RE, Anderson B, Wilkinson
149. Hariprasad R, Kumar L, Janga D, Kumar S, Vijayaraghavan M. Growing EJ, et al. Primary ovarian sarcoma: Analysis of prognostic variables
teratoma syndrome of ovary. Int J Clin Oncol. 2008;13:83–87. and the role of surgical cytoreduction. Cancer. 1998;82:1731–1737.
DOI: 10.1002/ijgo.12615

FIGO CANCER REPORT 2018

Update on the diagnosis and management of gestational


trophoblastic disease

Hextan Y.S. Ngan1,* | Michael J. Seckl2 | Ross S. Berkowitz3 | Yang Xiang4 | 


François Golfier5 | Paradan K. Sekharan6 | John R. Lurain7 | Leon Massuger8

1
Department of Obstetrics and
Gynecology, Queen Mary Hospital, University Abstract
of Hong Kong, Hong Kong, China Gestational trophoblastic disease (GTD) arises from abnormal placenta and is composed
2
Departments of Histopathology and Medical
of a spectrum of premalignant to malignant disorders. Changes in epidemiology of GTD
Oncology, Charing Cross Trophoblastic
Disease Center, Charing Cross Campus of have been noted in various countries. In addition to histology, molecular genetic
Imperial College London, London, UK
studies can help in the diagnostic pathway. Earlier detection of molar pregnancy by
3
Department of Obstetrics and
ultrasound has resulted in changes in clinical presentation and decreased morbidity
Gynecology, Division of Gynecologic
Oncology, Brigham and Women’s from uterine evacuation. Follow-­up with human chorionic gonadotropin (hCG) is
Hospital, Dana-Farber Cancer
essential for early diagnosis of gestational trophoblastic neoplasia (GTN). The duration
Institute, Harvard Medical School, Boston,
MA, USA of hCG monitoring varies depending on histology type and regression rate. Low-­risk
4
Department of Obstetrics and GTN (FIGO Stages I–III: score <7) is treated with single-­agent chemotherapy but may
Gynecology, Peking Union Medical College
Hospital, Chinese Academy of Medical require additional agents; although scores 5–6 are associated with more drug
Sciences and Peking Union Medical College, resistance, overall survival approaches 100%. High-­risk GTN (FIGO Stages II–III: score
Beijing, China
5
>7 and Stage IV) is treated with multiple agent chemotherapy, with or without adjuvant
Department of Obstetrics and Gynecology,
French Trophoblastic Disease Reference surgery for excision of resistant foci of disease or radiotherapy for brain metastases,
Centre, Lyon University Hospital, Claude achieving a survival rate of approximately 90%. Gentle induction chemotherapy helps
Bernard Lyon 1 University, Lyon, France
6 reduce early deaths in patients with extensive tumor burden, but late mortality still
Department of Obstetrics and
Gynecology, Institute of Maternal and Child occurs from recurrent resistant tumors.
Health, Medical College, Calicut, India
7
John I. Brewer Trophoblastic Disease KEYWORDS
Center, Northwestern University Feinberg Choriocarcinoma; Epithelioid trophoblastic tumor; FIGO Cancer Report; Gestational trophoblastic
School of Medicine, Chicago, IL, USA
8
neoplasia; Moles; Placental site trophoblastic tumor
Division of Gynecologic
Oncology, Department of Obstetrics and
Gynecology, Radboud University Medical
Centre Nijmegen, Nijmegen, Netherlands

*Correspondence
Hextan Ngan, Department of Obstetrics and
Gynecology, University of Hong Kong, Queen
Mary Hospital, Hong Kong, China.
Email: hysngan@hku.hk

1 |  INTRODUCTION premalignant partial (PHM) and complete hydatidiform mole (CHM),
as well as the malignant invasive mole, choriocarcinoma, placental
Gestational trophoblastic disease (GTD) is a group of uncommon site trophoblastic tumor (PSTT), and epithelioid trophoblastic tumor
conditions associated with pregnancy. Histologically, it includes the (ETT). The malignant forms can arise after any type of pregnancy and

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

Int J Gynecol Obstet 2018; 143 (Suppl. 2): 79–85 wileyonlinelibrary.com/journal/ijgo  |  79
|
80       Ngan ET AL.

are collectively known as gestational trophoblastic neoplasia (GTN). present.3 Hydropic spontaneous abortion may mimic the appearance
The GTD spectrum has recently been expanded to also include atypi- of partial mole.
cal placental site nodule (APSN) as 10%–15% may coexist with or A cyclin-­dependent kinase inhibitor p57 is encoded by the pater-
develop into PSTT/ETT.1 While PSTT, ETT, and APSN have more nally imprinted and maternally expressed gene and hence is absent in
varied production of the pregnancy hormone—human chorionic gon- CHM without the maternal genome. In contrast, PHM and nonmolar
adotropin (hCG)—all other forms of GTD produce this hormone very abnormal gestations with maternal genome have strong nuclear p57
well. Indeed, hCG is an excellent biomarker of disease progression, staining, which can be used to exclude complete mole. However, p57
response, and subsequent post-­treatment surveillance. Thus, a pla- cannot differentiate PHM from nonmolar gestations. Cytogenetics can
teaued or rising hCG level enables the early detection of progression help to differentiate CHM from PHM and hydropic spontaneous abor-
of CHM and PHM to GTN that occurs in 15%–20%, and 0.5%–5% tion. Typically, CHM is diploid and has 46,XX chromosomes with both
of cases, respectively.2,3 The use of this biomarker together with the X’s from paternal origin, whereas PHM is triploid with maternal and
development of highly effective therapies has transformed survival paternal genetic origin. Hydropic spontaneous abortion normally has
outcomes so that today nearly all women affected by GTN can expect 46,XX or XY from both parents.3
to be cured if managed properly. Rarely, invasive and metastatic moles can be diagnosed by removal
of the uterus or biopsy of a metastatic lesion.

2 |  EPIDEMIOLOGY
3.2 | Choriocarcinoma
Although epidemiological studies have reported a wide variation in the Grossly, the tumor is bulky with hemorrhagic and necrotic areas. Apart
incidence of hydatidiform mole, in most parts of the world it is 1 per from the uterus, it can be found in tubes, ovaries, lung, liver, spleen,
1000 pregnancies.4 In high-­income countries, the incidence of com- kidneys, bowel, or brain.3
plete mole is approximately 1–3 per 1000 pregnancies and the inci- Histologically, choriocarcinoma shows absence of chorionic villi
dence of partial mole is about 3 per 1000 pregnancies.3 Hydatidiform and presence of abnormal intermediate trophoblast and cytotropho-
mole appears to be caused by abnormal gametogenesis and fertiliza- blast, rimmed with syncytiotrophoblasts with areas of necrosis, and
tion,5,6 more frequent at the extremes of reproductive age (<15 and hemorrhage. Highly complex karyotypes have been reported and an
>45 years) and pregnancies at these ages are a risk factor for hydatidi- XX sex chromosome composition is seen in the majority.
form mole. The risk increases after age 35 and there is a 5–10 times
increased risk after 45 years. Teenagers have a two-­fold risk of hav-
3.3 | Placental site trophoblastic tumor
ing a molar pregnancy. There is an increasing risk for complete moles
with advancing maternal age.7 History of a previous molar pregnancy Grossly, PSTT appears as white-­tan to yellow nodular masses varying
increases the risk to 10 times that for sporadic moles. from 1–10 cm (average 5 cm) in the endomyometrium with half of the
The reported incidence of choriocarcinoma ranges from 1 in cases invading deep into the myometrium. Histologically, PSTT arises
40 000 pregnancies in North America and Europe, to 9.2 and 3.3 from the mononuclear intermediate trophoblast on the maternal side
2
per 40 000 pregnancies in Southeast Asia and Japan, respectively. of the placental bed. Tumor cells have irregular nuclear membranes,
Dietary deficiency of beta-­carotene and animal fat is considered to be hyperchromatic nuclei, and dense eosinophilic to amphophilic cyto-
etiological factor for complete mole, but not for partial mole.8 plasm. Most tumors have a low mitotic count. Chorionic villi are absent.
Tumor cells diffusely express human placental lactogen (hPL), MUC-­4,
HSD3B1, HLA-­G, and Mel-­CAM (CD146). Expression of hCG and inhi-
3 |  GENETICS AND PATHOLOGY
bin is focal. The proliferation index is generally modestly increased, with
Ki-­67 expressed in 10% to 30% of cells—higher than that of benign exag-
3.1 | Molar pregnancy
gerated placental site reaction.3 PSTT shows rare genetic imbalances.
Grossly, CHM consists of hydropic villi to semi-­transparent vesicles of
variable sizes with absence of normal placenta. Early complete hyda-
3.4 | Epithelioid trophoblastic tumor
tidiform moles have minimal or no gross evidence of abnormal villi.
Differential diagnoses include partial hydatidiform mole, hydropic Grossly, the tumor appears as white-­tan to brown discrete nodules
abortion, and early nonmolar gestation with some degree of tropho- or cystic hemorrhagic masses invading deep into surrounding tissues.
blastic hyperplasia. Histologically, complete mole has florid cistern Nearly half arise in the cervix or lower segment of the uterus and
formation, trophoblastic proliferation, and absence of fetal parts. some in the fundus and broad ligament.
Significant cytological atypia and mitotic figures are common. In the Histologically, ETT arises from the chorionic-­type intermediate
first trimester, CMH villi may not be enlarged but have a distinct pol- trophoblast. Islands of relatively uniform intermediate trophoblastic
ypoid appearance with abnormal villous stromal changes and mild cells with moderate amount of eosinophilic to clear cytoplasm and
to moderate trophoblastic hyperplasia. In contrast, such histologic round nuclei are surrounded by extensive necrosis and associated with
features are less marked in partial mole and fetal parts or cells are a hyaline-­like matrix. The mitotic count ranges from 0–9 per 10 HPF.
Ngan ET AL. |
      81

Extensive or “geographic” necrosis is often present. ETT may coexist


with other trophoblastic neoplasms. Box 2 Tools for investigation of gestational tropho-
blastic neoplasia.

4 | CLINICAL PRESENTATION, • Chest X-ray is appropriate to diagnose lung metastases and


INVESTIGATIONS, AND DIAGNOSIS can be used for counting the number of lung metastases to
evaluate the risk score. Lung CT may be used.
4.1 | Molar pregnancy • Liver metastases may be diagnosed by ultrasound or CT
scanning.
Patients usually present with second trimester vaginal bleeding and a
• Brain metastases may be diagnosed by MRI or CT scanning.
uterus greater than the gravid date. As diagnosis is often made in the first
trimester with ultrasound examination, complications such as hyperem-
esis gravidarum, pre-­eclampsia, and hyperthyroidism are less common. If pregnancy. Aside from abnormal vaginal bleeding, other clinical
there is vaginal passage of the gestational product, vesicles may be seen. presentations can include bleeding from metastatic sites such as
The typical honeycomb appearance of a complete mole is rarely the liver, spleen, intestines, lung, or brain; pulmonary symptoms;
seen, especially in the first trimester. Typically, there is absence of fetal and neurological signs from spine or brain metastasis.2 GTN should
parts, cystic appearance of the placenta, and a deformed gestational be considered in the differential diagnosis of patients with unusual
sac that may appear like a spontaneous abortion. Hence, some molar presentations and serum hCG should be performed as part of the
pregnancies are only diagnosed on histologic examination after evacu- workup of such patients.
ation for a spontaneous abortion.

5 | TREATMENT
4.2 | Gestational trophoblastic neoplasia
5.1 | Molar pregnancy
Postmolar GTN is usually diagnosed by hCG surveillance without
symptoms. At the FIGO Gynecology Oncology Committee meeting in Suction evacuation and curettage, ideally performed under ultra-
2000, the definition of postmolar GTN based on hCG-­level changes, sound guidance, is the preferred method of evacuation of a molar
9
histology, and specific investigations was agreed (Boxes 1 and 2). pregnancy independent of uterine size if maintenance of fertility is
desired. It is recommended that a 12–14 mm suction cannula is used
and that an intravenous oxytocin infusion is started at the onset of
4.3 | Human chorionic gonadotropin monitoring
suction curettage and continued for several hours postoperatively to
For monitoring of GTN, an hCG assay that can detect all forms of hCG enhance uterine contractility. Because the risk of bleeding increases
including beta-­hCG, core hCG, C-­terminal hCG, nicked-­free beta, beta with uterine size, blood for transfusion should be available when
core, and preferably the hyperglycosylated forms, should be used. A the uterus is greater than 16 weeks in gestational size. Rh immune
persistently low hCG level needs continuous monitoring as some may globulin should be given to Rh-­negative women at the time of molar
progress to GTN with rising hCG levels.10,11 To exclude a false-­positive evacuation as RhD factor is expressed on the trophoblast. Judicious
result, retest with another assay kit or a test for urine hCG may be used. use of appropriate evacuation equipment and techniques, access
to blood products, careful intraoperative monitoring, and early rec-
ognition and correction of complications results in improved out-
4.4 | Gestational trophoblastic neoplasia
comes.2,12 If there is no persistent bleeding, a second evacuation is
after nonmolar pregnancy
usually not needed.
As only about 50% of GTN follows molar pregnancy, the rest can Hysterectomy is an alternative to suction curettage if childbearing
occur after a spontaneous abortion, ectopic pregnancy, or a term is complete. In addition to evacuating the molar pregnancy, hyster-
ectomy provides permanent sterilization and decreases the need for
subsequent chemotherapy by eliminating the risk of local myometrial
Box 1 FIGO criteria for diagnosis of postmolar gesta-
tional trophoblastic neoplasia. invasion as a cause of persistent disease. Medical induction of labor
and hysterotomy are not recommended for molar evacuation, since
• When the plateau of hCG lasts for four measurements over a these methods increase maternal morbidity and the development of
period of 3 weeks or longer; that is, days 1, 7, 14, 21. postmolar GTN requiring chemotherapy.
• When there is a rise in hCG for three consecutive weekly Prophylactic administration of either methotrexate or actinomycin
measurements over at least a period of 2 weeks or more; D chemotherapy at the time of or immediately following molar evac-
days 1, 7, 14. uation is associated with a reduction in the incidence of postmolar
• If there is a histologic diagnosis of choriocarcinoma. GTN to 3%–8%. However, it should be limited to special situations in
Abbreviation: hCG, human chorionic gonadotropin. which the risk of postmolar GTN is much greater than normal or where
adequate hCG follow-­up is not possible.13
|
82       Ngan ET AL.

Follow-­up hCG monitoring every 1–2 weeks is essential for


5.3 | Gestational trophoblastic neoplasia
early diagnosis of and management of postmolar GTN. On the
other hand, postmolar GTN rarely occurs after the spontaneous Treatment of GTN is generally by chemotherapy. The best regimen
return of hCG levels to normal, allowing for a shortened follow-­up depends on stage and classification. In the 2000 FIGO staging and
period for most women. Hence, a single additional confirmatory classification (Tables 1 and 2), a risk score of 6 and below is classified
normal hCG measurement 1 month after first hCG normalization as low risk and above 6 is considered high risk.
is recommended for a PHM and monthly hCG measurements
should be obtained for only 6 months after hCG normalization for
5.3.1 | Low-­risk gestational trophoblastic neoplasia
a CHM.2,14 Termination of pregnancy is not indicated if acciden-
tal pregnancy occurs during surveillance after the hCG level has Patients with low-­risk GTN (WHO risk score 0–4) should be treated
returned to normal. In addition, data now show that it is safe to with one of the single agent methotrexate or actinomycin D protocols
recommend oral contraceptives. 15 listed in Box 3. The Cochrane Review in 2012, including 513 patients
The risk of recurrence is low (0.6%–2%) after one molar pregnancy, in five randomized controlled trials, showed that actinomycin D (Act-­
although much increased after consecutive molar pregnancies.16 D) appeared to be superior to methotrexate (MTX) (risk ratio [RR]
Mutations in NLRP7 and KHDC3L have been reported in women with 0.64; 95% confidence interval, [CI] 0.54–0.76).19 Methotrexate was
5,6
recurrent molar pregnancy. associated with significantly more treatment failure than actinomycin
D (RR 3.81; 95% CI 1.64–8.86).

5.2 | Coexisting normal pregnancy with mole


Chemotherapy should be changed to the alternative single agent if
Molar pregnancy rarely coexists with a normal pregnancy. The diag- there has been a good response to the first agent but the hCG level
nosis is usually made on ultrasound. Although there is a high risk of plateaus above normal during treatment, or if toxicity precludes an ade-
spontaneous abortion, about 40%–60% result in live births. The risk quate dose or frequency of treatment. If there is an inadequate response
of GTN in coexisting molar and normal pregnancy compared with
singleton molar pregnancy is increased from 15% to 20% to 27% to
46%.17,18 In the absence of complications and normal genetic and Box 3 First-­line single agent chemotherapy regimens for
ultrasound findings, pregnancy can proceed. low-­risk gestational trophoblastic neoplasia.

• MTX-FA 8-day regimen (50 mg MTX intramuscularly on days


T A B L E   1   FIGO staging and classification for gestational 1,3,5,7 with folinic acid 15 mg orally 24 hours after MTX on
trophoblastic neoplasia.
days 2,4,6,8); repeat every 2 weeks.
FIGO Stage Description • MTX 0.4 mg/kg (max. 25 mg) intravenously or intramuscularly
for 5 days every 2 weeks.
I Gestational trophoblastic tumors strictly confined to
the uterine corpus • Actinomycin D pulse 1.25 mg/m2 intravenously every
II Gestational trophoblastic tumors extending to the 2 weeks.
adnexae or to the vagina, but limited to the genital • Actinomycin D 0.5 mg intravenously for 5 days every 2 weeks
structures • Others: MTX 30–50 mg/m2 intramuscularly weekly, MTX
III Gestational trophoblastic tumors extending to the 300 mg/m2 infusion every 2 weeks
lungs, with or without genital tract involvement
Abbreviations: MTX-­FA, methotrexate–folinic acid.
IV All other metastatic sites

T A B L E   2   WHO scoring system based on prognostic factors.

WHO risk factor scoring with FIGO staging 0 1 2 4


Age <40 >40 — —
Antecedent pregnancy Mole Abortion Term
Interval from index pregnancy, months <4 4–6 7–12 >12
3 3 4 4 5
Pretreatment hCG mIU/mL <10 >10 –10 >10 –10 >105
Largest tumor size including uterus, cm — 3–4 ≥5 —
Site of metastases including uterus Lung Spleen, kidney Gastrointestinal tract Brain, liver
Number of metastases identified — 1–4 5–8 >8
Previous failed chemotherapy — — Single drug Two or more drugs

To stage and allot a risk factor score, a patient’s diagnosis is allocated to a Stage as represented by a Roman numeral I, II, III, or IV. This is then separated
by a colon from the sum of all the actual risk factor scores expressed in Arabic numerals e.g. Stage II:4, Stage IV:9. This Stage and score will be allotted for
each patient.
Ngan ET AL. |
      83

to the initial single agent, multiple agent chemotherapy as for high-­risk


5.3.3 | Ultra high-­risk gestational trophoblastic
disease should be initiated if there is a significant elevation in hCG level,
neoplasia and salvage therapy
development of metastasis, or resistance to sequential single agent che-
motherapy.3 Studies showed that change to single agent Act-­D gives a Among the high-­risk group as defined by the FIGO staging and clas-
good response rate of between 76% and 87% in patients with relatively sification, a subgroup with score of 13 or greater, as well as patients
low hCG levels3,20; as there are continuous updates on the cutoff level with liver, brain, or extensive metastases, do poorly when treated with
based on evolving data, physicians should refer to local guidelines from first-­line multiple agent chemotherapy. Similar findings have been
time to time. Otherwise, multiple agents should be considered. reported by others.25
Higher WHO risk score (5–6) and clinicopathologic diagnosis of For those with massive disease, starting with standard chemo-
choriocarcinoma are both associated with an increased risk of resis- therapy may cause sudden tumor collapse with severe bleeding, met-
tance to single agent chemotherapy. Lowering the threshold for the abolic acidosis, myelosuppression, septicemia, and multiple organ
use of multiple agent chemotherapy in these otherwise low-­risk failure, any or all of which can result in early death. To avoid this,
patients can be considered. the use of initial gentle rather than full-­dose chemotherapy seems
After the hCG level has returned to normal, consolidation with 2–3 logical. Indeed, induction etoposide 100 mg/m2 and cisplatin 20 mg/
more cycles of chemotherapy will decrease the chance of recurrence. m2 on days 1 and 2, repeated weekly for 1–3 weeks, before starting
3,21
The overall complete remission rate is close to 100%. normal chemotherapy appears to have eliminated early deaths in
one series26 with promising results now reported by others.25
For those patients with liver metastases, with or without brain
5.3.2 | High-­risk gestational trophoblastic neoplasia
metastases, or a very high-­risk score, EP (etoposide and platinum)/
Multiple agent chemotherapy regimens are used to treat high-­risk EMA or another more intensive chemotherapy regimen (Box 4), rather
GTN. The most commonly used is EMA-­CO (etoposide, methotrexate, than EMA-­CO, may yield a better response and outcome.23 For such
actinomycin D, cyclophosphamide, vincristine) (Table 3), although the high-­risk patients, a longer consolidation with four cycles of chemo-
Cochrane Database review failed to conclude what combination was therapy should be considered.
best.22 About 20% of patients fail EMA-­CO therapy but most can be In patients with brain metastases, an increase in the methotrexate
salvaged with further therapy; the overall survival rates for patients infusion to 1 g/m2 will help the drug cross the blood–brain barrier and
with high-­risk GTN are now running as high as 95%. A number of intrathecal methotrexate 12.5 mg may be used in some centers. This
adverse features that predict poorer outcomes, including liver and/or can be given at the time of CO when EMA-­CO is used, or with the EP
brain metastasis23,24 and the management of such patients together in the EP/EMA regimen. Some centers may give whole brain radio-
with salvage therapies are discussed below. therapy 3000 cGy in 200 cGy daily fractions concurrent with chemo-
therapy or use stereotactic or gamma knife radiation to treat existing
or residual brain metastases after chemotherapy.27 Patients failing
T A B L E   3   EMA-­CO (etoposide, methotrexate, actinomycin D, EMA-­CO are mostly salvaged with paclitaxel and etoposide alternat-
cyclophosphamide, vincristine) chemotherapy.
ing with paclitaxel and cisplatin (TE/TP) or with EP/EMA. In China,
Regimens the 5FU-­based FAEV regimen is also an effective salvage treatment.
For women failing EP/EMA or TE/TP, options include a number of
Regimen 1
other standard or high-­dose chemotherapy regimens with autologous
Day 1
Etoposide 100 mg/m2 intravenous infusion over
30 min
Actinomycin-­D 0.5 mg intravenous bolus Box 4 Salvage therapies.
2
Methotrexate 100 mg/m intravenous bolus
• EP-EMA (etoposide, cisplatin, etoposide, methotrexate and
200 mg/m2 intravenous infusion over 12 h
actinomycin-D)
Day 2
• TP/TE (paclitaxel, cisplatin/paclitaxel, etoposide)
Etoposide 100 mg/m2 intravenous infusion over 30 min
• MBE (methotrexate, bleomycin, etoposide)
Actinomycin-­D 0.5 mg intravenous bolus
• VIP or ICE (etoposide, ifosfamide, and cisplatin or
Folinic acid rescue 15 mg intramuscularly or orally every 12 h
carboplatin)
for four doses (starting 24 h after
beginning the methotrexate infusion)
• BEP (bleomycin, etoposide, cisplatin)
• FA (5-fluorouracil, actinomycin-D)
Regimen 2
• FAEV (floxuridine, actinomycin-D, etoposide, vincristine)
Day 8
• High-dose chemotherapy with autologous bone marrow or
Vincristine 1 mg/m2 intravenous bolus (maximum 2 mg)
stem cell transplant
Cyclophosphamide 600 mg/m2 intravenous infusion over 30 min
• Immunotherapy with pembrolizumab
The two regimens alternate each week
|
84       Ngan ET AL.

peripheral stem cell support (Box 4). Recent work suggests that check- multidisciplinary team of gynecology, gynecological oncology, medical
point immunotherapies such as pembrolizumab may also save some oncology, pathology, and the hCG laboratory. Work with a clear model
women.28 Finally, surgical salvage should not be overlooked. of care. Create a clinical guidelines committee, create a database, and
develop a website. Some form of annual funding will be needed for
the center to develop and maintain a database, website, patient infor-
5.4 | Role of surgery
mation, reading material, and nursing staff, and to allow presentations
Surgery may have an important role in the management of GTN. at national meetings. Try to establish central pathology review for the
Hysterectomy can be considered in uncontrolled uterine bleeding, whole region.
although it can often be avoided with the use of uterine artery embo-
lization. Laparotomy may be needed to stop bleeding in organs such
AU T HO R CO NT R I B U T I O NS
as the liver, gastrointestinal tract, kidneys, and spleen. Neurosurgery
is needed if there is bleeding into the brain or increased intracranial Each author contributed in writing different sections of the article as
pressure. The resection of an isolated drug-­resistant tumor may also well as critical appraisal of the whole article.
be curative.4,11

AC KNOW L ED G M ENTS
5.5 | Role of radiotherapy
This chapter updates the information published in the FIGO Cancer
Radiotherapy has a limited role in GTN, except in treatment of brain Report 2015 (Ngan HY, Seckl MJ, Berkowitz RS, Xiang Y, Golfier
metastasis, although its efficacy compared with intrathecal metho- F, Sekharan PK, Lurain JR. Update on the diagnosis and manage-
trexate is controversial.4,11 ment of gestational trophoblastic disease. Int J Gynecol Obstet.
2015;131(Suppl.2):S123–126).

5.6 | PSTT/ETT
CO NFL I C TS O F I NT ER ES T
Both PSTT and ETT are less chemosensitive than choriocarcinoma.
Hysterectomy is the primary mode of treatment in most cases and The authors have no conflicts of interest to declare.
surgery also plays an important role in metastatic disease. If fer-
tility preservation is desired, especially in a localized lesion, con-
REFERENCES
servative management such as uterine curettage, hysteroscopic
resection, and chemotherapy may be considered.29 Fertility preser- 1. Kaur B, Short D, Fisher RA, Savage PM, Seckl MJ, Sebire NJ. Atypical
placental site nodule (APSN) and association with malignant gesta-
vation is not suitable in diffuse lesions. In advanced stage, EP-­EMA
tional trophoblastic disease; a clinicopathologic study of 21 cases. Int
or TE/TP can be considered. Interval from antecedent pregnancy J Gynecol Pathol. 2015;34:152–158.
of more than 48 months seems to be the most significant adverse 2. Lurain JR. Gestational trophoblastic disease I: Epidemiology, pathol-
prognostic factor.30 ogy, clinical presentation and diagnosis of gestational trophoblastic
disease, and management of hydatidiform mole. Am J Obset Gynecol.
2010;203:531–539.
5.7 | Follow-­up 3. Seckl MJ, Sebire NJ, Berkowitz RS. Gestational trophoblastic disease.
Lancet. 2010;376:717–729.
After treatment of GTN, follow-­up hCG monitoring every month for at 4. Hancock BW, Seckl MJ, Berkowitz RS. Gestational Trophoblastic
Disease. 4th edn. ISSTD website; 2015. Available at: http://test.regis-
least 12 months is essential for surveillance of relapse. Reliable con-
traid.com/gtd-book.html. Accessed May 2, 2018.
traception must be used throughout this period. 5. Fisher RA, Hodges MD, Newlands ES. Familial recurrent hydatidiform
Future fertility, pregnancy, and offspring are not affected, although mole: A review. J Reprod Med. 2004;49:595–601.
psychosocial and sexual counseling may be needed for some patients. 6. Murdoch S, Djuric U, Mazhar B, et  al. Mutations in NALP7 cause
recurrent hydatidiform moles and reproductive wastage in humans.
Nat Genet. 2006;38:300–302.
7. Sebire NJ, Foskett M, Fisher RA, Rees H, Seckl M, Newlands E. Risk
6 | ESTABLISHMENT OF A (NATIONAL) of partial and complete relation hydatidiform molar pregnancy in rela-
GTD CENTER tion to maternal age. BJOG. 2002;109:99–102.
8. Berkowitz RS, Bernstein MR, Harlow BL, et  al. Case-­control study
of risk-­factors for partial molar pregnancy. Am J Obstet Gynecol.
Centralized care is needed for optimal management of a rare disease
1995;173:788–794.
like GTD. Without some type of centralization, treatment decisions 9. Ngan HY, Bender H, Benedet JL, Jones H, Montruccoli GC, Pecorelli
will be inconsistent. Centralized management can vary from only S. Gestational trophoblastic neoplasia, FIGO 2000 staging and classi-
hCG monitoring with treatment advice to patient referral. Creating fication. Int J Gynecol Obstet. 2003;83:175–177.
10. Cole LA. Hyperglycosylated hCG, a review. Placenta. 2010;31:
a center is not easy. It starts with sharing the idea with colleagues
653–664.
and promoting it at national meetings. Make sure you have support 11. Ngan HY, Kohorn EI, Cole LA, et al. Trophoblastic disease. Int J Gynecol
from the national obstetrics and gynecology governing body. Create a Obstets. 2012;119:S130–S136.
Ngan ET AL. |
      85

12. Berkowitz RS, Goldstein DP. Clinical practice. Molar pregnancy. N Engl 21. Lurain JR. Gestational trophoblastic disease II: Classification and man-
J Med. 2009;360:1639–1645. agement of gestational trophoblastic neoplasia. Am J Obstet Gynecol.
13. Wang Q, Fu J, Hu L, et  al. Prophylactic chemotherapy for hydatidi- 2011;204:11–18.
form mole to prevent gestational trophoblastic neoplasia. Cochrane 22. Alazzam M, Tidy J, Osborne R, Coleman R, Hancock BW, Lawrie TA.
Database Syst Rev. 2017;(9):CD007289. Chemotherapy for resistant or recurrent gestational trophoblastic
14. Coyle C, Short D, Jackson L, et  al. What is the optimal duration of neoplasia. Cochrane Database Syst Rev. 2016;(1):CD008891.
human chorionic gonadotrophin surveillance following evacuation of 23. Ahamed E, Short D, North B, Savage PM, Seckl MJ. Survival of women
a molar pregnancy? A retrospective analysis on over 20,000 consecu- with gestational trophoblastic neoplasia and liver metastases: Is it
tive patients. Gynecol Oncol. 2018;148:254–257. improving? J Reprod Med. 2012;57:262–269.
15. Costa HL, Doyle P. Influence of oral contraceptives in the develop- 24. Newlands ES, Holden L, Seckl MJ, McNeish I, Strickland S, Rustin GJ.
ment of post-­molar trophoblastic neoplasia–a systematic review. Management of brain metastases in patients with high-­risk gesta-
Gynecol Oncol. 2006;100:579–585. tional trophoblastic tumors. J Reprod Med. 2002;47:465–471.
16. Sebire NJ, Fisher RA, Foskett M, Rees H, Seckl MJ, Newlands ES. Risk 25. Bolze PA, Riedl C, Massardier J, et al. Mortality rate of gestational tro-
of recurrent hydatidiform mole and subsequent pregnancy outcome phoblastic neoplasia with a FIGO score of ≥13. Am J Obstet Gynecol.
following complete or partial hydatidiform molar pregnancy. BJOG. 2016;214:390.e1–390.e8.
2003;110:22–26. 26. Alifrangis C, Agarwal R, Short D, et  al. EMA/CO for High-­risk ges-
17. Sebire NJ, Foskett M, Paradinas FJ, et al. Outcome of twin pregnan- tational trophoblastic neoplasia: Good outcomes with induction
cies with complete hydatidiform mole and healthy co-­twin. Lancet. low-­dose etoposide-­cisplatin and genetic analysis. J Clin Oncol.
2002;359:2165–2166. 2013;31:280–286.
18. Lin LH, Maestá I, Braga A, et al. Multiple pregnancies with complete 27. Ghorani E, Kaur B, Fisher RA, et  al. Pembrolizumab is effective
mole and coexisting normal fetus in North and South America: A ret- for drug-­resistant gestational trophoblastic neoplasia. Lancet.
rospective multicenter cohort and literature review. Gynecol Oncol. 2017;390:2343–2345.
2017;145:88–95. 28. Neubauer NL, Latif N, Kalakota K, et al. Brain metastasis in gestational
19. Lawrie TA, Alazzam M, Tidy J, Hancock BW, Osborne R. First-­line che- trophoblastic neoplasia: An update. J Reprod Med. 2012;57:288–292.
motherapy in low-­risk gestational trophoblastic neoplasia. Cochrane 29. Zhao J, Lv WG, Feng FZ, et al. Placental site trophoblastic tumor: A
Database Syst Rev. 2016;(6):CD007102. review of 108 cases and their implications for prognosis and treat-
20. Prouvot C, Golfier F, Massardier J, et  al. Efficacy and safety of ment. Gynecol Oncol. 2016;142:102–108.
second-­line 5-­day dactinomycin in case of methotrexate fail- 30. Horowitz NS, Goldstein DP, Berkowitz RS. Placental site trophoblastic
ure for gestational trophoblastic neoplasia. Int J Gynecol Cancer. tumors and epithelioid trophoblastic tumors: Biology, natural history,
2018;28:1038–1044. and treatment modalities. Gynecol Oncol. 2017;144:208–214.
DOI: 10.1002/ijgo.12618

FIGO CANCER REPORT 2018

Role of imaging in the routine management of


endometrial cancer

Ming Yin Lin1 | Andrew Dobrotwir2 | Orla McNally3,4 | Nadeem R. Abu-Rustum5,6 | 


Kailash Narayan1,4,*

1
Gyne-Oncology Unit, Department of
Radiation Oncology and Cancer Imaging, Peter Abstract
MacCallum Cancer Centre, Melbourne, Endometrial cancer is the most common gynecologic cancer in women today. It is
Australia
2
surgically staged, and while surgery is the primary treatment modality, the identification
Department of Radiology, Royal Women’s
Hospital, Parkville, Australia of disease extent—in particular extrauterine spread—prior to surgery is important to
3
Gyne-Oncology Unit, Royal Women’s optimize treatment decision making. Ultrasound and MRI are useful for evaluating the
Hospital, Parkville, Australia
extent of local disease, while CT and PET are used for detecting lymph node or distant
4
Department of Obstetrics and
Gynecology, University of Melbourne, metastases. Diffusion-­weighted MRI has also been used for detecting small metastatic
Melbourne, Australia deposits in lymph nodes and omentum. Extrauterine soft tissue involvement can be
5
Gynecology Service, Department of detected by ultrasound, CT, MRI, and PET. Recently, intraoperative visualization
Surgery, Memorial Sloan Kettering Cancer
Centre, New York, NY, USA techniques, such as sentinel lymph node mapping, are increasingly used to avoid
6
Department of Obstetrics and extensive surgical staging without compromising treatment. Imaging is also used for
Gynecology, Weill Cornell Medical College,
planning adjuvant treatment and detection of postoperative residual disease in ­
New York, NY, USA
high-­risk patients, monitoring and detecting recurrent disease, and in post-­treatment
*Correspondence
surveillance of asymptomatic patients with high risk of relapse.
Kailash Narayan, Peter MacCallum Cancer
Centre, Melbourne, Australia.
Email: mahaguru@petermac.org KEYWORDS
Endometrial cancer; FIGO Cancer Report; MRI; PET; Radiology; Ultrasound

1 |  BACKGROUND application of local and/or regional treatment. Although endometrial


cancer is surgically staged, the identification of disease extent—in par-
Endometrial cancer is the most common gynecologic cancer seen in ticular extrauterine spread—prior to surgery is important to optimize
women today.1 It is more prevalent in high-­resource countries, but its treatment planning. This has been facilitated by noninvasive medical
incidence is rising in low-­resource countries as a result of rising obe- imaging technologies including ultrasound, X-­ray, computed tomogra-
sity and improved longevity. Historically, endometrial cancer is clas- phy (CT), magnetic resonance imaging (MRI), positron emission tomog-
sified according to histologic subtype, but recently—as a result of the raphy (PET), and, increasingly, co-­registered images such as PET-­CT or
Cancer Genome Atlas (TCGA)—a molecular-­based classification has PET-­MRI. More recently, intraoperative visualization techniques, such
been advocated owing to its superior prognostication.2 as sentinel lymph node (SLN) mapping are being used to avoid exten-
Endometrial cancer is curable, especially in the early stages. sive surgical staging without compromising treatment.
Endometrioid histology has better prognosis than nonendometrioid The aim of the present article is to describe the practical applica-
histologies. Surgery is the mainstay of treatment. Adjuvant radiother- tion of imaging in the routine diagnostic assessment, staging, treat-
apy and systemic chemotherapy play a role in selected cases. Accurate ment selection, adjuvant treatment planning, and post-­treatment
mapping of the extent of cancer spread is important for appropriate surveillance of endometrial cancer patients.

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

Int J Gynecol Obstet 2018; 143 (Suppl. 2): 109–117 wileyonlinelibrary.com/journal/ijgo  |  109
|
110       Lin ET AL.

2 |  CLINICAL PRESENTATION systematic pelvic and para-­aortic lymphadenectomy is associated


AND WORK-­U P with increased morbidity, more so in combination with radiotherapy
when the incidence of serious complications can be up to 26%.10,11
Most endometrial cancer occurs in postmenopausal women over the Thus, routine systematic pelvic and para-­aortic lymphadenectomy
age of 50 years (median age of 63 years) who commonly present with for staging purposes is not recommended.12 Staging lymphadenec-
vaginal bleeding. Less commonly the presentation is associated with tomy can be avoided in endometrioid adenocarcinoma histology
vaginal discharge, while abdominal distension and pain are usually where the FIGO grade is 1 or 2 and the tumor is limited to the inner
associated with advanced disease. half of the myometrium, cervical stroma is not involved and more
Evaluation of a woman suspected to have endometrial cancer recently, these cases are staged with the SLN mapping algorithm.13
involves detailed history-­taking including identifying risk factors, and The SLN mapping algorithm for staging has replaced lymphadenec-
physical examination including pelvic examination. Pelvic examination tomy in many practices worldwide and can be applied to almost all
involves estimation of the size and mobility of the uterus, detection of cases of apparent uterine-­confined endometrial cancer regardless
any vaginal extension of pathology, and direct inspection of the geni- of FIGO grade or histotype.14 Following a normal peritoneal sur-
tal tract to exclude other common local causes of bleeding. First-­line vey, any suspicious lymph node should be removed and sent for
imaging is a pelvic ultrasound. This should begin with transabdominal histologic examination. In the absence of SLN mapping, myometrial
ultrasound to assess the pelvic organs, followed by transvaginal ultra- invasion can be identified preoperatively using MRI to evaluate the
sound to assess endometrial thickness and any extension of abnor- need for lymphadenectomy.15 Where MRI is not available, intraop-
mality in the cervix. Endometrial thickness greater than 4 mm in a erative frozen section of the suspected region of the myometrium
postmenopausal woman is highly suspicious of an endometrial pathol- can be used to determine myometrial invasion in patients with
ogy, warranting further evaluation that can be done with endometrial grade 1–2 histology.16
sampling. In a meta-­analysis reviewing the accuracy of endometrial The aim of successful endometrial cancer surgery, when possible,
sampling devices, the Pipelle endometrial sampler (Pipelle de Cornier, is definitive treatment or, if surgery alone is not curative, to remove
Paris, France) was found to be highly sensitive for the detection of gross disease and accurately stage the patient in order to direct post-
endometrial carcinoma, with detection rates of 99.6% and 91% for operative adjuvant therapies.
post-­ and premenopausal women respectively, and 81% for atypical In the majority of patients with grade 1 and 2 endometrioid histol-
endometrial hyperplasia. 3 ogy, preoperative imaging will not significantly change the basic man-
When office endometrial sampling is not possible, for example in agement or prognosis of these patients. However, imaging is helpful in
severe cervical stenosis or virgin patients, hysteroscopy with dilatation patients with suspected extrauterine disease where additional surgery
and curettage (D&C) is the next investigation of choice. Hysteroscopy is required for its removal or for planning of adjuvant treatment. It is
with D&C is indicated if ultrasound indicates the presence of endo- desirable to know the following information prior to the surgery:
metrial polyps that may contain premalignant or malignant cells that
could be missed by endometrial sampling alone, or to obtain more 1. Histology and grade.
tissue if the diagnosis of neoplasia from office biopsy is in question. 2. Myometrial invasion (in select cases where fertility preservation or
Hysteroscopy D&C should also be considered in women with bleeding ovarian preservation is considered).
who are taking tamoxifen.4 3. Presence of resectable gross disease outside the uterus (enlarged
ovary, nodes, and omental disease).
4. Presence of unresectable disease. The possibility of finding extrau-
3 |  STAGING AND TREATMENT SELECTION terine disease is higher in poorly differentiated endometrioid ade-
nocarcinoma and nonendometrioid carcinoma. In patients with
Once a diagnosis of endometrial cancer has been established, the serous histology, extrauterine disease may be present in almost
treatment of choice is total hysterectomy with bilateral salpingo-­ 50% of all cases.
oophorectomy and surgical staging in the majority of cases. Lymph
node status assessment is required for accurate staging and for treat- Ultrasound and MRI are used for evaluating the extent of local dis-
ment planning. ease, while CT and PET are used for detecting lymph node or distant
The detection of lymph node metastases in surgically staged metastases.17 Diffusion-­weighted MRI (DWI-­MRI) has also been used
patients varies between 8% and 33% depending on the quality for detecting small metastatic deposits in lymph nodes and omentum.
of node dissection, pathologic assessment protocols, histologic Extrauterine soft tissue involvement can be detected by transabdominal
subtype, and the clinical stage.5–8 Increasing lymph node involve- ultrasound, CT, MRI, and PET. Apart from preoperative staging, imaging
ment is associated with depth of myometrial invasion and histo- is also used for planning adjuvant treatment and for detection of post-
logic grade of tumor. The incidence of nodal metastases in patients operative residual disease in high-­risk patients (CT/PET-­CT), monitoring
with less than and greater than 50% myometrial invasion was 5% and detecting recurrent disease (ultrasound, CT, MRI, and PET/PET-­CT)
and 18%, respectively.5,9 Knowledge of lymph node status assists and in post-­treatment surveillance of asymptomatic patients with high
in deciding adjuvant treatment and in radiotherapy planning. A risk of relapse (PET/PET-­CT).
Lin ET AL. |
      111

3.1 | Low-­resource regions where medical imaging (A) (C)


is not available
In low-­resource conditions, it is reasonable to proceed with total hys-
terectomy and bilateral salpingo-­oophorectomy once the patient has
been evaluated by clinical pelvic examination and the histologic diag-
nosis established. In the vast majority of patients with endometrioid
carcinoma and well-­ to moderately differentiated histology with inva-
sion limited to the inner half of the myometrium or no myometrial (B) (D)

invasion, namely FIGO Stage 1A, total abdominal hysterectomy and


bilateral salpingo-­oophorectomy are usually curative. All other types
or stages of endometrial cancer may have locoregionally advanced
disease where lymph node sampling is recommended. In poorly differ-
entiated histology or where examination under anesthesia suggests
extrauterine disease, a chest X-­ray—if available—should be performed.
In the absence of SLN mapping, gross examination of myometrial
F I G U R E   1   Ultrasound scans of a normal uterus. Transvaginal
invasion in a cut hysterectomy specimen intraoperatively can be used ultrasound scans of (A) sagittal plane (long) and (B) axial plane (trans)
to guide lymph node sampling. Such gross examination of myometrial with normal, thin, uniform echogenic endometrium (arrowheads)
invasion correctly identified disease in 89% (64/72) of patients.18 A and subendometrial hypoechoic halo (solid arrows). Transabdominal
pathological review of the surgical specimen should be carried out ultrasound scans of endometrial carcinoma in sagittal plane (C) and
axial plane (D). Note heterogeneous echogenicity due to hemorrhage
and, if indicated and where possible, patients should be referred for
and necrosis (dashed arrows).
further treatment.

In general, a cut-­off value of 4–5 mm is indicative of a carcinoma, with


3.2 | Medium-­resource regions where
96% sensitivity and 61% accuracy based on a meta-­analysis by Smith-­
ultrasound and CT may be available
Bindman et al.20 Furthermore, the heterogeneous echogenicity caused
Ultrasonography plays a central role in the management of patients by areas of hemorrhage and necrosis (Fig. 1C,D) and an irregular
with gynecological disorders. It is widely available and is a relatively endometrial myometrial interphase that denotes myometrial invasion
inexpensive imaging tool. In certain circumstances, its diagnostic by cancer supports the diagnosis of endometrial cancer. Transvaginal
accuracy may match that of MRI. ultrasound may also detect cervical stromal invasion.
Transabdominal ultrasound can provide additional information,
mostly related to extrauterine disease.
3.2.1 | Ultrasound
The uterine cavity and endometrium as well as the adnexa can be
3.2.2 | Computed tomography
examined in great detail using transvaginal ultrasound. Transrectal
ultrasound can also provide similar information and is useful in elderly CT is less helpful in investigating abnormalities within the uterus.
patients with vaginal stenosis. Transabdominal ultrasound should be Figure 2A–D show the difference in image quality and the soft tissue
used in conjunction with transvaginal ultrasound if the uterus is large, definition between CT (Fig. 2A,B) and MRI (Fig. 2C,D). On CT, a gross
and to avoid missing large ovarian or tubal pathology. suspicious endometrial mass may be seen as a hypodense lesion, or an
Ultrasound examination of the uterus should be carried out sys- enlarged endometrial cavity that often cannot be distinguished from
tematically in a stepwise fashion.19 Using a transvaginal ultrasound benign lesions. However, CT has better multiplanar spatial resolution
probe, the cervix should be examined in a sagittal plane for tumor that is useful in visualizing the entire pelvic and abdominal cavity for
invasion into the cervical stroma. The medial parametria should also enlarged nodes (Fig. 2F) and gross soft tissue masses, as well as dis-
be examined, followed by the entire body of the uterus from cornu to tant metastases in the lungs (Fig. 2E).
cornu in a sagittal plane, and from the cervix to the fundus in an axial In the absence of MRI or PET, CT should be requested if extra-
plan. Tumor size should then be measured and, if possible, Gordon’s uterine disease is suspected in patients with high-­grade histol-
ratio measured at the area of deepest myometrial invasion.19 ogy, deep myometrial invasion, or a large uterus. Frequently, CT is
Normal endometrium in a postmenopausal woman is smooth, usu- ordered as a baseline investigation prior to histologic diagnosis of
ally less than 1 mm thick, and has a thin hypoechoic layer between endometrial cancer. The use of intravenous contrast is helpful in
it and the myometrium (Fig. 1A,B). Although ultrasound can detect identifying vascular structures and soft tissue metastases, and dis-
altered endometrium, it is not specific and histologic confirmation is tinguishing lymph nodes from small bowel loops. A lymph node with
always required. There is a higher likelihood of endometrial cancer with a maximum short axis diameter of greater than or equal to 10 mm
increasing thickness of the endometrium and postmenopausal status. is regarded as suspicious of metastatic disease. Nonetheless, the
|
112       Lin ET AL.

following which a patient may have total hysterectomy, bilateral


(A) (C) (E)
salpingo-­oophorectomy, and surgical staging. Commonly, following
histologic diagnosis, patients are referred to gynecologic oncology
specialist multidisciplinary clinics. Here histology and any available
imaging are reviewed. In some centers, all confirmed cases of endo-
(F) metrial cancer will have pelvic MRI prior to surgery. In other centers,
CT of the chest, abdomen, and pelvis is requested if prior scan is not
(B) (D)
available. All pretreatment imaging and pathology is reviewed at mul-
tidisciplinary meetings and individual patient management plans are
formulated. Diagnostic information related to ultrasound and CT has
been discussed in previous sections. Here we describe the role of MRI
in endometrial cancer.

F I G U R E   2   Computed tomography (A: sagittal plane, B: axial


plane) and MRI (C: sagittal T2, D: axial diffusion-­weighted) in the
3.3.1 | Magnetic resonance imaging
same patient with endometrial carcinoma distending the endometrial
cavity (dotted arrows) and deep myometrial invasion (solid arrows). Where available, MRI is the imaging modality of choice for the ana-
E: Axial CT chest with multiple “cannonball” pulmonary metastases tomical study of the pelvis and abdomen. MRI is best suited to detect
(dashed arrows). F: Coronal CT abdomen demonstrating multiple
and evaluate endometrial cancer within the endometrial cavity; tumor
para-­aortic necrotic lymph node metastases (arrowheads).
infiltration into myometrium, endocervix, and gross extension into
the parametria; and other pelvic tumor deposits. On T2-­weighted
overall reported sensitivity in detecting nodal metastasis is only images, endometrial cancer usually appears of intermediate signal
about 40%. In a study involving measurement of 125 positive and intensity. Unlike normal endometrium it does not have a uniform high
160 negative pelvic lymph nodes in 32 consecutive patients with signal owing to the presence of higher cellularity, necrosis, and hem-
node-­positive endometrial cancer, there was a positive correlation orrhage within the tumor. The surrounding myometrium is composed
between the size of the positive lymph node and the size of the of two distinct layers. The inner myometrial layer or “junctional zone”
metastasis (P<0.01).21 Whereas 68 of 125 (54%) positive lymph abuts the endometrium and appears as a low signal band, while the
nodes measured less than 10 mm in maximum short axis diameter, outer myometrium is more variable in appearance but usually of
46 of 160 (29%) negative lymph nodes measured more than 10 mm intermediate signal (Fig. 3). Post intravenous contrast, the inner-
in maximum short axis diameter.21 It was observed that patients most myometrial layer enhances uniformly during the early dynamic
with single metastatic nodes tend to have tumor deposits in nodes phase as a continuous line or “subendometrial stripe.” Disruption of
less than 10 mm. In patients with multiple positive nodes, 85% of the subendometrial stripe or frank breach of the junctional zone is
patients had at least one positive node greater than 10 mm.8 In indicative of myometrial invasion. An intact junctional zone and a
node-­positive endometrial cancer patients the incidence of a sin- continuous band of early subendometrial enhancement exclude deep
gle positive node was 25%–40% and multiple nodes were found myometrial invasion.
in 60%–75% of patients.8 Therefore, CT should correctly detect Depth of myometrial invasion is related to the incidence of nodal
more than 40% of node-­positive patients—higher than sensitivity metastasis. The incidence of nodal metastasis in patients with less
and specificity studies would suggest. Additionally, CT scan of the than 50% and greater than 50% myometrial invasion was reported
chest, abdomen, and pelvis as a preoperative investigation is useful to be 5% and 18%, respectively.5 A study comparing presurgical MRI
for excluding unexpected anatomy that may result in modification images with histology showed the value of MRI in predicting the pres-
of planned surgery. ence and depth of myometrial invasion (Fig. 4) and the potential utility

(A) (B) (C)


3.3 | High-­resource countries where modern imaging
is available
In countries where health care is streamlined and well-­resourced,
patients often present to the primary healthcare provider with symp-
toms prior to referral to a specialist or subspecialist. Transvaginal
ultrasound is typically scheduled and, following a suspected diagno-
sis of endometrial hyperplasia or cancer, patients are referred to a
F I G U R E   3   Normal zonal anatomy on T2-­weighted MRI in (A)
gynecologist for further management and workup. Patients undergo
sagittal, (B) axial, and (C) coronal planes demonstrating high signal
office Pipelle biopsy or examination under anesthesia and D&C to
(bright) endometrium (arrowheads), low signal (darker) junctional zone
obtain endometrial tissue samples. Once the histologic diagnosis or inner myometrium (solid white arrows), and intermediate signal
of endometrial cancer is established, a CT or MRI scan is requested (grey) outer myometrium (dotted arrows).
Lin ET AL. |
      113

If cervical invasion is suspected, additional slice orientation per-


(A) (B)
pendicular to the axis of the endocervical canal is recommended.25
The presence of an intact enhancing cervical mucosa excludes cervical
stromal invasion.26
Multiparametric MRI including thin-­section (3 mm) high-­resolution
multiplanar T2-­weighted images and simple modifications, such as
the addition of double oblique T2-­weighted, diffusion-­weighted, and
dynamic (30, 60, 120 seconds, and 4 minutes) contrast-­enhanced
C(i) C(ii) C(iii)
images, improves staging and treatment planning in patients with
endometrial carcinoma.26
Often these preoperative images are helpful not only in planning
the primary surgery, but also during deliberation of postoperative
adjuvant radiotherapy for the presence of diverticula or any other vari-
ation in normal anatomy.
Metastatic node detection with MRI is similar to a high-­quality
CT scan with variable sensitivity ranging from 38% to 89% and
specificity ranging from 78% to 99%.26 However, DWI-­MRI increas-
F I G U R E   4   Deep myometrial invasion on MRI: (A) Sagittal T2
demonstrating obvious deep myoinvasion with heterogeneous tumor ingly routine in many centers, and higher field strength 3T MRI may
obliterating the normal uterine zonal anatomy (dashed arrow) and significantly enhance the sensitivity of detecting metastatic lymph
invading cervical stroma (solid arrow). (B) Pitfall of MRI myometrial nodes (Fig. 5) by combining the size of node and relative apparent
invasion assessment: large polypoid tumor stretches and thins the diffusion coefficient values.27 Incorporating morphologic features of
myometrium (dashed arrow) with age-­related loss of normal zonal
nodal involvement—best seen at high-­resolution T2-­weighted MRI—
anatomy (solid arrow), pathology confirming no evidence of deep
myoinvasion. (C) Multiparametric MRI improving staging accuracy: include internal heterogeneity, spiculated nodal margins, necro-
equivocal depth of myoinvasion and cervical stromal involvement sis, and signal intensity comparable to that in the primary tumor,
on sagittal T2 (Ci). Dynamic T1 fat saturated post contrast (Cii) improves the accuracy of evaluation in patients with rectal cancer
shows disruption of subendometrial stripe at anterior midbody of and may be applicable to those with endometrial carcinoma.26 DWI-­
uterus (dotted arrow) confirming myoinvasion. Cervical mucosal
MRI is also sensitive in detecting early invasive extrauterine and
enhancement preserved posteriorly but disrupted anteriorly
omental disease (Fig. 6).
(arrowhead) confirming stromal invasion. Diffusion-­weighted imaging
(Ciii) highlights tumor extent and deep cervical stromal invasion but
absence of bladder wall invasion (arrowhead).
(A) (B)
of MRI in presurgical prognostication of patients who may be suitable
for more conservative treatment.15
The sensitivity of MRI to distinguish superficial from deep myo-
metrial invasion varies from nearly 60%–88%,22 and is limited in some
situations such as very superficial invasion in premenopausal women
or a large polypoid tumor extruding into the cervical canal,23,24 as well
as in technique and image quality. The combination of T2-­weighted
and dynamic contrast-­enhanced magnetic resonance images offers
high accuracy for staging endometrial cancer in the range of 83%–
(C) (D)
91% (personal communication, KN, May 2018). Endometrial cancer is
best examined in the sagittal plane, providing longitudinal views of the
uterus and cervix as well as surrounding structures such as bladder,
rectum, and loops of bowel.
T2-­weighted MRI is the key sequence for evaluating myometrial
invasion as it depicts the uterine zonal anatomy, with the intermediate-­
signal-­intensity tumor well delineated against the low-­signal-­intensity
junctional zone.
A minimum of at least two T2-­weighted sequences in the sagittal,
F I G U R E   5   Lymph node evaluation on MRI is enhanced by the
axial oblique, or coronal oblique orientation (short and long axis of the
combination of high resolution T2 (A) and T1 (B) anatomical imaging
uterine body) of the pelvis and one T1-­weighted enhanced sequence
and functional sequences including post contrast T1 fat saturated (C)
acquired at 2 minutes ±30 seconds after intravenous contrast injec- and diffusion-­weighted imaging (D) increasing nodal conspicuity and
tion is recommended. improving morphological assessment (arrowheads).
|
114       Lin ET AL.

primary surgery is planned. Surgery for endometrial cancer can range


(A) (C)
from a minimum of peritoneal assessment and washings, hysterec-
tomy (abdominal, laparoscopic, or robotic) with bilateral salpingo-­
oophorectomy, to systematic lymphadenectomy, omentectomy, and
removal of any extrauterine disease, if present.
SLN mapping involves injection of a colored dye or radioisotope
tracer at both the 9 and 3 o’clock positions or at four sites in all four
quadrants of the cervix (Fig. 7). Methylene blue was used previously,
but now indocyanine green is the preferred choice. A combination of
(B) (D)
superficial (1–3 mm) and deep (1–2 cm) cervical injection is adequate.
SLN mapping can effectively evaluate pelvic nodal status with 85.1%
to 98.1% sensitivity, with negative predictive value of up to 99.8% in
clinically Stage 1 endometrial cancer patients.30 Such a high level of
accuracy resulted from using a well-­defined “SLN surgical algorithm”
that was developed at the Memorial Sloan Kettering Cancer Center
and adopted in the 2014 NCCN Guidelines.31 The SLN algorithm
involves: (1) removal of SLN and suspicious lymph nodes regardless of
mapping; (2) if bilateral SLN is not identified then side-­specific pelvic
F I G U R E   6   Peritoneal metastatic nodular deposits (solid arrows)
and common iliac node dissection is carried out; and (3) uterine serosal
demonstrated on axial T2 (A) and diffusion-­weighted imaging (B)
sequences in the same patient. Omental cake (arrowheads) on axial and peritoneal evaluation and washings are carried out.
T2: (C) intermediate signal: grey and prominent restricted diffusion on The SLN algorithm is now commonly applied as a surgical stag-
diffusion-­weighted imaging; (D): high signal: bright. ing strategy in all histologies and grades of apparent uterine-­confined
endometrial cancer. With this approach, all patients will at least have
MRI is useful for observing extrauterine disease and any other struc-
bilateral pelvic nodes sampled and evaluated by a strict pathological
tural anatomical detail that may influence the extent of primary surgery.
protocol to help exclude extrauterine disease and increase precision in
the prescription of adjuvant therapy. Comparison of staging outcomes
3.3.2 | Positron emission tomography– from the SLN algorithm versus traditional lymphadenectomy has been
computed tomography reported by Ducie et al.,14 with encouraging results.
In clinically advanced stages of endometrial cancer where extra-
PET-­CT is usually employed in the study of metastatic lymph nodes. It
uterine disease is suspected on clinical examination or preliminary pel-
has high specificity in detecting metastatic nodes; however, its sensi-
vic ultrasound, CT or MRI, establishing the extent of disease by PET/
tivity is only modest and depends on the size of metastatic deposit.28
The detection rate of PET-­CT was only 12% in metastatic lesions
measuring 4 mm or less, but 100% when the deposit was 10 mm or
larger.29 It is not routinely employed in assessing intrauterine disease
although it can detect depth of myometrial invasion and extension
into the endocervix reasonably well. In advanced stages, PET-­CT was
helpful in detecting metastatic deposits in the ovary and omentum,
and distant spread. In general, if the baseline MRI or CT scan sug-
gests enlarged nodes and if one believes that removal of all positive
nodes during primary surgery will offer a survival advantage, then
PET-­CT may be helpful in detecting the most caudal metastatic lymph
node, therefore guiding the extent of nodal clearance in that patient.
Conversely, if PET-­CT shows extensive disease where surgery is
unlikely to be curative, then a simple (palliative) hysterectomy may be
appropriate as the best immediate symptomatic measure, while the
remainder of the disease can be managed using palliative hormone
therapy, chemotherapy and radiotherapy alone or in combination.
F I G U R E   7   Example of sentinel lymph node mapping with
indocyanine green cervical injection using the PINPOINT system.
The right external iliac sentinel lymph node is seen medial to the
4 |  SENTINEL LYMPH NODE MAPPING
external iliac vessels, and ventral to the internal iliac vessels. Also
demonstrated are lymphatic trunks from the right paracervix crossing
Once the diagnosis of endometrial cancer is established and no con- over the obliterated umbilical ligament on the way to the sentinel
traindication is found on preliminary clinical examination and imaging, lymph node.
Lin ET AL. |
      115

CT prior to surgery may greatly assist in deciding the extent of surgery recurrence is localized or disseminated. A long interval between
over a standard staging laparotomy. the primary treatment and isolated recurrence is usually associated
with a better prognosis.
In a retrospective analysis of 36 women previously treated for
5 | IMAGING IN PLANNING endometrial cancer, Belhocine et al.34 found the sensitivity and spec-
ADJUVANT RADIOTHERAPY ificity of PET–CT to be 96% and 78%, respectively. These authors
reported that the use of fluorodeoxyglucose (FDG)-­PET confirmed
Currently, the indications for postoperative adjuvant radiotherapy are32: recurrence in 88% of the cases, which was useful in localizing the site
of disease, and it detected asymptomatic recurrence in 12%. In 35% of
1. Patients with grade 1 and 2 histology with deep myometrial patients with confirmed recurrence, additional metastatic sites were
invasion and where lymph nodes were not evaluated. detected, significantly altering management. PET has more specific-
2. Patients with grade 3, clear cell, or serous histology, irrespective of ity, sensitivity, and accuracy compared with conventional imaging or
myometrial invasion, where lymph nodes were not evaluated. tumor markers.
3. All stage 3 and 4 patients.

All node-­positive patients and grade 3 histologies with deep myome- 7 | IMAGING AND TREATMENT OF
trial invasions are recommended to receive external beam pelvic radio- RECURRENT ENDOMETRIAL CANCER
therapy (EBRT) or abdominopelvic radiotherapy, the so-­called extended
field radiotherapy (EFRT). Patients with lymphovascular space invasion and lymph node metas-
In the era of conformal radiotherapy, as a minimum, CT images tases are at high risk of recurrence.35 Eighty-­seven percent of recur-
with or without contrast in the treatment position are used for radi- rences are seen by 3 years from the time of primary surgery.36
ation planning. Often, enlarged nodes are treated with an additional Twenty-­five percent of all recurrences were solitary vaginal vault
radiation dose (nodal boost) for potentially better control of the dis- recurrences and approximately 60% had distant failure with or with-
ease. It can be difficult to delineate the “at risk” nodal target volume out locoregional recurrence.37
and determine the upper level of the radiation therapy field where Solitary vaginal vault relapses can be successfully salvaged by
only involved nodes were removed without the complete node dis- surgery or combination of EBRT and intracavitary or interstitial
section, unless this was a sentinel node. Similarly, in high-­risk patients brachytherapy.38,39 The rate of successful salvage is substantially
where nodes were not evaluated, the use of PET-­CT, either fused improved and treatment complication minimized by proper patient
with the planning CT on the treatment planning system or a planning selection using PET/CT to exclude asymptomatic multiple metastases.
PET-­CT in the radiotherapy position is very useful. In the postopera- MRI is particularly helpful in visualizing the treatment volume and its
tive radiotherapy setting, the use of PET can result in modification of relationship to surrounding normal anatomical structures (Fig. 8).
the treatment plan as additional disease becomes evident in up to 35%
of patients.33
Occasionally, in poorly differentiated and high-­grade histology
that may have had a close resection margin, postoperative soft tissue
recurrences are seen in the interval between surgery and radiother-
apy. Some of these patients may require an extra radiotherapy boost
requiring interstitial brachytherapy. In such cases a pre-­radiotherapy
pelvic MRI is very useful for identifying the tumor requiring additional (Ai) (Aii)
radiotherapy and its relation with surrounding normal tissues.

6 | IMAGING IN SURVEILLANCE AND


FOLLOW-­U P (Bii)
(Bi)

The routine use of imaging in surveillance following initial treatment


is not indicated. However, radiological imaging should be considered
where a potentially salvageable recurrent disease is suspected clini- F I G U R E   8   Use of imaging in diagnosis, treatment planning, and
cally or in the post-­therapeutic restaging of selected patients at high restaging of recurrent endometrial cancer: (Ai) MRI (axial) shows
the vaginal vault mass encased within loops of bowel; and (Aii)
risk for relapse.
Corresponding slice on pre-­radiotherapy PET-­CT, performed to
MRI can assist in identifying the extent of local recurrence
confirm isolated recurrence. (Bi) Radiotherapy planning CT with
and appropriateness for surgical resection and radiotherapy. interstitial implant needles in situ; and (Bii) Six-­month post-­treatment
PET–CT has the additional benefit of determining if the extent of restaging PET-­CT.
|
116       Lin ET AL.

Isolated para-­aortic recurrences, although infrequent, are salvage- surgically staged endometrial adenocarcinoma of endometrioid his-
able. Twenty-­two of the 29 such recurrences could be salvaged, result- tology. Int J Gynecol Cancer. 2008;18:269–273.
10. Lewandowski G, Torrisi J, Potkul RK, et  al. Hysterectomy with
ing in median survival of 3.5 (1–15) years.40
extended surgical staging and radiotherapy versus hysterectomy
alone and radiotherapy in stage I endometrial cancer: A comparison
of complication rates. Gynecol Oncol. 1990;36:401–404.
11. Tinga DJ, Timmer PR, Bouma J, Aalders JG. Prognostic significance of
8 |  CONCLUSION
single versus multiple lymph node metastases in cervical carcinoma
stage IB. Gynecol Oncol. 1990;39:175–180.
The application of imaging in the management of endometrial cancer 12. Frost JA, Webster KE, Bryant A, Morrison J. Lymphadenectomy for
has become routine. It has resulted in better matching of patients the management of endometrial cancer. Cochrane Database Syst Rev.
2017;(10):CD007585.
with the appropriate treatment modality or a combination of treat-
13. Zahl Eriksson AG, Ducie J, Ali N, et al. Comparison of a sentinel lymph
ment regimens, resulting in improved locoregional control and
node and a selective lymphadenectomy algorithm in patients with
reduction in treatment-­related morbidity. Imaging has also helped endometrioid endometrial carcinoma and limited myometrial inva-
in determining the best salvage strategy for patients with recurrent sion. Gynecol Oncol. 2016;140:394–399.
endometrial cancer. 14. Ducie JA, Eriksson AGZ, Ali N, et al. Comparison of a sentinel lymph
node mapping algorithm and comprehensive lymphadenectomy in
the detection of stage IIIC endometrial carcinoma at higher risk for
nodal disease. Gynecol Oncol. 2017;147:541–548.
AUTHOR CONTRI B UTI O N S
15. Cade TJ, Quinn MA, McNally OM, Neesham D, Pyman J, Dobrotwir
A. Predictive value of magnetic resonance imaging in assessing
All authors contributed intellectual and scientific input into this man-
myometrial invasion in endometrial cancer: Is radiological staging
uscript. KN and MYL conceived and designed the paper. MYL, AD, sufficient for planning conservative treatment? Int J Gynecol Cancer.
NAR, OM, and KN contributed to manuscript writing, review and 2010;20:1166–1169.
approval of the final version. 16. Mariani A, Webb MJ, Keeney GL, Haddock MG, Calori G, Podratz KC.
Low-­risk corpus cancer: Is lymphadenectomy or radiotherapy neces-
sary? Am J Obstet Gynecol. 2000;182:1506–1519.
17. Epstein E, Blomqvist L. Imaging in endometrial cancer. Best Pract Res
CO NFLI CTS OF I NTE RE ST
Clin Obstet Gynaecol. 2014;28:721–739.
The authors have no conflicts of interest to declare. 18. Traen K, Holund B, Mogensen O. Accuracy of preoperative tumor
grade and intraoperative gross examination of myometrial invasion
in patients with endometrial cancer. Acta Obstet Gynecol Scand.
REFERENCES 2007;86:739–741.
19. Leone FP, Timmerman D, Bourne T, et al. Terms, definitions and mea-
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. surements to describe the sonographic features of the endometrium
2018;68:7–30. and intrauterine lesions: A consensus opinion from the International
2. Talhouk A, McAlpine JN. New classification of endometrial can- Endometrial Tumor Analysis (IETA) group. Ultrasound Obstet Gynecol.
cers: The development and potential applications of genomic-­based 2010;35:103–112.
classification in research and clinical care. Gynecol Oncol Res Pract. 20. Smith-Bindman R, Kerlikowske K, Feldstein VA, et  al. Endovaginal
2016;3:14. ultrasound to exclude endometrial cancer and other endometrial
3. Dijkhuizen FP, Mol BW, Brolmann HA, Heintz AP. The accuracy abnormalities. JAMA. 1998;280:1510–1517.
of endometrial sampling in the diagnosis of patients with endo- 21. Reich O, Winter R, Pickel H, Tamussino K, Haas J, Petru E. Does
metrial carcinoma and hyperplasia: A meta-­analysis. Cancer. the size of pelvic lymph nodes predict metastatic involvement in
2000;89:1765–1772. patients with endometrial cancer? Int J Gynecol Cancer. 1996;6:
4. Abdaal A, Mushtaq Y, Khasati L, et al. Post-­menopausal bleeding -­ Is 445–447.
transvaginal ultrasound a useful first-­line investigation in tamoxifen 22. Cabrita S, Rodrigues H, Abreu R, et al. Magnetic resonance imaging
users? Post Reprod Health. 2018;24:72–78. in the preoperative staging of endometrial carcinoma. Eur J Gynaecol
5. Creasman WT, Morrow CP, Bundy BN, Homesley HD, Graham Oncol. 2008;29:135–137.
JE, Heller PB. Surgical pathologic spread patterns of endometrial 23. Sanjuan A, Escaramis G, Ayuso JR, et al. Role of magnetic resonance
cancer. A Gynecologic Oncology Group Study. Cancer. 1987;60(8 imaging and cause of pitfalls in detecting myometrial invasion and
Suppl):2035–2041. cervical involvement in endometrial cancer. Arch Gynecol Obstet.
6. ASTEC study group, Kitchener H, Swart AM, Qian Q, Amos C, 2008;278:535–539.
Parmar MK. Efficacy of systematic pelvic lymphadenectomy in 24. Chung HH, Kang SB, Cho JY, et al. Accuracy of MR imaging for the
endometrial cancer (MRC ASTEC trial): A randomised study. Lancet. prediction of myometrial invasion of endometrial carcinoma. Gynecol
2009;373:125–136. Oncol. 2007;104:654–659.
7. Todo Y, Takeshita S, Okamoto K, Yamashiro K, Kato H. Implications 25. Kinkel K, Forstner R, Danza FM, et al. Staging of endometrial cancer
of para-­aortic lymph node metastasis in patients with endome- with MRI: Guidelines of the European Society of Urogenital Imaging.
trial cancer without pelvic lymph node metastasis. J Gynecol Oncol. Eur Radiol. 2009;19:1565–1574.
2017;28:e59. 26. Rauch GM, Kaur H, Choi H, et  al. Optimization of MR imaging for
8. Ayhan A, Tuncer ZS, Tuncer R, Yuce K, Kucukali T. Tumor status of pretreatment evaluation of patients with endometrial and cervical
lymph nodes in early endometrial cancer in relation to lymph node cancer. Radiographics. 2014;34:1082–1098.
size. Eur J Obstet Gynecol Reprod Biol. 1995;60:61–63. 27. Lin G, Ho KC, Wang JJ, et  al. Detection of lymph node metastasis
9. Chi DS, Barakat RR, Palayekar MJ, et  al. The incidence of pelvic in cervical and uterine cancers by diffusion-­weighted magnetic reso-
lymph node metastasis by FIGO staging for patients with adequately nance imaging at 3T. J Magn Reson Imaging. 2008;28:128–135.
Lin ET AL. |
      117

28. Narayan K, Hicks RJ, Jobling T, Bernshaw D, McKenzie AF. A com- 34. Belhocine T, De Barsy C, Hustinx R, Willems-Foidart J. Usefulness of
parison of MRI and PET scanning in surgically staged loco-­ regionally (18)F-­FDG PET in the post-­therapy surveillance of endometrial carci-
advanced cervical cancer: Potential impact on treatment. Int J Gynecol noma. Eur J Nucl Med Mol Imaging. 2002;29:1132–1139.
Cancer. 2001;11:263–271. 35. Narayan K, Khaw P, Bernshaw D, Mileshkin L, Kondalsamy-
29. Kitajima K, Murakami K, Yamasaki E, Kaji Y, Sugimura K. Accuracy of Chennakesavan S. Prognostic significance of lymphovascular space
integrated FDG-­PET/contrast-­enhanced CT in detecting pelvic and invasion and nodal involvement in intermediate-­ and high-­risk endo-
paraaortic lymph node metastasis in patients with uterine cancer. Eur metrial cancer patients treated with curative intent using surgery and
Radiol. 2009;19:1529–1536. adjuvant radiotherapy. Int J Gynecol Cancer. 2012;22:260–266.
30. Abu-Rustum NR. Update on sentinel node mapping in uterine can- 36. Sohaib SA, Houghton SL, Meroni R, Rockall AG, Blake P, Reznek RH.
cer: 10-­year experience at Memorial Sloan-­Kettering Cancer Center. J Recurrent endometrial cancer: Patterns of recurrent disease and
Obstet Gynaecol Res. 2014;40:327–334. assessment of prognosis. Clin Radiol. 2007;62:28–34; discussion
31. Barlin JN, Khoury-Collado F, Kim CH, et  al. The importance of 35–36.
applying a sentinel lymph node mapping algorithm in endome- 37. Sorbe B, Juresta C, Ahlin C. Natural history of recurrences in endome-
trial cancer staging: Beyond removal of blue nodes. Gynecol Oncol. trial carcinoma. Oncol Lett. 2014;8:1800–1806.
2012;125:531–535. 38. Sorbe B, Soderstrom K. Treatment of vaginal recurrences in endo-
32. Meyer LA, Bohlke K, Powell MA, et al. Postoperative radiation ther- metrial carcinoma by high-­dose-­rate brachytherapy. Anticancer Res.
apy for endometrial cancer: American Society of Clinical Oncology 2013;33:241–247.
clinical practice guideline endorsement of the American Society 39. Hardarson HA, Heidemann LN, dePont Christensen R, Mogensen O,
for Radiation Oncology evidence-­based guideline. J Clin Oncol. Jochumsen KM. Vaginal vault recurrences of endometrial cancer in
2015;33:2908–2913. non-­irradiated patients -­ Radiotherapy or surgery. Gynecol Oncol Rep.
33. Simcock B, Narayan K, Drummond E, Bernshaw D, Wells E, Hicks RJ. 2015;11:26–30.
The role of positron emission tomography/computed tomography 40. Narayan K, Lin MY, Bernshaw D, Khaw P, Kondalsamy-Chennakesvan
in planning radiotherapy in endometrial cancer. Int J Gynecol Cancer. S. Tailoring adjuvant radiotherapy in endometrial cancer. Indian J
2015;25:645–649. Gynecol Oncol. 2017;15:45.
DOI: 10.1002/ijgo.12616

FIGO CANCER REPORT 2018

Surgical pelvic anatomy in gynecologic oncology

Shailesh Puntambekar1,* | Ranjit Manchanda2

1
Galaxy Care Hospital, Pune, India
2
Abstract
Barts Cancer Institute, Queen Mary
University of London, London, UK Laparoscopy, with its many advantages, has revolutionized the field of pelvic surgery
and enabled surgeons to gain a far greater understanding of pelvic anatomy. As
*Correspondence
Shailesh Puntambekar, Galaxy Care Hospital, technology evolves, our understanding is continuously improving. This article is a
Pune, India.
tribute to the anatomy of the pelvis, which itself has resisted evolutionary changes and
Email: shase63@gmail.com
remained the same, whereas our understanding of how to view and approach various
organ structures has evolved.

KEYWORDS
FIGO Cancer Report; Gynecologic oncology; Hypogastric nerve; Laparoscopy; Pelvic anatomy;
Predictive anatomy

given to understanding the anatomy of the vicinity of these organs


1 | INTRODUCTION
rather than the organs themselves; for example, their precise location
and the approach to these locations, such as vessels, potential spaces,
The science of anatomy is a systematic discipline that involves obser-
and nerves. The present article focuses on pelvic anatomy, which is
vation, understanding, and experimentation to obtain knowledge of the
discussed and explained using various supporting laparoscopic images.
functioning of the human body. Surgery then is the practical application
of this science to treat a disease, remove a tumor or simply minimize
symptomatology, and improve function and quality of life. With advances
2 | PELVIC ANATOMY
in technology, our vision and ability to detail structures and organs have
improved, and consequently so has our understanding. Lessons in anat-
omy that began with studies of cadavers transitioned to live and active 2.1 | Sacral promontory
anatomy lessons taught during surgery, moving from open surgery to
The sacral promontory in its literal sense is the summit of the pel-
the current status of learning anatomy via minimally invasive surgery.
vis. Pathologic conditions of the pelvis may reach the abdomen and
Minimally invasive surgery provides a magnified view of anatomic struc-
beyond; however, their origin always lies at a level below the sacral
tures and is currently one of the best mediums to gain knowledge and
promontory. The sacral promontory is the most protuberant struc-
improve our understanding of anatomy as we know it today.
ture in the bony pelvis and can therefore be used as a fixed point of
Gynecologic malignancies manifest across the clinical spectrum,
reference for various surgeries (Figure S1). It can also form a fixed
from localized tumors to locally aggressive tumors in the pelvic region
reference point in conditions where the general anatomy of the pelvis
to advanced metastatic spread through the abdomen, lungs, thorax,
seems scarred or distorted. Hence, orientation of structures in the pel-
and even the brain. A thorough knowledge of pelvic anatomy is essen-
vis should always begin with identification of the sacral promontory.
tial for the management of most gynecological malignancies and helps
The sacral promontory is significant for the following reasons (Fig. 1):
guide the surgeon toward achieving the required outcome.
Gross anatomy of the pelvis—namely the bladder, uterus, fallopian • The common iliac vessels bifurcate into the internal and exter-
tubes, ovaries, rectum, and the muscles—has remained unchanged; nal iliac vessels at this level.
however, knowledge of the anatomy of various structures that sur- • The ureter crosses over from the lateral to the medial side at this
round these organs has evolved over time. Much emphasis has been level over the bifurcation of the iliac vessels.

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

86  |  wileyonlinelibrary.com/journal/ijgo Int J Gynecol Obstet 2018; 143 (Suppl. 2): 86–92
Puntambekar and Manchanda |
      87

• The superior hypogastric nerve plexus, as a parasympathetic artery here is that it has no branches on the anteromedial aspect of
nerve plexus, unites to form the left and right hypogastric nerves the vessels, which facilitates safer and easier ilio-­obturator lymph
at this level. At this level the nerve fibers of the plexus are seen node dissection (Fig. 2; Figure S4). The inferior epigastric artery
and, when traced inferiorly, form the hypogastric nerves. arises from the anterior aspect of the external iliac artery, which
• The sacral promontory forms the initiation point for transperito- travels above the transversalis fascia under the anterior abdominal
neal para-aortic lymph node dissection. wall. During port placement in laparoscopy, utmost care should be
taken to prevent damage to the vessel since this will lead to signifi-
Following identification of the sacral promontory, careful and skill- cant bleeding. Also, retraction of the vessels under the transversalis
ful dissection performed lateral to the sacral promontory opens the fascia also makes it difficult to achieve hemostasis. The origin of the
pararectal retroperitoneal space. The same landmark is also achieved inferior epigastric artery or the deep circumflex iliac vein inferiorly
by beginning the dissection medial to the infundibulopelvic ligament. adjacent to it is considered the caudal limit of the ilio-­obturator
This approach to the retroperitoneal space is also called the endome- lymph node dissection.
triotic approach or the medial approach. The lateral approach is also While dissecting the pararectal spaces using the lateral approach,
known as the oncological approach, which will be discussed later in the first structure to be visualized after dissecting the peritoneum is
the article. the external iliac vessels. This identification assists in what is known
The line of the small bowel mesentery crosses the sacral promon- as “predictive anatomy” and helps traverse deeper into the pelvis
tory on its way to the right sacroiliac joint. This is significant because during dissection.
reflection of the small bowel along this line helps clearly delineate pel-
vic organs from abdominal organs.
2.2.2 | Internal iliac vessels
After bifurcation of the common iliac vessels at the sacral promon-
2.2 | Vessels
tory, the internal iliac artery originates and then divides into anterior
The arbor vitae of the pelvis begin at the L1 level where the aorta and posterior divisions. The anterior division of the internal iliac artery
bifurcates into the common iliac vessels (Figure S2). The region of is the main blood supply to the vital organs of the pelvis, namely the
vascular importance lies at the level of the sacral promontory where bladder (superior vesical artery) and uterus (uterine artery) (Figure S5).
the common iliac vessels divide into the internal and external iliac The posterior division is of less significance as it pierces the presacral
vessels (Figure S3). The internal iliac vessels are the main blood sup- fascia and supplies the gluteal region.
ply to the pelvis and its organs; however, as we go further into the The anterior division of the internal iliac artery travels parallel to
peripheral and deeper regions, anastomosis of the finer vessel of the the ureter. The first branch of the anterior division is the uterine artery,
external and internal iliac vessels supplying common areas is seen. after which the internal iliac artery branches again as the superior ves-
ical artery at a plane higher than the uterine artery and then contin-
ues as the obliterated hypogastric artery to the anterior abdominal
2.2.1 | External iliac vessels
wall. The uterine artery is the only vessel to cross horizontally over
The external iliac artery travels along the iliopsoas muscles on its the ureter. This is of chief clinical significance because it helps easy
way to supply the lower limbs. The significance of the external iliac

F I G U R E   1   Structures seen at the level of the sacral promontory. F I G U R E   2   External iliac artery and vein remain parallel while
Bifurcation of the common iliac artery into internal and external iliac travelling along the border of the pelvis, remaining medial to the
artery, with the ureter crossing over at the bifurcation. psoas muscle and branching into the inferior epigastric artery branch.
|
88       Puntambekar and Manchanda

F I G U R E   3   Relationships: The ureter always lies medial to the


internal iliac artery. The internal iliac artery always lies between the
F I G U R E   4   The obturator nerves and vessels are the lower limits
ureter and the external iliac artery. The first branch of the internal
of dissection of pelvic lymph nodes and lie between the external and
iliac artery is the uterine artery, followed by the superior vesical
internal iliac vessels. The obturator artery is the only artery arising
artery, after which the vessel collapses to form the obliterated
laterally from the internal iliac artery.
hypogastric artery or ligament.
uterus. The uterine artery is best visualized in the pararectal space
identification of the uterine artery (Fig. 3). According to the rules of (discussed later).
dissection, dissection of tubular structures is done in a parallel direc- Since the ascending cervical branch is formed a few millimeters
tion to the long axis of the structure. Thus, the first structure to away before entering the uterus, this allows easy access to clamp and
obstruct during the dissection of the ureter is the uterine artery. coagulate the vessel during intrafascial hysterectomy. The descending
Since the uterine artery arises much further along from the divi- cervical branch can be avoided as it helps in formation of the stump
sion of the internal iliac artery, this gives a length of approximately post coagulation.
5–6 cm of internal iliac artery devoid of any branches. This can be The uterine artery lies anterior to the ureter, whereas the uter-
useful for ligation of the internal iliac vessels in cases of severe ine vein lies posterior to the ureter (Figure S9; Fig. 5). Consequently,
bleeding (Figure S6). the ureter lies in the fork between the uterine artery and the uterine
There is no direct blood vessel from the internal iliac artery sup- vein. This helps distinguish bleeding in the pararectal area. In cases
plying the ureter, which dispels the myth of segmental supply of the of arterial bleeding, lifting the ureter to close the opening of uterine
ureter. The ureter has its own mesentery and can be completely sepa- artery stops the bleeding; with venous bleeding, lifting the ureter will
rated from the major vessels of the pelvis, thus aiding better dissection not cease the bleeding but will give access to catch the uterine vein.
during radical hysterectomy. On occasion, a small branch from the uterine artery may supply
The internal iliac veins also lie parallel to the artery; hence caution the ureter near the ureteric tunnel. This needs to be identified and
must be taken to carefully ligate only the internal iliac artery and not
the vein. This can be done by tracing it in a retrograde manner 2–3 cm
from the origin of the uterine artery where a cleavage can safely be
created to separate the internal iliac artery from the vein.
The only lateral branch of the internal iliac artery is the obtu-
rator artery, which lies in the lateral paravesical space (discussed
later) (Figure S7). The obturator vessels lie exactly underneath
the obturator nerve. Thus, the lower limit of obturator lymph
node dissection is the appearance of the obturator nerve (Fig. 4).
Dissection beyond this leads to damage to the obturator vessels
and significant bleeding.

2.2.3 | Uterine artery
The uterine artery is the first branch to arise from the anterior
division of the internal iliac artery (Figure S8). It crosses over the
ureter horizontally and anteriorly then divides into descending and F I G U R E   5   The ureter lies in the fork between the uterine artery
ascending cervical branches before piercing the substance of the and the uterine vein.
Puntambekar and Manchanda |
      89

coagulated during tunnel dissection to prevent bleeding and obscu- between the organs and the pelvic walls. These spaces are virtual
ration of the field. spaces, which is to say that they are not naturally present but are cre-
ated or easily developed by dissection to access vital structures under-
neath or to separate or retract the organs during dissection.
2.3 | Nerves
The pelvic structures (uterus, rectum, vagina, urinary bladder) are
3.1 | Pararectal space
supplied by the autonomic nervous system. They are innervated by
motor and sensory sympathetic and parasympathetic nerves. The The retroperitoneal space lying lateral to the rectum on either side is
inferior hypogastric nerve (T10–L2) provides sympathetic nerves and known as the pararectal space. Pararectal spaces take the shape of a
the pelvic splanchnic nerve (Figures S2–S4) provides the parasym- curve of sacrum bone.3 The roof of the pararectal space comprises the
pathetic fibers. Together these fibers merge and form the inferior posterior leaf of the broad ligament. Its medial borders are formed by the
hypogastric plexus, which supplies both the uterus and the urinary rectum and laterally are bound by the internal iliac artery. Cranially the
bladder. Fujii1 described the anatomy of the inferior hypogastric space terminates at the uterine artery and the levator ani forms its floor.
plexus in great detail. The pelvic splanchnic and inferior hypogas- The ureter further divides the pararectal space into the medial and
tric nerve form a cross-­shaped plexus. Nerve-­sparing radical hyster- lateral pararectal spaces. The medial pararectal space is known as the
ectomy involves identifying the inferior hypogastric nerve and the Okabayashi space, whereas the lateral pararectal space is known as
plexus, dividing only the uterine branch, and preserving the bladder the Latzko space (Fig. 6). The Okabayashi space is named after the
branches from the plexus. This helps better preserve urinary bladder famous Japanese surgeon Hidekazu Okabayashi who demonstrated
function after surgery. the first nerve-­sparing radical hysterectomy in Kyoto Imperial Hospital
in 1921.
The Okabayashi space contains nerve fibers of the superior
3 | SPACES hypogastric plexus that traverse from the rectosigmoid junction and
combine to form the hypogastric nerve, and then further at the level
Classical anatomy describes pelvic spaces as coelomic in form or a of the uterovesical junction form a plexus finally innervating the
manifestation of spaces lined by folds of the peritoneum that later bladder. It holds clinical and surgical significance for nerve-­sparing
led to formation of cavities not occupied by organs and fully lined by radical hysterectomy, as well for nerve-­sparing fertility preserving
peritoneal tissue. It also describes the organs lying outside the perito- procedures in cases of endometriosis. The nerves can be seen on
neal cavity and folds of peritoneum as ligaments. However, the latest careful dissection of the Okabayashi space. The approach to the
updates on surgical anatomy have focused on understanding retro- pararectal space is defined by two approaches, namely the endo-
peritoneal spaces. These spaces lie beyond the peritoneum and are metriotic approach and the oncological approach—medial and lat-
approached by dissection of the peritoneum at various places in the eral approaches, respectively (medial and lateral with respect to the
pelvic cavity.2 Since the peritoneal cavity itself is devoid of any vessels infundibulopelvic ligament).
and nerves, the spaces have gained importance in providing access to In the medial approach, the incision on the peritoneum is made
the vessels for dissection and nerves for surgery. The retroperitoneal medial to the infundibulopelvic ligament. The first structure to be
spaces of the pelvis are named based on the location of the near- visualized here under the peritoneum is the ureter. The ureter then
est organs. Each space has its own importance. The retroperitoneal
spaces of the pelvis are classified as follows:

1. Bilateral:
a. Pararectal space
b. Paravesical space
2. Unilateral/midline:
a. Prevesical space
b. Rectovaginal space
c. Retrorectal or presacral space

The peritoneal epithelial lining roofs all retroperitoneal spaces


and the common floor is the levator ani muscles. If these spaces were
to be pictographically explained, they would appear as hollow avas-
F I G U R E   6   The pararectal space is divided by the ureter into the
cular pyramidal structures with the broad base lying over the levator
medial and lateral pararectal spaces. The medial pararectal space
ani muscle and the apex pointed inward toward the peritoneal cavity.
contains the superior hypogastric nerve for performing nerve-­sparing
The retroperitoneal spaces of the pelvis can also be described radial hysterectomy, while the lateral pararectal space is the best
as peritoneal depressions formed by reflections of the peritoneum place for dissection of the uterine artery.
|
90       Puntambekar and Manchanda

needs to be separated from the internal iliac artery. Following the ligament. While medial paravesical space dissection is performed to
rules of dissection, the space is dissected further—parallel and lateral achieve optimum oncological clearance, the lateral paravesical space
to the ureter—until the apex of the uterosacral ligament is reached. contains in itself the obturator and pelvic lymph nodes, which need to
During this process the uterine artery is encountered and dissected. be dissected during radical hysterectomy.
By medializing the ureter, the lateral pararectal space (i.e. the The limit of dissection for the medial paravesical space is the floor
Latzko space) is clearly opened and dissected from its origin. The uter- formed by the levator ani muscle. For the lateral paravesical space, the
ine artery can be easily identified and dissected here. By lateralizing limit of posterior dissection is the obturator nerve. The medial para-
the ureter from the fold of peritoneum or the posterior leaf of the vesical space also provides access for bladder dissection in conditions
broad ligament, the Okabayashi space is opened clearly and the para- of adherent uterovesical fold due to previous cesarean delivery and
sympathetic nerves (Figures S2–S4) can be seen on gentle dissection. sometimes for mobilization of the bladder during exenteration or ure-
This is helpful in nerve-­sparing radical hysterectomy. These principles teric reanastomosis.
are also applicable in surgery for endometriosis.
In the lateral approach, the incision on the peritoneum is made
3.3 | Prevesical space
lateral to the infundibulopelvic ligament and the first structures
visible here are the external iliac artery and vein. This area is the The prevesical space is a small midline retroperitoneal space that lies
best place for easy lymph node dissection. The internal iliac artery between the bladder and the anterior abdominal wall. It communi-
lies below the external iliac vein. Once identified it needs to be cates with the paravesical space on both sides and is enclosed later-
dissected free from the ureter by using the same principles of dis- ally by the lateral umbilical ligament, which is the continuation of the
section. The lateral approach also helps in easy opening of the lat- obliterated hypogastric artery onto the abdominal wall.
eral paravesical space for pelvic lymph node dissection (discussed The prevesical space primarily exposes the bladder neck and
later) (Figure S10). urethra, making it more significant in urogynecology for the manage-
ment of stress urinary incontinence and sling surgeries. However, it
is also useful in anterior exenteration, for example in selected cases
3.2 | Paravesical space
of localized recurrent cervical (or other gynecologic) cancers.
The paravesical space is the retroperitoneal space that lies lateral to
the urinary bladder and anterior and superior to the pararectal space.
3.4 | Rectovaginal space
It is enclosed medially by the bladder, laterally by the pelvic walls, and
inferiorly by the uterine artery (Fig. 7). The pararectal and paravesi- The retroperitoneal space lining the outside of the pouch of
cal space can mutually communicate with each other, and also with Douglas is known as the rectovaginal space. It is enclosed anteriorly
the prevesical space. Dissection of these spaces ensures easy and by the uterus and the posterior vaginal wall, posteriorly by the rec-
complete removal of the uterus and its attachments during radical tum, and laterally by the uterosacral and the Mackenrodt ligament
hysterectomy. (Fig. 8). The roof comprises the peritoneal reflections of the pouch
The paravesical space is divided into medial and lateral paravesical of Douglas and the floor is formed by the levator ani muscle. The
spaces by the obliterated hypogastric artery or the lateral umbilical Denonvilliers fascia is a two-­layered fascia present retroperitoneally

F I G U R E   8   Rectovaginal space: A retroperitoneal space lying


F I G U R E   7   The pararectal and paravesical spaces are divided between the rectum and the uterus under the Pouch of Douglas. It is
by the uterine artery. The boundaries of the pararectal space are covered by two layers of Denonvilliers fascia. Dissection performed
the uterine artery (anterior), internal iliac artery (lateral), and the between the two layers creates an avascular plane and completely
ureter (medial). separates the uterus from the rectum in radical surgeries.
Puntambekar and Manchanda |
      91

yet between the rectum and the vagina: one layer covers the rec- of uterus and the ureter inserting into the bladder4 (Fig. 9). This space
tum and the second layer covers the vagina. The vaginal veins are is lined by the cervicovesical fascia and contains parasympathetic
present underneath the Denonvilliers fascia, covering the vagina nerves innervating the bladder. Careful dissection at this space helps
(Figure S11). Hence, the avascular plane is created exactly between in nerve-­sparing radical hysterectomy.
the two layers of Denonvilliers fascia to dissect the rectovaginal The cervicovesical fascia splits into anterior and posterior leaves.
space. This gives rise to the dictum: the “fat belongs to the bladder The anterior leaf allows the ureter to insert into the bladder (Figure
and fat belongs to the rectum.” S12). The fascia here wraps around the ureter to form bladder pillars,
Rectovaginal space dissection is done until the levator ani is while the posterior leaf further communicates with the endopelvic fas-
reached. This facilitates taking a larger cuff of vagina in radical hys- cia at the level of the levator ani (Figure S13).
terectomy. It also helps in symmetric dissection of the vagina ante-
riorly as well as posteriorly as dissection anteriorly is done below
the level of ureter insertion. It is also helpful in posterior slings 4 | CONCLUSION
in urogynecology.
This article has discussed the newer concepts of pelvic spaces,
ureter and its own mesentery, and details of pelvic vasculature.
3.5 | Retrorectal or presacral space These new concepts will continue to help shape our understanding
A thin, small retroperitoneal space lying behind the rectum is cov- of pelvic anatomy.
ered by the mesorectum anteriorly and Waldeyer fascia posteriorly.
The presacral veins lie posterior to the Waldeyer fascia. If careful
AU T HO R CO NT R I B U T I O NS
avascular dissection is not done between the mesorectum and the
Denonvilliers fascia there could be severe bleeding from the injured SP and MR wrote the manuscript and reviewed the final version.
presacral veins.
The retrorectal space is useful in posterior exenteration for com-
plete mesocolic excision of the involved rectum. It is also useful in CO NFL I C TS O F I NT ER ES T

deep infiltrating endometriosis involving the rectum, sacrocolpopexy, The authors have no conflicts of interest to declare.
and presacral neurectomy.

REFERENCES
3.6 | Yabuki space
1. Fujii S. Anatomic identification of nerve-­sparing radical hyster-
The Yabuki space, unlike other pelvic spaces, is not lined by peritoneal ectomy: A step-­by-­step procedure. Gynecol Oncol. 2008;111:
S33–S41.
epithelial lining. The Yabuki fourth space, as it is also called, is a mid-
2. Mirilas P, Skandalakis J. Surgical anatomy of the retroperitoneal
line small retroperitoneal space confined within the anterior surface spaces part II: The architecture of the retroperitoneal space. Ann Surg.
2010;76:33–42.
3. Ceccaroni M, Pontrelli G, Spagnolo E, et al. Parametrial dissection during
laparoscopic nerve-­sparing radical hysterectomy: A new approach aims
to improve patients’ postoperative quality of life. Am J Obstet Gynecol.
2010;202:e1–e2.
4. Yabuki Y, Sasaki H, Hatakeyama N, Murakami G Discrepancies between
classic anatomy and modern gynecologic surgery on pelvic connective
tissue structure: Harmonization of those concepts by collaborative
cadaver dissection. Am J Obstet Gynecol. 2005;193:7–15.

S U P P O RT I NG I NFO R M AT I O N

Additional supporting information may be found online in the


Supporting Information section at the end of the article.

Figure S1. Sacral promontory: Summit of the pelvis. In conditions of


obscuration of anatomy, dissection can begin from here are a fixed
start point for the purpose of orientation and safety. The structure
under the harmonic is the ureter.
F I G U R E   9   Yabuki space: The retroperitoneal space under the
uterovesical fold of peritoneum. It is bordered by the ureter anteriorly
Figure S2. Bifurcation of the aorta. The aorta bifurcates into the left
and the anterior surface of the uterus posteriorly. It also contains
nerves passing along the ureter to supply the bladder. and right common iliac artery on either side. Mild traction has been
|
92       Puntambekar and Manchanda

given to the right common iliac artery exposing the sacral promontory Figure S6. Left uterine artery and its relationships.
underneath. This is done during para-­aortic lymph node dissection.
Figure S7. Obturator artery and its relationships.

Figure S3. Structures on the right lateral pelvic wall and pararectal Figure S8. Uterine artery.
space, seen from lateral to medial: external iliac artery; external iliac
Figure S9. The uterine artery–vein–ureter complex.
vein; internal iliac artery; ureter. The first structure to cross the ureter
from the internal iliac artery is the uterine artery. Figure S10. Lateral limit of lymph node dissection. Pelvic lymph node
dissection is done until the pubic bone is exposed. Anteriorly, when
Figure S4. External iliac artery vessels. From their origin, the exter-
the traced along the bone, the corona mortis can be seen; this is the
nal iliac artery and vein travel in a parallel fashion. Neither one has
only communication between the external and internal iliac veins.
branches on the anterior or lateral side for the first 6–8 cm. This is
useful in lymph node dissection. The first branch is the inferior epigas- Figure S11. Relationship of veins to the Denonvilliers fascia.
tric artery and vein.
Figure S12. Cervicovesical fascia.

Figure S5. Internal iliac artery and its branches with the ureter. Figure S13. Endopelvic fascia.
DOI: 10.1002/ijgo.12622

FIGO CANCER REPORT 2018

Enhanced recovery after surgery in gynecologic oncology

Gretchen Glaser1 | Sean C. Dowdy1,* | Abraham Peedicayil2

1
Division of Gynecologic Oncology, Mayo
Clinic, Rochester, MN, USA Abstract
2
Department of Gynecologic Oncology, CMC Enhanced recovery protocols consist of a bundle of concepts including early feeding,
Hospital, Vellore, TN, India
opioid-­sparing multimodal pain management, and euvolemia, with the overarching goal
*Correspondence of hastening postoperative recovery. Enhanced recovery after surgery has been shown
Sean C. Dowdy, Division of Gynecologic
to reduce hospital length of stay, reduce costs, and decrease perioperative opioid
Oncology, Mayo Clinic, Rochester, MN, USA.
Email: Dowdy.Sean@mayo.edu requirements in benign and oncologic gynecologic surgery. Interventions without
supporting evidence of benefit, such as the use of mechanical bowel preparation,
routine use of nasogastric tubes and surgical drains, caloric restriction, routine use of
intravenous opioid analgesics, and over-­vigorous intravenous hydration should be
discouraged to improve broader endpoints such as patient satisfaction and overall
recovery. Successful implementation requires engagement from a multidisciplinary
team including surgeons, anesthesiologists, nurses, and pharmacists.

KEYWORDS
Enhanced recovery after surgery; ERAS; Fast-track surgery; FIGO Cancer Report; Quality
improvement

1 |  INTRODUCTION and was first used in colorectal surgery and later adapted to other
surgical specialties.3–5
Surgery will always remain a form of intentional injury, resulting in The three main tenets of ERAS are early feeding, euvolemia, and
collateral damage to normal tissues due to trauma-­induced physio- multimodal pain control; these will be reviewed in the present article.
logical changes mediated by cytokines, nitric oxide, and free oxygen While in recent years there has been rapid adoption of ERAS in many
1
radicals. However, multimodal interventions to combat the surgi- surgical specialties, change has been difficult in some groups because
cal stress response are thought to reduce the deleterious effects of these tenets challenge traditional dogma. To achieve optimal results,
surgical injury and lead to enhanced recovery, decreased morbid- a proactive, structured, and multidisciplinary process for ERAS imple-
ity, and reduced costs.2 Traditionally, patients were starved over- mentation is needed.6 ERAS Society guidelines have been published
night, given opioid analgesia, nasogastric suction, and liberal use for gynecologic oncology and should be considered standard of care.7,8
of intravenous fluids until flatus. To facilitate recovery, adoption of
evidence-­based, best perioperative practices is required. Elements
of best practice include early feeding, avoidance of mechanical 2 | EARLY FEEDING
bowel preparation, utilization of preoperative patient education,
optimization of comorbid conditions, euvolemia, normothermia, use Patients with poor nutritional status should be rehabilitated prior
of minimally invasive surgery, prevention of surgical site infection to surgery. Patients who are nutritionally replete are allowed to
and venous thromboembolism, and assurance of optimal pain con- eat solids until midnight and clear liquids until 2 hours before sur-
trol while minimizing opioid use. Such perioperative optimization gery. Carbohydrate loading is recommended the morning of surgery
is known as fast track or enhanced recovery after surgery (ERAS), to prepare the body for the stress of surgery and minimize insulin

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

Int J Gynecol Obstet 2018; 143 (Suppl. 2): 143–146 wileyonlinelibrary.com/journal/ijgo  |  143
|
144       Glaser ET AL.

resistance.9 In a systematic review of randomized controlled trials in


4 | EUVOLEMIA
patients with gynecologic cancer, early feeding resulted in shorter hos-
pital stay and improved patient satisfaction.10 Patients should resume
In guidelines published by the ERAS Society, maintenance of euv-
a regular diet 4–6 hours after surgery to maintain normal nutritional
olemia in the perioperative period was recognized as a key factor
status and improve healing. Early feeding may result in a higher rate of
in the reduction of morbidity and mortality associated with gyneco-
nausea or even vomiting, which is generally self-­limited and responds
logic oncology surgery.7 This was the result of investigations show-
to antiemetics. This nausea is distinct from paralytic ileus and should
ing that while excess salt and fluid increases complications, very
not be treated as such. Although ERAS protocols do not increase the
restrictive fluid regimens also fail to improve clinical outcomes.18,19
rate of paralytic ileus, if ileus does develop, this should be managed as
Fluid overload can lead to peripheral and small bowel edema, pul-
standard by withholding food and using nasogastric suction.
monary congestion, and electrolyte abnormalities, and increases
Mechanical bowel preparation should be avoided. A recent meta-­
postoperative pain; whereas fluid restriction may decrease cardiac
analysis showed that use of mechanical bowel preparation does not
output and thus oxygen delivery to tissues, delaying healing and
reduce the rate of anastomotic leak, surgical site infection, intra-­
causing end organ dysfunction.
abdominal fluid collections, reoperation, mortality, or interfere with
Rather than define optimal fluid balance quantitatively, clinically
visualization or bowel mobilization.11 On the contrary, mechanical
relevant goals apply.20–22 Replacing insensible losses, maintaining per-
preparations may cause dehydration and electrolyte and metabolic
fusion and blood pressure, allowing permissive oliguria (20 mL/h), and
derangements. Retrospective studies have suggested, however, that
limiting boluses to 250 mL all factor into a reasonable approach to
nonabsorbable oral antibiotics, such as neomycin, may reduce the
volume administration for patients undergoing gynecologic surgery.23
incidence of surgical site infections (RR 0.57; 95% CI 0.43–0.76) with
This not only makes allowances for the normal secretion of antidi-
no effect on organ space infections or anastomotic leaks, although
uretic hormone with surgical stress, but also prevents extracellular
most studies were performed in colorectal rather than gynecologic
fluid overload that may take weeks to eliminate.24
surgery.12,13 A meta-­analysis of mechanical bowel preparation versus
Maintenance of euvolemia is a component of ERAS that can be
oral antibiotic plus mechanical bowel preparation,14 showed that the
difficult to execute given that fluid therapy is administered by mul-
combination was more effective in reducing total surgical site infec-
tiple departments including anesthesia and surgery, and decisions
tions (16% vs 7.2%, P<0.001). A recent ACS-­NSQIP study of over
regarding the administration of intravenous fluids are often made
40 000 patients undergoing colorectal surgery showed that oral anti-
by the most junior members of the care team.25 Furthermore, the
biotics alone were as good as a combination of antibiotic and mechan-
intraoperative time period is a critical time for volume administration.
ical bowel preparation.15 The current practice at Mayo Clinic is to omit
In one meta-­analysis of nearly 1000 randomized patients, the effect
the use of any bowel preparation in favor of adopting measures specif-
of euvolemia was greatly reduced if fluid therapy was implemented
ically targeted to reduce surgical site infection and anastomotic leak.
postoperatively rather than intraoperatively.26 These findings urge
These measures can result in significant improvements in morbidity
multidisciplinary cooperation at initiation and maintenance of any
and do not require bowel preparation with its accompanying risks
ERAS program.
and patient dissatisfaction.7,8 Implementing a bundle of interventions
(made up of 18 elements) can successfully reduce surgical site infec-
tion to very low rates without the use of bowel preparation.16
5 | IMPACT OF ERAS ON LENGTH OF STAY
AND COST OF CARE WITH IMPLEMENTATION
3 |  MULTIMO DAL, OPIOID-­S PARING
PAIN CONTROL Faster, more streamlined recovery of gynecologic oncology patients
has been shown to lead to decreased length of stay and cost reduc-
Prior to induction of anesthesia, it is recommended that patients be tions, as has been demonstrated in colorectal surgery.27 At Mayo
given acetaminophen, celecoxib, and gabapentin. Infiltration of the Clinic, ERAS afforded a 3-­day reduction in median length of stay (from
wound with local anesthetic significantly reduces the need for opioids 8 to 5 days) for patients undergoing complex cytoreductive proce-
immediately after surgery.17 Patients should be provided with aceta- dures (hysterectomy, lymphadenectomy, and omentectomy, as well
minophen, ibuprofen, or ketorolac, and given limited doses of oral as bowel resection, splenectomy, diaphragmatic resection, extensive
opioid if needed. Patients with persistent pain may receive opioids by cytoreduction, or all of these).22 The change was durable, with a similar
intravenous push, with initiation of patient controlled analgesia only if length of stay reported 3 years later, and also included a 90% reduc-
more than two doses of intravenous opioid are necessary. In patients tion in opioid requirements in the first 48 hours.17 Colorectal patients
undergoing laparotomy for advanced ovarian cancer, less than 5% of who underwent similarly complex procedures in hospitals throughout
patients at the Mayo Clinic require patient controlled analgesia using Alberta, Canada, also required two fewer days in the hospital on aver-
these guidelines as part of an enhanced recovery protocol. 17
While age (9.8 days pre-­ERAS vs 7.5 days post-­ERAS).28 This was accompa-
epidurals may be used, less than 5% of patients at the Mayo Clinic nied by no change in 30-­day readmission rates, readmission length
receive an epidural17; this is not a required element of ERAS protocols. of stay, or 30-­day postoperative complications. In addition, patient
Glaser ET AL. |
      145

satisfaction remained high, with over 90% of patients rating care as 6. De Groot JJ, van Es LE, Maessen JM, Dejong CH, Kruitwagen RF,
excellent or very good regarding patient education, pain management, Slangen BF. Diffusion of enhanced recovery principles in gynecologic
oncology surgery: Is active implementation still necessary? Gynecol
quality of care during hospitalization, and the discharge process.22
Oncol. 2014;134:570–575.
The reduction in cost of care with the implementation of ERAS has 7. Nelson G, Altman AD, Nick A, et  al. Guidelines for pre-­ and intra-­
been impressive. Exploration of financial impact for colorectal patients operative care in gynecologic/oncology surgery: Enhanced Recovery
in Canada revealed that for every US $1 invested in ERAS, US $3.8 was After Surgery (ERAS ®) Society recommendations -­ Part I. Gynecol
Oncol. 2016;140:313–322.
returned.29 This was consistent with 17 other studies, as examined in
8. Nelson G, Altman AD, Nick A, et  al. Guidelines for post-­operative
a health economics review of ERAS programs.30 Studies in gyneco- care in gynecologic/oncology surgery: Enhanced Recovery After
logic oncology have also shown reductions in cost. An early program Surgery (ERAS ®) Society recommendations – Part II. Gynecol Oncol.
incorporating some ERAS elements showed a US $5500 reduction in 2016;140:323–332.
9. Kahokehr AA, Sammour T, Sahakian V, Zargar-Shoshtari K, Hill AG.
median 30-­day postoperative hospital costs when compared with tra-
Influences on length of stay in an enhanced recovery programme after
ditional management.31 In more recent investigations employing the colonic surgery. Colorectal Dis. 2011;13:594–599.
entire ERAS spectrum, Kalogera et al.22 reported a 30-­day cost of care 10. Obermair A, Simunovic M, Isenring L, Janda M. Nutrition interven-
savings of more than US $7600 per patient for complex cytoreductive tions in patients with gynecological cancers requiring surgery. Gynecol
Oncol. 2017;145:192–199.
cases, and a US $3000 per patient reduction for staging laparotomies
11. Rollins KE, Javanmard-Emamghissi H, Lobo DN. Impact of mechanical
(hysterectomy, lymphadenectomy, and omentectomy only). Modesitt
bowel preparation in elective colorectal surgery: A metanalysis. World
et al.32 found a median cost reduction of almost US $2000 in their J Gastroenterol. 2018;24:519–536.
gynecologic oncology cohort. In our current healthcare climate, these 12. Bellows CF, Mills KT, Kelly TN, Gagliardi G. Combination of oral non-­
benefits cannot be underestimated. absorbable and intravenous antibiotics versus intravenous antibiotics
alone in the prevention of surgical site infections after colorectal sur-
gery: A meta-­analysis of randomized controlled trials. Tech Coloproctol.
2011;15:385–395.
6 | CONCLUSION 13. Cawich SO, Teelucksingh S, Hassranah S, Naraynsingh V. Role of oral
antibiotics for prophylaxis against surgical site infections after elec-
tive colorectal surgery. World J Gastrointest Surg. 2017;9:246–255.
ERAS protocols combine evidence-­based care components in the
14. Chen M, Song X, Chen LZ, Lin ZD, Zhang XL. Comparing mechani-
perioperative period enabling patients to recover more quickly and cal bowel preparation with both oral and systemic antibiotics versus
resume normal functions. This can be applied to the majority of mechanical bowel preparation and systemic antibiotics alone for the
patients undergoing gynecologic surgery. The successful implemen- prevention of surgical site infection after elective colorectal surgery:
A meta-­analysis of randomized controlled clinical Trials. Dis Colon
tation of an ERAS program requires the active participation of the
Rectum. 2016;59:70–78.
patient and a multidisciplinary team effort. 15. Garfinkle R, Abou-Khalil J, Morin N, et  al. Is there a role for oral
antibiotic preparation alone before colorectal surgery? ACS-­NSQIP
analysis by coarsened exact matching. Dis Colon Rectum. 2017;60:
AUT HOR CONTRI B UTI O N S 729–737.
16. Johnson MP, Kim SJ, Langstraat CL, et al. Using bundled interventions
SD created the concept for this chapter. GG, AP, and SD contributed to reduce surgical site infection after major gynecologic cancer sur-
to the writing. All authors edited and approved the final version of gery. Obstet Gynecol. 2016;127:1135–1144.
the manuscript. 17. Kalogera E, Bakkum-Gamez JN, Weaver AL, et al. Abdominal inci-
sion injection of liposomal bupivacaine and opioid use after
laparotomy for gynecologic malignancies. Obstet Gynecol.
2016;128:1009–1017.
CO NFLI CTS OF I NTE RE ST
18. Lobo DN, Bostock KA, Neal KR, Perkins AC, Rowlands BJ, Allison SP.
The authors have no conflicts of interest to declare. Effect of salt and water balance on recovery of gastrointestinal func-
tion after elective colonic resection: A randomised controlled trial.
Lancet. 2002;359:1812–1818.
19. MacKay G, Fearon K, McConnachie A, Serpell MG, Molloy RG,
REFERENCES
O’Dwyer PJ. Randomized clinical trial of the effect of postoperative
1. Bessey PQ. Metabolic response to critical illness. In: Wilmore DW, intravenous fluid restriction on recovery after elective colorectal sur-
Cheung LY, Harken AH, Holcroft JW, Meakins JL, eds. Scientific gery. Br J Surg. 2006;93:1469–1474.
American Surgery. New York: Scientific American Inc; 1995:1–31. 20. Phan TD, D’Souza B, Rattray MJ, Johnston MJ, Cowie BS. A ran-
2. Kehlet H. Multimodal approach to control postoperative pathophysi- domised controlled trial of fluid restriction compared to oesophageal
ology and rehabilitation. Br J Anaesth. 1997;78:606–617. Doppler-­guided goal-­directed fluid therapy in elective major col-
3. Wind J, Polle SW, Fung Kon Jin PH, et  al. Systematic review of orectal surgery within an Enhanced Recovery After Surgery program.
enhanced recovery programmes in colonic surgery. Br J Surg. Anaesth Intensive Care. 2014;42:752–760.
2006;93:800–809. 21. Gomez-Izquierdo JC, Trainito A, Mirzakandov D, et al. Goal-­directed
4. Zargar-Shoshtari K, Hill AG. Optimization of perioperative care for fluid therapy does not reduce primary postoperative ileus after elec-
colonic surgery: A review of the evidence. ANZ J Surg. 2008;78:13–23. tive laparoscopic colorectal surgery: A randomized controlled trial.
5. Kalogera E, Dowdy SC. Enhanced recovery pathway in gynecologic Anesthesiology. 2017;127:36–49.
surgery: Improving outcomes through evidence-­based medicine. 22. Kalogera E, Bakkum-Gamez JN, Jankowski CJ, et al. Enhanced recov-
Obstet Gynecol Clin North Am. 2016;43:551–573. ery in gynecologic surgery. Obstet Gynecol. 2013;122:319–328.
|
146       Glaser ET AL.

23. Dowdy SC, Nelson G. Enhanced recovery in gynecologic oncol- 28. Nelson G, Kiyang LN, Crumley ET, et al. Implementation of enhanced
ogy -­ A sea change in perioperative management. Gynecol Oncol. recovery after surgery (ERAS) across a provincial healthcare sys-
2017;146:225–227. tem: The ERAS Alberta colorectal surgery experience. World J Surg.
24. Plank LD, Connolly AB, Hill GL. Sequential changes in the metabolic 2016;40:1092–1103.
response in severely septic patients during the first 23 days after the 29. Thanh NX, Chuck AW, Wasylak T, et al. An economic evaluation of the
onset of peritonitis. Ann Surg. 1998;228:146–158. enhanced recovery after surgery (ERAS) multisite implementation pro-
25. Gramlich LM, Sheppard CE, Wasylak T, et  al. Implementation of gram for colorectal surgery in Alberta. Can J Surg. 2016;59:415–421.
enhanced recovery after surgery: A strategy to transform surgical care 30. Stowers MD, Lemanu DP, Hill AG. Health economics in enhanced
across a health system. Implement Sci. 2017;12:67. recovery after surgery programs. Can J Anaesth. 2015;62:219–230.
26. Rahbari NN, Zimmermann JB, Schmidt T, Koch M, Weigand MA, 31. Gerardi MA, Santillan A, Meisner B, et  al. A clinical pathway for
Weitz J. Meta-­analysis of standard, restrictive and supplemen- patients undergoing primary cytoreductive surgery with rectosig-
tal fluid administration in colorectal surgery. Br J Surg. 2009;96: moid colectomy for advanced ovarian and primary peritoneal cancers.
331–341. Gynecol Oncol. 2008;108:282–286.
27. Lemanu DP, Singh PP, Stowers MD, Hill AG. A systematic review to 32. Modesitt SC, Sarosiek BM, Trowbridge ER, et al. Enhanced recovery
assess cost effectiveness of enhanced recovery after surgery pro- implementation in major gynecologic surgeries: Effect of care stan-
grammes in colorectal surgery. Colorectal Dis. 2014;16:338–346. dardization. Obstet Gynecol. 2016;128:457–466.
DOI: 10.1002/ijgo.12621

FIGO CANCER REPORT 2018

Cancer in pregnancy

Matthys H. Botha1,* | Shalini Rajaram2 | Kanishka Karunaratne3

1
Department of Obstetrics and
Gynecology, Stellenbosch University and Abstract
Tygerberg Hospital, Stellenbosch, South Africa The incidence of cancer in pregnancy is increasing. The most frequent malignancies
2
Department of Obstetrics and
include breast and cervical cancers. Diagnosis may be complicated by late presentation.
Gynecology, University College of Medical
Sciences and Guru Teg Bahadur Hospital, Imaging during pregnancy should consider risks to the fetus. Diagnostic work-­up,
Delhi, India
including tumor markers, can be influenced by the physiology of pregnancy. Treatment
3
National Cancer Institute of Sri Lanka,
of cancer can often be safely administered with good maternal and fetal outcomes.
Maharagama, Sri Lanka
Chemotherapy, radiotherapy, and surgery must be adapted to the pregnancy state.
*Correspondence
Counselling and emotional support are an essential part of management.
Matthys H. Botha, Department of Obstetrics
and Gynecology, Stellenbosch University,
Stellenbosch, South Africa. KEYWORDS
Email: mhbotha@sun.ac.za Cancer in pregnancy; Cancer treatment; Chemotherapy; FIGO Cancer Report;
Pregnancy; Radiotherapy

1 |  INTRODUCTION from other registries, and cervical and ovarian cancer incidence was
lower.2 In an international cohort of 1170 woman diagnosed with
Cancer in pregnancy, although uncommon, has been increasing in cancer during pregnancy, the most common invasive cancers in preg-
recent years as demonstrated by various epidemiological studies. nancy were breast cancer (39%, n=462), followed by cervical (13%,
The Danish registry showed a rise in the proportion of cancer asso- n=147), lymphoma (10%, n=113), ovarian (7%, n=88), and leukemia
ciated with pregnancy from 5.4% (n=572) to 8.3% (n=1052) over a (6%, n=68).4 A population-­based study from Sweden found that the
30-­year period—the most common cancers being melanoma, cervical, most common cancer during pregnancy was melanoma (25%, n=232),
1
and breast cancer. The increased incidence could not be explained followed by breast (15%, n=139), cervical (15%, n=139), and ovarian
by advanced maternal age alone. An Australian study on pregnancy-­ cancer (6%, n=54).5
associated cancer reported 1798 cancers over a 14-­year period, 499 The change in treatment and outcome of cancer in pregnancy was
during pregnancy and 1299 in the postpartum period, giving a crude reflected in the international cohort study.3 For every five years of the
incidence of 137.3 per 100 000 pregnancies.2 There was a statisti- study (1996–2016), treatment increased by 10% (95% CI 5–15) and
cally significant increase in pregnancy-­associated malignancy during use of chemotherapy increased by 31% (95% CI 20–43). Live birth
this period and the number of mothers aged over 35 years increased rates increased and preterm births decreased. Maternal survival was
from 13.2% to 23.6%. However, age explained only a 14% increase similar to nonpregnant women treated for cancer and encouraging
in incidence. The postpartum period (up to 12 months after delivery) fetal, neonatal, and early childhood outcomes were observed in this
was included, bearing in mind a delay in diagnosis during pregnancy or study. As such, oncologic treatment is possible during pregnancy,
considering it part of the cancer-­in-­pregnancy continuum. often without significantly endangering maternal or fetal safety.
In a recent Italian population-­based linkage study, breast cancer
was the most common cancer (32%, n=479) and the risk of developing
a pregnancy-­related cancer increased significantly with age, from 60 2 | DIAGNOSIS
per 100 000 for women younger than 30 years to 265 per 100 000 for
women older than 40 years.3 Melanoma, breast, and thyroid cancers Diagnosis of cancer is vital for successful treatment regardless of
were more common in the Australian population than those reported pregnancy status. Diagnosis of cancer in pregnancy is, unfortunately,

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium,
provided the original work is properly cited.
© 2018 The Authors. International Journal of Gynecology & Obstetrics published by John Wiley & Sons Ltd on behalf of International Federation of Gynecology and
Obstetrics

Int J Gynecol Obstet 2018; 143 (Suppl. 2): 137–142 wileyonlinelibrary.com/journal/ijgo  |  137
|
138       Botha ET AL.

often delayed; this is in part because many symptoms of malignancy uterine corpus and ovaries, pregnancy has various effects on benign
are similar to the symptoms of pregnancy, including nausea/vomit- conditions that may mimic malignancy.
ing, breast changes, abdominal pain, anemia, and fatigue. Breast
changes and the pregnant uterus may make physical examination
3 | SPECIFIC CANCERS
difficult. In addition, the clinician may be more hesitant to assign the
appropriate tests because of concerns that laboratory results may
3.1 | Cervical cancer
be inaccurate or that radiologic testing is harmful. Owing to its rar-
ity, cancer might not be considered in the differential diagnosis. This Cervical cancers are best diagnosed by cytology in early pregnancy,
delay in diagnosis can lead to late presentation, complex treatment, but may also present with abnormal bleeding, vaginal discharge, and
and poor prognosis.6 abdominopelvic pain.16 Cervical cancers in pregnancy generally pre-
sent as Stage I disease at diagnosis. Pregnant women are three times
more likely to have Stage I disease than nonpregnant women with
2.1 | Laboratory testing
cervical cancer.17
Tumor markers should be used with caution owing to pregnancy-­ Management of preinvasive disease (CIN 1 to CIN 3) can be
induced elevation. The sensitivity and specificity of tumor markers deferred until 6–8 weeks after delivery. However, it is recommended
may be lower during pregnancy.7 Physiological changes in pregnancy that a colposcopy is performed in each trimester to assess lesion size
and accompanying alteration of commonly used laboratory values and disease progress. Colposcopy can be challenging during pregnancy
may complicate the diagnosis of malignancy. Hemoglobin and hema- owing to increased vascularity and an increase in genital edema.18
tocrit values are typically lower, whereas alkaline phosphatase and Immediate definitive treatment, regardless of gestational age, is
lactate dehydrogenase are typically higher in pregnancy. CA 15-­3 generally appropriate for invasive cancer in the following settings19:
used in breast cancer, CA 125 used in epithelial ovarian cancer, and
alpha-­fetoprotein used in germ cell tumors are physiologically ele- 1. Documented lymph node metastases.
vated in pregnancy.8,9 2. Progression of disease during the pregnancy.
3. Patient choice to terminate the pregnancy.

2.2 | Imaging
Stage IA1 disease can be managed by conization and is best done
Imaging in pregnancy to diagnose and stage cancer may create con- between 12 and 20 weeks of pregnancy.19 The term “coin biopsy” is
flict between maternal benefit and fetal risk. Therefore, the following sometimes used in pregnancy to highlight that the incision should not be
issues need to be taken into account when choosing the appropriate deep enough to cause damage to the fetal membranes. This is best per-
imaging technique: formed in theatre with adequate anesthesia. Prophylactic cerclage may
be an option both for prevention of premature labor and management of
1. Safety of the fetus. operative bleeding.
2. Risk of metastasis. In stages IA2–IB1 (less than 2 cm) disease, conization or simple
3. Viability of the fetus. trachelectomy can be performed because parametrial extension is
seen in less than 1% of women. Simple trachelectomy is a less com-
Radiation exposure above 100 mGy is associated with fetal plicated procedure and is defined as excision of the cervix 1 cm above
malformation and childhood cancer.10 If X-­rays are needed, proper the tumor border.20 However, it is necessary to assess lymph node
abdominal shielding should be provided. Mammogram images are status by laparoscopic pelvic lymphadenectomy before conservative
increasingly difficult to interpret owing to physiological hypervascu- surgery. In the event of node-­positive disease, the woman should be
larity and density of the breast tissue.11 Computed tomography (CT told that immediate treatment is recommended. Laparoscopic lymph-
scan) is best avoided during pregnancy because of the unaccept- adenectomy is best performed before 20 weeks of pregnancy.17,19 The
able cumulative radiation and contrast doses.12 Positron-­emission second international consensus guidelines recommend this treatment
tomography (PET) imaging in pregnancy is debatable owing to the in pregnancies of less than 22–25 weeks.19 Abdominal and vaginal
13
risk to the fetus of radiation exposure. radical trachelectomy have also been performed during pregnancy,
Ultrasound as a tool for diagnosis and staging is used worldwide. with variable pregnancy outcomes. Abdominal radical trachelectomy
It is noninvasive and helps to perform guided biopsies of the breast is technically more challenging and is associated with significant
and lymph nodes. blood loss and prolonged surgery (3.5–7.5 hours, median 5.3 hours).21
Magnetic resonance imaging (MRI) is safe in all trimesters of preg- Obstetric outcome is poor and, therefore, radical trachelectomy is not
nancy. It is the imaging technique of choice for diagnosis and staging.14 recommended during pregnancy.19
Histopathology of tissues provides definitive diagnosis of tumor Stage IB1 tumors larger than 2 cm may be treated with neoad-
type and grading. The pathologist should always be informed of the juvant chemotherapy (NACT) with or without pelvic lymphadenec-
patient’s pregnancy status to avoid incorrect diagnosis resulting from tomy. Chemotherapy is relatively safe in the second trimester
pregnancy-­associated tissue changes.15 Aside from the changes to the although there is a higher risk of preterm labor, premature rupture of
Botha ET AL. |
      139

membranes, and fetal growth restriction. NACT stabilizes the tumor Treatment is as for nonpregnant women with the goal of achieving
until fetal maturity is achieved so that cesarean delivery and radical local control and preventing distant metastases. Modified radical mas-
hysterectomy can be performed. In women with advanced pregnancy tectomy with axillary staging is the treatment of choice in the first tri-
(greater than 22–25 weeks), pelvic lymphadenectomy is not possible mester. Adjuvant chemotherapy can be started in the second trimester
and those with Stage IA1 to Stage IB1 disease can postpone defini- and radiation therapy given postpartum.28,29 Breast conserving sur-
tive treatment until fetal maturity is achieved without compromising gery can be done safely in the second and third trimesters. Experience
survival. NACT can also be given for locally advanced cancers and in in using sentinel lymph node biopsy (SLNB) in pregnancy is limited.
advanced pregnancy to preserve pregnancy until optimal fetal survival Blue dye is contraindicated but technetium-­99m has been used safely.
(35–36 weeks) is reached. Cesarean delivery followed by radical sur- Adjuvant chemotherapy with anthracycline-­based regimes, such as
gery or definitive chemotherapy/radiotherapy showed good obstetric various combinations of cyclophosphamide, doxorubicin, and fluoro-
and oncological outcome.22 uracil, can be started in the second and third trimester. There are insuf-
Cesarean delivery is the preferred choice for delivery of the fetus ficient data to recommend taxanes, but paclitaxel can be used after
in the presence of bulky tumors. Vaginal delivery risks the possibility of the first trimester if indicated by disease status. Trastuzumab is con-
catastrophic bleeding and implant metastases in vaginal tears or epi- traindicated and oligohydramnios/anhydramnios have been reported.
siotomy scars. In locally advanced tumors a lower segment transverse Endocrine treatments including tamoxifen, aromatase inhibitors, and
cesarean delivery should be avoided owing to the risk of cutting or LHRH analogues are contraindicated in pregnancy due to the high risk
tearing into tumor tissue. A classical incision will minimize blood loss of birth defects. If a woman becomes pregnant while on tamoxifen,
and avoid the large tumor vessels. pregnancy termination should be advised.28,29
Where pregnancy preservation is not desired or in advanced cases,
pregnancy termination and treatment as in nonpregnant women
3.3 | Ovarian cancer
is advocated. In early gestation (less than 12 weeks), spontaneous
abortion occurs after pelvic radiation. In second trimester cases, hys- Adnexal masses are not uncommon in pregnancy and occur in roughly
terotomy followed by definitive chemoradiation is preferred because 1 in 600 to 1 in 1500 pregnancies. The majority of these are benign
obstetric complications are fewer. and only 1%–3% may be malignant. The most commonly encountered
ovarian malignancy is germ cell, followed by sex cord stromal tumors,
borderline tumors, and lastly invasive epithelial cancers.30 The most
3.2 | Breast cancer
common symptom is abdominal or pelvic pain and one-­third of ovar-
Breast cancer, although uncommon, is the most prevalent malig- ian cancers are diagnosed incidentally. The majority of tumors pre-
nancy encountered in pregnancy and the postpartum period. The sent as Stage I, with a mean age of 25.8 years and 31.6 years in germ
difficulty and delay in diagnosis may be attributed to pregnancy-­ cell tumors and invasive epithelial cancers, and mean size of 18 and
related changes in the breast and diagnostic challenges that allow 12 cm, respectively.31,32
these cancers to go undetected until the first postpartum year. Transvaginal and abdominal ultrasound have high sensitivity
Breast cancers in pregnancy are usually larger in size, node posi- and specificity in the diagnosis of ovarian masses. However, MRI
tive, Stage II or III, high-­grade invasive ductal cancer, and usually has the best diagnostic accuracy in the diagnosis of ovarian malig-
ER/PR/HER2/neu-­negative.23,24 Prognosis is related to tumor nancy in pregnancy.33 MRI (without contrast) and diffusion-­weighted
characteristics and delay in starting treatment and not to preg- imaging are useful in assessing the extent of peritoneal disease and
nancy alone. nodal metastases. Tumor markers are not useful as CA 125, alpha-­
When breast cancer is suspected in pregnancy, delay in diagno- fetoprotein, and beta hCG are elevated in pregnancy. Inhibin B, anti-­
sis should be avoided. Mammography and ultrasound are the best Müllerian hormone, HE4, CA 19-­9, and lactate dehydrogenase are not
imaging modalities; mammography has a sensitivity of more than 80% elevated in pregnancy and can be used for diagnosis.
despite pregnancy-­associated breast changes.25 Ultrasound can be Early-­stage ovarian cancer can be treated surgically in the second
used to assess the extent of the disease in breast and lymph nodes and and third trimester with surgical staging, salpingo-­oophorectomy,
for guiding biopsies.26 Core needle biopsy is the preferred modality. omentectomy, peritoneal biopsies, and evaluation of suspicious
Experience with MRI in pregnancy is limited and gadolinium-­enhanced lymph nodes. Both laparoscopy and laparotomy are accepted pro-
contrast is contraindicated in the first trimester because it crosses the cedures. Care should be taken to avoid ovarian rupture and spillage.
placenta. A recent study of breast MRI showed 98% sensitivity in Paclitaxel/carboplatin-­based chemotherapy can be given safely in
diagnosis of pregnancy-­associated breast cancer and changed treat- the second and third trimester in epithelial ovarian cancer, stop-
ment modality in 28% of women.27 Comprehensive staging studies are ping before 37 weeks to avoid myelosuppression in the neonate.
needed because breast cancer in pregnancy may present in advanced Bevacizumab is not recommended during pregnancy. Vaginal delivery
stage. Chest X-­ray with abdominal shielding to evaluate the lungs and is ideal in early-­stage ovarian cancer treated surgically. In germ cell
ultrasound for liver involvement are safe in pregnancy. Bone scans can tumors, a BEP regime is considered too toxic because fetal growth
be performed postpartum, but if a woman is symptomatic, noncon- restriction and neonatal complications are high. Weekly paclitaxel
trast skeletal MRI is safe.25 and cisplatin is recommended as per ESMO guidelines.29 Advanced
|
140       Botha ET AL.

cancers (Stage III and IV) detected in the first trimester and early Trastuzumab is generally contraindicated in pregnancy because of
second trimester will need complete debulking surgery and adju- HER2 receptors on the kidneys of the fetus, resulting in oligo-­ or anhy-
vant chemotherapy. In the late second and third trimesters, NACT is dramnios and fetal lung hypoplasia. The antifolates, such as metho-
given until fetal maturity is reached. Cesarean delivery followed by trexate, are also contraindicated.
complete cytoreductive surgery is feasible. Although pregnancy out- The risk of congenital malformation is linked directly to gesta-
comes are good, prematurity, fetal loss due to surgical complications, tional age and before 12 weeks there is a risk for abnormalities of the
and fetal growth restriction do occur. Oncological outcomes are the eyes, ears, and blood systems; the risk decreases significantly after
same as in nonpregnant women.34 complete organogenesis.
Chemotherapy may cause a significant reduction in maternal blood
production, leading to low platelet counts and risk of overwhelming
4 |  CANCER TREATMENT IN PREGNA NCY
infection. When chemotherapy is used, timing of delivery should be
planned carefully. Elective delivery should not be planned within three
4.1 | Radiotherapy
weeks after chemotherapy. For this reason, chemotherapy should not
Radiotherapy in a pregnant patient should be planned carefully. be administered after 37 weeks of pregnancy owing to risk of sponta-
The radiation oncologist and the physicist must minimize direct and neous onset of labor.
indirect sources of radiation to the fetus. The fetal dose should not Long-­term follow-­up of children born to mothers who received
exceed 50–100 mGy.35 The risks at various times of fetal develop- chemotherapy during pregnancy does not indicate an increased risk
ment are summarized in Table 1. of congenital abnormalities or mental delay. The number of children
Scatter and leakage radiation are a concern even with proper with long-­term follow-­up is still small and the data should be inter-
shielding, although adequate shielding can minimize risks. Shielding preted with caution. Potential risks include concern for cardiac func-
of the gravid uterus, especially in advanced pregnancy, can be diffi- tion in children exposed to anthracyclines during the fetal period.
cult owing to the heavy materials used. Some obstetric experts advise Anthracyclines are commonly used for breast cancer treatment and
regular clinical and ultrasound examinations to determine the lie of have a direct effect on cardiac function. In a follow-­up study of 17
the fetus to get the position of the fetal head out of the potential field children, no changes in electrocardiogram or echocardiography could
of radiotherapy; for example, in the case of chest radiotherapy, the be found after the use of anthracyclines.
fetus should be in a cephalic position and external cephalic version
may be indicated.
4.3 | Surgery
Cancer surgery in pregnancy may be indicated for diagnosis, treat-
4.2 | Chemotherapy
ment, and staging. Surgery is urgent but not an emergency and may be
After the first trimester, most chemotherapeutic agents can be used delayed until fetal maturity is established without compromising total
with relative safety. Transplacental transport of chemotherapeutic care. As a principle, surgical procedures are best undertaken in the
agents differs widely, with some agents such as paclitaxel crossing second trimester to prevent spontaneous abortion.38,39
the placenta at a low rate, anthracyclines crossing the placenta at an Surgery in the second trimester is technically less complicated com-
intermediate rate, and carboplatin at a high rate.8,9 Despite this, car- pared with in the third trimester owing to the size of the uterus. When
boplatin administered after 12–14 weeks of pregnancy seems to do surgery is performed after 28 weeks, tocolytics should be admin-
little harm to the developing fetus.37 istered to prevent premature labor and corticosteroids to enhance
lung maturity. Handling of the uterus should be kept to a minimum.
T A B L E   1   Risks to the fetus of radiotherapy during pregnancy.a Unilateral or bilateral oophorectomy can be carried out safely after
the first trimester. Whenever possible, regional anesthesia is preferred
Gestational age (weeks) Risks
over general anesthesia owing to the potential risk of aspiration.40
Preimplantation (1) Lethality A lateral tilt during surgery may help to prevent aortocaval com-
Organogenesis (2–7) Lethality, gross malformations, growth pression.41 The patient should be consented for emergency cesarean
retardation, sterility, cataracts, other
delivery in the third trimester if fetal compromise is encountered.
neuropathology, malignant disease
Therapeutic surgery can be performed in any trimester.
Early fetal (8–15) Lethality, gross malformations, growth
retardation, mental retardation,
sterility, cataracts, malignant disease
Midfetal (16–25) Gross malformations, growth 5 | PLACENTAL AND FETAL
retardation, mental retardation, TUMOR INVOLVEMENT
sterility, cataracts, malignant disease
Late fetal (>25) Growth retardation, sterility, cataracts, Metastatic disease to the placenta and the fetus is rare. The most
malignant disease likely tumors to metastasize to the placenta include melanomas and
a
Adapted from Stovall et al.36 hematological malignancies. In all cases where malignant spread is
Botha ET AL. |
      141

possible, the placenta should be submitted for careful histologic evalu- information in a clear, evidence-­based, and unbiased manner, giv-
ation. The fetus should be examined carefully at birth and at regular ing due consideration to the patient’s values, concerns, beliefs, and
intervals after birth for any signs of metastatic disease. priorities in life.

AU T HO R CO NT R I B U T I O NS
6 | COUNSELLING
MB, SR, and KK each contributed to the research and writing of the
Cancer during pregnancy represents both a psychological and biologi- manuscript.
cal dilemma given that treatment should be directed to save two lives:
maternal and fetal. Using a multidisciplinary approach, counselling can
help to reduce the distress of the patient and her family. It is essential CO NFL I C TS O F I NT ER ES T
that the obstetrician, oncologist, pediatrician, and psychotherapist take
The authors have no conflicts of interest to declare.
leading roles. The patient and her family should be actively involved in
the decision-­making process, which will enhance confidence and support.

REFERENCES
6.1 | Breaking bad news 1. Eibye S, Kjaer SK, Mellemkjaer L. Incidence of pregnancy-­associated
cancer in Denmark, 1977–2006. Obstet Gynecol. 2013;122:608–617.
Receiving bad news is painful for any patient.42 Counselling should 2. Lee YY, Roberts CL, Dobbins T, et  al. Incidence and outcomes of
always include information on the ongoing pregnancy and impact of pregnancy-­associated cancer in Australia, 1994–2008: A population-­
the disease on the mother and baby.43 Certain guidelines should be based linkage study. BJOG. 2012;119:1572–1582.
3. Parazzini F, Franchi M, Tavani A, Negri E, Peccatori FA. Frequency
adhered to42:
of pregnancy related cancer: A population based linkage study in
Lombardy, Italy. Int J Gynecol Cancer. 2017;27:613–619.
 1. Assess the mental state of the patient. 4. de Haan J, Verheecke M, Van Calsteren K, et  al. Oncological man-
 2. Ensure privacy. agement and obstetric and neonatal outcomes for women diagnosed
with cancer during pregnancy: A 20-­year international cohort study of
 3. Take adequate time to assess the situation.
1170 patients. Lancet Oncol. 2018;19:337–346.
 4. Be honest. 5. Andersson TM, Johansson AL, Fredriksson I, Lambe M. Cancer during
 5. Provide accurate information. pregnancy and the postpartum period: A population-­based study.
 6. Show empathy. Cancer. 2015;121:2072–2077.
 7. Arrange for family members to be present. 6. Voulgaris E, Pentheroudakis G, Pavlidis N. Cancer and pregnancy: A
comprehensive review. Surg Oncol. 2011;20:e175–e185.
 8. Provide evidence-based treatment options.
7. Han SN, Lotgerink A, Gziri MM, Van Calsteren K, Hanssens M, Amant
 9. Inform about other supportive services. F. Physiologic variations of serum tumor markers in gynecologi-
10. Clearly indicate that the patient has the final decision regarding cal malignancies during pregnancy: A systematic review. BMC Med.
their care. 2012;10:86.
8. Sarandakou A, Protonotariou E, Rizos D. Tumor markers in bio-
11. Briefly explain the process by which the diagnosis was reached.
logical fluids associated with pregnancy. Crit Rev Clin Lab Sci.
12. Provide varied methods to convey the information, for example, 2007;44:151–178.
written material and video. 9. Moore RG, Miller MC, Eklund EE, Lu KH, Bast RC Jr, Lambert-
Messerlian G. Serum levels of the ovarian cancer biomarker HE4 are
decreased in pregnancy and increase with age. Am J Obstet Gynecol.
6.2 | Counselling about prognosis 2012;206:349.e1–349.e7.
10. McCollough CH, Schueler BA, Atwell TD, et  al. Radiation exposure
Patients require information about the prognosis to make treatment and pregnancy: When should we be concerned? Radiographics.
decisions. Most patients want specific and honest information about 2007;27:909–917; discussion 917–918.
11. Ayyappan AP, Kulkarni S, Crystal P. Pregnancy-­associated
the prognosis:
breast cancer: Spectrum of imaging appearances. Br J Radiol.
2010;83:529–534.
1. Specific: median survival. 12. Wang PI, Chong ST, Kielar AZ, et al. Imaging of pregnant and lactating
2. General: “I think your chances are good.” patients: Part 2, evidence-­based review and recommendations. AJR
Am J Roentgenol. 2012;198:785–792.
3. Statistical: average time gain.
13. Almuhaideb A, Papathanasiou N, Bomanji J. 18F-­FDG PET/CT imag-
4. Exceptional cases: survival against the odds. ing in oncology. Ann Saudi Med. 2011;31:3–13.
14. Michielsen K, Vergote I, Op de Beeck K, et al. Whole-­body MRI with
A realistic and honest approach is essential to maintain the diffusion-­weighted sequence for staging of patients with suspected
patient’s confidence and support. Providing too much information ovarian cancer: A clinical feasibility study in comparison to CT and
FDG-­PET/CT. Eur Radiol. 2014;24:889–901.
using medical jargon, hiding news, false reassurance, and a pater-
15. Amant F, Deckers S, Van Calsteren K, et  al. Breast cancer in preg-
nalistic approach disregarding the patient’s concerns all reduce their nancy: Recommendations of an international consensus meeting. Eur
confidence.44 The basis of shared decision making is to exchange J Cancer. 2010;46:3158–3168.
|
142       Botha ET AL.

16. Sekine M, Kobayashi Y, Tabata T, et al. Malignancy during pregnancy in 30. Leiserowitz GS, Xing G, Cress R, Brahmbhatt B, Dalrymple JL, Smith
Japan: An exceptional opportunity for early diagnosis. BMC Pregnancy LH. Adnexal masses in pregnancy: How often are they malignant?
Childbirth. 2018;18:50. Gynecol Oncol. 2006;101:315–321.
17. Morice P, Uzan C, Gouy S, Verschraegen C, Haie-Meder C. 31. Blake EA, Kodama M, Yunokawa M, et  al. Feto-­maternal outcomes
Gynaecological cancers in pregnancy. Lancet. 2012;379:558–569. of pregnancy complicated by epithelial ovarian cancer: A systematic
18. Hunter MI, Monk BJ, Tewari KS. Cervical neoplasia in pregnancy. Part review of literature. Eur J Obstet Gynecol Reprod Biol. 2015;186:97–105.
1: Screening and management of preinvasive disease. Am J Obstet 32. Kodama M, Grubbs BH, Blake EA, et  al. Feto-­maternal outcomes
Gynecol. 2008;199:3–9. of pregnancy complicated by ovarian malignant germ cell tumor:
19. Amant F, Halaska MJ, Fumagalli M, et al. Gynecologic cancers in preg- A systematic review of literature. Eur J Obstet Gynecol Reprod Biol.
nancy: Guidelines of a second international consensus meeting. Int J 2014;181:145–156.
Gynecol Cancer. 2014;24:394–403. 33. Thomassin-Naggara I, Fedida B, Sadowski E, et  al. Complex US
20. Rob L, Skapa P, Robova H. Fertility-­sparing surgery in patients with adnexal masses during pregnancy: Is pelvic MR imaging accurate for
cervical cancer. Lancet Oncol. 2011;12:192–200. characterization? Eur J Radiol. 2017;93:200–208.
21. Capilna ME, Szabo B, Becsi J, Ioanid N, Moldovan B. Radical tra- 34. Fruscio R, de Haan J, Van Calsteren K, Verheecke M, Mhallem M,
chelectomy performed during pregnancy: A review of the literature. Amant F. Ovarian cancer in pregnancy. Best Pract Res Clin Obstet
Int J Gynecol Cancer. 2016;26:758–762. Gynaecol. 2017;41:108–117.
22. Ricci C, Scambia G, De Vincenzo R. Locally advanced cervical cancer 35. Shaw P, Duncan A, Vouyouka A, Ozsvath K. Radiation exposure and
in pregnancy: Overcoming the challenge. A case series and review of pregnancy. J Vasc Surg. 2011;53(Suppl.1):28S–34S.
the literature. Int J Gynecol Cancer. 2016;26:1490–1496. 36. Stovall M, Blackwell CR, Cundiff J, et al. Fetal dose from radiotherapy
23. Middleton LP, Amin M, Gwyn K, Theriault R, Sahin A. Breast carci- with photon beams: Report of AAPM Radiation Therapy Committee
noma in pregnant women: Assessment of clinicopathologic and Task Group no. 36. Med Phys. 1995;22:63–82.
immunohistochemical features. Cancer. 2003;98:1055–1060. 37. Cordeiro CN, Gemignani ML. Gynecologic malignancies in pregnancy:
24. Johansson ALV, Andersson TM, Hsieh CC, et al. Tumor characteristics Balancing fetal risks with oncologic safety. Obstet Gynecol Surv.
and prognosis in women with pregnancy-­associated breast cancer. Int 2017;72:184–193.
J Cancer. 2018;142:1343–1354. 38. Mazze RI, Kallen B. Appendectomy during pregnancy: A Swedish reg-
25. Case AS. Pregnancy-­associated breast cancer. Clin Obstet Gynecol. istry study of 778 cases. Obstet Gynecol. 1991;77:835–840.
2016;59:779–788. 39. Cohen-Kerem R, Railton C, Oren D, Lishner M, Koren G. Pregnancy
26. Yang WT, Dryden MJ, Gwyn K, Whitman GJ, Theriault R. Imaging of outcome following non-­obstetric surgical intervention. Am J Surg.
breast cancer diagnosed and treated with chemotherapy during preg- 2005;190:467–473.
nancy. Radiology. 2006;239:52–60. 40. Ezri T, Szmuk P, Evron S, Geva D, Hagay Z, Katz J. Difficult airway in
27. Myers KS, Green LA, Lebron L, Morris EA. Imaging appearance and obstetric anesthesia: A review. Obstet Gynecol Surv. 2001;56:631–641.
clinical impact of preoperative breast MRI in pregnancy-­associated 41. Kendrick JM, Neiger R. Intraoperative fetal monitoring during nonob-
breast cancer. AJR Am J Roentgenol. 2017;209:W177–W183. stetric surgery. J Perinatol. 2000;20:276–277.
28. National Comprehensive Cancer Network. NCCN Clinical Practice 42. Girgis A, Sanson-Fisher RW. Breaking bad news. 1: Current best
Guidelines in Oncology (NCCN guidelines). Breast Cancer. 2017. advice for clinicians. Behav Med. 1998;24:53–59.
https://www.nccn.org. Accessed February 1, 2018. 43. Alder J, Bitzer J. Psychooncologic care in young women facing cancer
29. Zagouri F, Dimitrakakis C, Marinopoulos S, Tsigginou A, Dimopoulos and pregnancy. Recent Results Cancer Res. 2008;178:225–236.
MA. Cancer in pregnancy: Disentangling treatment modalities. ESMO 44. Barraclough J. Cancer and Emotion: A Practical Guide to Psycho-
Open. 2016;1:e000016. Oncology. Chichester: John Wiley & Sons; 1999.

You might also like