You are on page 1of 159

Clinical Lymphatic Mapping

in Gynecologic Cancers
Clinical Lymphatic Mapping
in Gynecologic Cancers
Second Edition

Edited by
Charles F. Levenback, MD
Professor (Retired), Department of Gynecologic Oncology
University of Texas MD Anderson Cancer Center, Houston, Texas

Ate G.J. van der Zee, MD, PhD


Professor of Gynecological Oncology and CEO
University Medical Center Groningen, The Netherlands

Robert L. Coleman, MD, FACOG, FACS


Chief Scientifc Offcer, US Oncology Research, The US Oncology
Network, McKesson Specialty Health, The Woodlands, Texas

Boca Raton London New York

CRC Press is an imprint of the


Taylor & Francis Group, an informa business
Second edition published 2022
by CRC Press
2 Park Square, Milton Park, Abingdon, Oxon, OX14 4RN
and by CRC Press
6000 Broken Sound Parkway NW, Suite 300, Boca Raton, FL 33487–2742
© 2022 Taylor & Francis Group, LLC
First edition published 2004
CRC Press is an imprint of Informa UK Limited
This book contains information obtained from authentic and highly regarded sources. While all
reasonable efforts have been made to publish reliable data and information, neither the author[s]
nor the publisher can accept any legal responsibility or liability for any errors or omissions that may
be made. The publishers wish to make clear that any views or opinions expressed in this book by
individual editors, authors or contributors are personal to them and do not necessarily refect the
views/opinions of the publishers. The information or guidance contained in this book is intended
for use by medical, scientifc or health-care professionals and is provided strictly as a supplement to
the medical or other professional’s own judgement, their knowledge of the patient’s medical history,
relevant manufacturer’s instructions and the appropriate best practice guidelines. Because of the
rapid advances in medical science, any information or advice on dosages, procedures or diagnoses
should be independently verifed. The reader is strongly urged to consult the relevant national drug
formulary and the drug companies’ and device or material manufacturers’ printed instructions, and
their websites, before administering or utilizing any of the drugs, devices or materials mentioned in
this book. This book does not indicate whether a particular treatment is appropriate or suitable for
a particular individual. Ultimately it is the sole responsibility of the medical professional to make
his or her own professional judgements, so as to advise and treat patients appropriately. The authors
and publishers have also attempted to trace the copyright holders of all material reproduced in this
publication and apologize to copyright holders if permission to publish in this form has not been
obtained. If any copyright material has not been acknowledged please write and let us know so we
may rectify in any future reprint.
All rights reserved. No part of this book may be reprinted or reproduced or utilised in any form
or by any electronic, mechanical, or other means, now known or hereafter invented, including
photocopying and recording, or in any information storage or retrieval system, without permission
in writing from the publishers.
For permission to photocopy or use material electronically from this work, access www.
copyright.com or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive,
Danvers, MA 01923, 978–750–8400. For works that are not available on CCC please contact
mpkbookspermissions@tandf.co.uk
Trademark notice: Product or corporate names may be trademarks or registered trademarks and are
used only for identifcation and explanation without intent to infringe.
British Library Cataloguing-in-Publication Data
A catalogue record for this book is available from the British Library
Library of Congress Cataloging-in-Publication Data
Names: Levenback, Charles, editor. | Zee, Ate Gerard Jan van der, editor. | Coleman, Robert L., editor.
Title: Clinical lymphatic mapping in gynecologic cancers / edited by Charles Levenback,
Ate G.J. van der Zee, Robert L. Coleman.
Description: Second edition. | Boca Raton : CRC Press, [2022] | Includes bibliographical references
and index.
Identifers: LCCN 2021037615 (print) | LCCN 2021037616 (ebook) | ISBN 9781032186436 (hardback) |
ISBN 9781032186443 (paperback) | ISBN 9781003255536 (ebook)
Subjects: MESH: Genital Neoplasms, Female—surgery | Breast Neoplasms—surgery | Lymph Node
Excision—methods | Sentinel Lymph Node Biopsy
Classifcation: LCC RC280.G5 (print) | LCC RC280.G5 (ebook) | NLM WP 145 | DDC 616.99/465—dc23
LC record available at https://lccn.loc.gov/2021037615
LC ebook record available at https://lccn.loc.gov/2021037616
ISBN: 978-1-032-18643-6 (hbk)
ISBN: 978-1-032-18644-3 (pbk)
ISBN: 978-1-003-25553-6 (ebk)
DOI: 10.1201/9781003255536
Typeset in Times
by Apex CoVantage, LLC
For Ginny
Charles F. Levenback

To my parents, Biny Van der Zee-Muntingh and Martin van der Zee, for their love
and long-lasting support. To my wife, Joukje van der Naalt, and my children,
Wierd, Heleen, and Hannah, for bringing so much joyful distraction into my life.
Ate G.J. van der Zee

To my parents, Marlene and Gary Beatty and Robert and Sharon Coleman, for their
unending encouragement, support, love, and guidance. To my wife, Fay, whose
love, insight, and daily selfess sacrifce strengthen and inspire me. To my children,
Jay, Christina, Kay, Joe, Mary, and Teresa, and grandchildren (so far . . . ), Noelle,
Jason, Nate, Marten, Lily, and Eli, who bring true color, joy, and happiness to
my life. To my patients, whose charity of self makes this endeavor possible.
Robert L. Coleman
Contents

List of contributors.......................................................................................................................................................................... ix
Preface ............................................................................................................................................................................................ xi

1 The history of lymphatic mapping: a gynecologic perspective.......................................................................................... 1


Charles F. Levenback

2 Lymphatic anatomy: microanatomy and physiology.......................................................................................................... 9


Erin K. Crane and Charles F. Levenback

3 Lymphatic anatomy: lymphatics of the vulva ................................................................................................................... 15


Anca Chelariu-Raicu and Robert L. Coleman

4 Lymphatic anatomy: lymphatics of the cervix .................................................................................................................. 23


Anca Chelariu-Raicu and Katherine C. Kurnit

5 Lymphatic anatomy: lymphatics of the uterus.................................................................................................................. 29


Jennifer J. Mueller and Nadeem R. Abu-Rustum

6 Lymphatic anatomy: lymphatics of the ovary ................................................................................................................... 35


Ate G.J. van der Zee

7 Lymphatic anatomy: lymphatics of the breast and axilla ................................................................................................ 39


James V. Fiorica

8 Modalities of detection of sentinel nodes in lymphatic mapping..................................................................................... 43


Blanca Segarra, Nuria Agusti, and Pedro T. Ramirez

9 Ultrastaging of the sentinel node ........................................................................................................................................ 55


Celien P.H. Vreuls, Ate G.J. van der Zee, and Paul J. van Diest

10 Sentinel lymph node biopsy of the vulva............................................................................................................................ 65


Maaike Oonk and Ate G.J. van der Zee

11 Sentinel lymph node mapping in cervical cancer ............................................................................................................. 71


David Cibula and Martina Borcinova

12 Sentinel lymph node mapping in breast cancer ................................................................................................................ 83


Angelena Crown and Mary L. Gemignani

13 Sentinel lymph node biopsy of the endometrium .............................................................................................................. 99


Jennifer J. Mueller and Nadeem R. Abu-Rustum

14 Impact of sentinel lymph node mapping on quality of life..............................................................................................111


Mariana Antonella Nigra, Pedro T. Ramirez, and Larissa Meyer

15 Radiation oncology considerations ....................................................................................................................................117


Gwendolyn Joyce McGinnis and Anuja Jhingran

vii
viii Contents

16 Special considerations ........................................................................................................................................................ 129


Michael Frumovitz

17 Clinical trial design ............................................................................................................................................................ 135


Scott Jordan and Brian M. Slomovitz

Index .............................................................................................................................................................................................143
Contributors

Nadeem R. Abu-Rustum, MD Anuja Jhingran, MD


Gynecology Service Radiation Oncology
Department of Surgery University of Texas MD Anderson Cancer Center
Memorial Sloan Kettering Cancer Center Houston, Texas
New York, New York
Scott Jordan, MD, FACOG
Nuria Agusti, MD Florida International University and Broward Health
IRTA Cabrils Fort Lauderdale, Florida
Barcelona, Spain
Katherine C. Kurnit, MD, MPH
Martina Borcinova, MD Gynecologic Oncology
Gynecologic Oncology Center NCH Medical Group
Department of Obstetrics and Gynecology Arlington Heights and University of Chicago Hospitals
First Faculty of Medicine Chicago, Illinois
Charles University and General University Hospital
Prague, Czech Republic Gwendolyn Joyce McGinnis, MD
Radiation Oncology
Anca Chelariu-Raicu, MD University of Texas MD Anderson Cancer Center
Department of Gynecologic Oncology & Reproductive Medicine Houston, Texas
University of Texas MD Anderson Cancer Center
Houston, Texas Larissa Meyer, MD
Department of Gynecologic Oncology & Reproductive Medicine
David Cibula, MD, PhD, FCMA University of Texas MD Anderson Cancer Center
Gynecologic Oncology Center Houston, Texas
Department of Obstetrics and Gynecology
First Faculty of Medicine Jennifer J. Mueller, MD
Charles University and General University Hospital Gynecology Service
Prague, Czech Republic Department of Surgery
Memorial Sloan Kettering Cancer Center
Erin K. Crane, MD, MPH
New York, New York
Department of Gynecologic Oncology
Levine Cancer Institute Mariana Antonella Nigra, MD
Charlotte, North Carolina Department of Gynecologic Oncology
Sanatorio Allende
Angelena Crown, MD
Cordoba, Argentina
Breast Service
Department of Surgery Maaike Oonk, MD, PhD
Memorial Sloan Kettering Cancer Center Department of Gynecologic Oncology,
New York, New York University Medical Center Groningen
James V. Fiorica, MD Groningen, The Netherlands
Sarasota Memorial Hospital
Pedro T. Ramirez, MD
Florida State University College of Medicine
Department of Gynecologic Oncology & Reproductive Medicine
Sarasota, Florida
University of Texas MD Anderson Cancer Center
Michael Frumovitz, MD, MPH Houston, Texas
Department of Gynecologic Oncology & Reproductive Medicine
University of Texas MD Anderson Cancer Center Blanca Segarra, MD
Houston, Texas Department of Gynecologic Oncology & Reproductive Medicine
University of Texas MD Anderson Cancer Center
Mary L. Gemignani, MD, MPH Houston, Texas
Breast Service
Department of Surgery Brian M. Slomovitz, MD, MS, FACOG
Memorial Sloan Kettering Cancer Center Florida International University and Broward Health
New York, New York Fort Lauderdale, Florida

ix
Preface

The editors spent a large part of their surgical training per- Our ultimate goals remain unchanged from 20 years ago, to
forming regional lymphadenectomy for women with vulvar, improve the outcomes for women who have trusted us with
endometrial, and cervical cancer. This huge diagnostic sur- their care during one of the greatest challenges of their lives.
gical effort appeared to be of no beneft to the survival of A large part of the story of the development of sentinel node
approximately 80% of women who had negative nodes, while biopsy is the importance of time-honored clinical skills like
simultaneously being associated with signifcant negative long- knowledge of surgical anatomy, careful surgical technique,
term side effects. When we learned of the pioneering work and patient safety to improve surgical outcomes. In addition
of Dr. Donald Morton regarding sentinel lymph node biopsy the editors have spent signifcant efforts during their careers
in patients with cutaneous melanoma, we independently real- to build global collaborative clinical study groups to enable
ized the technique could be easily adapted to vulvar and other the performance of carefully designed clinical trials on the
gynecologic cancer patients. That started for us a two-decade implementation of innovative surgical techniques, which is a
journey to apply sentinel node biopsy to our patient popula- challenge, especially in relatively rare cancers such as most
tion. On that journey we have made many friends around the gynecologic cancers.
world, trained numerous other physicians, and, most impor- Even as new targeted and immunotherapies become avail-
tantly, improved outcomes for women suffering from gyne- able, the aforementioned skills remain of critical importance
cologic cancer. We published the frst edition of this book to to patient outcomes. We hope that our work inspires new
create an easy-to-use resource for gynecologic oncologists investigators to continue the never-ending journey of continu-
incorporating sentinel lymph node biopsy into their practice. ous study and improvement of our core clinical knowledge
Incredibly, due to the hard work and collaboration of gyne- base and skills for the beneft of our patients and their families.
cologic oncologists worldwide, regional lymphadenectomy has
been largely eliminated from the routine care of women with Charles F. Levenback
vulvar, endometrial, and cervical cancer. As we near the end Ate G.J. van der Zee
of our careers, we decided to publish a second edition as an up- Robert L. Coleman
to-date reference for those practicing gynecologic oncology.

xi
1
The history of lymphatic mapping: a gynecologic perspective

Charles F. Levenback

CONTENTS
Introduction ...................................................................................................................................................................................... 1
Cadaver studies of lymphatic anatomy ............................................................................................................................................ 1
Halsted model for surgical management of solid tumors ................................................................................................................ 3
In vivo injection of dyes................................................................................................................................................................... 3
Early efforts at sentinel node identifcation ..................................................................................................................................... 4
Progress toward the modern lymphatic mapping concept ............................................................................................................... 6
Safety and surgical innovation ............................................................................................................................................... 7
References........................................................................................................................................................................................ 7

Since the frst edition of this book there has been an explo-
Introduction sion in knowledge about patient safety and the risks associated
with the introduction of new procedures and medical devices
In the new millennium, it is commonly believed that the into surgical practice. In this chapter, we will explore lessons
answers to cancer questions will result from innovations in learned from the introduction of sentinel lymph node biopsy
molecular biology that are brought from the bench to the bed- into gynecologic oncology surgical practice. Subsequent chap-
side through the work of translational researchers. However, ters will review the lymphatic anatomy of the female lower
the story of the development of lymphatic mapping serves as genital tract and breast and the current state of knowledge
a reminder to us that, even now, some advances in cancer care regarding lymphatic mapping for tumors at these sites.
still come from the more traditional routes of careful study of
anatomy, clinical observation, and literature research.
The history of the development of lymphatic mapping is also
a humbling lesson in how important innovations in medicine can
Cadaver studies of lymphatic anatomy
diffuse slowly. The term ‘sentinel node’ was frst used in 1960,1
yet even today the full potential of this concept has not been real- Early anatomists learned about lymphatic anatomy from
ized. Initial reports of success with the sentinel node concept cadavers. This work was complicated by the fact that lym-
were largely ignored or misinterpreted. Old ideas can be diffcult phatic channels are diffcult to see and very fragile and by
to abandon, and new ideas are not always what they appear. the fact that tissue had to be extensively putrefed or fxed to
The relative infrequency of gynecologic tumors is an addi- be studied. Painstaking dissections led to the production of
tional impediment to the development of lymphatic mapping remarkable drawings of lymphatic anatomy. These illustra-
in our specialty. Fortunately, we can learn a lot from the expe- tions were frequently reproduced by subsequent investigators
rience of others in treating more common diseases. I saw the with small improvements (Figures 1.1 and 1.2). Anatomists
lymphatic mapping procedure for the frst time when a patient developed a variety of aids to help visualize the lymphatic
presented to MD Anderson Cancer Center in 1992 with syn- channels, including a number of mercurial compounds that,
chronous primary tumors: a 2-cm squamous cell cancer of the when injected into various soft tissues, canalized the lym-
cervix and a thin cutaneous melanoma of the anterior thigh. phatics. This technique most likely contributed to the depic-
Standard treatment at the time would have required inguinal, tions of lymphatic anatomy that erroneously show vulvar
femoral, and pelvic lymphadenectomy that would have surely lymphatics crossing the labial-crural fold, best represented
left the patient with signifcant lymphedema. After extensive by Sappey’s illustration (Figure 1.3). In the early twentieth
consultation, the surgical oncologist performed lymphatic century, the French gynecologists Leveuf and Godard2 stud-
mapping and sentinel node biopsy. I was amazed to see lym- ied the lymphatic anatomy of the cervix using injection of
phatic channels and the blue sentinel lymph node. Up to this Gerotti blue into the cervices of neonatal cadavers. They
time any type of preoperative or intraoperative direct imag- found that the dye drained in a highly reproducible way to a
ing of lymphatic channels and lymph nodes was not possible. lymph node usually found in the obturator space or close to
When I saw the blue node in the groin, it was clear that the the iliac vessels. They named this the ‘principal’ lymph node
same technique could be applied to carcinoma of the vulva. (Figure 1.4).

DOI: 10.1201/9781003255536-1 1
2 Clinical Lymphatic Mapping

FIGURE 1.3 Vulvar lymphatics depicted by P.C. Sappey (1879). Note


that lymphatics cross the labial-crural fold and buttocks. (Reproduced
from Parry-Jones19 with permission.)

FIGURE 1.1 Lymphatics of the abdomen and pelvis (1787) by P.


Mascagni. (Reproduced from Plentl and Friedman4 with permission from
Elsevier.)

FIGURE 1.2 Lymphatics of the abdomen and pelvis. (From a nine- FIGURE 1.4 Leveuf and Godard’s2 description (1923) of lymphatic
teenth-century French textbook.) drainage of the cervix. (Courtesy of Dr. Daniel Dargent.)
The history of lymphatic mapping 3

Halsted model for surgical


management of solid tumors
The great anatomist of the second century, Galen, believed that
cancer was a local process resulting from a systemic disease
of bodily fuids and humors. In the West, this concept was not
challenged until the seventeenth century, when Valsalva pro-
posed that cancer spread from primary tumors to lymph nodes.3
This understanding of cancer and the metastatic process ulti-
mately led to the Halstedian model for surgical management
of solid tumors. In this model, the primary tumor, the primary
regional lymph nodes, and the second-echelon regional nodes,
as well as all of the intervening lymphatic channels – including
the fne cutaneous lymphatics – were resected en bloc. This
approach was integrated into the treatment of gynecologic can-
cers by several gynecologic surgeons working in multiple sites
on the European continent. Rupprecht, Basset, Stoeckel, and
Bucura are credited with simultaneous descriptions in 1912
of what we now refer to as radical vulvectomy and bilateral
inguinal femoral lymphadenectomy.4 The next generation of
gynecologic cancer surgeons, including Twombly,5 Taussig,6
and Way,7 championed radical vulvectomy and bilateral ingui-
nal femoral lymphadenectomy, with only minor revisions for
patients with vulvar cancer, because of the dramatic improve-
ment in survival compared with less radical or nonsurgical
treatments. Stanley Way summarized the surgical philosophy
of the day in 1951 when he stated, “The most important steps
in the operation are undoubtedly the very wide removal of the
vulva and the resection of the lymph nodes, certainly as far as
the node of Cloquet”8 (Figure 1.5). Way added that the morbid-
ity and mortality of the procedure were justifed and that the
operation “represents the fnal achievement of surgery in this
disease.” Within a generation, however, the radicality of surgi-
cal treatment of the vulva would be diminishing, and today,
no indications remain for radical vulvectomy and bilateral
inguinal femoral lymphadenectomy for the primary treatment
of vulvar cancer.
Although Way was incorrect in his assessment, he did make
a prophetic plea for the establishment of special treatment cen-
ters for patients with vulvar cancer because of its rarity, the
diffculty of radical surgery for vulvar cancer, and the need
for experienced medical and nursing care. Way foreshadowed
both the formation of the subspecialty of gynecologic oncol-
ogy and the establishment of comprehensive cancer centers.

In vivo injection of dyes


The next advances in the study of lymphatic anatomy and the
development of lymphatic mapping came from in vivo studies.
Intraoperative observation of the lymphatics has always been
hampered by two facts: The lymphatics are essentially invis-
ible, and the lymph is colorless. Two of the frst investigators to
use living subjects for study were Stephen Hudack and Phillip
McMaster.9 They experimented with injecting a variety of vital FIGURE 1.5 Depiction of inguinal nodes (a) and pelvic nodes (b) by
dyes into the skin of a variety of mice, rats, and rabbits. They Stanley Way8 in 1951. The relationship of the nodal groups, including
then used themselves and six other volunteers as experimen- direct clitoral drainage to ‘deep femoral’ nodes and contralateral labial
tal subjects to study the cutaneous lymphatics of the skin and drainage, envisaged by Way in schematic form (c – redrawn for clarity).
4 Clinical Lymphatic Mapping

lymphatic drainage patterns. Using very fne needles, small


quantities of vital blue dyes, and photographic magnifcation,
they were able to demonstrate the fne lymphatic capillaries of
the skin. They varied the type of dye and the volume injected
and measured the rate of lymph transport (up to 15 cm in 5
min) and the impact of infamed skin, heat, and wheal forma-
tion on lymph transport. In their examination of the effect of
wheal formation, they studied one individual who responded
to “a frm stroke with a blunt instrument,” with formation of
a wheal within 2–3 min. The lymphatics in the region of the
wheal became less effective at draining the dye.
Clinicians began to experiment with injection of a variety of
dyes to help visualize lymph nodes. Gray10 injected sky-blue
dye into freshly resected skin to study cutaneous lymphatics.
Zeit and Wilcoxon11 injected India ink into the cervices of
patients before surgery to assist in the visualization of pelvic
lymph nodes. The authors observed that this technique aided
in teaching a relatively new procedure, radical hysterectomy
and pelvic lymphadenectomy. Injections of 0.5 mL of India ink
into the cervix at 3 and 9 o’clock at least 8 h prior to surgery
stained all of the pelvic nodes black. Braithwaite12 injected
indigo carmine into the cecum in an attempt to link appen-
dicitis and duodenal ulcers. Weinberg and Greaney13 injected
sky-blue dye into the peritoneum during radical gastrectomy
to assist the pathologists in identifcation of draining lymph
nodes in the specimen.
Eduard Eichner systematically studied the anatomy of the
female lower genital tract using intraoperative injections of blue
dyes. In a remarkable 2-year period, he published three papers
that described anatomic fndings after injections in the cervix,
ovary, uterine fundus, vulva, and vagina14–16 (Figure 1.6). He
supplemented his work with a fourth study published in 1958,
of patients with cervical stumps.17 Eichner experimented with
obstructing lymphatic fow at various points – for example, he
tied off the infundibulopelvic ligament to see how this would
alter lymphatic fow. Eichner made several important observa-
tions; however, he also made errors that contributed to mis-
conceptions about lymphatic drainage of the genital tract, in
particular the vulva. Eichner engaged in a series of injections
of various vulvar sites. Unlike Hudack and McMaster, who
used intradermal injections, Eichner used deep subcutaneous FIGURE 1.6 (a) Lymphatic drainage of the cervix as depicted by
injections. Eichner was unable to demonstrate dye in the ingui- Eduard Eichner17 in 1958 (reproduced with permission); (b) key aspects
nal nodes of his subjects; rather, he found dye in the pelvic of depiction.
nodes. Presumably, the dye was following the prevascular lym-
phatics of the deep vessels of the vulva directly to the pelvis
rather than following the superfcial cutaneous lymphatics that negative, the remaining lymph nodes of the neck would also be
drain the groin. This work suggested direct lymphatic drain- negative and full neck dissection could be avoided.
age from the vulva to the pelvis. It took several decades to The individual who combined the two elements of the mod-
demonstrate that this very rarely, if ever, occurs. ern approach, lymphatic mapping and sentinel node iden-
tifcation, is Ramon Cabanas. Cabanas, along with Manuel
Riveros and Ramiro Garcia, frst described lymphography
of the penis in a 1967 report of an anatomic study performed
Early efforts at sentinel node identification
while Cabanas was at the National University in Asuncion,
The individual most frequently credited with coining the Paraguay.18 Paraguay has one of the highest incidences of
phrase ‘sentinel node’ is Ernest Gould.1 He proposed using the penile carcinoma in the world. Cabanas later did a fellowship
lymph node found at the junction of the anterior and posterior in urologic oncology at Memorial Sloan-Kettering Cancer
facial veins for frozen-section analysis to determine the need Center. Over an 8-year period, Cabanas amassed a series of
for a full neck dissection. His hypothesis, based on fndings 80 patients with penile cancer in whom he used lymphography
in 28 patients with parotid cancer, was that if this node was to identify the sentinel lymph node in the groin (Figure 1.7).
The history of lymphatic mapping 5

FIGURE 1.7 (Continued)

He found that the sentinel lymph node was always located


among the superfcial inguinal nodes, that the sentinel node
was involved with disease in all patients who had metastases,
and that the sentinel node was the only positive node in 12
cases. Cabanas suggested that only patients with a positive
sentinel lymph node required the more extensive (and morbid)
procedure of complete lymphadenectomy.
In the 1970s, Cabanas was conducting his studies of sentinel
nodes in patients with penile cancer, while gynecologic oncol-
ogists were attempting to fnd ways to reduce the morbidity of
radical vulvectomy and bilateral inguinal femoral lymphad-
enectomy. Parry-Jones19 performed in vivo injections of blue
dye to show that vulvar lymphatics do not cross the buttock
and labial-crural fold, as had been depicted by Sappey. This
observation permitted surgeons to reduce the radicality of the
operation while still adhering to Halstedian principles. In con-
trast to Eichner, Parry-Jones used intradermal injections and
found in all cases that the dye was arrested in the groin and did
not reach the pelvic nodes. This conficted with his hypothesis
that there is direct lymphatic drainage from the vulva to the
pelvis. Rather than reject his hypothesis, Parry-Jones rejected
FIGURE 1.7 (a and b) Cabanas’s technique for penile lymphography. his methods. After experimenting with several compounds, he
Blue dye injected into the phallus of the penis is taken up by cutaneous
settled on Imferon injected into the subcutaneous tissue of the
lymphatics. A cut-down is performed, and the dorsal lymphatic is cana-
lized and injected with ethiodized oil. (c) Plain radiographs identify both vulva 24 h prior to surgery. Iron particles found in the iliac
the lymphatic channel and ethiodized oil deposited in the sentinel node. lymph nodes confrmed for Parry-Jones his hypothesis that the
(Reproduced from Cancer with permission18.) vulva has lymphatic drainage directly to the pelvic nodes.19
6 Clinical Lymphatic Mapping

Other investigators took other avenues to reduce the mor- Donald Morton developed the frst important clinical applica-
bidity of radical vulvectomy and bilateral inguinal femoral tion of lymphoscintigraphy by using colloidal gold to identify
lymphadenectomy. Wharton et al.20 suggested that the risk of the primary nodal drainage basin in patients with cutaneous
lymph node metastases in patients with less than 5-mm inva- melanoma at sites on the body with ambiguous lymphatic
sion was so low that lymphadenectomy could be omitted in drainage patterns, primarily the trunk (Figure 1.8).
these patients. This approach was rapidly abandoned, how- At the time of Morton’s studies, there was a major debate
ever, following the subsequent publication of several reports of regarding the management of patients with early-stage mela-
lymph node metastases in patients with less than 5-mm inva- noma. The vast majority of these patients, 90%, have nega-
sion.21 Morris22 suggested that patients with well-lateralized tive nodes, and the effcacy of regional lymphadenectomy in
lesions could be treated with hemivulvectomy. This work the 10% with positive nodes was the subject of intense debate.
represented one of the earliest departures from the practice of Some clinicians recommended a complete lymphadenectomy
performing radical vulvectomy for the treatment of invasive in all patients with stage I disease, while others recommended
vulvar cancer and was a clear departure from the Halstedian observation. Morton set out to determine whether the senti-
concept of en bloc resection. nel node as described by Cabanas and others could be iden-
Philip DiSaia focused the attention of the gynecologic oncol- tifed intraoperatively. His group studied various dyes in a
ogy community on the devastating effect of radical vulvectomy feline model29 and selected isosulfan blue as the best choice
on body image and sexual function. In 1979, he described com- for intraoperative lymphatic mapping. In 1992, Morton et al.30
bining less radical resection of the primary tumor with a less described their results in 223 patients with cutaneous mela-
radical approach to groin dissection, as suggested by Cabanas. noma. A total of 237 lymphatic basins were mapped, and the
DiSaia proposed radical local excision and superfcial inguinal sentinel node was identifed in 82% of them. More than 3000
lymphadenectomy for selected patients with vulvar cancer.23 lymph nodes were removed, of which 8% were sentinel, and in
DiSaia introduced the term ‘sentinel nodes’ to the gynecologic only two cases did nonsentinel nodes contain metastatic dis-
oncology community in North America. He suggested that the ease when the sentinel nodes were disease free. This paper is
superfcial inguinal lymph nodes were the sentinel nodes of one of the most cited references in modern surgical oncology
the vulva. This is the location where Cabanas always found the and prompted an explosion of interest in this technique.
sentinel nodes in patients with penile cancer, and DiSaia added
his own observation that the femoral nodes were never positive
if the superfcial inguinal nodes were negative. DiSaia relied
on location relative to anatomic landmarks to identify ‘senti-
nel nodes’ – a technique similar to that described by Gould in
1960 – rather than relying on landmarks in combination with
lymphatic mapping, as described by Cabanas. DiSaia funda-
mentally misrepresented the sentinel node concept; however, it
was embraced by gynecologic oncologists interested in reduc-
ing the morbidity of groin dissection. Berman et al.24 reported
no groin relapses in a group of 50 cervical cancer patients; how-
ever, these outstanding results could not be reproduced. In the
Gynecologic Oncology Group (GOG) protocol 75, reported by
Stehman et al,25 almost 8% of the 121 patients with vulvar can-
cer suffered a groin recurrence following superfcial inguinal
lymphadenectomy with negative nodes. This compares with
a recurrence rate of less than 1% following inguinal femoral
lymphadenectomy with negative nodes in a group of more than
300 patients participating in GOG protocol 37.26 On the basis
of the GOG results, gynecologic oncologists have abandoned
superfcial inguinal lymphadenectomy as it was described by
DiSaia in 1978.

Progress toward the modern


lymphatic mapping concept
Injection of radionuclides into human subjects to localize
regional lymph nodes was frst reported in 1953 by Sherman
and Ter-Pogossian.27 Numerous radiolabeled compounds FIGURE 1.8 Lymphoscintogram of a patient with a cutaneous mela-
noma of the midback. Note lymphatic drainage to three of four potential
have since been used for this purpose. The frst gynecologic lymphatic basins. (From Nieweg O, Jansen L, Uren RF, et al, Instructive.)
application of lymphoscintigraphy was in 1982, when Iversen Cases: Case 4: Drainage to Multiple Basins. In Nieweg O, Essner R,
and Aas28 studied vulvar lymphatic drainage in a group of 52 Reintgen D, eds, Lymphatic Mapping and Probe Applications in Oncology.
patients, most of whom had cervix cancer. A group led by (Marcel Dekker, Inc.: New York, 2000) 292–4, with permission.)
The history of lymphatic mapping 7

In rapid succession, several large series of lymphatic map- are ultrasafe and still provide high satisfaction to consum-
ping and sentinel node biopsy in patients with breast cancer ers. High-reliability organizations (HROs) operate in high-
and melanoma were published from centers around the world, risk environments by being highly sensitive to operations,
essentially all confrming Morton et al.’s observations. A deferring to expertise, and not trying to simplify complex
decade later, reports have been published describing lymphatic operations, in addition to a preoccupation with failure and
mapping and sentinel node identifcation in patients with head a commitment to resilience. The lessons of HROs are being
and neck, endocrine, gastrointestinal, genitourinary, and applied in health care, especially in surgery and anesthesiol-
reproductive cancers. ogy. For example, the World Health Organization Surgical
The impact of Morton’s work cannot be overstated. Between Safety Checklist has transformed how surgical procedures are
Cabanas’s landmark 1978 study and Morton’s in 1992 there conducted. Risk-adjusted data, such as the National Surgical
were 16 citations in PubMed with ‘sentinel node’ in the title Quality Improvement Database (NSQIP), allow accurate mea-
or abstract. Vulvar cancer was the frst disease site other than surement of new protocols to reduce surgical site infections.
penile, melanoma, and breast to be cited associated with ‘sen- Minimally invasive surgery and enhanced recovery protocols
tinel node.’31 The frst reference to sentinel node biopsy in have reduced the surgical length of stay dramatically. Low-
endometrial cancer was in 1996.32 From 1995 to 2020 there risk anesthesia for outpatient procedures is ultrasafe.
have been 365 citations with sentinel and vulvar in the title, as Organizations are also looking more carefully at how
well as 554 with sentinel and endometrial in the title. The cor- surgical innovations are introduced to the operating room.
responding numbers for breast cancer and melanoma are 7515 Marcus et al.39 noted that new devices and procedures are
and 3957. For all these disease sites the number of citations is usually associated with a spike in complications that are hid-
still increasing.33 den from patients and the public. The introduction of sentinel
Dr. Morton was one of the founders of the International node biopsy in vulvar cancer is notable because most patients
Sentinel Node Society in 1998. Numerous validation trials in with a negative sentinel node do not get adjuvant therapy, and
melanoma were conducted under his leadership. The sixth edi- groin relapses have a very high mortality rate. This differed
tion of the AJCC Cancer Staging Manual for breast cancer from breast cancer, where many node-negative patients still
included substages for micro-metastases in regional lymph received some type of adjuvant therapy. For example, in GOG
nodes, a change forced by the adoption of sentinel lymph node 173 there were 13 false-negative cases. These occurred in ten
biopsy.34 By 2001 there were over 17,000 analytic melanoma member institutions, and seven of the cases were accession
cases that resulted in the incorporation of sentinel lymph node cases 1, 2, or 3. Fortunately these patients also had an ingui-
biopsy into the American Joint Committee on Cancer (AJCC) nal femoral lymphadenectomy. However, in situations where
staging of melanoma.35 The current staging manuals and treat- there is a false-negative sentinel node and no regional lymph-
ment guidelines for breast cancer and melanoma depend com- adenectomy or adjuvant therapy, morbidity and mortality go
pletely on the status of sentinel lymph nodes. up sharply.
Staging and consensus guidelines for the management of Finally, Chassin and Loeb, writing on behalf of The Joint
gynecologic cancers increasingly incorporate sentinel lymph Commission, noted, “high reliability organizations do not tolerate
node biopsy. The NCCN Vulvar Squamous Cell Carcinoma the existence of behaviours that suppress the reporting of safety
Guidelines Version 2.202136 indicate that inguinal femoral concerns and perpetuate the existence of unsafe conditions.”40
lymphadenectomy should only be performed when a senti- The aviation industry has been especially adept at the use of sim-
nel lymph node cannot be identifed. The NCCN Guidelines ulators, checklists, and anonymous error reporting to reduce pre-
Version 1.2021 Endometrial Carcinoma Principles for ventable commercial airline deaths. The safety standards cannot
Evaluation and Surgical Staging state that “sentinel lymph be circumvented based on seniority or other privilege.
node mapping may be considered” as a staging procedure.37 The future of sentinel lymph node biopsy is bright. All the
Similarly, Cerc-C38 (NCCN guidelines version 1.2021 tools of quality and safety should be used to avoid some of the
Cervical Cancer) recommends “bilateral pelvic lymphadenec- pitfalls of the past, as well as those yet to be discovered.41
tomy (with or without sentinel lymph node mapping)” as the
surgical management of appropriate patients.
The outcome of all of this work is an enormous shift in surgi- REFERENCES
cal management of solid tumors. Regional lymphadenectomy
1. Gould EA, Winship T, Philbin PH, et al. Observations
has been eliminated from most aspects of surgical oncology,
on a ‘sentinel node’ in cancer of the parotid. Cancer.
including gynecologic oncology. The morbidity associated 1960;13:77–8.
with regional lymphadenectomy, including wound infection, 2. Leveuf J, Godard H. Les lymphatiques de l’utérus. Rev
bleeding, and lymphedema, have decreased. Long-term out- Chir. 1923;219–48.
comes, including survival, are increasing with more accurate 3. Borgstein P, Meijer S. Historical perspective of lymphatic
staging and improved treatment options. tumour spread and the emergence of the sentinel node con-
cept. Eur J Surg Oncol. 1998;24:85–9.
Safety and surgical innovation 4. Plentl AA, Friedman EA. Lymphatics of the cervix uteri.
In: Lymphatic System of the Female Genitalia. Philadelphia
During the same period of the development of sentinel WB Saunders; 1971. pp. 75–115.
lymph node biopsy, health care leaders have observed indus- 5. Twombly G. The technique of radical vulvectomy for carci-
tries such as commercial aviation and nuclear energy that noma of the vulva. Cancer. 1953;3:516–30.
8 Clinical Lymphatic Mapping

6. Taussig FJ. Cancer of the vulva: an analysis of 155 cases. inguinal lymphadenectomy and modifed radical hemivul-
Am J Obstet Gynecol. 1940;40:764–73. vectomy: a prospective study of the Gynecologic Oncology
7. Way S. Carcinoma of the vulva. Am J Obstet Gynecol. Group. Obstet Gynecol. 1992;79:490–7.
1960;79:692. 26. Homesley HD, Bundy BN, Sedlis A, et al. Radiation therapy
8. Way S. Carcinoma of the vulva. In: Malignant Disease of the versus pelvic node resection for carcinoma of the vulva with
Female Genital Tract. Philadelphia: Blakiston; 1951. pp. 27–8. positive groin nodes. Obstet Gynecol. 1986;68:733–40.
9. Hudack S, McMaster P. The lymphatic participation in 27. Sherman A, Ter-Pogossian M. Lymph-node concentration
human cutaneous phenomena. J Exp Med. 1933;57:751–74. of radioactive colloidal gold following interstitial injection.
10. Gray J. The relation of lymphatic vessels to the spread of Cancer. 1953;6:1238–40.
cancer. Br J Surg. 1938;6:462–95. 28. Iversen T, Aas M. Lymph drainage from the vulva. Gynecol
11. Zeit PR, Wilcoxon G. In vivo coloring of pelvic lymph nodes Oncol. 1983;16:179–89.
with India ink. Am J Obstet Gynecol. 1950;59:1164–6. 29. Wong JH, Cagle LA, Morton DL. Lymphatic drainage of
12. Braithwaite L. Flow of lymph from iliocecal angle. Br J skin in a sentinel lymph node in a feline model. Ann Surg.
Surg. 1923;11:7. 1991;214:637–41.
13. Weinberg JA, Greaney EM. Identifcation of regional lymph 30. Morton DL, Wen DR, Wong JH, et al. Technical details
nodes by means of a vital staining during surgery of gastric of intraoperative lymphatic mapping for early stage mela-
cancer. Surg Gyn Obst. 1950;90:561–5. noma. Arch Surg. 1992;127:392–9.
14. Eichner E, Bove ER. In vivo studies on the lymphatic drain- 31. Levenback C, Burke TW, Gershenson DM, et al.
age of the human ovary. Obstet Gynecol. 1954;3:287–97. Intraoperative lymphatic mapping for vulvar cancer. Obstet
15. Eichner E, Goldberg I, Bove ER. In vivo studies with direct Gynecol. 1994;84(2):163–7.
sky blue of the lymphatic drainage of the internal genitals 32. Burke TW, Levenback C, Tornos C, et al. Intraabdominal
of women. Am J Obstet Gynecol. 1954;67:1277–86. lymphatic mapping to direct selective pelvic and para-
16. Eichner E, Mallin LP, Angell ML. Further experience with aortic lymphadenectomy in women with high-risk endo-
direct sky blue in the in vivo studies of gynecologic lym- metrial cancer: results of a pilot study. Gynecol Oncol.
phatics. Am J Obstet Gynecol. 1955;69:1019–26. 1996;62(2):169–73.
17. Eichner E, Rubinstein L. Cervical stump lymphatics. Obstet 33. Balch CM. Detection of melanoma metastases with the
Gynecol. 1958;12:521–7. sentinel node biopsy: the legacy of Donald L. Morton, MD
18. Riveros M, Garcia R, Cabanas R. Lymphadenectomy of the (1934–2014). Clin Exp Metastasis. 2018;35(5–6):425–9.
dorsal lymphatics of the penis. Cancer. 1967;20:2026–31. 34. Singletary SE, Allred C, Ashley P, et al. Staging sys-
19. Parry-Jones E. Lymphatics of the vulva. J Obstet Gynaecol tem for breast cancer: revisions for the 6th edition of the
Br Commonwealth. 1963;70:751–65. AJCC Cancer Staging Manual. Surg Clin North Am.
20. Wharton J, Fletcher G, Delclos L. Invasive tumors of the 2003;83(4):803–19.
vagina: clinical features and management. In: Coppleson 35. Balch CM, Soong SJ, Gershenwald JE, et al. Prognostic fac-
M, editor, Gynecologic Oncology: Fundamental Principles tors analysis of 17,600 melanoma patients: validation of the
and Clinical Practice. New York: Churchill-Livingstone; American Joint Committee on Cancer melanoma staging
1981. pp. 345–59. system. J Clin Oncol. 2001;19(16):3622–34.
21. Parker RT, Duncan I, Rampone J, et al. Operative manage- 36. NCCN Vulvar Squamous Cell Carcinoma Guidelines
ment of early invasive epidermoid carcinoma of the vulva. Version 2.2021. Available from: www.nccn.org.
Am J Obstet Gynecol. 1975;123:349–55. 37. NCCN Guidelines Version 1.2021, endo-C, p. 1. Available
22. Morris JM. A formula for selective lymphadenectomy: its appli- from: www.nccn.org.
cation to cancer of the vulva. Obstet Gynecol. 1977;50:152–8. 38. NCCN guidelines version 1.2021 Cervical Cancer.
23. DiSaia PJ, Creasman WT, Rich WM. An alternate approach Available from: www.nccn.org.
to early cancer of the vulva. Am J Obstet & Gynecol. 39. Marcus RK, Lillemoe HA, Caudle AS. Facilitation of sur-
1979;133:825–32. gical innovation: is it possible to speed the introduction of
24. Berman ML, Keys H, Creasman W, et al. Survival and pat- new technology while simultaneously improving patient
terns of recurrence in cervical cancer metastatic to periaor- safety? Ann Surg. 2019;270(6):937–41.
tic lymph nodes (a Gynecologic Oncology Group study). 40. Chassin MR, Loeb JM. High-reliability health care: getting
Gynecol Oncol. 1984;19:8–16. there from here. Milbank Q. 2013;91(3):459–90.
25. Stehman FB, Bundy BN, Dvoretsky PM, et al. Early stage 41. Levenback CF. Editorial, International Journal of
I carcinoma of the vulva treated with ipsilateral superfcial Gynecologic Oncology, in Press.
2
Lymphatic anatomy: microanatomy and physiology

Erin K. Crane and Charles F. Levenback

CONTENTS
Introduction ...................................................................................................................................................................................... 9
Early studies of the lymphatic system ............................................................................................................................................. 9
Lymph and lymphatic vessels ........................................................................................................................................................ 10
Physiology...................................................................................................................................................................................... 10
Lymph nodes .................................................................................................................................................................................. 10
Immunology of regional lymph nodes........................................................................................................................................... 11
Surgical anatomy of the lymphatic system .................................................................................................................................... 12
Summary ........................................................................................................................................................................................ 13
References...................................................................................................................................................................................... 13

demonstrated hydrostatic and oncotic pressures within the


Introduction lymphatic system.5
At the start of the twentieth century, the clinical use of sur-
Formed by an intricate network of lymphatic channels and gery became more popular, and intraoperative observations
lymph nodes, the human lymphatic system performs many cru- stimulated interest in lymphatic function. Braithwaite observed
cial functions, including maintenance of interstitial and intra- that pigmented lymph nodes from an infected appendix could
vascular equilibrium, fltration of immunogens and particulate be followed along the superior mesenteric artery to the duo-
matter, and transport of lipids and immune cells. Lymphatics denum and coined the term ‘glands sentinel’ after studying
are paramount in facilitating the immune response to patho- omental lymph node drainage from an intestine.6 In 1938,
gens and malignancy and directing interactions between Gray studied freshly resected skin using thorium oxide injec-
immune cells and antigens, ultimately determining whether to tions (Thorotrast), which permitted a detailed study of skin
engage in an immune response or tolerance. The purpose of lymphatics.7 He further defned histologic criteria for identi-
this chapter is to provide an overview of the lymphatic system fying lymphatics, including 1) endothelial lining, 2) frequent
with an emphasis on how it relates to lymphatic mapping. semilunar valves, 3) connection of lymph vessels to lymph
glands, and 4) the presence of coagulated lymph or nothing in
the lymph channels.7 In the 1950s, Kinmoth injected contrast
Early studies of the lymphatic system into lymphatics, giving rise to the lymphoscintigram.8
Through injection of thorium oxide into freshly resected
Lymphatic vessels were frst detailed in ancient Greek writ- breast tumors, Gray concluded that “the mode of spread of
ings of Hippocrates and Aristotle in the fourth century BC as cancer to lymph glands is generally by means of lymphatic
“fbers which take a position between blood vessels and nerves emboli of cancer cells carried along the lymph stream.”7 The
and which contain a colorless liquid.”1 As cadaveric dissec- infuential William Halsted supported this concept in his
tions became more accepted, knowledge of the human lym- work on axillary lymph node dissections for breast cancer,
phatic system expanded. In the mid-1600s, Bartholin coined suggesting that malignancy spreads in an organized fashion
the term ‘lymphatics,’ also recognizing lymphatic dysfunction from lymph nodes to viscera.7,9 This view was debated by
in ascites and edema.2 In the early 1700s, the French surgeon many, particularly Tyzzer, who published the discovery of
Petit described lymphatic spread of breast cancer to axil- metastatic disease in patients in the absence of regional lymph
lary lymph nodes.3 Virchow similarly described the involve- node metastases.10 While our modern understanding of malig-
ment of lymphatics in cancer, suggesting a role in fltration.4 nancies incorporates concepts of both lymphatic and hema-
Despite advances made by cadaveric dissection, lymphatic togenous dissemination, the basis of these historic works has
study in vivo contributed the most knowledge to understand- established lymph node assessment as an essential procedure
ing the details of the lymphatic system. In the 1890s, Starling in surgical oncology.

DOI: 10.1201/9781003255536-2 9
10 Clinical Lymphatic Mapping

Lymph and lymphatic vessels Physiology


Lymphatics permeate the human body, located in nearly every The body circulates 8–12 L of lymphatic fuid daily,13 much
organ except bone marrow, cartilage, and the cornea. It was of which occurs passively and depends on several factors,
thought that the central nervous system lacked lymphatics, including hydrostatic pressure, osmotic pressure, diffusion and
until recently in 2015, when Louveau et al. demonstrated lym- convection, and chemotaxis. Terminal lymphatics remain col-
phatic vessels lining the dura within the brain.11 lapsed under normal conditions until an increase in interstitial
The lymphatic system begins with small lymphatic capil- pressure causes the anchoring flaments to pull the endothe-
laries (terminal lymphatics), which function as sites of fuid lial cells apart, allowing an infux of lymph into the terminal
exchange. Terminal lymphatics have blind ends and are lined capillary. The larger channels containing smooth muscle then
by a single layer of overlapping endothelial cells, with fenes- contract and propel lymph fuid and particulate matter forward
trations ranging in size from 10 to 25 μm12 (Figure 2.1). These through the lymph system, until it returns into the circula-
cells are anchored by collagen flaments to the surrounding tion. The lymphatic system lacks a central pump and relies on
connective tissue. A combination of diapedesis, hydrostatic valves to prevent backfow of lymph. Lymphatic vessels have
forces, and colloidal gradients guides particles and fuid into very low pressure and are sensitive to small changes, requiring
the gaps between overlapping endothelial cells. It has been only 2–4 cm of H2O to initiate rhythmic contractions.18 Gray
hypothesized that the contraction and relaxation of the fla- massaged tissue after injection to accelerate lymphatic fow.
ments facilitate permeation through the endothelial cells, form- Many clinicians still perform external massage during lym-
ing microvalves between the lymphatics and interstitium.13 phatic mapping to enhance dispersal of dye.
Afferent (collecting) lymph channels are connected to the In general, lymph fuid is clear and colorless, resembling
terminal lymphatics. Unlike terminal lymphatics, collecting diluted plasma. The volume, rate of fow, and composition of
lymphatic vessels lack flamentous attachments and possess lymph vary with site and activity. The rate of lymphatic fow is
smooth muscle fbers and valves. The thicker afferent chan- of particular clinical interest in sentinel node mapping and has
nels coalesce to form larger lymphatics that ultimately drain been characterized in various studies by employing lymphos-
to regional lymph nodes. Larger afferent channels contain an cintigraphy. In lymphoscintigraphy, a tagged colloid is imaged
inner layer of circular smooth muscle and may be located in as it transits through lymphatics. Lymphatic fow varies based
close proximity to blood vessels or contain vasa vasorum for on tissue type; for example, lymph travels slower in skin as
nutritional support13 (Figure 2.1). Secondary valves prevent compared to skeletal muscle and is cleared much faster when
lymphatic backfow and delineate ‘lymphangions,’ or individ- injected intradermally versus subcutaneously.19 In conditions
ual units with contractile properties.14 such as edema or lymphedema, lymphatic fow slows, whereas
More recently, various lymphatic endothelial markers, during exercise or infammation fow rate increases. Anatomic
including PROX1 (prospero homeobox protein), LYVE1 (lym- location affects fow rate, with the lowest rate observed in the
phatic vessel endothelial hyaluronan receptor 1), and VEGFR3 head and neck and the highest rate in the lower extremity.20
(vascular endothelial growth factor receptor-3), have aided in From a gynecologic perspective, the primary tumor in
the histologic detection of lymphatic channels and advanced patients with vulvar, cervical, or uterine cancers is relatively
our understanding of lymphangiogenesis and lymphatic close to the regional lymph node bed, and therefore both dye
anatomy.15–17 and radionuclide are transferred quickly to the sentinel node
or nodes. In endometrial cancer, for example, time from injec-
tion of indocyanine green in the cervix to pelvic lymph node
visualization approximates 5 minutes and lasts up to 1 hour.21

Lymph nodes
Spread throughout the body are small, bean-shaped lymph
nodes (Figure 2.2). Lymph nodes are highly complex organs
which function as the primary antigen recognition site in the
body and act as flters for particulate matter, antigens, and can-
cer cells.
Afferent lymphatics transport lymphatic fuid into lymph
nodes, which enters the medullary sinus – a common site for
small metastases. The lymph fuid fows through the medulla
and germinal centers of the lymph node and then exits the
FIGURE 2.1 Blind terminal sacs of lymphatic capillaries with a single efferent channel. The lymphatics of the lower extremities
overlapping layer of endothelial cells. These are 10–25 μm in diameter
and pelvis coalesce into large collecting ducts and ultimately
and allow interstitial fuid to pass in and out of the lymphatic capillary.
(Modifed from Kam C, Uren RF. Microanatomy and physiology of the form the thoracic duct. The thoracic duct and right lym-
lymphatic system. In: Nieweg O, et al, eds, Lymphatic Mapping and Probe phatic duct join the venous system at the junction of the left
Applications in Oncology [Marcel Dekker: New York, 2000] 1–22.) subclavian and jugular veins (Figure 2.3). Consequently, the
Anatomy: microanatomy and physiology 11

left supraclavicular lymph nodes represent a common site for


metastases from gynecologic cancers.
The afferent lymphatics transport lymphocytes and antigen-
presenting cells (APCs) into the lymph nodes, which are then
directed by stromal chemokines toward their designated loca-
tions and roles.22 Dendritic cells present antigens to T-cells
within the lymph node cords, whereas follicular dendritic cells
present antigens to B-cells within the lymph node follicles.23
Lymphocytes that do not encounter antigens exit the lymph
node and continue on to other lymph nodes.
Lymph node metastases occur when detached malignant
cells enter the lymphatic circulation and drain through regional
lymph nodes. Some tumors such as sarcomas spread primar-
ily hematogenously with a low rate of lymph node metastases,
while others, such as carcinoids, may produce larger lymph
node metastases than the primary tumor size itself. In gyne-
cologic malignancies such as endometrial adenocarcinomas
and squamous cell carcinomas of the vulva and cervix, lymph
node status is the single most important prognostic factor.

Immunology of regional lymph nodes


Within the last 10 years, our knowledge of the immune
FIGURE 2.2 Anatomy of a lymph node. Lymph nodes flter debris from response to cancer has grown exponentially. Paradoxically,
lymph and are a critical site of antigen presentation and immune activa- lymph nodes and the immune system can both inhibit and
tion. (Modifed from Kam C, Uren RF. Microanatomy and physiology of permit the propagation of metastatic disease. Dendritic cells
the lymphatic system. In: Nieweg O, et al, eds, Lymphatic Mapping and bearing tumor antigens may migrate from the periphery into
Probe Applications in Oncology [Marcel Dekker: New York, 2000] 1–22.) the lymph node, or antigens may drain directly into the node
itself, where APCs within the node such as macrophages, fol-
licular dendritic cells, or B-cells may detect them and induce
an immunologic response – or conversely – immunogenic
tolerance.24
In one study where melanoma cells were injected directly
into lymph nodes, a CD8+ cytotoxic response resulted in cell
death.25 Another group found that lymphocytes harvested
from lymph nodes were less likely to inhibit melanoma cell
line growth when taken closer to the primary tumor, whereas
more distant lymph nodes were more effective at inducing
apoptosis.26 These observations support the belief that after
an initial robust immune response, eventually the continued
exposure to tumor antigens results in exhaustion, with antigen
tolerance and immunosuppression.24 Additionally, the sentinel
nodes play an important role in the ‘premetastatic niche’ and
are primed by tumor-secreted factors such as vascular endo-
thelial growth factor (VEGF), which induce lymphangiogen-
esis and augment lymphatic fow, thus preparing the “soil for
the seed.”27
In the feld of gynecologic oncology, a similar microenvi-
ronment of relative immune suppression has been observed
in sentinel lymph nodes. In cervical cancer, Heeren and col-
leagues found that in comparison to nonmetastatic tumor-
draining lymph nodes, those with metastases had molecular
profles more associated with immune suppression.28 An anal-
ysis of dendritic cells in sentinel and nonsentinel lymph nodes
FIGURE 2.3 Main lymphatic collecting trunks have valves that ensure
unidirectional fow. (Modifed from Kam C, Uren RF. Microanatomy and
in endometrial and cervical cancer indicated that while the
physiology of the lymphatic system. In: Nieweg O, et al, eds, Lymphatic dendritic cells in the sentinel nodes had a higher concentration
Mapping and Probe Applications in Oncology (Marcel Dekker: New of markers of immune stimulation, those with metastases had
York, 2000) 1–22.) a lower number of mature dendritic cells, suggesting relative
12 Clinical Lymphatic Mapping

immune suppression.29 Concordantly, Baiocchi and colleagues nodes.33 Fortunately, the wide acceptance of sentinel lymph
demonstrated that the size of the sentinel lymph node metas- node mapping in uterine, cervical, and vulvar cancer has nul-
tasis was associated with the risk of nonsentinel lymph node lifed the importance of lymph node count.
metastases.30 This suggests that perhaps a smaller antigenic While a full lymphadenectomy with a higher lymph node
load may allow the sentinel node to retain its immunogenic- count may portend an improved diagnostic sensitivity, it also
ity. This may also account for the observation that the pres- causes an increased risk of surgical complications. Such com-
ence of isolated tumor cells within sentinel nodes is generally plications may range from incisional infections to lymphocyst
clinically insignifcant in a range of malignancies. Fattorossi et formation. While most lymphocysts may undergo expectant
al. described differences in lymphocyte composition between management, some require drainage, surgical intervention,
lymph nodes more proximal to the tumor site than the more or even sclerotherapy. Extensive lymphadenectomy may also
distal ones in patients with endometrial and cervical cancer.31 result in lymphedema, most commonly experienced in the
The notion that sentinel nodes eventually become over- lower extremities in patients with gynecologic malignancies.
whelmed by antigens while more distant lymph nodes remain Surgically induced lymphedema occurs as a result of lymphatic
immunocompetent supports the practice of sentinel lymph disruption, with subsequent pooling of lymph and increased
node mapping in gynecologic malignancies, in addition to the lymphatic pressure required to bypass the obstructed site
concept that full lymphadenectomy is not superior to sentinel (Figure 2.5). Interventions such as compression hose, sequen-
lymph node mapping. tial compression devices, massage, and simple elevation all
Much more work is required in these areas to understand can help control lymphedema but cannot fully reverse it. In
fully the metastatic process in sentinel lymph nodes and how a large review of 1000 lymphadenectomies for gynecologic
to use this knowledge in treatment strategies. malignancies, Querleu et al. reported 71 lymphocysts, 15% of
which required intervention.34 In vulvar cancer, full inguino-
femoral lymphadenectomy harbors a 30% lymphedema rate,
Surgical anatomy of the lymphatic system
Lymphatic channels are usually not visible during surgery, but
with the advent of lymphatic mapping, often a blue or green
channel becomes visible (Figure 2.4). Similarly, lymph nodes
vary in size and consistency so that some, but not all, nodes
are visible or palpable during surgery. Lymphatics may be
disrupted during surgery without any adverse consequences,
owing to a large network of low-pressure lymphatic vessels
with extensive collateral fow.
Surgeons often note a wide variation in the number of
regional lymph nodes described in surgical pathology reports.
Historically, a higher lymph node yield corresponded to a
higher sensitivity in detecting lymph node metastases and, as
some suggested, even a better prognosis.32 Perhaps as impor-
tant, lymph node yield is often interpreted as an indication of
surgical skill. However, lymph node count not only varies by
surgeon but also by pathologist and method of isolating lymph

FIGURE 2.5 Various degrees of lymphedema. Mild lymphedema


(a) causes little effect on lifestyle. Moderate lymphedema (b) requires
FIGURE 2.4 Larger lymphatic trunks may be seen during lymphatic compression devices and massage; however, most patients continue to
mapping. function well. Severe lymphedema (c) is debilitating.
Anatomy: microanatomy and physiology 13

Summary
Our understanding of lymph node physiology and immunol-
ogy is ever-expanding. Lymph nodes, especially sentinel
nodes, are the site of the initial immune response, but also
play an important role in immune tolerance and immuno-
genic escape. Lymphatic mapping and sentinel node biopsy
provide an opportunity to narrow the focus of basic science
and translational, as well as clinical, investigation. As such,
future research should focus on exploiting the differences in
sentinel and nonsentinel nodes and harnessing the potential of
immunology.

REFERENCES
1. Crivellato E, Travan L, Ribatti D. The Hippocratic trea-
tise ‘on glands’: the frst document on lymphoid tissue and
lymph nodes. Leukemia. 2007;21:591–2.
2. Rusznyak I. [Signifcance of lymphatic circulation]. Orv
Hetil. 1959;100:1857–63.
3. Olson JS. Bathsheba’s Breast: Women, Cancer, and History.
1st ed. Baltimore, MD: The Johns Hopkins University
Press; 2005. pp. 27–45.
4. Loukas M, et al. The lymphatic system: a historical per-
spective. Clin Anat. 2011;24:807–16.
5. Taylor AE. Capillary fuid fltration. Starling forces and
lymph fow. Circ Res. 1981;49:557–75.
6. Braithwaite L. Flow of lymph from iliocecal angle. Br J
Surg. 1923;11:7.
7. Gray J. The relation of lymphatic vessels to the spread of
cancer. Br J Surg. 1938;26:462–95.
8. Liapis C, Balzer K, Benedetti-Valentinni F. Vascular
Surgery. Berlin: Springer; 2007.
9. Halsted WS, Dahlen G, Ohman AE. The results of opera-
tions for the cure of cancer of the breast performed at
the Johns Hopkins Hospital from June, 1889, to January,
1894. The Johns Hopkins Hospital Reports. Ann Surg.
1894;20(5):497–555.
10. Tyzzer EE. Factors in the production and growth of tumor
metastases. J Med Res. 1913;28:309–32;301.
11. Louveau A, et al. Structural and functional features
of central nervous system lymphatic vessels. Nature.
2015;523:337–41.
12. Tanis PJ, Nieweg OE, Valdes Olmos RA, et al. Anatomy
and physiology of lymphatic drainage of the breast from
the perspective of sentinel node biopsy. J Am Coll Surg.
2001;192:399–409.
13. Breslin JW, et al. Lymphatic vessel network structure and
physiology. Compr Physiol. 2018;9:207–99.
14. Mislin H. Active contractility of the lymphangion
and coordination of lymphangion chains. Experientia.
1976;32:820–2.
15. Wigle JT, et al. An essential role for Prox1 in the induc-
tion of the lymphatic endothelial cell phenotype. EMBO J.
FIGURE 2.5 (Continued) 2002;21:1505–13.
16. Thiele W, Rothley M, Schmaus A, et al. Flow cytometry-
with an even higher incidence in patients requiring adjuvant based isolation of dermal lymphatic endothelial cells from
radiation.35 Indeed, sentinel lymph node mapping offers a newborn rats. Lymphology. 2014;47:177–86.
signifcant decrease in such complications, which will be dis- 17. Jussila L, et al. Lymphatic endothelium and Kaposi’s sar-
cussed in more detail in subsequent chapters. coma spindle cells detected by antibodies against the
14 Clinical Lymphatic Mapping

vascular endothelial growth factor receptor-3. Cancer Res. 27. Qian CN, et al. Preparing the ‘soil’: the primary tumor
1998;58:1599–604. induces vasculature reorganization in the sentinel lymph
18. Aukland K, Reed RK. Interstitial-lymphatic mechanisms node before the arrival of metastatic cancer cells. Cancer
in the control of extracellular fuid volume. Physiol Rev. Res. 2006;66:10365–76.
1993;73:1–78. 28. Heeren AM, et al. Nodal metastasis in cervical cancer occurs
19. Modi S, Stanton AW, Mortimer PS, et al. Clinical assess- in clearly delineated felds of immune suppression in the pel-
ment of human lymph fow using removal rate constants of vic lymph catchment area. Oncotarget. 2015;6:32484–93.
interstitial macromolecules: a critical review of lympho- 29. Kara PP, et al. Analysis of dendritic cells in sentinel lymph
scintigraphy. Lymphat Res Biol. 2007;5:183–202. nodes of patients with endometrial and patients with cervi-
20. Uren RF, Howman-Giles RB, Thompson JF. Demonstration cal cancers. Int J Gynecol Cancer. 2009;19:1239–43.
of second-tier lymph nodes during preoperative lymphos- 30. Baiocchi G, Salani R. Meeting report from the 19th meet-
cintigraphy for melanoma: incidence varies with primary ing of the International Gynecologic Cancer Society
tumor site. Ann Surg Oncol. 1998;5:517–21. (IGCS) 2019: summary of selected abstracts and meeting
21. Choi HJ, et al. Time-lapse imaging of sentinel lymph node highlights. Gynecol Oncol. 2019;155:389–92.
using indocyanine green with near-infrared fuorescence 31. Fattorossi A, et al. Lymphocyte composition of tumor
imaging in early endometrial cancer. J Gynecol Oncol. draining lymph nodes from cervical and endometrial can-
2016;27:e27. cer patients. Gynecol Oncol. 2004;92:106–15.
22. Gasteiger G, Ataide M, Kastenmuller W. Lymph node – an 32. Abu-Rustum NR, et al. Is there a therapeutic impact
organ for T-cell activation and pathogen defense. Immunol to regional lymphadenectomy in the surgical treat-
Rev. 2016;271:200–20. ment of endometrial carcinoma? Am J Obstet Gynecol.
23. Willard-Mack CL. Normal structure, function, and histol- 2008;198:457:e451–5; discussion 457:e455–6.
ogy of lymph nodes. Toxicol Pathol. 2006;34:409–24. 33. Cormier B, Sauthier P, Lussier C, et al. Determinants of
24. Sleeman JP. The lymph node pre-metastatic niche. J Mol lymph node count in endometrial cancer surgical staging.
Med (Berl). 2015;93:1173–84. Int J Gynecol Cancer. 2012;22:1361–6.
25. Preynat-Seauve O, et al. Extralymphatic tumors pre- 34. Querleu D, et al. Audit of preoperative and early compli-
pare draining lymph nodes to invasion via a T-cell cross- cations of laparoscopic lymph node dissection in 1000
tolerance process. Cancer Res. 2007;67:5009–16. gynecologic cancer patients. Am J Obstet Gynecol.
26. Farzad Z, et al. Lymphocytes from lymph nodes at different 2006;195:1287–92.
distances from human melanoma vary in their capacity to 35. Huang J, Yu N, Wang X, et al. Incidence of lower limb
inhibit/enhance tumor cell growth in vitro. Melanoma Res. lymphedema after vulvar cancer: a systematic review and
1997;7(Suppl 2):S59–65. meta-analysis. Medicine (Baltimore). 2017;96:e8722.
3
Lymphatic anatomy: lymphatics of the vulva

Anca Chelariu-Raicu and Robert L. Coleman

CONTENTS
The vulva........................................................................................................................................................................................ 15
Embryology of the vulva and the groin................................................................................................................................ 15
Topography of the vulva....................................................................................................................................................... 15
Topography of the inguinal lymphatics................................................................................................................................ 15
Superfcial inguinal lymphatics ............................................................................................................................................ 17
Femoral (deep) inguinal lymphatics..................................................................................................................................... 17
Lymphatic drainage .............................................................................................................................................................. 18
Natural history and patterns of spread ........................................................................................................................................... 18
Illustration of the anatomy of the groin ......................................................................................................................................... 19
Summary ........................................................................................................................................................................................ 22
References...................................................................................................................................................................................... 22

The vulva (Figure 3.1). Superfcially, these structures consist of, or are
covered by, a keratinized, stratifed, squamous epithelium to
In certain respects, understanding the anatomy of the vulva, its the level of the vestibule, where the epidermis becomes a non-
targeted nodal basin, and its directed, physiologic lymphatic keratinized squamous mucous membrane. The vulvar struc-
drainage should provide the clearest insight into why lymphatic tures are situated atop the superfcial perineal fascia, a caudal
mapping as a surgical procedure garners clinical relevance. continuation of the abdominal Scarpa’s layer. Support is given
Detailed study of this anatomy coupled with clinical correlates to the vulva through loose attachment to this fascia. Deep to
of patients with vulvar carcinoma has dictated surgical care, of this layer are the erectile and muscular contents of the genital
which lymphatic mapping and sentinel lymph node biopsy have foor (Figure 3.2). The deep or inferior fascia of the urogenital
become standard procedures over en bloc nodal resection. This diaphragm, on which the clitoris and ischiocavernosus, bul-
section will highlight the anatomy of the vulva and the groin, bocavernosus, and deep transverse perineal muscles lie, is an
drawing on embryologic and postmortem studies, which have important surgical landmark indicating the deep margin of
challenged the traditional concepts of nodal location and thus locoregional resection. This fascial layer becomes continuous
are important to surgical concepts guiding management. with the fascia lata laterally.

Embryology of the vulva and the groin Topography of the inguinal lymphatics
Embryologically, the external female genitalia arise from undif- The landmarks and topographic anatomy of the groin are illus-
ferentiated tissues that comprise the genital tubercle, labio- trated in Figure 3.3. The same keratinizing squamous epithe-
scrotal swellings, and urogenital folds.1 In the absence of fetal lium that covers the vulva covers the groin. Deep to the skin is
androgens, further differentiation of these tissues forms the the subcutaneous tissue, which is divided by the superfcial, or
external female phenotype, which is distinguishable 12 weeks Camper’s, fascia. Dorsal to this layer is fatty tissue containing
after fertilization. Ambiguity of this differentiation can result the superfcial inguinal nodes and the superfcial vasculature,
from the presence of exogenous or endogenous sex steroids dur- such as the superfcial external circumfex vessels, the super-
ing development; however, the null phenotype is female. fcial inferior epigastric vessels, and the superfcial external
pudendal vessels. Removal of this layer reveals the deep femo-
Topography of the vulva ral fascia, which is continuous laterally with the fascia lata of
the thigh and medially with the fascia of the adductor longus
The vulva comprises the mons pubis, the clitoris, the labia muscle, and which is coplanar with the inferior fascia of the
majora and minora, the vestibule (urethral meatus and urogenital diaphragm. The fossa ovalis is demarcated by the
hymenal remnant), the perineal body, the associated erectile falciform, or Hey’s, ligament and is perforated by the great
tissues and muscles, and the supporting subcutaneous tissues saphenous vein and other small vessels. As described later, the

DOI: 10.1201/9781003255536-3 15
16 Clinical Lymphatic Mapping

FIGURE 3.1 The external female genitalia are highlighted with the major anatomic landmarks.

FIGURE 3.2 The deep vulvar tissues are positioned in relation to the deep perineal fascia (urogenital diaphragm).

FIGURE 3.3 The topographic anatomy of the groin is presented (right side).
Anatomy: lymphatics of the vulva 17

perforate covering over the fossa is most aptly termed the crib- Nicklin et al. detailed this nodal group location in reference
riform lamina. Below the femoral fascia lies the femoral ves- to anatomic-surgical landmarks, such as the anterior superior
sels and nerve, along with the deep musculature of the groin. iliac spine and the pubic tubercle, using lymphangiography.5
The femoral or deep inguinal nodes are located along the fem- They noted that the most lateral node in this group was never
oral vein and usually within the fossa ovalis. The most supe- positioned in the outer 15% of the right inguinal ligament or the
rior node is commonly referred to as the node of Rosenmüller outer 20% of the left inguinal ligament. A subsequent study by
or Cloquet. Micheletti et al. correlated this observation anatomically and
Lymphatic drainage of the vulva (and the lower third of the embryologically as being associated with the superfcial exter-
vagina) goes principally to the groin by traversing the subcuta- nal circumfex vasculature.3,6 In their study of preterm fetuses,
neous tissue medial to the labial-crural fold. The node groups nodal tissue was identifed around these vessels and never lat-
are situated in the subcutaneous tissue overlying the femoral eral to the crossing of the medial border of the sartorius muscle
triangle (Scarpa’s triangle) bounded by the inguinal ligament and the inguinal ligament. Rob et al. proposed another distribu-
cephalad, the sartorius muscle inferior-laterally, and the adduc- tion of superfcial nodes into three groups: superfcial medial
tor longus medially Anatomically, these nodes are categorized group (Sf-M), located above and medially from the femoral
as ‘superfcial’ and ‘deep’ according to their location in refer- vein and medially to the great saphenous vein; superfcial inter-
ence to the fascia lata and position in the fossa ovalis (Figure mediate group (Sf-IM), including nodes in the vicinity of the
3.4). Traditional illustrations of the fossa ovalis depict it as cov- saphenous and femoral veins and superior-laterally from them;
ered by a perforated membrane contiguous with the fascia lata and superfcial lateral group (Sf-L) as the group of nodes in
called the cribriform fascia. However, embryologic studies have the outer third of the groin.7 The authors reported the highest
documented that this ‘fascia’ is actually distinct from the fascia percentage of nodes in the medial group (50%), followed by the
lata (femoral portion) and develops in midgestation (23–24 fetal intermediate group (34.7%). The surgical implications of these
weeks) from the connective tissue of the fossa ovalis.2–4 Through fndings support limiting the lateral extent of skin and groin
condensation, the connective tissue’s superfcial layer becomes nodal dissection, which in turn may reduce operative morbidity
a thickened membrane; therefore, it is more correctly termed by not disrupting important potential collateral lymphatic fow.
cribriform ‘lamina.’ This distinction is important and relevant
to the anticipated location of the inguinal nodes, as embryologic
Femoral (deep) inguinal lymphatics
nodal buds, which make up the deep femoral chain, arise from
superfcial nodes in this same connective tissue. The femoral or deep nodes, as mentioned, develop embryo-
logically from a nodal mass, which is situated by 11 weeks’
gestation on top of the femoral fascia. During gestation, the
Superficial inguinal lymphatics
connective tissue of the fossa proliferates and differentiates,
The superfcial inguinal nodal group primarily receives afferent dividing the nodal bundle into individual nodes. A few, usually
channels from the vulvar tissues traversing the labia and mons one to fve in total, migrate in the fossa to lie along the femoral
veneris. This group contains approximately ten nodes, which vein and become the femoral nodes.6 Cadaveric studies of the
lie in the subcutaneous tissue in or around the fossa ovalis and groin have found that these nodes are most often found at or
the saphenous vein and its tributaries. Detailed topographic below the junction of the saphenous vein with the femoral vein
studies of this nodal group have demonstrated that these nodes (Figure 3.5). In one-third of cases, the nodal tissue is located
lie uniformly within Scarpa’s triangle. Medially and inferi- cephalad to this point.2 All nodal tissue appears to be bound
orly, the superfcial bundle is limited by the femoral triangle. by the margins of the fossa ovalis. No nodal tissue is found

FIGURE 3.4 The superfcial inguinal nodes are illustrated in relationship to the femoral fascia and fossa ovalis.
18 Clinical Lymphatic Mapping

FIGURE 3.5 The relationship between the deep inguinal nodes and the femoral triangle is depicted.

beneath the fascia lata in any direction, and it is never located


lateral to the femoral artery. Interestingly, Cloquet’s node in Natural history and patterns of spread
their series was identifed in just 46% of the dissections and
was unilateral in 30%, rendering it an imperfect marker of Although many tumor cell types can arise within the vulva or
cephalad nodal extent. It is hypothesized that this node’s loca- its associated adnexal structures, most primary carcinomas of
tion is also variably identifed as the most distal node in the the vulva are squamous cell and arise from the epithelium.12
external iliac chain. Rob et al. reported 16.1% of deep inguinal The most frequent sites of involvement are the labia minora,
nodes from all the nodes dissected in the groin.7 clitoris, fourchette, perineal body, and medial labia majora.
The classic pattern of growth and spread is local extension and
lymphovascular embolization to either regional or distant sites.
Lymphatic drainage With the exception of vulvar melanoma, the pattern of spread
The clinically important features of in vivo vulvar lymphatic is relatively predictable. Local growth of the primary carci-
drainage, frst described by Eichner and later by Parry-Jones, noma generally occurs in a radial pattern, as well as invad-
are that ipsilateral fow from the vulva does not cross the labial- ing deep into the underlying subcutaneous tissues. Objective
crural fold laterally and that, in general, only midline struc- measures of this growth have been standardized with creation
tures (clitoris and perineum) have bilateral efferent drainage.8,9 of the TNM staging system.13 In this manner, the tumor is
Previously observations regarding the direct communication of described in reference to size (T1 ≤2 cm, T2 >2 cm), involve-
the clitoral lymphatics to the pelvis, although this is clinically ment of the distal midline structures (T3: urethra, vagina, and
very rare, have been confrmed by a more recent study. The pre- anus), and invasion of the midline organs (T4: bladder mucosa,
viously mentioned results showed that the risk of contralateral upper urethra, and rectal mucosa) or bony fxation.
nonsentinel metastasis in the case of bilateral sentinel mapping Once invasion occurs, the tumor can spread via the dermal
with unilateral positivity is very low.10 Ordered lymphatic fow lymphatics to the regional nodes following the previously
from the vulva generally goes to the superfcial group frst, fol- described pattern of fow. There appears to be a threshold with
lowed by drainage to the femoral nodes. From there, the efferent regard to the likelihood of groin metastatic disease based on
channels pass under the inguinal ligament in the femoral canal the depth of invasion. Kelley and colleagues reported that the
to communicate with the external iliac and pelvic nodal chains. frequency of metastatic disease among 24 patients with 1 mm
Lymphatic mapping studies in women undergoing groin dissec- or less of invasion (measured as depth of deepest invasion from
tion for cancer of the vulva have confrmed the variable location the most proximal adjacent rete peg) was 0%.14 In contrast,
of the sentinel node, although it is most commonly a medi- tumors with >1 mm invasion have a risk of nodal metastases
ally located superfcial node near the fossa ovalis.11 Confusion of up to 34%.15 This observation has led to a subclassifcation
regarding the existence of direct vulvar lymphatic drainage to the of stage I disease (IA: T1 lesion with 1-mm invasion or less,
pelvis has been confounded by different in vivo mapping tech- IB: T1 lesion with greater than 1-mm invasion), with the clini-
niques. However, studies that have used intradermal injection cal implication being that nodal dissection may safely be dis-
demonstrate exclusive primary lymphatic drainage to the groin. missed in these patients. Another group investigated in their
However, studies using deep subcutaneous injections have shown study an alternative method of measuring the depth of invasion
direct drainage to the pelvis. Clinical experience has indicated (from the basement membrane of the deepest adjacent tumor-
that the best candidates for lymphatic mapping of cutaneous free rete ridge to the deepest point of invasion) and evaluated
tumors are those with small, superfcial tumors that essentially its accuracy by comparing it with the conventional method of
have exclusive lymphatic drainage via cutaneous lymphatics. measuring and clinical outcome. Interestingly, the fndings
Anatomy: lymphatics of the vulva 19

of this study showed that by using the alternative measur-


ing method 19% of patients with FIGO stage IB tumor were Illustration of the anatomy of the groin
downstaged to FIGO stage IA, and in consequence would have
spared these patients from groin surgery.16 However, the alter- There is no other anatomic area of concern to gynecologic
native method of measuring needs further clinical validation; oncologists where so much confusion reigns regarding anat-
therefore, the conventional one remains the standard. omy and nomenclature as the groin. The nomenclature issues
The current management of early vulvar cancer is separate have been explored in detail by several authors.2,3,21 Confusion
resection of the primary tumor and the at-risk groin(s).17–19 regarding nomenclature has been compounded by errors in
Careful inspection of en bloc tumor resections and biopsies by illustration of groin anatomy that are propagated by new com-
Cherry and Glücksmann demonstrated that tumor lymphatic ment on the subject. This section will review the efforts of pre-
emboli were present in only 19% of cases.20 Clinically, recur- vious illustrators to depict groin anatomy, as well as identify
rences in the retained skin bridges among patients undergo- the most accurate images.
ing separate incision removal of the primary lesion and the In 1971, Plentl and Friedman offered a new set of illustra-
regional lymphatics are uncommon. This observation would tions of the groin that were widely imitated22 (Figure 3.6a–c).
support the hypothesis that lymphatic metastases occur as a In Figure 3.6a, the general location of the superfcial ingui-
result of embolization rapidly and most often without interval nal nodes are accurate; however, the number (fve) is fewer
deposition of viable tumor. than the eight to ten commonly found. In all the fgures, the

FIGURE 3.6 (a, b, and c) Lymphatic anatomy of the right groin as demonstrated in 1971 by Plentl and Friedman27 (redrawn for added clarity).
20 Clinical Lymphatic Mapping

superfcial and femoral nodes are shown as the same size. It is 1979 to depict the location of the superfcial inguinal nodes25
common for the nodes to vary in size; sentinel nodes are usu- (Figure 3.7a). This fgure shows the nodes as being rather large
ally at least 1 cm in size. and shows ‘sentinel’ nodes in multiple sites. Current map-
In the cross-section in Figure 3.6c, the patient is very thin, ping experience suggests that sentinel nodes are usually at
as there is almost no fat between the skin and Camper’s fas- the medial edge of the femoral triangle and rarely lateral to
cia. This fgure shows the superfcial nodes very close to the the saphenous vein. DiSaia et al. also introduced a new cross-
skin. In other patients, the ‘superfcial’ inguinal nodes can section illustration of the groin (Figure 3.7b). This illustration
be very ‘deep’ to the skin due to a thick layer of subcutane- shows seven nodes in a single cross-section, with the femo-
ous fat. This overlooked observation contributed to the poor ral nodes larger than the superfcial nodes, the femoral nodes
results observed with external beam radiation therapy by lateral to the femoral artery, and the cribriform fascia as a
the Gynecologic Oncology Group.23 The cribriform fascia is continuous thick structure. This illustration has been updated
depicted as a bold black line separating the superfcial and to portray more accurately cribriform fascia as fenestrated
femoral nodes. As described earlier, this structure is more (Figure 3.8). In addition, the traditionally described ‘femo-
appropriately described as a lamina. ral’ nodes are now within the fossa ovalis, and the distinc-
Figures drawn by Cabanas24 to illustrate the location of sen- tion of these nodes as ‘below’ the cribriform fascia has been
tinel nodes in the male groin were modifed by DiSaia et al. in de-emphasized.

FIGURE 3.7 (a and b) Sentinel lymph nodes and inguinal femoral anatomy as illustrated by DiSaia et al.25 (redrawn for added clarity).

FIGURE 3.8 Further update of cross-sectional anatomy of the groin (redrawn from Ref. 28 for added clarity).
Anatomy: lymphatics of the vulva 21

Other texts have taken a different approach (Figure 3.9a–b). fascia follows an undulating path with the appearance of all the
In Figure 3.9a, the lymphatics of the vulva appear to cross lymph nodes ‘above’ this structure. Rouzier et al.26 attempted
the labial-crural fold, contrary to the work of Parry-Jones.7 In to illustrate the levels of the groin with schematic diagrams that
Figure 3.9b, Camper’s fascia is not shown, and the cribriform show the extent of various surgical approaches (Figure 3.10).

FIGURE 3.9 (a and b) Vulvar and inguinal lymphatic anatomy (redrawn from Ref. 29 for added clarity).

FIGURE 3.10 Operative (A) and cross-sectional (B) anatomy of the groin. a, fascia lata; b, fossa ovalis; c, great saphenous vein; d, superfcial epigas-
tric vein; e, inguinal ligament. The compartments of the groin are (1) superfcial medial inguinal nodes, (2) medial femoral nodes, and (3) superfcial
lateral inguinal nodes. Sentinel nodes are most commonly found in compartment 1 and sometimes in compartment 3. Femoral sentinel nodes are rarely,
if ever, seen. (Redrawn from Ref. 26 for added clarity.)
22 Clinical Lymphatic Mapping

13. AJCC. Cancer Staging Manual. Atlanta, GA: Lippincott-


Summary Raven; 1997.
14. Kelley JL, Burke TW, Tornos C, et al. Minimally invasive
The anatomic considerations of the vulva and groin continue vulvar carcinoma: an indication for conservative surgical
to play a vital role in the evolution of contemporary manage- therapy. Gynecol Oncol. 1992;44:240–4.
ment strategies. Lymphatic mapping and sentinel node pro- 15. Burke TW, Levenback C, Coleman RL, et al. Surgical ther-
cedures have led to a better understanding of in vivo groin apy of T1 and T2 vulvar carcinoma: further experience with
anatomy, particularly since the blue dye procedure allows radical wide excision and selective inguinal lymphadenec-
clear visualization of lymphatic channels and lymph nodes. It tomy. Gynecol Oncol. 1995;57:215–20.
is anticipated that future study will further elucidate important 16. Yoder BJ, Rufforny I, Massoll NA, et al. Stage IA vulvar
selection criteria whereby patients will undergo individualized squamous cell carcinoma: an analysis of tumor invasive
curative extirpative surgery with minimal morbidity. characteristics and risk. Am J Surg Pathol. 2008;32(5):765–
72. https://doi.org/10.1097/PAS.0b013e318159a2cb. PMID:
18379417.
REFERENCES 17. van den Einden LC, Massuger LF, Jonkman JK, et al. An
1. Langman J. Urogenital system. In: Langman J, editor, alternative way to measure the depth of invasion of vulvar
Medical Embryology. Baltimore, MD: William & Wilkins; squamous cell carcinoma in relation to prognosis. Mod
1981. pp. 255–9. Pathol. 2015;28(2):295–302.
2. Borgno G, Micheletti L, Barbero M, et al. Topographic 18. Berman ML, Soper JT, Creasman WT, et al. Conservative
distribution of groin lymph nodes. J Reprod Med. surgical management of superfcially invasive stage I vulvar
1990;35:1127–9. carcinoma. Gynecol Oncol. 1989;35:352–7.
3. Micheletti L, Preti M, Zola P, et al. A proposed glossary 19. Hacker NF, Berek JS, Lagasse LD, et al. Individualization
of terminology related to the surgical treatment of vulvar of treatment for stage I squamous cell vulvar carcinoma.
carcinoma. Cancer. 1998;83:1369–75. Obstet Gynecol. 1984;63:155–62.
4. Micheletti L, Levi AC, Bogliatto F, et al. Rationale and 20. Cherry C, Glucksmann A. Lymphatic embolism and lymph
defnition of the lateral extension of the inguinal lymphad- node metastasis in cancers of vulva and of uterine cervix.
enectomy for vulvar cancer derived from an embryological Cancer. 1955;8:564.
and anatomical study. J Surg Oncol. 2002;81:19–24. 21. Levenback C, Morris M, Burke TW, et al. Groin dissection
5. Nicklin JL, Hacker NF, Heintze SW, et al. An anatomi- practices among gynecologic oncologists treating early vul-
cal study of inguinal lymph nodes topography and clinical var cancer. Gynecol Oncol. 1996;62:73–7.
implications for the surgical management of vulval cancer. 22. Plentl AA, Friedman EA. Clinical signifcance of the vul-
Int J Gynecol Cancer. 1995;5:128–33. var lymphatics. In: Lymphatic System of Female Genitalia.
6. Micheletti L, Levi AC, Bogliatto F. Anatomosurgical impli- Philadelphia, PA: W.B.Saunders; 1971. pp. 27–50.
cations derived from an embryological study of Scarpa’s tri- 23. Stehman FB, Bundy BN, Thomas G, et al. Groin dissec-
angle with particular reference to groin lymphadenectomy. tion versus groin radiation in carcinoma of the vulva: a
Gynecol Oncol. 1998;70:358–64. Gynecologic Oncology Group study. Int J Radiat Oncol
7. Rob L, Robova H, Pluta M, et al. Further data on senti- Biol Phys. 1992;24:389–96.
nel lymph node mapping in vulvar cancer by blue dye and 24. Cabanas RM. An approach for the treatment of penile car-
radiocolloid Tc99. Int J Gynecol Cancer. 2007;17(1):147–53. cinoma. Cancer. 1977;39:456–66.
8. Parry-Jones E. Lymphatics of the vulva. J Obstet Gynecol 25. DiSaia PJ, Creasman WT, Rich WM. An alternate approach
Br Commonwealth. 1963;70:751–65. to early cancer of the vulva. Am J Obstet Gynecol.
9. Eichner E, Mallin LP, Angell ML. Further experience with 1979;133:825–32.
direct sky blue in the in vivo studies of gynecologic lym- 26. Rouzier R, Haddad B, Dubernard G, et al. Inguinofemoral
phatics. Am J Obstet Gynecol. 1955;69:1019–26. dissection for carcinoma of the vulva: effect of modifca-
10. Woelber L, Eulenburg C, Grimm D, et al. The risk of con- tions of extent and technique on morbidity and survival. J
tralateral non-sentinel metastasis in patients with primary Am Coll Surg. 2003;196:442–50.
vulvar cancer and unilaterally positive sentinel node. Ann 27. Plentl AA, Friedman EA. Lymphatics of the cervix uteri.
Surg Oncol. 2016;23(8):2508–14. In: Plentl AA, Friedman EA, editors, Lymphatic System of
11. Levenback C, Coleman RL, Burke TW, et al. Intraoperative Female Genitalia. Philadelphia: WB Saunders; 1971. pp.
lymphatic mapping and sentinel node identifcation with 75–115.
blue dye in patients with vulvar cancer. Gynecol Oncol. 28. DiSaia P. Invasive cancer of the vulva. In: DiSaia P,
2001;83:276–81. Creasman WT, editors, Clinical Gynecologic Oncology. St
12. Wilkinson E. Premalignant and malignant tumors of the Louis, MO: Mosby; 2002.
vulva. In: Kurman R, editor, Blaustein’s Pathology of the 29. Doherty M. Clinical anatomy of the pelvis. In: Copeland
Female Genital Tract. New York: Springer-Verlag; 1994. L, editor, Textbook of Gynecology. Philadelphia, PA: WB
pp. 87–129. Saunders; 1993. pp. 48–84.
4
Lymphatic anatomy: lymphatics of the cervix

Anca Chelariu-Raicu and Katherine C. Kurnit

CONTENTS
Topographic anatomy of the cervix ............................................................................................................................................... 23
Topographic anatomy of the cervical lymphatics .......................................................................................................................... 24
Lateral trunk ......................................................................................................................................................................... 24
Posterior trunk ...................................................................................................................................................................... 25
Anterior trunk ....................................................................................................................................................................... 25
The natural history of cervix cancer relevant to its lymphatic anatomy ........................................................................................ 26
Summary ........................................................................................................................................................................................ 26
References...................................................................................................................................................................................... 27

Although the uterine cervix and the uterine corpus differ ana- The cervix is composed of fbrous, elastic, and smooth mus-
tomically and functionally, their respective lymphatic systems cular tissue. It is more fbrous than the corpus, with smooth
are similar. The mucosa, muscularis, and serosa have distinct muscle comprising just 15% of the body of the cervix, primar-
lymphatic fow, but within each layer, fow between the cervix ily at the endocervix. It is lined by columnar and squamous
and the corpus is continuous. We have chosen to discuss the epithelium. The endocervix is lined by a single layer of mucin-
uterine cervix and uterine corpus separately given that tumors producing columnar epithelium, which also lines the many
arising in the cervix are treated differently from tumors arising endocervical glands. In this respect, the mucin-producing
in the corpus. Understanding and exploitation of the normal apparatus is not truly glandular but is a complex infolding of
lymphatic anatomy of the uterine cervix have led to refne- the mucosal surface. The distal extent of this mucosa joins
ment and individualization of treatment for the diseased cer- stratifed, nonkeratinizing squamous epithelium on or near the
vix. Further elucidation of how disease affects the lymphatic exocervix. The location of this abrupt transformation is vari-
framework of the cervix and lymphangiogenic processes will able and changes continuously during the reproductive years,
be important in clarifying the natural history of cervical can- responding to the lower vaginal pH after puberty.3 In this
cer and increasing our therapeutic acumen. process, columnar epithelium undergoes metaplastic change
to become more resilient squamous epithelium.4 It is in this
transformation zone that most preinvasive and invasive cervi-
cal lesions arise. Distally, the exocervical tissue is continuous
Topographic anatomy of the cervix with the vaginal epithelium.
Anatomically, the cervix is defned as the region of the uterus
from the isthmus to its vaginal termination. Depending on
a woman’s age and on uterine and cervical factors, the size
of the cervix varies with respect to the size of the corpus.1
In general, the cervix ranges from 2 to 4 cm in length in the
nulligravid woman. It is connected to the vagina through an
oblique fbrous attachment where approximately one-third
of the anterior wall and approximately one-half of the pos-
terior wall are exposed to the vagina (infravaginal cervix)2
(Figure  4.1). The vaginal portion, or exocervix, is convex.
Centrally located within the exocervix is the external cervical
os. The size of this opening to the uterus varies depending on a
woman’s age and history of parturition. Proximal to the exter-
nal os is the elliptical endocervical canal, which terminates
at the internal cervical os. Here the cervix joins the uterine
isthmus. Anteriorly and posteriorly, the supravaginal cervix is
covered by the parietal peritoneum. FIGURE 4.1 Topographic anatomy of the cervix.

DOI: 10.1201/9781003255536-4 23
24 Clinical Lymphatic Mapping

FIGURE 4.2 The cervix is supported by the cardinal and uterosacral ligaments, which blend with the fascia of the pelvic diaphragm.

The paired uterosacral and cardinal ligaments primarily When the stromal lymphatics reach the serosa, they inter-
support the cervix and uterus. The uterosacral ligaments attach face through a series of lacunae, which go on to invest the
at the base of the uterus and run in the rectouterine perito- entire corpus.5 These major efferent systems coalesce in the
neal folds to the sacrum. The cardinal ligaments are thickened cervix to form three dominant extravisceral trunks (anterior,
bands of fbrous tissue in continuation with the endopelvic lateral, and posterior). It should be noted that these cervical
fascia that extend to the lateral pelvic sidewall (Figure 4.2). lymphatic vessels often share terminal nodes with other sites,
Within these bands also run the nerves and vessels to the cer- such as the uterine corpus and the vagina. However, it is rele-
vix and pelvic viscera. vant that there is little intermixing of lymph; rather, separate
lymphatic trunks from these sites run in parallel to common
basins and particular terminal nodes. Valves within these
trunks also prevent retrograde fow under normal conditions.
Topographic anatomy of the cervical lymphatics
The lymphatic vessels of the endocervical mucosa are found
Lateral trunk
immediately below the cuboidal/columnar glandular epithe-
lium and are irregular in their arrangement. The fragile indi- The largest and most signifcant pathway of the extravisceral
vidual vessels coalesce to form channels, which fow out from cervical drainage comes from the lateral trunk. There are
the mucosa within the plicae palmatae, generally perpendicu- three branches within this trunk, each of which drains into
lar to the axis of the cervical canal. Interfacing with the stroma, specifc nodal basins within the pelvis (Figure 4.3). The upper
the channels fatten and angle to run parallel with the endocer- branches of the lateral trunk are two to three large vessels that
vical canal, where they combine further to make larger chan- course from the cervical serosa cephalad and anterior along
nels. Ultimately, these vessels converge upon intermediate the path of the uterine vessels, crossing over the ureter and
trunks located within the stromal vasculature. Within the cer- obliterated umbilical artery, and coming to rest in the inter-
vical stroma, a dense and regular lattice of lymphatic vessels is iliac nodal basin at the bifurcation of the external and inter-
found. Near the mucosal surface, lymphatics grossly outnum- nal iliac vessels. The lateral-trunk drainage is considered the
ber vascular channels and seemingly bear no consistent rela- dominant drainage of the cervix and was referred to by Leveuf
tionship with blood vessels or each other. Circumferentially, as and Godard as “la voie principale.”6 The large node in this
these channels coalesce into larger trunks, they course more location is consistent clinically; however, in its absence, a sec-
regularly with the cervical veins, often surrounding them to ondary node is often found in the common iliac chain. On the
the serosa. A small number of reports have documented the right, this secondary node is often located on the common iliac
existence of a valvular system within stromal lymphatics that vein; on the left, it can be found between the iliopsoas muscle
prevents retrograde fow in all but disease states. and common iliac artery.
Anatomy: lymphatics of the cervix 25

FIGURE 4.3 The dominant lateral lymphatic trunk drains into several lower pelvic nodal basins. (Modifed from Plentl and Friedman.)5

The path of the upper branches of the lateral trunk to the is described in the classic, cadaver-based anatomic studies.
iliac nodes can be interrupted by small parametrial lymph Clinical reports rarely comment on these lymph nodes, and
nodes. These lymph nodes are diffcult to study. They can eas- gluteal nodes appear to be a rare site for metastatic disease.
ily be overlooked in cadaver studies. In vivo, they are not seen The lower branches of the lateral trunk continue on towards
during radical hysterectomy. They are very close to the cer- the presacral (subaortic) lymph nodes, as described later.
vix and therefore are not imaged well on lymphoscintigraphy
because of the large amount of residual radioactivity in the
Posterior trunk
cervix. They are not seen well on blue dye studies either, again
because of their small size, their proximity to the cervix, and The posterior collecting trunk primarily drains towards the
the fact that they are not seen on dissection. subaortic nodal basin. This trunk arises from the posterior cer-
The equally important middle branches of the lateral trunk vix and courses along the uterosacral ligament to the lateral
serve as a dominant pathway to the obturator lymph nodes, rectal fascia, continuing anteriorly over the sacral promontory
also referred to by some as the posterior interiliac nodes. The or posteriorly to the superior rectal nodes (Figure 4.4). Direct
course of the lymph in this case is along the middle cardinal drainage to the aortic nodes can be demonstrated in vessels
ligament to the posterior aspect of the internal iliac vessel. from this trunk, which travel with the ureter to the pelvic brim,
Arising from the lateral dorsal cervix are the lower branches where they diverge into the common iliac, subaortic, and aor-
of the lateral trunk. Some of these pass posteriorly around the tic nodal basins.
ureter and course either dorsally to the uterosacral ligament,
terminating in the inferior gluteal nodes, or cephalad and ven-
Anterior trunk
trally, terminating in the superior gluteal nodes. Still others
course posteriorly along the sacral hollow, terminating in the The anterior trunk leaves the anterior cervix and courses along
sacral and subaortic nodes directly. the posterior surface of the bladder to the superior vesical
The lower branches of the lateral trunk serve as a pathway artery. From there it travels to the obliterated umbilical vessel
to the superior and inferior gluteal lymph nodes. This pathway and terminates in the distal interiliac nodal basins anteriorly.
26 Clinical Lymphatic Mapping

TABLE 4.1
Likelihood of Lymph Node Metastases in Patients with Early-Stage
Cervical Cancer
Stage % of patients with metastases
IA1 0.3
IA2 1–7
IB1 16
IB2 32
IIA 25
IIB 27–43

at increased risk of developing cervical cancer.11,12 Most cer-


tainly, the shift from carcinoma in situ to invasive cancer is the
result of an accumulation of genetic errors and the interaction
of HPV DNA with the host genome.9,13–16 Cervical cancer is
believed to progress through a state of microscopic invasion
into the stroma and radial growth on the surface. Ultimately,
a locus is formed that, in general, grows locally to invade the
deeper stroma and, later, the paracervical and parametrial tis-
sues. If left untreated, the disease will expand through these
tissues to involve the lateral pelvic side wall. In addition, it
may grow to involve the bladder, rectum, or both.
Cancer arising from the cervix has been reported to extend
into the uterine corpus in 10%–30% of cases and into the uter-
ine adnexa in 0.5%–1.6% of cases.17–19 Considering that as
many as 17% of patients with early-stage cervix cancer have
FIGURE 4.4 The posterior lymphatic trunk courses along the uterosac- lymph node metastases,20–22 lateral spread from the pelvic
ral ligament to drain into the common iliac, subaortic, aortic, and superior
lymph nodes (obturator, internal iliac, and external iliac lymph
gluteal nodes. A blue lymph node is shown at a common location to fnd
sentinel lymph nodes in patients with cervical cancer. Sentinel nodes may nodes) to the pelvic sidewall, to the common iliac lymph nodes,
also be found in obturator, common iliac, presacral, and paraaortic sites. and then the paraaortic lymph nodes was thought to be the
(Modifed from Plenty and Friedman.)5 rule.20 However, studies using sentinel lymph node mapping
have demonstrated that any of the pelvic lymph node groups,
and even the paraaortic lymph nodes, may contain the frst
Separate but parallel vessels run along the cardinal ligament, draining lymph node.23 Still, it is uncommon to fnd isolated
where they meet without intermixing with the lateral trunk paraaortic lymphatic spread in the absence of metastatic pel-
and drain into that trunk’s distribution. vic disease.20,24 Thus, as a group, the pelvic lymph nodes are
still the most commonly involved sites of lymphatic spread.20
The likelihood of nodal involvement by disease stage among
The natural history of cervix cancer patients with early-stage disease is shown in Table 4.1.
Hematogenous spread of disease is relatively unusual and
relevant to its lymphatic anatomy most frequently involves the lungs, bones, intraabdominal vis-
Most cancers arising in the uterine cervix are carcinomas of cera, or distant lymphatics.25 One study indicated that patients
its epithelium. The predominant histologic subtype is squa- with hematogenous metastasis had a 5.3-fold higher risk of
mous; however, an increasing proportion of adenocarcinoma, death compared to those with lymphatic metastasis.26 Variant
comprising approximately 25% of cases, has been observed.7,8 cell types, such as neuroendocrine or glassy cell carcinoma,
Cervical carcinomas are largely believed to develop from a may be associated with distant disease in the absence of local
preinvasive lesion, carcinoma in situ, over a number of years. spread. Current therapy for these variants typically involves
However, another model has been proposed which hypothe- multimodality strategies designed to address the tumor’s
sizes that carcinoma in situ may develop rapidly following a potential for local and distant metastasis.27,28
human papillomavirus (HPV) infection and not develop from
low-grade dysplasia.9 Even though many of the molecular
events driving the transformation from carcinoma in situ to Summary
invasive cancer are not well known, the link between geni-
tal HPV infections and cervical cancer was demonstrated as Lymphatic anatomy of the cervix is surprisingly complex.
early as the 1980s by Harold zur Hausen.10 Some data suggest Pelvic lymph nodes actually refer to several nodal basins
that the presence of HPV and a high viral load place patients immediately adjacent to one another. Lymphatic mapping
Anatomy: lymphatics of the cervix 27

studies have the potential to help understand the clinical sig- 15. Wistuba, II, Montellano FD, Milchgrub S, et al. Deletions
nifcance, if any, of the various anatomic variations. of chromosome 3p are frequent and early events in the
pathogenesis of uterine cervical carcinoma. Cancer Res.
1997;57(15):3154–8.
REFERENCES 16. Wentzensen N, Sherman ME, Schiffman M, et al. Utility
1. Bartoli JM, Moulin G, Delannoy L, et al. The normal of methylation markers in cervical cancer early detec-
uterus on magnetic resonance imaging and variations tion: appraisal of the state-of-the-science. Gynecol Oncol.
associated with the hormonal state. Surg Radiol Anat. 2009;112(2):293–9.
1991;13(3):213–20. 17. Reyes C, Murali R, Park KJ. Secondary involvement of the
2. Ferenczy A, Wright T. Anatomy and histology of the cervix. adnexa and uterine corpus by carcinomas of the uterine
In: Kurman RK, editor, Blaustein’s Pathology of the Female cervix: a detailed morphologic description. Int J Gynecol
Genital Tract. New York: Springer-Verlag; 1994. pp. 185–201. Pathol. 2015;34(6):551–63.
3. Singer A. The uterine cervix from adolescence to the meno- 18. Sutton GP, Bundy BN, Delgado G, et al. Ovarian metas-
pause. Br J Obstet Gynaecol. 1975:82:1–99. tases in stage IB carcinoma of the cervix: a Gynecologic
4. Reich O, Regauer S, McCluggage WG, et al. Defning the Oncology Group study. Am J Obstet Gynecol. 1992;166(1
cervical transformation zone and squamocolumnar junc- Pt 1):50–3.
tion: can we reach a common colposcopic and histologic 19. Kim MJ, Chung HH, Kim JW, et al. Uterine corpus involve-
defnition? Int J Gynecol Pathol. 2017;36(6):517–22. ment as well as histologic type is an independent predictor
5. Plentl A, Friedman E. Lymphatics of the cervix uteri. In: of ovarian metastasis in uterine cervical cancer. J Gynecol
Lymphatic System of Female Genitalia. Philadelphia, PA: Oncol. 2008;19(3):181–4.
WB Saunders; 1971. pp. 75–115. 20. Hopkins MP, Morley GW. Stage IB squamous cell cancer
6. Leveuf J, Godard H. Les lymphatiques de l’utérus. Rev of the cervix: clinicopathologic features related to survival.
Chir. 1923:3:219–48. Am J Obstet Gynecol. 1991;164(6 Pt 1):1520–7; discussion
7. Smith HO, Tiffany MF, Qualls CR, et al. The rising inci- 7–9.
dence of adenocarcinoma relative to squamous cell carci- 21. Sevin BU, Nadji M, Averette HE, et al. Microinvasive car-
noma of the uterine cervix in the United States – a 24-year cinoma of the cervix. Cancer. 1992;70(8):2121–8.
population-based study. Gynecol Oncol. 2000;78(2):97–105. 22. Chen B, Wang L, Ren C, et al. The effect of neoadjuvant
8. van der Horst J, Siebers AG, Bulten J, et al. Increasing chemotherapy on lymph node metastasis of FIGO stage
incidence of invasive and in situ cervical adenocarci- IB1-IIB cervical cancer: a systematic review and meta-
noma in the Netherlands during 2004–2013. Cancer Med. analysis. Front Oncol. 2020;10:570258.
2017;6(2):416–23. 23. Levenback C, Coleman RL, Burke TW, et al. Lymphatic
9. Nedjai B, Reuter C, Ahmad A, et al. Molecular progres- mapping and sentinel node identifcation in patients with
sion to cervical precancer, epigenetic switch or sequential cervix cancer undergoing radical hysterectomy and pelvic
model? Int J Cancer. 2018;143(7):1720–30. lymphadenectomy. J Clin Oncol. 2002;20(3):688–93.
10. zur Hausen H. Papillomaviruses causing cancer: evasion 24. Balaya V, Mathevet P, Magaud L, et al. Predictive factors
from host-cell control in early events in carcinogenesis. J of unexpected lymphatic drainage pathways in early-stage
Natl Cancer Inst. 2000;92(9):690–8. cervical cancer. Gynecol Oncol. 2019;154(1):102–9.
11. Josefsson AM, Magnusson PK, Ylitalo N, et al. Viral load 25. Delgado G, Bundy BN, Fowler WC, et al. A prospective sur-
of human papilloma virus 16 as a determinant for develop- gical pathological study of stage I squamous carcinoma of
ment of cervical carcinoma in situ: a nested case-control the cervix: a Gynecologic Oncology Group Study. Gynecol
study. Lancet (London, England). 2000;355(9222):2189–93. Oncol. 1989;35(3):314–20.
12. Moberg M, Gustavsson I, Wilander E, et al. High viral 26. Kim K, Cho SY, Kim BJ, et al. The type of metastasis
loads of human papillomavirus predict risk of invasive cer- is a prognostic factor in disseminated cervical cancer. J
vical carcinoma. Br J Cancer. 2005;92(5):891–4. Gynecol Oncol. 2010;21(3):186–90.
13. Park TW, Fujiwara H, Wright TC. Molecular biology of 27. Lagasse LD, Creasman WT, Shingleton HM, et al. Results
cervical cancer and its precursors. Cancer. 1995;76(10 and complications of operative staging in cervical cancer:
Suppl):1902–13. experience of the Gynecologic Oncology Group. Gynecol
14. Muller CY, O’Boyle JD, Fong KM, et al. Abnormalities of Oncol. 1980;9(1):90–8.
fragile histidine triad genomic and complementary DNAs 28. Cohen JG, Kapp DS, Shin JY, et al. Small cell carcinoma of
in cervical cancer: association with human papillomavirus the cervix: treatment and survival outcomes of 188 patients.
type. J Nat Cancer Inst. 1998;90(6):433–9. Am J Obstet Gynecol. 2010;203(4):347 e1–6.
5
Lymphatic anatomy: lymphatics of the uterus

Jennifer J. Mueller and Nadeem R. Abu-Rustum

CONTENTS
Introduction .................................................................................................................................................................................... 29
References...................................................................................................................................................................................... 33

Introduction the myometrium in a regular and perpendicular pattern. The


lymphatics travel to the uterine serosa. Along this journey to
The lymphatic anatomy of the uterine corpus compared to the serosa, they anastomose with other lymphatics and enlarge
that of other solid organs is complex, and we are indebted to in diameter. Anastomotic connections between the corpus and
the pioneering clinicians and anatomists for their ingenuity cervix within the mucosal and muscular layers develop in this
and devotion in studying and revealing its intricacies across manner. The lymphatic trunks arising from this vast network
a historic body of literature. A portion of early works focused condense to form the major collecting channels that drain the
on lymphatic mapping in the normal-appearing uterus, while uterus extraviscerally. They are joined by the serosal lymphat-
another portion focused on lymphatic mapping of uterine can- ics. This creates a dominant confuence of lymph towards the
cers to establish the path of tumor emboli. In the former, repro- lateral margins of the uterus.
ducible patterns of lymphatic drainage were established, and Gynecologist and anatomist Dr. Erle Henriksen described
in the latter, an enhanced understanding of metastatic lym- the major lymphatic drainage patterns of the uterine corpus.2,3
phatic spread in endometrial cancer emerged. In his study of 420 uterine cancer necropsies, Henriksen
In this section, we will review key early work that describes described a high degree of variability in lymphatic channels,
the lymphatic drainage pathways of the uterine corpus. We with frequent anomalous and anastomotic relationships to
will review common patterns of metastatic lymph node spread other channels and nodes.3 Anatomists predating Henriksen
in endometrial cancer based on necropsy studies and the cur- also described a highly irregular, anastomosing endometrial
rent literature describing lymphatic spread patterns in endo- lymphatic network that maintains lymphatic continuity across
metrial cancer. all regions of the uterus.4
The lymphatics of the uterus are complex, given the asso- Henriksen described three major lymphatic channels drain-
ciated distinct anatomic regions (e.g., cervix, uterine body, ing the cervix. The frst major channel exits the cervix and
fundus, and fallopian tubes) and tissue layers (e.g., endome- connects to paracervical, external iliac, and obturator lymph
trium, myometrium, and serosa). The most challenging is nodes. The second courses along in a similar manner to con-
the endometrial lymphatic anatomy due to valves within the nect to internal iliac lymph nodes. The third major channel
lymphatics that preclude dye study and the changes that occur travels back and forth within the uterosacral folds, connect-
in cycling endometrium. In the 1980s, Blackwell and Fraser ing to presacral lymph nodes. These channels were observed
described the superfcial lymphatics of the endometrium intercommunicating between interiliac and presacral nodal
using curettage specimens from 42 normally cycling women.1 basins. Henriksen also described three main channels drain-
They used electron and light microscopy ultrastructure data ing the corpus. The upper corpus lymphatics join ovarian ves-
to three-dimensionally reconstruct endometrial lymphatic sels and drain paraaortic nodal basins below the renal vessels.
architecture. Lymphatics originate as blind vessels in the The middle and lower corpus (in close association with cervi-
endometrium. These capillaries extend to the endomyometrial cal lymphatic channels) coalesce and course along the uterine
interface and enlarge and coalesce with frequent anastomo- vessels, draining paracervical, interiliac, and obturator nodes.
ses. Ultrastructural reconstruction suggests the blind termi- The fundus lymphatic channels follow the round ligament to
nal ends serve an absorptive function by drawing fuid into the inguinal nodal basins.
the lymphatic system, which could provide a ready conduit Plentl and Friedman4 described four key lymphatic drain-
for lymphatic dissemination of endometrial cancer, even in age routes for the uterus in an excellent textbook review of
early stages. As lymphatic capillaries traverse the endome- this topic: 1) A fundal/ovarian route in which lymphatics travel
trium, they enlarge in diameter and coalesce to form lacunae, cephalad to infrarenal paraaortic lymph nodes. 2) A fallopian
which lie parallel to the muscularis layer of the myometrium. tube/uterine cornu route in which lymphatics anastomose
Lymphatic channels emerge from the lacunae and perforate with ovarian lymphatic channels to drain aortic and superior

DOI: 10.1201/9781003255536-5 29
30 Clinical Lymphatic Mapping

gluteal lymph nodes. 3) A mid and lower uterine body route In summary, uterine lymphatic drainage pathways consis-
in which lymphatics condense in the broad ligament. A longi- tently follow an ovarian drainage pathway to the aortic and
tudinal channel named ‘the anastomosis of Poirier’ connects gluteal nodal region, a round ligament drainage pathway to
subserosal corpus lymphatics with the cervix in this region. the inguinofemoral nodal region, and a cervical/midbody uter-
The lymphatic channels within the broad ligament pass over ine drainage pathway to the external iliac and obturator nodal
the obliterated hypogastric artery and terminate to interiliac region, as well as the common, internal iliac, and presacral
nodes. 4) A round ligament route in which lymphatics drain to nodal region (Figure 5.1).
femoral lymph nodes, which was deemed less important. This early work prompted additional questions for research-
Lecuru et al. injected a colored dye into the uterus of 11 ers. Which nodal basins are the most critical to surgically eval-
female cadavers, 5 within the corpus and 6 within the cervix, uate? Are there dominant drainage pathways early in cancer
and mapped lymphatic drainage to determine any difference progression? Does endometrial tumor location infuence nodal
in these patterns for cervical versus uterine malignancies.5 The spread of disease?
cervical injections drained primarily to internal iliac nodes, Henriksen was among the frst to describe lymph node spread
with no paraaortic connections. The corpus injections drained patterns in cervical and uterine carcinomas in a series of 64
to nodes posterior to the external iliac vessels in the obturator patients with varying stages of disease, some treated with radi-
region primarily and, less commonly, to the paraaortic region. ation.3 Tumor metastases were noted via gross anatomic study

FIGURE 5.1 Main lymphatic drainage pathways of the uterus. The left side illustrates the three main lymphatic drainage pathways of the uterine
corpus. The frst pathway drains the cervical and mid/lower uterine lymphatics to pelvic nodal basins along the iliac and obturator vessels. The most
common pattern in this pathway is the upper or ventral paracervical pathway. Lymphatic trunks condense in the paracervix and cross medial to lateral
over the cranial part of the obliterated umbilical ligament. The less common pattern is the lower or dorsal paracervical pathway. These trunks condense
in the paracervix and traverse dorsally in the meso-ureter to drain the presacral common iliac nodes. The second pathway drains the fundal portion
of the uterus, including the serosa, which joins with the ovarian vessels and drains to paraaortic nodal basins. The third pathway (and least dominant)
drains the mid-uterine lymphatics, which travel within the round ligament to the inguinal nodal basins. The right side illustrates the uterine serosal
lymphatics. Lymphatic capillaries traverse the endometrium and enlarge and coalesce as lacunae on their journey through the myometrium to the
serosa. Frequent anastomotic connections between the corpus and cervix in the muscularis and mucosal layers develop and help form a vast network
of interconnected channels that drain the uterus extraviscerally.
Anatomy: lymphatics of the uterus 31

alone. Nodal metastases were located in external, internal, and histologic grade, and depth of myometrial invasion. This
common iliac regions, as well as obturator, presacral, aortic, helped to rationalize comprehensive pelvic and paraaortic
and inguinal regions. Henriksen later studied 188 endometrial lymphadenectomy as a surgical staging procedure in early-
cancer necropsies and made several key observations. First, stage endometrial cancer.16–18
in 15% of patients, only aortic nodes were involved, with no Sentinel lymph node (SLN) mapping emerged by combin-
other distant metastases. Second, tumor emboli in a lymphatic ing our understanding of lymphatic drainage pathways with
channel or node were seen to interfere with lymphatic fow locations of occult nodal spread in endometrial cancer. SLN
and created shunts to alternative channels. This collateraliza- mapping allows the surgeon to see lymphatic channels drain
tion led to unpredictable sites of metastases and bypassed the an individual uterus in real time. Live demonstrations of SLN
primary nodal basins typically involved. Third, widespread mapping and SLN data generated over the years have expanded
pelvic nodal metastases without aortic nodal involvement were our knowledge of endometrial cancer nodal spread and have
observed, and contrarily, widespread aortic nodal metastases helped surgeons observe pelvic drainage using a modern
without pelvic nodal involvement were also observed.3 approach. The method identifes nodal areas at risk, avoids
Taken as a whole, this body of research rationalized the removal of less likely affected nodal basins, and ultimately,
importance of lymph node evaluation in endometrial cancer. increases surgical precision for this disease. The frst such
Since then, researchers have sought to understand the risk of study of selective lymph node biopsy in endometrial cancer
nodal metastases development and the relative incidence based was conducted by Burke et al. in 1996. The authors described
on nodal basin involvement, an outgrowth of this early work. the feasibility of using a fundal injection of blue dye to target
In 1989, Ayhan et al.6 published a key paper on nodal metas- select nodes for biopsy in 15 patients with high-grade endome-
tasis in clinical stage I endometrial cancer, noting a 15% rate trial cancer.19 Only many years later was the concept of SLN
of pelvic and 9% rate of paraaortic nodal metastasis in a series mapping for endometrial cancer revived. Some SLN mapping
of 106 patients. They also observed that specifc uterine risk studies have annotated the specifc locations of SLNs. Jewell
factors, including increasing tumor grade, presence of lymph- et al. reported on 217 surgically staged patients with endome-
vascular space invasion, increasing depth of myometrial inva- trial cancer of various histologies. The majority of SLNs were
sion, and lower uterine segment involvement, correlated with located in the external and internal iliac nodal basins (63%),
an increasing incidence of pelvic and paraaortic nodal metas- and the remaining nodes were in the obturator, common iliac,
tases. This work has been recapitulated multiple times in other and aortic nodal basins.20 The prospective FIRES trial ana-
studies showing the same relationship between nodal metasta- lyzed surgically staged patients with endometrial cancer and
ses and these uterine risk factors.7–10 found that the external iliac (38%) and obturator (25%) nodal
Mariani et al. examined routes of lymphatic spread in 112 basins were the most common locations for SLNs, followed by
surgically staged patients with clinical stage I endometrial the paraaortic (below the IMA), internal iliac, and presacral
cancer and reported that external iliac lymph nodes, fol- nodal basins.21
lowed by obturator lymph nodes, were the most commonly Cervical tracer injection for SLN mapping in endometrial
involved nodes across all patients assessed.11 In patients with cancer has become the most popular injection site, as it is
tumors involving the cervix, there was a signifcant increase in effective in mapping pelvic lymphatics and is easy to adopt
common iliac nodal metastases. The authors found paraaor- and perform in a consistent manner. Injecting this portion
tic nodal metastases in 35 patients (31%), and all but 3 (9%) of the uterus is an approach supported by data from the nec-
had concurrent pelvic nodal metastases. Based on their data, ropsy studies mentioned earlier. A cervical injection allows for
Mariani and colleagues concluded that paraaortic nodal dis- tracer uptake within the predominant drainage channels of the
semination occurs via the common iliac nodes when the cer- corpus, although this practice has been criticized due to less
vix is involved. When the cervix is not involved, paraaortic frequent tracer uptake in the aortic nodal basins.22,23
nodal dissemination occurs via routes shared by the external A cervical injection of dye leads to two primary drainage
iliac and obturator lymph nodes, an uncommon event. routes, as described in historical anatomic textbooks and
Matsumoto et al.12 compared the lymphatic spread of 27 confrmed by live intraoperative demonstrations reported by
endometrial, 25 cervical, and 58 ovarian node-positive cancers Abu-Rustum and the Memorial Sloan Kettering Cancer Center
and described their major drainage pathways. Endometrial group. The most common route emerges from the lateral uter-
cancers metastasized to both pelvic (primarily iliac and obtu- ine body and crosses over the obliterated umbilical ligament
rator) and aortic (primarily above the inferior mesenteric artery to drain into external iliac and obturator lymph nodes (upper
[IMA]) nodal basins, although pelvic nodal metastasis was the or ventral paracervical pathway). The less common pathway
dominant pattern, with few direct aortic metastases (7%). This travels within the mesoureter cephalad to drain common
was in contrast to ovarian cancer, which metastasized to pelvic iliac and presacral lymph nodes (lower or dorsal paracervical
and aortic nodal basins equally. Cervical cancers metastasized pathway).24 Figures 5.2 and 5.3 illustrate the two major pelvic
primarily to pelvic nodal basins. SLN mapping patterns following a cervical injection of dye in
Research has demonstrated the prognostic signifcance of endometrial cancer. In 2017, Geppert and colleagues published
nodal spread in endometrial cancer;13–15 however, locating a their work with mapping uterine lymphatics after either a fun-
positive lymph node can be challenging. Metastatic lymph dal or cervical tracer injection and confrmed these observa-
nodes are often only detected microscopically, the lymphatic tions, noting the lower paracervical drainage pathway was
drainage of the corpus is complex, and the presence and loca- more common.25 Interesting work by Bollino et al.26 demon-
tion of nodal metastases can be infuenced by tumor location, strated high-risk endometrial cancers, as opposed to low-risk
32 Clinical Lymphatic Mapping

FIGURE 5.2 Most common pelvic sentinel lymph node mapping pattern following a cervical injection of dye in endometrial cancer. Lymphatic chan-
nels condense in the paracervix and emerge from the lateral uterine body and cross over the obliterated umbilical ligament to drain into the external
iliac and obturator lymph nodes. This is also termed the upper or ventral paracervical pathway.

FIGURE 5.3 Less common pelvic sentinel lymph node mapping pattern following a cervical injection of dye in endometrial cancer. Lymphatic chan-
nels condense in the paracervix but do not cross over the obliterated umbilical ligament. Rather, these channels travel cephalad within the meso-ureter
to drain common iliac and presacral lymph nodes. This is also termed the lower or dorsal paracervical pathway.
Anatomy: lymphatics of the uterus 33

subtypes, more frequently drained via the upper paracervical early-stage endometrial carcinoma: randomized clinical
pathway. For a comprehensive discussion on SLN mapping in trial. J Natl Cancer Inst. 2008;100(23):1707–16.
endometrial cancer, see Chapter 13. 14. Barton DP, Naik R, Herod J. Effcacy of systematic pel-
vic lymphadenectomy in endometrial cancer (MRC
ASTEC Trial): a randomized study. Int J Gynecol Cancer.
REFERENCES 2009;19(8):1465.
1. Blackwell PM, Fraser IS. Superfcial lymphatics in the 15. Yokoyama Y, Maruyama H, Sato S, et al. Indispensability
functional zone of normal human endometrium. Microvasc of pelvic and paraaortic lymphadenectomy in endometrial
Res. 1981;21(2):142–52. cancers. Gynecol Oncol. 1997;64(3):411–17.
2. Henriksen E. The lymphatic spread of carcinoma of the cer- 16. Hirahatake K, Hareyama H, Sakuragi N, et al. A clinical
vix and of the body of the uterus. A study of 420 necropsies. and pathologic study of para-aortic lymph node metastasis
Am J Obstet Gynecol. 1949;58(5):924–42. in endometrial carcinoma. J Surg Oncol. 1997;65(2):82–7.
3. Henriksen E. The lymphatic dissemination in endome- 17. Chuang L, Burke TW, Tornos C, et al. Staging laparotomy
trial carcinoma. A study of 188 necropsies. Am J Obstet for endometrial carcinoma: assessment of retroperitoneal
Gynecol. 1975;123(6):570–6. lymph nodes. Gynecol Oncol. 1995;58(2):189–93.
4. Plentl AA, Friedman EA. Lymphatics of the cervix uteri. 18. Larson DM, Johnson KK. Pelvic and para-aortic lymph-
In: Lymphatic System of the Female Genitalia: The adenectomy for surgical staging of high-risk endometri-
Morphologic Basis of Oncologic Diagnosis and Therapy. oid adenocarcinoma of the endometrium. Gynecol Oncol.
Philadelphia: WB Saunders; 1971. pp. 75–115. 1993;51(3):345–8.
5. Lecuru F, Neji K, Robin F, et al. Drainage lymphatique de 19. Burke TW, Levenback C, Tornos C, et al. Intraabdominal
l’uterus. Resultats preliminaires d’une etude experimentale. lymphatic mapping to direct selective pelvic and para-
J Gynecol Obstet Biol Reprod (Paris). 1997;26(4):418–23. aortic lymphadenectomy in women with high-risk endo-
6. Ayhan A, Yarali H, Urman B, et al. Lymph node metastasis metrial cancers: results of a pilot study. Gynecol Oncol.
in early endometrium cancer. Aust N Z J Obstet Gynaecol. 1996;62(2):169–73.
1989;29(3 Pt 2):332–5. 20. Jewell EL, Huang JJ, Abu-Rustum NR, et al. Detection of
7. Creasman WT, Morrow CP, Bundy BN, et al. Surgical patho- sentinel lymph nodes in minimally invasive surgery using
logic spread patterns of endometrial cancer. A Gynecologic indocyanine green and near-infrared fuorescence imag-
Oncology Group Study. Cancer. 1987;60(8 Suppl):2035–41. ing for uterine and cervical malignancies. Gynecol Oncol.
8. Chi DS, Barakat RR, Palayekar MJ, et al. The incidence of 2014;133(2):274–7.
pelvic lymph node metastasis by FIGO staging for patients 21. Rossi EC, Kowalski LD, Scalici J, et al. A comparison
with adequately surgically staged endometrial adenocar- of sentinel lymph node biopsy to lymphadenectomy for
cinoma of endometrioid histology. Int J Gynecol Cancer. endometrial cancer staging (FIRES trial): a multicen-
2008;18(2):269–73. tre, prospective, cohort study. Lancet Oncol. 2017;18(3):
9. Creasman WT, Ali S, Mutch DG, et al. Surgical- 384–92.
pathological fndings in type 1 and 2 endometrial cancer: 22. Rossi EC, Jackson A, Ivanova A, et al. Detection of sentinel
an NRG Oncology/Gynecologic Oncology Group study on nodes for endometrial cancer with robotic assisted fuores-
GOG-210 protocol. Gynecol Oncol. 2017;145(3):519–25. cence imaging: cervical versus hysteroscopic injection. Int
10. Mueller JJ, Nobre SP, Braxton K, et al. Incidence of pel- J Gynecol Cancer. 2013;23(9):1704–11.
vic lymph node metastasis using modern FIGO staging and 23. Rossi EC. Current state of sentinel lymph nodes for
sentinel lymph node mapping with ultrastaging in surgi- women with endometrial cancer. Int J Gynecol Cancer.
cally staged patients with endometrioid and serous endo- 2019;29(3):613–21.
metrial carcinoma. Gynecol Oncol. 2020;157(3):619–23. 24. Abu-Rustum NR. Sentinel lymph node mapping for endo-
https://doi.org/10.1016/j.ygyno.2020.03.025. metrial cancer: a modern approach to surgical staging. J
11. Mariani A, Webb MJ, Keeney GL, et al. Routes of lym- Natl Compr Canc Netw. 2014;12(2):288–97.
phatic spread: a study of 112 consecutive patients with 25. Geppert B, Lonnerfors C, Bollino M, et al. A study on uter-
endometrial cancer. Gynecol Oncol. 2001;81(1):100–4. ine lymphatic anatomy for standardization of pelvic senti-
12. Matsumoto K, Yoshikawa H, Yasugi T, et al. Distinct nel lymph node detection in endometrial cancer. Gynecol
lymphatic spread of endometrial carcinoma in compari- Oncol. 2017;125(2):256–61.
son with cervical and ovarian carcinomas. Cancer Lett. 26. Bollino M, Geppert B, Lonnerfors C, et al. Pelvic senti-
2002;180:83–9. nel lymph node biopsy in endometrial cancer: a simplifed
13. Benedetti Panici P, Basile S, Maneschi F, et al. Systematic algorithm based on histology and lymphatic anatomy. Int J
pelvic lymphadenectomy vs. no lymphadenectomy in Gynecol Cancer. 2020;30(3):339–45.
6
Lymphatic anatomy: lymphatics of the ovary

Ate G.J. van der Zee

CONTENTS
Introduction .................................................................................................................................................................................... 35
Lymphatic anatomy........................................................................................................................................................................ 35
Distribution of pelvic and aortic lymph node metastasis in patients with ovarian cancer ............................................................. 35
Sentinel node detection in ovarian cancer...................................................................................................................................... 36
References...................................................................................................................................................................................... 37

Introduction large collecting trunks that leave the ovarian hilus within the
mesovarium to form the subovarian plexus, which is composed
Comparable to the other major gynecologic malignancies, of efferent trunks from the uterine corpus, fallopian tube, and
information on the lymphatics of the ovary can be obtained ovary. Mixing of lymph that has originated from each organ
from three different types of studies: 1) descriptive anatomy does occur. Efferent vessels from this plexus drain in a cepha-
reports in which the lymphatic anatomy is studied with or lad direction along with the ovarian blood vessels to terminate
without previous injection of different types of dyes; 2) studies in the aortic nodes. In about 25% of women, a collateral trunk
on the distribution of pelvic and aortic lymph node metastases exists which bypasses the subovarian plexus. It reaches the
in patients with early-stage ovarian cancer who underwent pel- pelvic wall within the folds of the broad ligament, terminating
vic and aortic lymph node evaluation; and 3) studies in which in the uppermost interiliac nodes.3
lymphatic mapping by injection of a tracer has been performed
in patients with early-stage ovarian cancer. However, in com-
parison to cervical, endometrial, and vulvar cancer, the num-
ber of this third category of studies in ovarian cancer is very Distribution of pelvic and aortic lymph node
limited. This lack of studies on lymphatic mapping may well
metastasis in patients with ovarian cancer
be due to the fact that lymphogenous pathways are not always
of primary relevance in the dissemination of ovarian cancers. When one looks at ovarian cancer, pelvic and aortic nodes
The most common and earliest mode of dissemination is by could also be primary and sentinel ones, in view of the fre-
exfoliation of cells that implant along the surfaces of the peri- quent observations of pelvic and aortic lymph node metastasis
toneal cavity. However, in 1985, the International Federation alone in patients with ovarian cancer. In a study by Matsumoto
of Gynecologists and Obstetricians (FIGO) modifed the stag- et al., in which systematic pelvic and aortic lymph node dissec-
ing for ovarian cancer, in part to refect the prognostic signif- tion was performed, the incidence of aortic lymph node metas-
cance of metastatic spread to the pelvic or paraaortic lymph tasis alone in node-positive ovarian cancer patients (all FIGO
nodes.1 Especially in patients with disease macroscopically stages) was 21%,4 a fgure which is consistent with fgures pre-
confned to one of the ovaries, survival decreases signifcantly viously reported (15%–55%).5–8 Overall, these data show that
when retroperitoneal extension is found, thereby emphasizing direct metastasis to the aortic lymph nodes frequently occurs
the need for proper evaluation of the lymph nodes, and con- in ovarian cancer. Matsumoto and colleagues also showed that
sequently justifying more aggressive adjunctive therapy when metastasis at both pelvic and aortic lymph nodes is common
positive nodes are found.2 in ovarian cancer, again in keeping with previous studies in
ovarian cancer. However, the incidence of aortic lymph node
metastasis was not related to the incidence of positive common
iliac and sacral nodes, a subgroup which could be assumed
Lymphatic anatomy to be affected when aortic lymph node metastasis spreading
The tunica albuginea of the ovary is essentially devoid of lym- from pelvic nodes occurs. In combination with an incidence
phatics, while the parenchymal lymphatics comprise a rich of pelvic lymph node metastasis alone in patients with ovarian
network, whose presence is related to developing follicles. cancer of 17%, these data clearly indicate two separate ways of
Lymph from the ovary is drained peripherally by six to eight lymphatic spread in ovarian cancer. Therefore, it appears that

DOI: 10.1201/9781003255536-6 35
36 Clinical Lymphatic Mapping

30% of patients with presumed early-stage cancer will expe-


rience an upstaging of the disease after comprehensive lym-
phatic and peritoneal staging.17 Pelvic and paraaortic lymph
node dissection is therefore part of a comprehensive surgical
staging in early disease to defne the accurate stage that will
have signifcant implications on the choice and indication of
the adequate adjuvant systemic treatments. The incidence
of lymph node metastasis in apparent early-stage epithelial
ovarian cancer (EOC) (all histologic grades) is estimated to
be around 14%–15%;18–19 approximately 35% of patients have
only pelvic-positive nodes, 37% have only paraaortic-positive
nodes, and the remaining 28% have both pelvic and paraaor-
tic involvement.20 In particular, patients with apparent early-
stage cancer might be candidates for sentinel node detection
in order to obviate the side effects associated with full pelvic
and paraaortic lymphadenectomy in patients with, in principle,
a low likelihood of metastatic lymph nodes. A recent review
indicates a frequency of approximately 3% metastatic nodes in
these patients, which stresses the need for alternative staging
techniques, i.e., sentinel node detection rather than full lymph-
adenectomy in these patients.21
As already mentioned in the introduction of this chapter,
limited data are available on lymphatic mapping in ovarian
cancer. Until recently, only retrospective studies on small
series of patients with a variety of tracers and injection sites
were published. From the available retrospective literature,
the most promising technique seems to be injection into the
infundibulopelvic, as well as the proper ovarian ligament.
Indocyanine green seems to be an excellent tracer for suc-
FIGURE 6.1 Lymphatic drainage of the ovary and fallopian tube. cessful sentinel node detection of ovarian tumors, which can
(Modifed from Plentl and Friedman.3)
be used during laparoscopic or robotic surgery. The detec-
tion rates and true positive rates of studies support further
investigation of the technique (for reviews, see Refs. 22 and
metastasis in ovarian cancer may take two separate routes: one 23). In a prospective phase II study, Lago et al. recently
via the broad ligament to the internal and external iliac and showed the feasibility and safety of sentinel node detec-
obturator lymph nodes and one via the infundibulopelvic liga- tion for ovarian cancer staging. In their protocol 99mTc and
ment to the aortic lymph nodes (Figure 6.1). indocyanine green were injected intraoperatively (either at
The pattern of lymphatic spread in ovarian cancer macro- laparotomy or laparoscopy) into both the utero-ovarian and
scopically confned to one ovary is controversial with respect infundibulopelvic ligament stump.24 The Sentinel Lymph
to whether contralateral lymph node metastases do occur. In Nodes in Early-Stage Ovarian Cancer trial (SELLY) is an
a retrospective study in this category of patients, Cass et al. ongoing prospective, phase II, single-arm study that includes
found positive pelvic nodes alone in 50%, positive aortic patients with presumed stages I–II epithelial ovarian cancer
nodes alone in 36%, and both in 14% of cases, while iso- planned for immediate or delayed minimally invasive com-
lated contralateral lymph node metastases were seen in 4%.9 prehensive staging. An interim analysis on the frst 31 patients
Comparable low fgures have also been reported by others,10–13 was recently published and showed that the detection rate of
while several studies did not fnd contralateral metastases.14,15 the sentinel node in early-stage ovarian cancer is low when
Based on the lymphatic anatomy, it is hard to imagine a way patients are submitted to delayed-staging surgery. However,
of contralateral spread. All the studies that have reported con- the interim analysis also shows that the sentinel node proce-
tralateral metastasis were retrospective, and it may well be dure with injection of indocyanine green in the ovarian ped-
that a lesion such as occult disease in the ipsilateral ovary has icle is feasible, but technically challenging. Preliminary data
been missed. on its diagnostic accuracy indicate that the sentinel node tech-
nique with intraoperative indocyanine green has the potential
to provide reliable and useful information on nodal status and
may allow the avoidance of systematic lymphadenectomy in
Sentinel node detection in ovarian cancer
the majority of apparent early-stage patients.25 However, more
More than 20% of patients with epithelial ovarian cancer will mature data from this trial and others need to be awaited and
be diagnosed at an apparent early stage (I–II), with the dis- until then the application of the sentinel node technique in
ease macroscopically confned to the pelvis.16 Attributed to ovarian cancer patients should only be used within the protec-
the extrapelvic vascular supply of the adnexa, approximately tion of clinical trials.
Anatomy: lymphatics of the ovary 37

REFERENCES 14. Burghardt E, Girardi F, Lahousen M, et al. Patterns of pel-


vic and paraaortic lymph node involvement in ovarian can-
1. FIGO. Changes in gynecologic staging by the International
cer. Gynecol Oncol. 1991;40:103–6.
Federation of Gynecologists and Obstetricians. Am J Obstet
15. Benedetti Panici P, Greggi S, Maneschi F, et al. Anatomical
Gynecol. 1990;162:610–11.
and pathological study of retroperitoneal nodes in epithelial
2. Carnino F, Fuda G, Ciccone G, et al. Signifcance of
ovarian cancer. Gynecol Oncol. 1993;51:150–4.
lymph node sampling in epithelial carcinoma of the ovary.
16. Young RC, Walton LA, Ellenberg SS, et al. Adjuvant ther-
Gynecol Oncol. 1997;65:467–72.
apy in stage I and stage II epithelial ovarian cancer. Results
3. Plentl A, Friedman E. Lymphatic System of the Female
of two prospective randomized trials. N Engl J Med.
Genitalia. Vol. 2. Philadelphia, PA: WB Saunders; 1971.
1990;322:1021–7.
4. Matsumoto K, Yoshikawa H, Yasugi T, et al. Distinct lym-
17. Timmers PJ, Zwinderman K, Coens C, et al. Lymph node
phatic spread of endometrial carcinoma in comparison with
sampling and taking of blind biopsies are important ele-
cervical and ovarian carcinomas. Cancer Lett. 2002;180:83–9.
ments of the surgical staging of early ovarian cancer. Int J
5. Chen SS, Lee L. Incidence of para-aortic and pelvic lymph
Gynecol Cancer. 2010;20:1142–7.
node metastases in epithelialcarcinoma of the ovary.
18. Berek JS, Crum C, Friedlander M. Cancer of the ovary,
Gynecol Oncol. 1983;16:95–100.
fallopian tube, and peritoneum. Int J Gynaecol Obstet.
6. Onda T, Yoshikawa H, Yokota H, et al. Assessment of
2012;119(Suppl 2):S118–29.
metastases to aortic and pelvic lymph nodes in epithelial
19. Ledermann JA, Raja FA, Fotopoulou C, et al. Newly diag-
ovarian carcinoma. A proposal for essential sites for lymph
nosed and relapsed epithelial ovarian carcinoma: ESMO
node biopsy. Cancer. 1996;78:803–8.
Clinical Practice Guidelines for diagnosis, treatment and
7. Sakai K, Kamura T, Hirakawa T, et al. Relationship
follow-up. Ann Oncol. 2013;24(Suppl 6):vi24–32.
between pelvic lymph node involvement and other dis-
20. Ledermann JA, Luvero D, Shafer A, et al. Gynecologic
ease sites in patients with ovarian cancer. Gynecol Oncol.
Cancer InterGroup (GCIG) consensus review for mucinous
1997;65:164–8.
ovarian carcinoma. Int J Gynecol Cancer. 2014;24(9 Suppl
8. Tsumura N, Sakuragi N, Hareyama H, et al. Distribution
3):S14–19.
pattern and risk factors of pelvic and para-aortic lymph
21. Lago V, Minig L, Fotopoulou C. Incidence of lymph node
node metastasis in epithelial ovarian carcinoma. Int J
metastases in apparent early-stage low-grade epithelial
Cancer. 1998;79:526–30.
ovarian cancer: a comprehensive review. Int J Gynecol
9. Cass I, Li AJ, Runowicz CD, et al. Pattern of lymph node
Cancer. 2016;26:1407–14.
metastases in clinically unilateral stage I invasive epithelial
22. Uccella S, Gisone B, Ghezzi F. Laparoscopic senti-
ovarian carcinomas. Gynecol Oncol. 2001;80:56–61.
nel node detection with ICG for early ovarian cancer:
10. Wu P, Qu J, Lang J, et al. Lymph node metastasis of ovarian
description of a technique and literature review. EJOGR.
cancer: a preliminary survey of 74 cases of lymphadenec-
2018;221:193–4.
tomy. Am J Obstet Gynecol. 1986;155:1103–8.
23. Mach P, Kimmig R, Buderath P. The role of sentinel-
11. Petru E, Lahousen M, Tamussino K, et al. Lymphadenec-
node biopsy in ovarian cancer. Minerva Ginecol.
tomy in stage I ovarian cancer. Am J Obstet Gynecol.
2020;72(6):399–403.
1994;170:656–62.
24. Lago V, Bello P, Montero B, et al. Sentinel lymph node
12. Onda T, Yoshikawa H, Yasugi T, et al. Patients with ovar-
technique in early-stage ovarian cancer (SENTOV):
ian carcinoma upstaged to stage III after systematic lymph-
a phase II clinical trial. Int J Gynecol Cancer.
adenectomy have similar survival to stage I/II patients
2020;30(9):1390–6.
and superior survival to other stage III patients. Cancer.
25. Uccella S, Nero C, Vizza E, et al. Sentinel-node biopsy in
1998;83:1555–60.
early-stage ovarian cancer: preliminary results of a pro-
13. Walter AJ, Magrina JF. Contralateral pelvic and aortic
spective multicentre study (SELLY). Am J Obstet Gynecol.
lymph node metastasis in clinical stage I epithelial ovarian
2019;221(4):324.e1–324.e10.
cancer. Gynecol Oncol. 1999;74:128–9.
7
Lymphatic anatomy: lymphatics of the breast and axilla

James V. Fiorica

CONTENTS
References...................................................................................................................................................................................... 42

Lymphatic mapping of the breast is altering the long-standing artery. It has three branches, which are the anterior and pos-
approach to the breast cancer treatment model: radical mas- terior humoral circumfex arteries and the subscapular artery.
tectomy with complete axillary lymphadenectomy. It is a dra- The largest of the axillary branches is this subscapular artery.
matic departure from Halsted’s modality of 100 years ago.1 This artery branches into the subscapular circumfex and the
The status of the axillary lymph node has consistently been thoracodorsal arteries, which are associated with the central
shown to be the most signifcant prognostic factor in patients and subscapular lymph nodes, which are described in more
with breast cancer.2 The breast lies within the superfcial fascia detail later in this chapter.
of the anterior thoracic wall. It is situated between the second The internal mammary artery medially (65%) and the lat-
and sixth ribs and the sternal edge and midaxillary line. The eral thoracic artery (35%) supply blood to the breast (Figure
posterior surface of the breast ends abruptly at the chest wall, 7.1). The cephalic vein serves as a landmark separating the
where it reaches the pectoralis major fascia. It is composed of pectoralis major muscle from the deltoid muscle. The vein
skin, parenchyma, and stroma. The stroma and connective tis- travels through the deltopectoral triangle and pierces the cla-
sue are intertwined with blood vessels, nerves, and lymphatics. vipectoral fascia, joining the axillary vein. Branches of the
Beneath the nipples are fve to ten milk ducts which connect to brachial plexus are located throughout the course of the axilla.
fve to ten additional ducts, each draining an individual breast The long thoracic nerve is located on the medial wall of the
lobe. Each lobe is composed of 20–40 lobules, which in turn axilla, arising in the neck from the ffth, sixth, and seventh
connect to 10–100 tubulosaccular units called alveoli. The roots of the brachial plexus. It innervates the serratus anterior
subcutaneous connective tissue surrounds glands and extends muscle, which permits raising the arm above the shoulder. The
as septa between the lobes and lobules, providing support for intercostobrachial nerve is the lateral cutaneous branch of the
the glandular elements. Cooper’s ligaments are suspensory second intercostal nerve and the joining of the medial cutane-
structures that insert perpendicular to the dermis. ous nerve of the arm, supplying the skin of the foor of the
The pectoralis major muscle is a fan-shaped muscle with two axilla and the upper medial aspect of the arm.
divisions. The clavicular division originates from the clavicle The lymphatic system of the breast is just as intricate and
and can be easily distinguished from the larger costosternal complicated. The subaerolar lymphatic plexus of Sappey com-
division that originates from the sternum and costal cartilage municates to the vertical lymphatics in a subepithelial and
of ribs 2–6. These fbers converge on the greater tubercle of subdermal manner. The unidirectional lymphatics fow from
the humerus. The pectoralis minor muscle is located beneath this superfcial subaerolar plexus through the lactiferous duct
the pectoralis major muscle and arises from the external sur- lymphatics to the perilobar and deep subcutaneous lymphatic
face of ribs 2–5. The posterior suspensory ligaments extend plexus. This deep lymphatic plexus drains in a centrifugal
from the deep surface of the breast to the deep pectoral fascia. direction to the axillary nodes (97%) and the internal mam-
The subscapular muscle arises from the frst rib near the costo- mary nodes (3%). The axillary nodes are divided into six
chondral junction and extends laterally to insert on the inferior groups: the lateral (axillary) group lies medial and posterior
surface of the clavicle. to the axillary vein (four to six nodes), the external mammary
The axillary sheath extends from the neck to the axilla and group (anterior or pectoral) lies along the lower border of the
is surrounded by a layer of fascia. This sheath contains the pectoralis minor muscle adjacent to the lateral thoracic ves-
great vessels and nerves of the upper extremities. The axillary sels (four to fve nodes), the subscapular (posterior) group lies
artery is divided into three parts. The frst segment, located along the posterior wall of the axilla at the lateral border of the
medial to the pectoralis minor muscle, has the supreme tho- scapula (six to seven nodes), and the central group is embed-
racic branch. This segment supplies the frst and second inter- ded in the fat of the axilla behind the pectoralis minor muscle
costal spaces. The second segment, located posterior to the and is the common shallow palpable group of nodes (three to
pectoralis minor muscle, contains the thoracoacromial trunk four nodes). The apical (subclavicular) nodes are located at
and the lateral thoracic artery branches. Located lateral to the the upper border of the pectoralis minor muscle and receive
pectoralis minor muscle is the third segment of the axillary lymph from all areas (6–12 nodes). Two additional pathways

DOI: 10.1201/9781003255536-7 39
40 Clinical Lymphatic Mapping

FIGURE 7.1 Primary blood supply to the breast.

FIGURE 7.2 Primary lymph nodes of the breast and axilla.

are the transpectoral and the retropectoral (Rotter’s nodes). The internal mammary nodes are located in the retrosternal
The Rotter’s interpectoral group is located between the pecto- interspaces within 2 cm of the sternal margin and transverse
ralis major and minor muscles (one to four nodes) and passes the internal mammary vessels. The right internal mammary
to the central and apical nodes directly. Prepectoral nodes nodes enter the internal mammary lymphatic trunk, which
refer to single lymph nodes, which are found in the subcutane- terminates in the subclavicular nodal group. The left internal
ous tissue within the breast (Figure 7.2). mammary chain enters the main thoracic duct.3 The surgeon
Lymphatics of the breast and axilla 41

TABLE 7.1 and tumor biology in the current era of more conservative sur-
Classifcation of Lymph Nodes Draining the Breast gery and newer systemic neoadjuvant therapies. The risk of
metastases and death increases with both breast cancer size at
Nodal group Anatomic name Level Number of nodes
detection and number of axillary lymph nodes involved.9 The
Axillary External mammary I 4–5 breast lymphatic level approach is a more focused and less rad-
Subscapular I 6–7 ical dissection to remove the disease.10 Additionally, sentinel
Lateral I 4–6 node biopsy will reduce complications from more extensive
Central II 3–4 surgical procedures or adjuvant therapies. If women do have
Rotter’s II 1–4 micrometastasis in the supraclavicular nodes, they are classi-
Apical III 6–12 fed as having stage IV disease.
Internal mammary – 6–8
With proper injection techniques and timing, certain
anatomic landmarks are helpful for identifying the sentinel
Prepectoral nodes – 1
node. If one draws a line from the lateral border of the pec-
Deltopectoral – 1–2
toralis major muscle and another at the lateral border for the
Supraclavicular (Stage IV) – 2–3
latissimus dorsi muscle in the axilla, these lines will outline
the outer borders of the axillary limits for dissection. A tan-
gential line is drawn at the axillary hairline in a perpendicu-
should be familiar with the anatomy of the breast’s lym- lar, anterior-to-posterior direction. Another line can then be
phatic system to avoid false-negative results of sentinel nodes. drawn through the axis of the axilla, through the center point
Patients can be undertreated with poor oncologic outcomes if of the hairline. The intersecting lines mark the center of a
high false-negative rates occur.4 5-cm circle on the axilla. Ninety-four percent of the SLNs
The level of nodes is often described as level I, II, or III are within this circle. The remaining 6% are in the level II
in relationship to the pectoralis minor muscle. Nodal tissue location (Figure 7.3). Once the SLN is identifed on the skin,
lateral to the minor muscle is known as level I (external mam- an accurate incision may be made, overlying the area of high-
mary, lateral, and subscapular). Those lymphatics beneath est activity with the gamma probe. Internally, the anatomic
the muscle are level II (central ± subscapular), and the lymph landmarks are the central axillary vein and the third branch
nodes on the medial edge of the pectoralis minor muscle are
known as level III (apical) lymph nodes. During the standard
dissection, level I and a portion of level II nodes are removed5
(Table 7.1).
Advocates of axillary dissection contend that this method
renders regional control of axillary disease. Since the time of
Halsted, the status of the regional nodal basin has remained
the single most important variable in predicting prognosis.6
Recent thinking supports the idea that microscopic disease
may be cured with adjuvant chemotherapy with or without
node dissection. Some physicians advocate abandoning the
axillary lymph node dissection. The development of intra-
operative lymphatic mapping and selective lymphadenectomy
has permitted the mapping of lymphatic fow from the primary
tumor and the identifcation of the sentinel lymph node (SLN)
in the regional basin.
SLN biopsy for breast cancer may obviate the need for rou-
tine axillary node dissections in most node-negative patients
and still provide prognostic information to the patient and cli-
nician. The assignment of levels I, II, and III was relatively
arbitrary and has led to a high (15%) prevalence of skip metas-
tasis.7 Data from the American College of Surgeons Oncology
Group 20011 Trial suggested that patients with even a stage
T1 or T2 tumor along with one or two positive lymph nodes
can be spared axillary lymph node dissection.8 With lym-
phatic mapping, variations of breast lymph drainage have been
apparent. Direct lymphatic drainage from primary sites has
been noted to level II and III axillary nodes, subclavian nodes,
and supraclavicular nodes. More accurate staging can improve
survival by identifying patients who will gain with either the
surgical procedure itself (complete dissection) or the accompa-
nying adjuvant therapy. Tumor size and nodal status still have
a signifcant infuence on overall mortality independent of age FIGURE 7.3 Location of sentinel lymph nodes of the axilla.
42 Clinical Lymphatic Mapping

of the intercostal nerve beneath the clavipectoral fascia. 4. Goel V, Raju K, Dasu S, et al. Awareness of lymphatic sys-
The vein may be found easily, and when the nerve crosses tem to decrease false negative sentinel node rate in breast
over the vein, four quadrants are defned within the circle cancer. Indian J Surg Oncol. 2019;10:673–5.
described earlier. The node is considered an SLN if it is blue 5. Nathanson SD, Wachna DL, Gilman D, et al. Pathways
or has a blue-stained afferent lymphatic vessel leading to it. of lymphatic drainage from the breast. Ann Surg Oncol.
In addition, the node is sentinel if it has an in vivo radioactiv- 2001;8:837–43.
ity (radioactivity in the SLN/neighboring non-SLN) of 3:1 or 6. Heerdt AS. Lymphatic mapping and sentinel lymph node
an ex vivo activity of 10:1.11 biopsy for breast cancer.  JAMA Oncol.  2018;4(3):431.
The mapping techniques vary slightly from center to center. https://doi.org/10.1001/jamaoncol.2017.4000.
However, the results have attained similar high success rates 7. Veronesi U, Rilke F, Luini A, et al. Distribution of axillary
in skilled hands. Sensitivity and diagnostic accuracy rates node metastases by level of invasion. An analysis of 539
have been greater than 95%, with false-negative (skip) rates cases. Cancer. 1987;59:682–7.
8. Marino MA, Avendeno D, Zapata R, et al. Lymph node
of 0%–10%. These results suggest that lymphatic mapping has
imaging in patients with primary breast cancer: concurrent
the potential to change the standards for surgical management
diagnostic tools. Oncologist. 2020;25:e231–42.
of breast cancer.12
9. Saadatmand S, Bretveld R, Siesling S, et al. Infuence of
Newer studies under investigation include a radioactive
tumour stage at breast cancer detection on survival in mod-
infrared activated fducial refection. Preliminary fnd-
ern times: population based study in 173797 patients. BMJ.
ings show that this refective guided excision may more
2015;351:h4901–10.
accurately refect the status of the axilla after neoadjuvant 10. Yuan Q, Hou J, He Y, et al. Minimize the extent and
chemotherapy.13 morbidity of axillary dissection for node-positive breast
cancer patients: implementation of axillary lymph node
dissection based on breast lymphatic level. BMC Cancer.
REFERENCES 2021;21:293–302.
1. Chua B, Ung O, Boyages J. Competing considerations in 11. Cox C. Techniques for lymphatic mapping in breast carci-
regional nodal treatment for early breast cancer. Breast J. noma. In: Whitman D, Reintgen D, editors, Radioguided
2002;8:15–22. Surgery. Austin, TX: Landes Biosciences; 1999. pp. 72–82.
2. Manca G, Rubello D, Tardelli E, et al. Sentinel lymph node 12. Reintgen M, Kerivan L, Reintgen E, et al. Breast lymphatic
biopsy in breast cancer: indications, contraindications, and mapping and sentinel lymph node biopsy: state of the art
controversies. Clin Nucl Med. 2016;41:126–33. 2015. Clin Breast Cancer. 2016;16:155–65.
3. Bland K, Klimberg V, Copeland EM, et al.  Anatomy of 13. Taback B, Jadeja P, Ha R. Enhanced axillary evaluation
the breast, axilla, chest wall, and related metastatic sites. using refective-guided sentinel lymph node biopsy: a pro-
In: The Breast Comprehensive Management of Benign spective feasibility study and comparison with controver-
and Malignant Diseases. Philadelphia, PA: Elsevier; 2018. sial lymphatic mapping techniques. Clin Breast Cancer.
pp. 20–36. 2018;18:e869–74.
8
Modalities of detection of sentinel nodes in lymphatic mapping

Blanca Segarra, Nuria Agusti, and Pedro T. Ramirez

CONTENTS
Introduction.................................................................................................................................................................................... 43
Mapping tracers ............................................................................................................................................................................. 43
Blue dye agents .................................................................................................................................................................... 44
Colloids and lymphoscintigraphy......................................................................................................................................... 44
Indocyanine green ................................................................................................................................................................ 45
Empty lymph node packet syndrome ...................................................................................................................... 45
Technique by disease site............................................................................................................................................................... 46
Vulvar cancer........................................................................................................................................................................ 46
Sentinel lymph node technique................................................................................................................................ 46
Endometrial cancer............................................................................................................................................................... 47
Dose of ICG on the SLN mapping................................................................................................................................................. 48
Cervical cancer..................................................................................................................................................................... 48
Ovarian cancer...................................................................................................................................................................... 49
Safety.................................................................................................................................................................................... 49
Blue dye................................................................................................................................................................... 49
Technetium .............................................................................................................................................................. 49
Indocyanine green.................................................................................................................................................... 49
Mapping in special settings .................................................................................................................................................. 50
Pregnancy ................................................................................................................................................................ 50
Obesity..................................................................................................................................................................... 50
Novel modalities ............................................................................................................................................................................ 50
Intraoperative identifcation techniques ............................................................................................................................... 50
[99mTechnetiumTc] tilmanocept ............................................................................................................................... 50
Carbon nanoparticles............................................................................................................................................................ 50
India ink................................................................................................................................................................... 51
Superparamagnetic iron........................................................................................................................................... 51
Imaging-enhanced modalities .............................................................................................................................................. 51
Contrast-enhanced ultrasound ................................................................................................................................. 51
Conclusion ..................................................................................................................................................................................... 51
References...................................................................................................................................................................................... 51

Introduction Mapping tracers


The landscape of lymphatic mapping tracers has evolved The three conventional tracers currently used in gyneco-
over the years, with broader expansion in the feld of gyneco- logic surgery are technetium-99m radiocolloid (99mTc), vari-
logic oncology. Newer substances are associated with higher ous blue dyes, and indocyanine green (ICG). Historically,
detection rates, improved side effect profles, and lower cost. the SLN detection was usually performed by peritumoral
Mapping substances range in their abilities and use, given their or intratumoral injection of a tracer. In most tumors, blue
different compositions. The ideal mapping substance should dye associated with 99mTc-nanocoloid albumin has been the
have a high detection rate with the least possible cost and num- most commonly used agent in gynecologic cancer because
ber of side effects. In the setting of sentinel lymph node (SLN) this combination increases detection rates.1 Recently, ICG, a
mapping, this would entail a low false-negative rate (negative nonradioactive tracer based on fuorescence, detectable by its
SLNs on pathology in the setting of a positive lymph node light emission after excitation with a specifc wavelength, has
in the remainder of the lymph node dissection) to minimize been used more commonly, becoming the standard tracer in
undertreatment of patients. many institutions.

DOI: 10.1201/9781003255536-8 43
44 Clinical Lymphatic Mapping

Blue dye agents • Low patient-absorbed radiation dose, minimiz-


ing patient exposure to doses of harmful radiation.
The two substances most commonly used as blue dyes are Although the half-life is relatively short, the dose is
methylene blue and isosulfan blue (sometimes referred to as enough to detect positive nodes more than 24 hours
patent blue). The substances are functionally interchangeable post-injection.
when used as biotracers and with equal effcacy; however,
• Low cost per patient doses. The availability of
their different chemical structures explain critical differences
radioisotope-based medicines, as well as the costs
between the elements. Methylene blue is an organic chloride
of production and research, transportation, and stor-
salt with a molecular mass of 319.8 g/mol. It may not be an
age, signifcantly infuence use. Generator-produced
ideal tracer because it is very poorly absorbed when injected
radioisotopes are still the most commonly used.
intradermally and easily diffuses into the surrounding tissue,
Their costs, higher than those of radioisotopes
causing substantial tissue stain without specifc targeting of
obtained in a reactor, are compensated for by their
the sentinel node. However, some claim that among its benefts
wide application.
is the fact that it is cheaper, more readily available, and has a
better side effect profle than isosulfan blue. It should be noted • Widespread commercial availability.
that methylene blue has been associated with higher rates of
anaphylaxis. Another mapping agent in this category is iso- Planar imaging is the most commonly applied preoperative
sulfan blue. It has a molecular mass of 566.7 g/mol and is a imaging for sentinel node identifcation. Lymphoscintigraphy
2,5-disulfonic acid isomer of patent blue. Approximately 50% is a safe, reproducible, and noninvasive technique utiliz-
of isosulfan blue is weakly bound to serum proteins, and it is ing radionuclides to image regional lymph node drainage.1
rapidly transported through the lymphatics after intradermal The lymphatic channels are best appreciated in a composite
injection, making it less likely to diffuse into the surrounding dynamic image from the individual progressive frames. This
tissue. The structure of both blue dyes contains both hydro- process usually takes 20 minutes in total. Delayed scans are
phobic and hydrophilic regions. The hydrophilic areas allow then performed at 2.5–3 hours after injection of the radiocol-
for intravenous administration, as the substances dissolve in loid tracer. These scans should include all node felds that can
water and blood. The hydrophobic regions serve as high-affnity receive drainage from the injection site. Each static acquisition
binding sites for oncotic proteins that circulate throughout the may last 5–10 minutes to guarantee that even very weak senti-
lymphovascular system. Blue dyes can only be seen in the vis- nel nodes are detected.2 Another often used imaging modality
ible light spectrum. This difference contributes to the overall is the single photon emission computed tomography (SPECT)
accessibility and low cost of blue dye technology. overlaid with traditional computed tomography (CT) scan –
SPECT/CT. This tool allows surgeons to determine the ana-
tomic location of sentinel nodes, and the images correlate with
Colloids and lymphoscintigraphy the anatomic location of the sentinel node location. SPECT/
Scintigraphy tracks a radioactive tracer in a living body and CT appears to offer a signifcant improvement in sentinel node
provides a view of the position and concentration of a radioiso- detection and anatomic localization when compared with lym-
tope within the body. 99mTc is responsible for more than 80% of phoscintigraphy because it provides more accurate imaging of
applications in nuclear medicine. Technetium derives its name pathological lesions and enables correction for absorption of
from the Latin word for “artifcial,” as it was the frst artif- radiation in the patient’s body.3
cially formed element. While 99mTc, where “m” stands for a The intraoperative detection of the sentinel node relies not
metastable isotope, can be found in nature, the majority of the only on the visual inspection of the lymphatic basin to iden-
world’s supply is manufactured. 99mTc products have different tify the ‘blue node’ but also on the assessment of the radioac-
biological absorptive properties depending on their oxidative tive colloid in the sentinel node, or ‘hot node,’ through the aid
states. 99mTc-labeled radiopharmaceuticals are used in 85% of a gamma detection device. Detection devices used intra-
of all nuclear medicine procedures because of the following operatively are surgical gamma probes and portable imaging
benefts: gamma cameras. These portable systems might reduce the rate
of unidentifed positive SLNs. In summary, the performance
• Half-life time of approximately 6 hours. This shorter of a gamma detection probe can best be described by the fol-
half-life allows for the capture of an increased lowing quality criteria:4
number of emissions over a short period, creating
an emission density that can form high-resolution • Spatial selectivity at 30 cm (radial sensitivity distri-
images. bution): This describes the width of the measurement
cone, and it has a strong infuence on the detection of
• Small particles that fow into secondary lymph nodes
lymph nodes in the presence of nontarget radiation
more rapidly, therefore enabling the identifcation of
with a broad measurement cone. The importance is
an increased number of nodes.
because the background signal can exceed the target
• Gamma photon emission energy of 140 keV is ideal signal of the lymph node.
both for detection by diagnostic gamma camera
• Spatial resolution: This determines the minimal
imaging and for gamma probe detection. This energy
distance needed between two lymph nodes to be
level provides adequate tissue penetration and man-
detected separately. In the axillary, inguinal, and
ageable imaging technologies.
Modalities of detection 45

iliac regions, spatial resolution higher than 25 mm in the radiation count compared with the basal count (at least
is recommended. For the SLN procedure, high spa- fve to ten times greater than background radioactivity).
tial resolution probes are required to ensure a more
accurate localization. Where more precise spatial
Indocyanine green
localization of activity is needed, collimation of the
detector may be required. ICG is a sterile, anionic, water-soluble tricarbocyanine dye
• Shielding (sensitivity at the probe housing): The leak that shows diffuse fuorescence in the near-infrared spectrum
sensitivity should not exceed 0.1% of the system’s when illuminated by 806-nm light.8 Its biological half-life is
sensitivity. Intraoperative probes are intended for 150–180 seconds. It is odorless, unstable in solution, soluble
directional counting and must have adequate shield- in water and methanol, and insoluble in all other organic sol-
ing on the back and sides. vents.1 ICG emits light with a wavelength of approximately 820
• Sensitivity: It is defned by the count rate per unit of nm that is captured using a specifc video camera device that
activity and is determined at the tip of the probe. It enables it to be displayed in the visible-light spectrum. The
may be increased by widening the energy window organic structure of ICG allows for it to be safely delivered
and/or by reducing collimation and shielding. intravenously. Following intravenous injection, ICG is rapidly
bound to plasma proteins, especially lipoproteins, and distrib-
• Energy discrimination: High-energy resolution
uted throughout the lymphovascular systems.9 ICG metabolism
enables using two different radioisotopes at a time
has no extrahepatic or enterohepatic circulation. Therefore,
and distinguishing the signal of the patient’s radia-
ICG is absorbed from the plasma exclusively by hepatic paren-
tion from those of background radiation.
chymal cells and secreted entirely into the unconjugated bile.
• Display and acoustics: A clear correlation between Intracervical ICG distributes in the lymphatic system, where
the tone and the measurement signal. A digital or it binds to lipoproteins and drains via lymphatic channels and
analog display is recommended. nodes. From the lymph nodes, fltered lymph is propelled back
• The shape of the probe: A slender and kinked probe to the blood circulation via the subclavian vein to maintain tis-
is desirable. sue fuid homeostasis.8
Intravenous and submucosal administration are the two
Until the introduction of ICG as a mapping tracer, blue dye main routes available.9 There is currently no consensus regard-
was considered the standard mapping tracer. However, tech- ing the concentration and volume of ICG to be infused. These
niques such as radioactive colloid injections and lymphoscin- range from 1.25 to 5 mg (single image sequence) for intrave-
tigraphy have provided a viable alternative as an added tool nous and from 1.25 to 5 mg/mL (4–10 mL) for intracervical
when using blue dye in the setting where ICG is not available. submucosal injection. Nevertheless, the maximum dosage
The primary advantage of using radiocolloid and lympho- must never exceed 2 mg/kg (intravenous) or 5 mg (intersti-
scintigraphy is the improvement in detecting lymph nodes that tially).8 The standard dose commonly administered in clinical
are outside the routine anatomic landmarks of dissection, and practice (0.1–0.5 mg/mL/kg) is well below the toxicity level.
consequently, the reduction of the failure rate of SLN identif- ICG should be injected at a concentration of 1.25 mg/mL.
cation. In addition, using the gamma probe, the surgeon may A 25-mg vial with ICG powder is diluted in 20 mL of sterile
explicitly target those areas suggesting the presence of a sen- aqueous water. If the ICG solution is stored at 4°C, ICG should
tinel node that may have been missed due to poor blue dye be used within 1–2 days after the preparation.10 The timing
uptake and have not been identifed by lymphoscintigraphy. of ICG injection in gynecologic surgery during laparotomy is
Close cooperation between nuclear medicine specialists, sur- not well established; however, we recommend the infusion be
geons, pathologists, and in many cases radiologists and medi- performed before the incision. In laparoscopic and robotic-
cal physicists is required to assure the highest success rate of assisted surgery, injection is performed before insertion of a
detection.5 uterine manipulator and/or docking. When the endometrium
The frst stage of SLN identifcation is preoperative lym- is the site of injection, the tubes are sealed before injection.
phoscintigraphy. It is performed in a 1- or 2-day protocol Although there are not enough data to defne the optimal time
with the injection of 99mTc-labeled radiocolloid on the day of to begin dissection, Rossi et al. suggest waiting 10 minutes
the surgery, 4–6 hours before the planned surgery, or a day between injection and lymph node dissection.11 When con-
before, 16–20 hours before the scheduled surgery, respectively. sidering the learning curve, it has been demonstrated that
The preparations are based on the protocol used, 15–20 MBq surgeons require 30 procedures to achieve the detection and
(1-day protocol) or 37–74 MBq (2-day protocol). The protocol false-negative rates necessary to perform this technique by
choice depends on the logistics and experience of the team.6 minimally invasive methods.12
Intraoperative lymphoscintigraphy is usually performed 120
minutes after administration of the radiocolloid (the static
phase of examination). In some instances, to establish the Empty lymph node packet syndrome
direction of lymphatic drainage and to facilitate identifying Removal of presumed mapped lymph nodes that on fnal
the SLN, a dynamic exam is performed immediately after the pathology are determined to be only lymphatic trunks or adi-
administration of the radioactive tracer.7 It is recommended to pose tissue without nodal tissue is a phenomenon that seems to
perform a quantitative measure of the radioactivity in the sen- be more associated with increased ICG utilization. The FILM
tinel node with the gamma probe in order to detect an increase study13 was designed to determine whether ICG is superior to
46 Clinical Lymphatic Mapping

isosulfan blue dye in detecting SLNs in women with clinical of sentinel nodes mainly when the use of lymphatic mapping
stage I endometrial or cervical cancer who underwent cura- and SLN biopsy alone is planned. The injection of radiocol-
tive surgery. This study described a 5%–6% rate of presumed loid should be followed by imaging (lymphoscintigraphy and/
“nodes” with bright signal that were not confrmed to be or SPECT-CT) and surgery within 1–6 hours.
lymph nodes on pathology. More recently, Thomaier and col- The use of radiotracers requires complex logistics, and
leagues14 evaluated whether the rate of empty packet dissec- despite the need for radiocolloids during preoperative plan-
tion changed with increasing surgeon experience. In total, 236 ning, this technology is based on intraoperative acoustic
patients undergoing SLN mapping during minimally invasive feedback and is unable to provide real-time visual guidance.
hysterectomy for either endometrial cancer (85%) or complex More recently, ICG has been used for lymphatic mapping
atypical hyperplasia (15%) were evaluated. The study showed at multiple solid tumor sites. The greatest advantage of this
that the proportion of empty packet dissections declined with technique using the fuorescent agent is that it can be per-
an increasing amount of procedures. The amount of SLNs formed in a one-step, all intraoperative procedure, thus reduc-
removed did not change as surgeon expertise improved. The ing patient discomfort associated with the current two-step
authors concluded that the rate of empty packet SLN dissec- procedure, including preoperative injections of radiocolloid.
tions using indocyanine green dye decreases with an increas- Furthermore, theoretically, the lymphatic channels and SLNs
ing number of procedures and that this stabilizes after 30 in vulvar cancer are often located in a relatively superfcial
procedures. When the surgeon is concerned about this phe- location in the groin when compared to other tumors; there-
nomenon, it is recommended for the tissue to be sent to the fore, near-infrared fuorescence imaging could be particularly
pathologist to confrm that the sample contains lymph node useful for this indication. Nevertheless, this technique requires
tissue rather than adipose tissue. special equipment (near-infrared imaging cameras) that is not
currently available at most centers.
Crane et al.17 published the frst clinical study evaluating the
technical feasibility of intraoperative near-infrared fuores-
Technique by disease site cence imaging for SLN detection in vulvar cancer. The authors
reported the successful use of ICG alone at a concentration
Vulvar cancer
of 0.5 mg/mL in conjunction with an intraoperative imaging
SLN mapping has been proposed as an alternative to inguino- system for the SLN procedure in vulvar cancer. In that study,
femoral lymph node dissection in early-stage vulvar cancer. involving ten patients, 26 of 29 SLNs (90%) were detected in
Mapping for vulvar cancer has been traditionally performed vivo by near-infrared fuorescence. Furthermore, lymphatic
using a dual-modality approach: 1) preoperative 99mTc injec- channels could be visualized in 5 of 16 groins (31%) contain-
tion with lymphoscintigraphy intraoperative gamma probe ing SLNs. In conclusion, radiocolloid tracers alone or in com-
detection or 2) intraoperative peritumoral vulvar injection bination with blue dye remain the standard for the detection of
with blue dye. Radiocolloid tracers should be used alone or SLNs in vulvar cancer in most institutions.
with blue dye. In patients where lymphoscintigraphy did not
identify a sentinel node in the groin(s) of interest, the addi-
Sentinel lymph node technique
tion of blue dye should be used. A meta-analysis performed by
the Cancer Care Ontario group reported a 63% overall detec- To perform SLN mapping with blue dye and 99mTc nanocolloid,
tion rate with the use of blue dye alone compared to 85% with it is recommended that the combination of preoperative imag-
the use of radiocolloid alone.15 The overall detection rate with ing and the use of a gamma probe is most effective in detecting
the combination of blue dye and radiocolloid was 86.9%. The the sentinel node within the inguinofemoral region.
false-negative rates with blue dye alone and radiocolloid alone The day before surgery, a total of 1–2.5 mCi of 99mTc in
were 9.3% and 10.4%, respectively, but when both techniques 2–4 mL of volume may be injected. Mapping substances are
were combined, it lowered this rate to 6.6%. Given these fnd- injected intradermally using a four-point injection technique
ings, the group recommended the use of radiocolloid tracers usually at the 2, 5, 7, and 10 o’clock positions.18 It is important
alone or in combination with blue dye for SLN mapping of to note that tracers should not be injected intratumorally, but
early-stage vulvar cancer. rather into normal epithelium, just lateral to the gross edge
The introduction of radiotracers allowed improved localiza- of the tumor. To access the superfcial dermal lymphatics that
tion of the SLN as well as better reliability. Puig-Tintore et al.16 drain to the groin, it is important to perform an intradermal
published the feasibility of lymphoscintigraphy combined with injection. A deeper injection might lead the tracers to the lym-
a handheld gamma probe and isosulfan blue in detecting the phatic channels into the pelvis and therefore not represent the
SLN in vulvar cancer. In their study the introduction of radio- true lymphatic mapping of the primary vulvar tumor.
tracers allowed an improved localization rate of the SLN to The following day, after induction of anesthesia, a total of 4 mL
98%, and the authors concluded that the combination of the of blue dye is injected at the same locations. During the surgery,
two methods is the best approach. Therefore, the use of radio- handheld gamma-ray detection is used to confrm that lympho-
tracers enhances the localization of the SLN and gives better scintigraphy marked the area of greatest activity in the groin.
results than when the blue dye technique alone is used. There This is important to consider prior to making the groin incision in
is insuffcient evidence to make recommendations regarding order to tailor the location and size of the incision. The scarpa fas-
preoperative lymphoscintigraphy. However, it seems to facili- cia is incised, followed by dissection through the underlying tis-
tate the presence, location (unilateral vs. bilateral), and number sue in an attempt to identify lymphatic channels. Once the groin
Modalities of detection 47

lymphatic basin has been exposed, the surgeon should follow the with 97.5% and 81.3% unilateral and bilateral SLN detection
tinted blue dye channels. With the intermittent use of the gamma rates, respectively. Seventy patients received all three tracers
probe, the sentinel node is identifed and removed. The sentinel with 93.5% and 80.5% unilateral and bilateral detection rates,
node is excised separately, and measurements are again repeated respectively. No signifcant differences were observed between
ex vivo. After removal of the frst sentinel lymph node, the groin the two groups in unilateral or bilateral detection rates or in
is reexamined for radioactivity. If radioactivity is detected at a the identifcation of lymph node metastasis.20
level greater than 10% of the frst excised SLN, the dissection is The major advantage with the use of blue tracers is that they
continued in search of additional SLNs. do not require dedicated and often costly equipment. Blue dye
The feasibility of near-infrared fuorescence-guided SLN is injected in the operating room while the patient is under
biopsy in vulvar cancer has been previously demonstrated using anesthesia. A spinal needle is used to inject 4 mL of dye at
several ICG-based tracer formulas. For the detection of SLN, a rate of 5–10 seconds per quadrant to optimize lymphatic
the near-infrared fuorescence signal is measured percutane- penetration and minimize deep pelvic tissue staining.21 The
ously prior to skin incision19 and continuously during the sur- injection consists of 1 mL of a 10-mg/m: blue dye solution for
gical procedure. To verify nodal removal, the surgical wound a total dose of 40 mg. This dose is injected in the cervix super-
should be reexamined using fuorescence imaging as well as fcially (2–3 mm cervical submucosa) and deep (3–4 mm) at
the other techniques such as radiotracing/imaging and visual 3 and 9 o’clock positions for four injections in total. For cen-
inspection for blue dye and palpation if applicable. With this ters wishing to use radiolabeled colloid as the tracer of choice
technique the sentinel node should also be examined ex vivo. for SLN mapping, the most commonly used is 99mTc. It should
be injected before surgery (generally the day before) into a
radio-protected setting. Four cervical injections of 0.2 mL (20
Endometrial cancer
MBq each) of unfltered technetium sulfur colloid (Nanocis;
There are three different techniques for SLN uterine cancer CIS Bio International, Saclay, France) are recommended with
mapping based on the site of injection: subserosal uterine a 25-gauge spinal needle at the 3, 6, 9, and 12 o’clock posi-
fundus, cervical, or endometrial through hysteroscopy. Given tions the day or morning before surgery. Scintigraphic images
the obstacles of the subserosal and hysteroscopic techniques, are obtained with a triple-head gamma camera (Irix; Marconi
such as the fact that they are more technically challenging, Corporation, Cleveland, OH, USA) 2 hours after injection and
less reproducible, and more expensive than the cervical injec- then every 30 minutes.
tion, these approaches have not gained popularity. There is In some centers, ICG is preferred over the combination of
evidence that cervical injection achieves a higher detection other tracers, or is at least as useful as the blue dye and radio-
rate and a similar anatomic nodal distribution as the hystero- labeled colloids.22 The results of the FILM study13 served as the
scopic injection for SLN mapping.11 It provides excellent dye basis for an application to the Food and Drug Administration
penetration to the region of the main uterine lymphatic trunks (FDA) for on-label use of interstitial injection of ICG com-
that condense in the parametria and appear in the broad liga- bined with near-infrared imaging for lymphatic mapping. It
ment leading to the pelvic and, occasionally, paraaortic senti- was the frst prospective, randomized, phase III, multicenter,
nel nodes. The cervix is easily accessible and rarely distorted noninferiority trial that showed how ICG improved SLN iden-
or scarred from prior procedures. tifcation rates (74% rate of detection of at least one SLN with
Regarding tracer options, several studies have evaluated the isosulfan blue dye compared with 96% with ICG and 31% rate
use of ICG alone or in combination with blue dye and 99mTc. of bilateral sentinel nodes with blue dye and 78% with ICG).
A prospective randomized study aimed to evaluate whether There are three different options for direct cervical injection,
three tracers (blue dye, ICG, and 99mTc) performed better than although the most commonly used is the two-injection tech-
two (ICG and 99mTc) tracers. One hundred and ffty-seven nique, which is often preferred to the four-quadrant injection
patients were included. Eighty patients received ICG and 99mTc options (Figure 8.1). After general anesthesia is performed,

FIGURE 8.1 Common cervical injection sites for mapping uterine cancer. (From Abu-Rustum NR, Rob L, Sentinel lymph node identifcation for
early-stage cervical and uterine cancer, in Abu Rustum NR, Barakat RR, Levine DA, eds, Atlas of Procedures in Gynecologic Oncology 3E, CRC
Press, 2013, with permission.)
48 Clinical Lymphatic Mapping

FIGURE 8.2 (a) Most common location of SLNs (blue arrow) following a cervical injection. (b) Less common location of SLNs (green arrow) usu-
ally seen when lymphatic trunks are not crossing over the umbilical ligament but following the mesoureter cephalad to the common iliac and presacral
region. (From Abu-Rustum NR, Rob L, Sentinel lymph node identifcation for early-stage cervical and uterine cancer, in Abu Rustum NR, Barakat
RR, Levine DA, eds, Atlas of Procedures in Gynecologic Oncology 3E, CRC Press, 2013, with permission.)

ICG is injected in the cervix deep (3–4 cm) and superfcially adherence to the SLN algorithm.25 Moreover, standardization
(2–3 mm cervical submucosa) at 3 and 9 o’clock positions, of surgical technique and surgical-quality assessment tools are
for a total of four injections.22 Surgeons should avoid diffuse critical to the conduct of clinical trials. Specifc mandatory
smearing. The uterine body lymphatic trunks commonly cross and prohibited steps of SLN dissection in endometrial cancer
over the obliterated umbilical artery, with the most common have been identifed and validated based on consensus among a
locations of pelvic SLNs medial to the external iliac, ventral to large number of international experts.
the hypogastric, or in the superior part of the obturator region
(Figure 8.2). Surgeons should open the retroperitoneum and
Cervical cancer
search for the lymph node by activating the infrared button on
the camera head. The primary mode used is the near-infrared SLN mapping in cervical cancer initially evaluated the util-
mode, but two additional modes may also be used: spy mode ity of blue dye and/or radiocolloid. In this setting, while the
(black and white mode) or color segmented fuorescence mode patient is under anesthesia, an injection of isosulfan blue
in the form of a color gradient. Surgeons can switch back and (Lymphazurin) or methylene blue 1% is administered. With
forth between the different modes as often as needed. a spinal needle a total of 4 mL of blue dye is injected directly
into the cervical stroma next to the lesion. A tenaculum may
be used to assist in the stromal injection. Two milliliters per
injection for each side are administered at the 3 and 9 o’clock
Dose of ICG on the SLN mapping
positions, which correspond to the position of the parametria.
Papadia et al. designed a study to evaluate the impact of dif- The 12 o’clock injection may be eliminated in order to avoid
ferent doses of ICG (high dose: 5 mg/mL and a volume of 8 staining the vesicocervicovaginal space. It is recommended
mL and low dose: 1.25 mg/mL and a volume of 4 mL) on the that the injection be performed after the laparotomy has been
SLN mapping in endometrial cancer. A higher concentration performed and the surgical feld exposed. The procedure is
and volume of ICG are associated with a larger number of planned through laparotomy, given that the minimally inva-
retrieved SLNs.23 sive approach is no longer considered after the results of the
The key to successful SLN mapping is the adherence to an LACC trial, which demonstrated worse disease-free survival
SLN algorithm. Incorporating an SLN mapping algorithm for minimally invasive radical hysterectomy when compared
reduces the false-negative rate to 2%.24 A reasonable SLN detec- to the open approach.26 The sentinel node identifcation and
tion rate varies from practice to practice, but a detection rate of removal is performed frst. Preoperative lymphoscintigraphy
80%–90% or higher is desired.12 SLN mapping for endometrial is performed using radiolabeled fltered 99mTc. It is injected
cancer is becoming an established standard of care in many directly into two quadrants of the stroma of the cervix using
centers worldwide, while increasing surgeon experience and a spinal needle closest to the area of a normal cervix/tumor
a corresponding increase in detection rates of 90% or greater, interface. In patients who have undergone a prior cone biopsy,
in addition to a decrease in false-negative rates as a result of the injection is given into the bed of the cone on the day before
Modalities of detection 49

surgery. ICG is increasingly becoming part of the standard of exceed 1.5%.30 Anaphylaxis during anesthesia is often a clinical
care, given its high detection rate, higher signal-to-background diagnosis presenting, in decreasing frequency, with cardiovas-
ratio, improved cost-effectiveness, and decreased adverse cular collapse, erythema, angioedema, bronchospasm (severe
effects and toxicity. ICG should be injected with a similar or transient), urticaria and/or rash, gastrointestinal symptoms,
technique as explained earlier for endometrial cancer. and pulmonary edema.31 It is important to understand that dur-
ing anesthesia, anaphylaxis develops 90% of the time within 10
minutes after intravenous administration of the causal agent.32
Ovarian cancer
However, a time delay of 15–30 minutes may be observed in
The detection of SLN in ovarian cancer has been evaluated in cases of anaphylactic reaction to blue dyes, refecting the fact
the setting of early disease and is currently in the experimental that these agents are being administered intradermally rather
phase. After the frst publication in 1991,27 there have been a than intravenously. Adverse reactions to isosulfan blue have
number of small series with small numbers of patients. Most been confrmed as a rare event, less than 2%.33
studies are exploratory and evaluate the ovarian lymphatic Another systemic manifestation seen after intradermal injec-
drainage, although in many cases these have been performed tions of isosulfan blue is an acute transient or a more prolonged
in patients who had benign,27 borderline, cervical or endome- decline in oxygen saturation as measured by pulse oximetry.
trial27 tumors instead of ovarian cancer. The main diffculties Coleman et al. provided a detailed review of the etiology of this
in performing SLN mapping in early ovary cancer include 1) phenomenon.34 Pulse oximetry is a noninvasive modality provid-
a large and complex area of lymphatic drainage, 2) the need ing continuous estimations of peripheral tissue oxygen saturation.
for intraoperative injection, and 3) the injection site (it is not It functions via determination of the concentrations of oxyhemo-
possible to perform a peritumoral or intratumoral injection due globin and reduced hemoglobin species by measuring the absor-
to the risk of tumor rupture). The only systematic review on bance of light at two wavelengths, 660 and 940 nm, respectively.
the feasibility of SLN in epithelial ovarian cancer has been Isosulfan blue has an absorbance peak of 646 nm. The dye can
published recently.28 The data thus far confrm that the tech- also infuence pulse oximetry measurements because it causes
nique appears feasible with a detection rate of approximately abnormal hemoglobin species.35 Fortunately, this phenomenon is
90%, although there are important limitations, such as low transient and typically lasts 3–5 minutes. The inaccuracy of the
sample size, injection of the tracer in different anatomical pulse oximeter in this setting has been confrmed by obtaining
areas, and the use of different tracers (radiotracer, methylene arterial blood sampling during the acute fall in SpO2, a fnding
blue, and ICG combined or alone). This review concluded that which confrms adequate and reassuring oxygen saturation.
the best tracer is ICG and the two best injection sites are the
infundibulo-pelvic ligament and the uterovarian ligament.
Technetium
Five studies have assessed the combination of a radiotracer
and blue dye. The detection rate was 88.1% when a radiocol- Radiation protection is based on three principles, which
loid (with or without blue dye) was used (N = 109). Three stud- have been described by the International Commission on
ies combined patent blue with 99mTc, and the number of SLNs Radiological Protection.36 These include the following: justi-
that have been identifed only with patent blue dye were 100%, fcation – the application of radiation is justifed only when the
83%, and 20%, respectively.28 When ICG alone was used, the beneft to the exposed individual outweighs the negative con-
detection rate was 93.3% (N = 15).29 Based on the published sequences; optimization – ensure that the radiation dose to the
literature, it appears that, with the exception of patent blue, all patient and the general public is ‘as low as reasonably achiev-
other tracers have resulted in a high detection rate of SLNs. able’; and limitation – boundaries should be placed on risks to
The utero-ovarian ligament and the infundibulopelvic ligament individuals to ensure that the risks do not exceed acceptable
have been used as the site of injection in the majority of stud- values. The determinant of the radiation dose to which the
ies. Injection of the tracer should be performed, preferably with patient is exposed is the degree of clearance from the site of
the adnexa still in situ, both in the suspensory ligament and the injection and the lymph node. The clearance of radiocolloids
infundibulopelvic ligament. With this approach, the detection from the interstitial space is very slow; therefore, the largest
rate has exceeded 90%. In two studies, an injection into the dose of radiation is delivered to the site of injection.
ovarian cortex appeared to be less sensitive (71.4%, N = 21).28 It should be emphasized that according to the International
Ideally, an interval of 10–15 minutes should be allowed after Atomic Energy Agency guidelines, the permitted annual dose
injection before starting SLN mapping; this provides the tracer is 1 mSv. The maximal allowed doses of radiation exposure,
enough time to spread to the lymph nodes (detection rate >90%). according to the National Council on Radiation Protection
(NCRP) recommendations, are 500 mSv for skin per year for
the exposed personnel. Considering this dose limit, a surgeon
Safety could perform more than 9000 sentinel node procedures with-
out exceeding the dose limits.37
Blue dye
Approximately 50% of isosulfan blue in aqueous solution is
weakly bound to serum proteins, leading to its affnity for lym-
Indocyanine green
phatic channels. Its primary excretion is biliary (90%), and thus ICG has an excellent safety profle with a high affnity for albu-
patients with hepatobiliary insuffciency may be at an increased min, and therefore there is almost no leakage of the compound
risk of complications. The overall complication rate should not into surrounding tissue. The liver absorbs and excretes the
50 Clinical Lymphatic Mapping

substance but does not metabolize it into toxic by-products;1 ICG SLN mapping seems to be feasible in pregnant cervical
for that reason it exhibits low toxicity with few side effects cancer patients, and no adverse events from ICG injection were
(36 out of 21,278 cases; 0.17%), which include shock symp- recorded.45
toms (0.02%), nausea (0.08%), vascular pain (0.04%), and fever
(0.02%).38 The incidence of mild adverse reactions have been
Obesity
previously described as 0.15%, and 0.05% for severe adverse
reactions, with no deaths after 1923 procedures.39 One should Two independent studies published by Tanner et al.46 and
note that there are a number of contraindications for the use of Eriksson et al.21 suggested the effect of increased body mass
ICG, such as iodine allergy, liver disease, dialysis, renal fail- index (BMI) on SLN detection. The objective of the frst study
ure, uremia, or previously documented anaphylactic reaction was to identify the patient, tumor, and surgeon factors associ-
(1 of 42,000 patients). It should also be used with caution in ated with successful SLN in patients with endometrial cancer
elderly patients with decreased physiological function.39 There and complex atypical hyperplasia. The authors showed the
are no data available describing the signs, symptoms, or labo- decrease of the rate of successful bilateral mapping with blue
ratory fndings accompanying overdosage. for 58% of patients with a BMI ⩾ 30 kg/m2. While this rate
with ICG was always superior to isosulfan blue, the variabil-
ity was more pronounced at higher BMIs. The Eriksson et al.
Mapping in special settings study21 aimed to determine the impact of obesity on the rate
of successful SLN mapping in patients with endometrial can-
Pregnancy
cer undergoing robotic surgery and compared SLN detection
SLN mapping procedures are feasible in pregnant patients. rates using ICG versus blue dye. With increasing BMI, there
SLN should be considered the standard of care for melanoma was a signifcant decrease in successful bilateral mapping
and breast cancer in the clinically negative axilla. Isosulfan rates for both the ICG and blue-dye groups; however, the use
blue should only be given during pregnancy when benefts out- of ICG resulted in better bilateral and overall mapping rates
weigh the risks because it is systemically absorbed after sub- compared with the use of blue dye in all BMI groups. To sum-
cutaneous injection.40 Methylene blue has not been found to be marize, these studies concluded that visualization of SLNs is
safe and effective in pregnant women according to the FDA, improved when ICG was used in comparison to blue dye in
and this labeling has not been approved. It can cause fetal obese patients.
harm when administered to a pregnant woman. Intraamniotic
injection during the second trimester has been associated
with neonatal intestinal atresia and fetal death. This drug also Novel modalities
caused adverse developmental outcomes in animals when
administered orally during organogenesis.41 Intraoperative identification techniques
Regarding the use of 99mTc as a widely radioactive tracer
[99mTechnetiumTc] tilmanocept
in SLN, the American Society of Clinical Oncology main-
tains pregnancy as a contraindication for this radiotracer.42 [99mTechnetiumTc] tilmanocept (trade name Lymphoseek) is
However, the European Association of Nuclear Medicine a molecule that binds receptors in reticuloendothelial cells in
and the Society of Nuclear Medicine and Molecular Imaging the nucleus and can be evaluated via an intraoperative hand-
concluded that the use of 99mTc is justifed by the low risks held gamma detector.47 Lymphoseek has been approved in the
of the procedure, considering the limited doses.43 It has been United States and Europe for SLN mapping of breast cancer,48
calculated that when using a nanocolloid with a short half- melanoma,49 and head and neck cancer,50 with sensitivities of
life and large particle size, such as 99mTc, and due to the >97% for detecting SLNs. Fluorescent-labeled tilmanocept is
accumulation of the nanocolloid in the lymph node itself, potentially advantageous over sulfur colloids, as it is better tol-
the fetal radiation exposure is <5 mGy, even in the inguinal erated by patients and is cleared from the injection site fvefold
lymph nodes. It is also recommended that in pregnancy one faster than sulfur colloid.
should use the single-day protocol, as the administered dose Preclinical animal model trials have been successful in
is lower, the time between admission and surgery is shorter, endometrial cancer, with SLN identifcation via robotic sur-
and the detection rate does not differ from the 2-day pro- gery, by tagging the [99mTc] tilmanocept to near-infrared fuo-
tocol. Therefore, when maternal outcome may beneft from rescent dye, as well as with gallium-68 to allow for assessment
an SLN, it should not be withheld because of fear for fetal on positron emission tomography (PET). During robotic or
radiation exposure.44 laparoscopic surgery, fuorescent-labeled tilmanocept does
It is generally not recommended for pregnant personnel to not require the expertise of radiodetection equipment. Even
perform lymphatic mapping when using radiocolloid, given though preoperative [99mTc] tilmanocept imaging may not be
that one should always aim for the radiation dose to be ‘as low regularly used, cross-sectional imagery can be particularly
as reasonably achievable.’ The distance to maintain for a preg- useful in patients with high BMI.
nant staff member should be at least 1 meter from the patient.
It is recommended that before introducing the sentinel node
Carbon nanoparticles
procedure, all personnel should be informed about the proper
handling of material and safety procedures.5 As it pertains to Carbon nanoparticles are 150-nm particles that can be injected
ICG, although the FDA has classifed it as a category C drug, into the anatomical area of interest and drain through the
Modalities of detection 51

lymphatic vessels (diameter, 120–500 nm). They are then visu- contrast-enhanced ultrasound-guided core biopsy of the SLN
alized directly as a black stain in the lymphatic channels and could identify metastatic nodes preoperatively and reduce the
SLN. Carbon nanoparticles are advantageous in that nuclear number of surgical SLN biopsies in patients with breast cancer.
medicine is not required, they are relatively inexpensive, and Eleven prospective studies and one retrospective study with
no special equipment is required. Overall, they are well tol- 1520 participants were included. The SLN identifcation and
erated; a major side effect is skin staining.51 More recently, localization rate for contrast-enhanced ultrasound-guided skin
carbon nanoparticle use was studied in endometrial cancer in marking had varying success (70%–100% detection rate).55 The
a prospective single-center trial of 115 patients.52 The tracer introduction of super-resolution imaging, ultrafast ultrasound
was shown to have a comparable detection rate to those of imaging, and improved microbubble transit may improve the
currently used methods in endometrial cancer and a safe and visualization of lymphatics and SLNs with this technique.
effective profle for SLN mapping. The results revealed that Implementing new technology into clinical practice is
carbon nanoparticles achieved a total detection rate of 96.5%, challenging. The risk associated with the application of new
and the cervical injection route was associated with 100% sen- techniques or procedures makes it diffcult to replace the
sitivity and negative predictive value. Although more studies traditional and already accepted surgical technique. To over-
are needed to confrm these fndings, this colorimetric imag- come this scenario, Marcus et al.56 aimed to assess the safety
ing tracer provides a feasible alternative for centers lacking and effcacy outcomes of an institutional structured evalua-
fuorescent imaging systems. tion process specifcally focused on the introduction of sur-
gical innovations. A multidisciplinary specialized team of
surgical quality offcers composed the Continuous Quality
India ink
Improvement Team (CQIT) and performed structured reviews
India ink is an injectable colloid suspension of carbon par- of suggested new surgical procedures before being evaluated
ticles that dyes SLNs black. In a study in early-stage cervical by the Value Analysis Team business. The results showed the
cancer,53 India ink was used in combination with radiocolloid review process improved the safety and effciency; no device-
and blue dye to determine whether adding this colloid would related complications were observed, and the time between
help identify parametrial sentinel nodes pathologically. The product proposal and trial safety decreased from a mean of
authors performed lymphatic mapping and SLN biopsy in 20 260 to 99 days (P = 0.014).
women undergoing radical hysterectomy or trachelectomy
using a ‘triple injection’ technique. Pathologic processing of
parametrium and nodal tissue was then performed to identify
Conclusion
India ink in specimens. India ink alone identifed only a single
pelvic sentinel node (0.3%) not detected with blue dye and/or As we gather additional information in support of the util-
radiocolloid in this study. However, the addition of this ink ity of lymphatic mapping and sentinel node detection in the
was useful to detect the true sentinel nodes and not second- management of gynecologic cancers, we strive to pursue this
echelon nodes detected by blue dye that had already passed approach to decrease patient morbidity and improve diagnostic
through sentinel nodes. accuracy. While the focus of most perfusion mapping studies
in gynecologic oncology has been on technetium colloids and
blue dye, with the new modality of identifcation of the sen-
Superparamagnetic iron
tinel nodes, it has improved detection rates while decreasing
Superparamagnetic iron is a tracer that can be injected pre- cost and side effects. Alternative approaches are actively being
operatively for intraoperative magnetic detection. In 2016, studied in both gynecologic and nongynecologic cancers.
Karakatsanis54 performed a study aimed to compare the
effcacy of this novel tracer in sentinel node biopsy in breast
cancer with 99mTc and patent blue in a multicenter prospective REFERENCES
trial and performed a meta-analysis of all published studies. 1. Moukarzel LA, Feinberg J, Levy EJ, et al. Current and
Seven studies were included, with a total of 1118 cases per- novel mapping substances in gynecologic cancer care. Int J
formed and 2300 nodes retrieved. No difference was observed Gynecol Cancer. 2020;30(3):387–93.
in the detection rates per case in any of the studies between the 2. Hiram SC. The role of nuclear medicine. Sentinel Lymph
superparamagnetic iron and conventional methods. Node Biopsy. Breast Cancer Res. 2001;3(2):104–8.
3. Zalewski K, Benke M, Mirocha B, et al. Technetium-99m-
based radiopharmaceuticals in sentinel lymph node biopsy:
Imaging-enhanced modalities gynecologic oncology perspective. Curr Pharm Des.
Contrast-enhanced ultrasound 2018;24(15):1652–75.
4. Wengenmair KJ. Quality Criteria of Gamma Probes:
Contrast-enhanced ultrasound is a technique that employs Requirements and Future Developments. Berlin,
the injection of perfuorobutane microbubbles to visualize Heidelberg: Spring; 2005.
lymphatic channels on ultrasound and facilitate the identif- 5. Giammarile F, Bozkurt MF, Cibula D, et al. The EANM
cation of SLNs for subsequent biopsy or resection with skin clinical and technical guidelines for lymphoscintigraphy
marking or wire localization. This method has been studied and sentinel node localization in gynaecological cancers.
in a systematic review and meta-analysis to evaluate whether Eur J Nucl Med Mol Imaging. 2014;41(7):1463–77.
52 Clinical Lymphatic Mapping

6. Mount MG, White NR, Nguyen CL, et al. Evaluating one newly diagnosed uterine cancer undergoing robotic surgery.
day versus two days preoperative lymphoscintigraphy pro- Ann Surg Oncol. 2016;23(8):2522–8.
tocols for sentinel lymph node biopsy in breast cancer. Am 22. National Comprehensive Cancer Network. Uterine
Surg. 2015;81(5):454–7. Neoplasms (Version1.2020) [cited 2020 Feb 10]. Available
7. Petersen LJ, Pedersen RD, Skindhoj S, et al. Early dynamic from: www.nccn.org/professionals/physician_gls/default.
versus late static lymphoscintigraphy for the identifcation aspx#uterine.
of sentinel lymph nodes in breast cancer. Clin Nucl Med. 23. Papadia A, Buda A, Gasparri ML, et al. The impact of dif-
2011;36(12):1098–101. ferent doses of indocyanine green on the sentinel lymph-
8. Ferreira H, Smith AV, Wattiez A. Application of indocya- node mapping in early stage endometrial cancer. J Cancer
nine green in gynecology: review of the literature. Surg Res Clin Oncol. 2018;144(11):2187–91.
Technol Int. 2019;34:282–92. 24. Barlin JN, Khoury-Collado F, Kim CH, et al. The impor-
9. Reinhart MB, Huntington CR, Blair LJ, et al. Indocyanine tance of applying a sentinel lymph node mapping algorithm
green: historical context, current applications, and future in endometrial cancer staging: beyond removal of blue
considerations. Surg Innov. 2016;23(2):166–75. nodes. Gynecol Oncol. 2012;125(3):531–5.
10. Mindt S, Karampinis I, John M, et al. Stability and degrada- 25. Abu-Rustum NR. Update on sentinel node mapping
tion of indocyanine green in plasma, aqueous solution and in uterine cancer: 10-year experience at Memorial
whole blood. Photochem Photobiol Sci. 2018;17(9):1189–96. Sloan  – Kettering Cancer Center. J Obstet Gynaecol Res.
11. Rossi EC, Jackson A, Ivanova A, et al. Detection of sentinel 2014;40(2):327–34.
nodes for endometrial cancer with robotic assisted fuores- 26. Ramirez PT, Frumovitz M, Pareja R, et al. Minimally inva-
cence imaging: cervical versus hysteroscopic injection. Int sive versus abdominal radical hysterectomy for cervical
Gynecol Cancer. 2013;23(9):1704–11. cancer. N Engl J Med. 2018;379(20):1895–904.
12. Khoury-Collado F, Glaser GE, Zivanovic O, et al. 27. Vanneuville G, Mestas D, Le Bouedec G, et al. The lym-
Improving sentinel lymph node detection rates in endome- phatic drainage of the human ovary in vivo investigated
trial cancer: how many cases are needed? Gynecol Oncol. by isotopic lymphography before and after the menopause.
2009;115(3):453–5. Surg Radiol Anat. 1991;13(3):221–6.
13. Frumovitz M, Plante M, Lee PS, et al. Near-infrared fuo- 28. Dell’Orto F, Laven P, Delle Marchette M, et al. Feasibility
rescence for detection of sentinel lymph nodes in women of sentinel lymph node mapping of the ovary: a systematic
with cervical and uterine cancers (FILM): a randomised, review. Int J Gynecol Cancer. 2019;29(7):1209–15.
phase 3, multicentre, non-inferiority trial. Lancet Oncol. 29. Angelucci M, Corrado G, Mancini E, et al. Laparoscopic
2018;19(10):1394–403. indocyanine green sentinel lymph node mapping in early
14. Thomaier L, Jager L, Stone R, et al. Risk of empty lymph ovarian cancer. A pilot study and review of the literature.
node packets in sentinel lymph node mapping for endome- Ann Surg Oncol. 2016;28:23–8.
trial cancer using indocyanine green. Int J Gynecol Cancer. 30. Ramirez PT, Levenback C. Sentinel nodes in gynecologic
2019;29(3):513–17. malignancies. Curr Opin Oncol. 2001;13(5):403–7.
15. Covens A, Vella ET, Kennedy EB, et al. Sentinel lymph 31. Fisher M, Baldo B. Anaphylaxis during anaesthesia: current
node biopsy in vulvar cancer: systematic review, meta- aspects of diagnosis and prevention. Eur J Anaesthesiol.
analysis and guideline recommendations. Gynecol Oncol. 1994;11(4):263–84.
2015;137(2):351–61. 32. Moss J. Adverse drug reactions caused by Histamine. ASA
16. Puig-Tintoré LM, Ordi J, Vidal-Sicart S, et al. Further data Refr Cours Anesthesiol. 1992;20:155–68.
on the usefulness of sentinel lymph node identifcation and 33. Cimmino VM, Brown AC, Szocik JF, et al. Allergic reac-
ultrastaging in vulvar squamous cell carcinoma. Gynecol tions to isosulfan blue during sentinel node biopsy – a com-
Oncol. 2003;88(1):29–34. mon event. Surgery. 2001;130(3):439–42.
17. Crane L, Themelis G, Arts H, et al. Intraoperative near- 34. Coleman RL, Whitten CW, O’Boyle J, et al. Unexplained
infrared fuorescence imaging for sentinel lymph node decrease in measured oxygen saturation by pulse oximetry
detection in vulvar cancer: frst clinical results. Gynecol following injection of Lymphazurin 1%(isosulfan blue)
Oncol. 2011;120(2):291–5. during a lymphatic mapping procedure. J Surg Oncol.
18. Koh W-J, Greer BE, Abu-Rustum NR, et al. Vulvar cancer, 1999;70(2):126–9.
version 1.2017, NCCN clinical practice guidelines in oncol- 35. Scheller MS, Unger RJ, Kelner MJ. Effects of intrave-
ogy. J Natl Compr Canc Netw. 2017;15(1):92–120. nously administered dyes on pulse oximetry readings.
19. Schaafsma BE, Verbeek FP, Peters AA, et al. Near-infrared Anesthesiology (Philadelphia).1986;65(5):550–2.
fuorescence sentinel lymph node biopsy in vulvar cancer: 36. Protection ICoR. 1990 recommendations of the interna-
a randomised comparison of lymphatic tracers. BJOG. tional commission on radiological protection. Ann ICRP.
2013;120(6):758–64. 1991;21(1–3):1–201.
20. Kessous R, How J, Abitbol J, et al. Triple tracer (blue dye, 37. Najaf M, Nedaie H, Lahooti A, et al. Radiation exposure
indocyanine green, and Tc99) compared to double tracer of the surgeons in sentinel lymph node biopsy. Iran J Radiat
(indocyanine green and Tc99) for sentinel lymph node detec- Res. 2012;10(1):53–7.
tion in endometrial cancer: a prospective study with ran- 38. Lecuru FR, McCormack M, Hillemanns P, et al. SENTICOL
dom assignment. Int J Gynecol Cancer. 2019;29(7):1121–5. III: an international validation study of sentinel node biopsy
21. Eriksson AGZ, Montovano M, Beavis A, et al. Impact of in early cervical cancer. A GINECO, ENGOT, GCIG and
obesity on sentinel lymph node mapping in patients with multicenter study. Int J Gynecol Cancer. 2019;29(4):829–34.
Modalities of detection 53

39. Hope-Ross M, Yannuzzi LA, Gragoudas ES, et al. Adverse blue dye for identifcation of sentinel lymph nodes in clini-
reactions due to indocyanine green. Ophthalmology. cally node-negative cutaneous melanoma. Ann Surg Oncol.
1994;101(3):529–33. 2013;20(2):680–8.
40. Khera SY, Kiluk JV, Hasson DM, et al. Pregnancy- 50. Agrawal A, Civantos FJ, Brumund KT, et al. [99m Tc]
associated breast cancer patients can safely undergo lym- Tilmanocept accurately detects sentinel lymph nodes
phatic mapping. Breast J. 2008;14(3):250–4. and predicts node pathology status in patients with oral
41. Drugs.com. Contraindications in Methylene Blue Injection squamous cell carcinoma of the head and neck: results
2018 [updated 2018 Jan 11] [cited 2020 Feb 3]. Available of a phase III multi-institutional trial. Ann Surg Oncol.
from: www.drugs.com/pro/methylene-blue-injection.html# 2015;22(11):3708–15.
s-34070-32020. 51. Zhang L, Huang Y, Yang C, et al. Application of a carbon
42. Lyman GH, Somerfeld MR, Bosserman LD, et al. Sentinel nanoparticle suspension for sentinel lymph node mapping
lymph node biopsy for patients with early-stage breast can- in patients with early breast cancer: a retrospective cohort
cer: American Society of Clinical Oncology clinical prac- study. World J Surg Oncol. 2018;16(1):112.
tice guideline update. J Clin Oncol. 2016;35(5):561–4. 52. Zuo J, Wu LY, Cheng M, et al. Comparison study of
43. Giammarile F, Alazraki N, Aarsvold JN, et al. The EANM laparoscopic sentinel lymph node mapping in endome-
and SNMMI practice guideline for lymphoscintigraphy and trial carcinoma using carbon nanoparticles and lym-
sentinel node localization in breast cancer. Eur J Nucl Med phatic pathway verifcation. J Minim Invasive Gynecol.
Mol Imaging. 2013;40(12):1932–47. 2019;26(6):1125–32.
44. de Haan J, Vandecaveye V, Han SN, et al. Diffculties with 53. Frumovitz M, Euscher ED, Deavers MT, et al. ‘Triple injec-
diagnosis of malignancies in pregnancy. Best Pract Res tion’ lymphatic mapping technique to determine if parame-
Clin Obstet Gynaecol. 2016;33:19–32. trial nodes are the true sentinel lymph nodes in women with
45. Papadia A, Mohr S, Imboden S, et al. Laparoscopic indocya- cervical cancer. Gynecol Oncol. 2012;127(3):467–71.
nine green sentinel lymph node mapping in pregnant cervical 54. Karakatsanis A, Christiansen PM, Fischer L, et al. The
cancer patients. J Minim Invasive Gynecol. 2016;23(2):270–3. Nordic SentiMag trial: a comparison of super paramagnetic
46. Tanner EJ, Sinno AK, Stone RL, et al. Factors associated iron oxide (SPIO) nanoparticles versus Tc99 and patent blue
with successful bilateral sentinel lymph node mapping in in the detection of sentinel node (SN) in patients with breast
endometrial cancer. Gynecol Oncol. 2015;138(3):542–7. cancer and a meta-analysis of earlier studies. Breast Cancer
47. Surasi DS, O’Malley J, Bhambhvani P. 99mTc-tilmanocept: Res Treat. 2016;157(2):281–94.
a novel molecular agent for lymphatic mapping and sen- 55. Moody AN, Bull J, Culpan A-M, et al. Preoperative sen-
tinel lymph node localization. J Nucl Med Technol. tinel lymph node identifcation, biopsy and localization
2015;43(2):87–91. using contrast enhanced ultrasound (CEUS) in patients with
48. Wallace AM, Han LK, Povoski SP, et al. Comparative breast cancer: a systematic review and meta-analysis. Clin
evaluation of [99m Tc] tilmanocept for sentinel lymph node Radiol. 2017;72(11):959–71.
mapping in breast cancer patients: results of two phase 3 56. Marcus RK, Lillemoe HA, Caudle AS, et al. Facilitation of
trials. Ann Surg Oncol. 2013;20(8):2590–9. surgical innovation: is it possible to speed the introduction
49. Sondak VK, King DW, Zager JS, et al. Combined analysis of new technology while simultaneously improving patient
of phase III trials evaluating [99m Tc] tilmanocept and vital safety? Ann Surg. 2019;270(6):937–41.
9
Ultrastaging of the sentinel node

Celien P.H. Vreuls, Ate G.J. van der Zee, and Paul J. van Diest

CONTENTS
Introduction .................................................................................................................................................................................... 55
Frozen section ................................................................................................................................................................................ 55
Imprint cytology............................................................................................................................................................................. 56
Serial sectioning............................................................................................................................................................................. 57
Immunohistochemistry .................................................................................................................................................................. 57
Fine-needle aspiration cytology ..................................................................................................................................................... 58
Flow cytometry .............................................................................................................................................................................. 58
Molecular techniques ..................................................................................................................................................................... 59
The optimal standard protocol ....................................................................................................................................................... 60
Summary remarks .......................................................................................................................................................................... 60
References...................................................................................................................................................................................... 61

investigation, immunohistochemistry (IHC), imprint cytology,


fne-needle aspiration cytology, fow cytometry, and molecular
Introduction analysis. Several review papers on the pathology of the SN
The main task for the pathologist is to examine sentinel nodes have been published.1,5,9–11 The aim of this chapter is to provide
(SNs) for possible metastasis.1–5 This examination has to be an up-to-date discussion of the virtues and faws of these dif-
done with more attention (‘ultrastaging’) than examination of ferent methods and to present practical guidelines for the SN
routinely removed lymph nodes with the usual central hema- investigation in gynecologic cancers.
toxylin and eosin (H&E) section. A false-negative SN assess-
ment leading to omission of lymph node dissection may lead
to untreatable locoregional tumor outgrowth in tumor-bearing
Frozen section
lymph nodes that have been left behind. The SN evaluation
will usually be done postoperatively. If indicated, a complete As just briefy mentioned, it would be an advantage to the
lymph node dissection would then be performed in a second patient and surgeon if the regional lymph node dissection,
session. However, it is advantageous for the patient and sur- when necessary, could be performed in the same operating
geon if the regional lymph node dissection, when necessary, session as the SN procedure and the excision of the primary
could be performed in the same operating session as the SN tumor, obviating the need for a second operation. However,
procedure and the excision of the primary tumor. This requires there is also a logistic drawback: the time for possible sub-
accurate and effcient intraoperative assessment of the SN by sequent lymph node dissection needs to be reserved in all
the pathologist. patients, while it will be used in only some of the patients.
Size measurement of SN metastases in gynecologic cancer Accurate and effcient intraoperative assessment of the SN
follows the thresholds of breast cancer and melanoma. This by the pathologist may be done by frozen-section analysis,
means that metastases smaller than 0.2 mm are denoted iso- usually a single H&E frozen section. The next paragraphs will
lated tumor cells (ITCs), those 0.2–2 mm are micrometastases, make clear that without step sections and IHC, up to 15%–20%
and those >2 mm in size are macrometastases. In breast cancer of metastases may be missed in patients with breast cancer or
cells may be displaced by biopsy procedures and transported cutaneous melanoma. Therefore, the theoretical sensitivity for
to the SN, leading to epithelial groups in SNs that do not rep- detection of metastases by single H&E frozen section analysis
resent true metastases.6,7 Displacement in gynecologic cancers is (under optimal conditions) probably not higher than 80%–
has not been studied so far. Measuring the size of metastases 85% and lower in more diffcult conditions, as is often the case
may be diffcult when multiple smaller separate but neighbor- with frozen sections.
ing clusters are present, for which Gabor Cserni has provided In our initial study on 54 breast cancer patients,12 27/31 SN
guidelines.8 metastases were detected by the frozen section procedure.
There are several ways for postoperative and intraopera- Sensitivity was 87%, specifcity 100%, positive predictive
tive SN evaluation. These include traditional histopathologic value 100%, and negative predictive value (NPV) 91%. These

DOI: 10.1201/9781003255536-9 55
56 Clinical Lymphatic Mapping

data compare very well with some other early studies.13–15 recent studies, results were comparable. Rychlik et al. reported
However, in a larger follow-up study, sensitivity dropped to a sensitivity of 81% and NPV of 98% (excluding ITC).31 Balaya
about 60% in our hands when the percentage of lobular carci- et al. described a sensitivity of 42% and NPV of 90%, or 56%
nomas had increased, and frozen sections were also routinely and 94% if ITCs were excluded.32 Dostalek et al. reported a
analyzed by less experienced pathologists, leading to some sensitivity of 47% and NPV of 93% (excluding ITCs).33 These
judgment errors.16 These observations have been confrmed studies indicate that the yield of frozen section analysis in cer-
by others.17,18 Obviously, the ability of the frozen sections to vical cancer is not optimal.
detect metastases is higher for macrometastases than micro- In our view, the disadvantage of potentially missing some
metastases, let alone ITCs.18 Smaller metastases undetected by smaller metastases due to loss of tissue can be outweighed
frozen section that appear in the fnal paraffn H&E or IHC by the advantage of performing full lymph node dissection
sections will usually necessitate lymph node dissection in a during the initial operation in SN-positive cancer patients
second operation. and by the increased number of lymph node metastases
Veronesi et al.19 described a combination of intraoperative found by the more detailed investigation of the SN compared
step frozen sections with rapid IHC to arrive at an intraop- to usual locoregional lymph nodes. The operative delay of
erative fnal diagnosis in breast cancer, correctly predicting a 15–20 min caused by the frozen-section investigation may
metastasis-free SN in 95.4% of cases.20 This procedure was, be acceptable, since during this time the primary tumor can
however, very labor-intensive and led to a signifcant increase be excised.
in the operating time (up to 1 h). Zurrida et al.21 devised a
similar but quicker intraoperative method, requiring about 40
min, in which pairs of sections were taken every 50 µm for
the frst 15 sections and every 100 µm thereafter, sampling the
Imprint cytology
entire node. This extensive intraoperative frozen examination Imprint slides can simply be produced from lymph nodes by
correctly predicted an uninvolved axilla in 95.3% of cases. frmly pressing the cut surface to a glass slide. In a postopera-
Ultraquick IHC protocols are mandatory for this.22 In another tive and intraoperative setting, the May-Grünwald-Giemsa and
study using extensive intraoperative sampling taking up to 60 Quickdiff stains, respectively, allow detection of metastatic
sections per SN,14 H&E section analysis by an experienced cells. Imprint cytology has previously been described as help-
pathologist proved more important than IHC, and this method ful in the detection of lymph node metastases of prostate34 and
had the accuracy of a defnitive histologic examination. The breast cancer35,36 Fisher et al.36 detected breast cancer axillary
frozen-section procedure should not be essentially different lymph node metastases intraoperatively in imprints of 18/21
for cervical adenocarcinomas and vulvar and cervical squa- (86%) patients. In an intraoperative study in breast cancer,12
mous cell carcinomas.23 imprints were prepared from all cut SN surfaces, showing a
In frozen-section analysis, care must be taken that the tis- sensitivity of imprints for detection of SN metastases of 62%
sue is frozen fat (as by freezing it with a cooled fat weight on with a specifcity of 100%. The sensitivity of the imprints was
top of it) and that cutting is performed cautiously to prevent signifcantly lower than that of frozen sections. In addition,
inadvertent tissue loss as much as possible. Intraoperative step there were no SNs in which the imprints convincingly showed
sections are time-consuming and lead to excessive tissue loss, metastases while the frozen sections did not. Therefore, we
and extra sections for the different levels need to be cut imme- found that imprints did not have more value than the intraop-
diately for IHC.19 Nevertheless, it is unavoidable that some SN erative, frozen-section SN investigation.
material will be lost; theoretically, this may lead to missing However, other authors have had much better experience.
some smaller SN metastases. The chance of this occurring Motomura et al.37 described a sensitivity of intraoperative
will naturally depend on the size and the distribution of these imprint cytology of breast cancer SNs of 91%, a result which
metastases. However, since the likelihood of second-echelon was better than H&E frozen-section analysis. Turner et al.38
lymph nodes containing metastases usually correlates to the used a combination of frozen-section analysis and imprints,
size of the SN metastasis,24,25 this risk may be acceptable. The resulting in a high sensitivity. Ratanawichitrasin et al.39
percentage of SN metastases discarded during tissue process- observed an imprint sensitivity of 82% compared to H&E fnal
ing is probably quite low when proper measures to prevent sections (no IHC), a result which is comparable to the experi-
tissue loss are taken, since the percentage of SN metastases ence of Cserni.40 The imprint method therefore seems to be
found in our initial breast cancer study12 was comparable to reasonably useful for intraoperative SN evaluation.
that in other breast cancer studies without an intraoperative Interestingly, the perception of the surgeon of the impor-
procedure.26–28 Moreover, it has to be realized that for the aver- tance of false-negative and false-positive results appears to
age lymph node, only about 1% of the total node volume is be changing. Reasoning from the fact that, traditionally, the
actually looked at under the microscope, even when making complete lymph node dissection is the standard therapy, one
step sections and performing IHC. could argue that false-positive results are not really a prob-
The value of the frozen section has especially been studied lem and that false-negative results are practically a bigger
in cervical cancer. In the recent SENTIX trial, frozen sections problem since a second operation will then be necessary.
failed to detect 54% of positive lymph nodes (pN1), including However, the growing reliability and importance of the SN
28% of cases with macrometastases and 90% with microme- procedure has put more emphasis on the impact of false-
tastases.29 In the cervical cancer study of Roy et al., frozen positive results, since they would lead to an ‘unnecessary’
section analysis missed 42% of the SL metastases.30 In more lymph node dissection.
Ultrastaging of the sentinel node 57

From the average lymph node, several thousand sections can


Serial sectioning be produced. As this obviously confronts the pathologist with
an unacceptably high workload, a compromise has to be found
The SNs are fxed in neutral buffered formaldehyde and com- between workload and sensitivity, which will unavoidably lead
pletely embedded after lamellating them according to their to missing some tumor cells. This compromise is best achieved
size. Various protocols for lamellating have been advocated. by taking step sections at regular intervals, increasing the
Our preferred way is as follows: SNs smaller than 0.5 cm are percentage of metastases found in SNs with about 8%,42,43
processed and paraffn-embedded intact, those between 0.5 depending on the protocol used (Table 9.1). In vulvar squa-
and 1 cm are halved, and those larger than 1 cm are lamel- mous cell cancer,23,44–47 the yield of step sectioning and IHC
lated into pieces of approximately 0.5 cm in size (Figure 9.1). It seems to be lower. In vulvar cancer, De Hullu et al.44 detected
has been suggested that tumor cells preferentially enter the SNs by step sectioning and IHC in 102 sentinel lymph nodes that
through the contralateral side of the hilum of lymph nodes.38 were negative on routine examination 4 additional metastases
If true, this would have important consequences, since bisect- (4%; 95% confdence interval [CI], 1%–9%). Three of the four
ing the long axis of the SN from the outer capsule to the hilum metastases were observed in patients whose lymph nodes were
would produce two cut surfaces of the midline region where the negative on routine examination. One additional metastasis
frst single tumor cell or small groups of tumor cells would be was found in a patient who already had another SN with meta-
readily visible. However, experimental evidence for this theory static disease. In squamous cell cervical cancer, Levenback et
is lacking, and, in practice, it is quite diffcult to identify the al.23 detected in 31 patients with no positive SNs on routine
hilum. Therefore, in our opinion, there is suffcient evidence to H&E processing no micrometastases with serial step section-
recommend this as the current optimal protocol (see Table 9.3). ing. Ten patients with negative SNs on routine H&E and serial
Future studies may help to make SN sampling more effcient step sectioning had their SNs submitted to cytokeratin IHC
when the hilum approach is better validated. It remains there- analysis. No metastases were identifed with this technique.
fore common practice to lamellate perpendicular to the long One patient with a positive SN on routine H&E staining had a
axis to allow for maximum visualization of the SN surface.11,41 micrometastasis found by serial sectioning of the contralateral
sentinel node.23
In endometrial cancer, ‘ultrastaging’ protocols increased the
number of patients with lymph node metastases from 10.6% to
15.2%, not dissecting the yield from step sections and IHC.48
Step sections are obviously necessary only when the frst
section appears to be tumor negative. In cutting the step rib-
bons, one section of each ribbon should be mounted for
H&E staining, while the rest of the ribbon should be saved
for eventual IHC (see later). Although mucin histochemical
stains (periodic acid–Schiff [PAS] and Alcian blue) may be
useful for detecting adenocarcinoma metastases, especially in
primary lobular breast cancer, IHC is a much more sensitive
method (see later).

Immunohistochemistry
FIGURE 9.1 Lymph node metastasis at routine histopathologic exami-
nation (hematoxylin and eosin staining, ×200 microscope magnifcation). IHC is a generally applicable and cost-effective technique that
has proven to be very useful for detection of tumor cells in
TABLE 9.1 lymph nodes (especially micrometastases and ITCs) by dem-
onstration of proteins that are, within the context of the tissue
Cumulative Number of Patients with Breast Cancer Sentinel Node
analyzed, tumor specifc (Figures 9.2 and 9.3). Overall, IHC
Metastases Found with Each Additional Level (250-mm Intervals)
increases the percentage of metastases found in non-SN lymph
in a Total of 86 Patients
nodes by, on average, 20%,1 the gain being highest in lobular
H&E % H&E IHC % IHC breast cancer. For the SN, the range of conversion of the SN
  positive positive positive positive
to positive by ultrastaging (however, with quite varying proto-
Level 1 69 80 74 86 cols) is 2%–20%, with an average of 11% (Table 9.2).
Level 2 71 83 77 90 Different antibodies have been used for breast cancer, of
Level 3 73 85 81 94 which CAM5.2 seems to be the one most widely used. In gen-
Level 4 75 87 84 98 eral, little background staining is seen with this antibody, and
Level 5 76 88 86 100 its sensitivity is about 100%. Caveats are epithelial and meso-
thelial inclusions and macrophages that have eaten keratin.
Abbreviations: HE, hematoxylin/eosin staining; IHC, CAM5.2 immunohisto- Staining of sinus-lining (dendritic) cells may sometimes be
chemistry.
observed, especially when using automated immunostainers,
Source: Adapted from Torrenga et al.42 but sinus-lining cells are easily recognized by their dendritic
58 Clinical Lymphatic Mapping

TABLE 9.2
Overview of Different Gynecologic Cancer Studies Evaluating
the Conversion Rate of SNs to Positive by IHC
No. of % Converted to
patients positive by
Study Organ converted IHC
de Hullu 2000 44 36 vulva 3/23 13
Terada 200049 73 vulva 2/14 4
Molpus 200150 75 vulva 2/18 11
Levenback 200047 21 cervix 0/31 0
Moore 2003 51 29 vulva 0/29 0
Van der Zee 200852 403 vulva 68/163 42
Euscher 200853 48 cervix 1/33 19
FIGURE 9.2 Lymph node metastasis confrmed with immunohisto- Devaja 201154 60 vulva 0/5 0
pathologic examination (AE1/3 immunohistochemical staining ×100 Ballester 201155 125 endometrium 8/16 50
microscope magnifcation).
Levenback 201256 418 vulva 30/132 23
Koskas 201357 187 endometrium 18/38 47
Kim 201358 508 endometrium 23/64 36
Naoura 201559 180 endometrium 17/41 41
Touhami 2015 60 268 endometrium 19/43 44
Salvo 201761 188 cervix 6/27 22
Overall 197/677 29%

Fine-needle aspiration cytology


A few studies have explored the possibility of ultrasonographi-
cally guided fne-needle aspiration biopsy (FNAB) to detect SN
metastases. Motomura et al.64 examined the axillae of 60 patients
with breast cancer, in whom a hot spot was detected by gamma
probe, by ultrasonography. Preoperative diagnosis of SN metasta-
FIGURE 9.3 Scattered keratin-positive cells in a sentinel lymph node. The sis by gamma probe and ultrasonographically guided FNAB was
clinical signifcance of this fnding in gynecologic cancers is not known compared with the histologic results of SN. SNs were visualized
(AE1/3 immunohistochemical staining ×400 microscope magnifcation). by ultrasonography in 29 of 60 patients (48%). The sensitivity,
specifcity, and overall accuracy of ultrasonography in the diag-
nosis of SN metastasis were 50%, 92%, and 77%, respectively.
Of 14 patients with positive results by ultrasonography, 4 had
morphology. CAM5.2 is also useful for metastases of adeno- positive and 2 had negative cytology. The combination of ultra-
carcinomas from most other sites. EMA and other MUC1 anti- sonography and ultrasonographically guided FNAB for visu-
bodies have too low a specifcity, since they may also stain alized nodes had a sensitivity of 79%, specifcity of 93%, and
plasma cells and macrophages, and the sensitivity of EMA is overall accuracy of 86%. Blind FNAB in the hot spot was not
also low, since breast cancers and other adenocarcinomas can useful in the detection of SN metastasis in patients whose SNs
well be EMA negative. CEA and NCRC11 are specifc but not failed to be detected by ultrasonography. Gamma probe and ultra-
very sensitive. sonographically guided FNAB is therefore a potentially useful
In the overview of Dundr et al. on cervical cancer, lymph method for preoperative detection of SN metastasis. In patients
node involvement was detected only after ultrastaging in 21% with positive SN FNAB, further SN biopsy is no longer indicated,
of patients.62 and complete axillary lymph node dissection can be performed as
For squamous cell carcinoma metastases, AE1/3 is a reli- a primary procedure. One feasibility study on ultrasound-guided
able antibody. It has high specifcity and sensitivity. Van den FNAB in vulvar cancer was recently published showing agree-
Brekel et al.63 found additional non-SN lymph node metasta- ment with fnal histology in 37 of 43 cases (86%), with sensitivity
ses in 3/13 head and neck squamous cell carcinoma patients and specifcity 77% and 100%, respectively.65
(23%) with this antibody. This antibody can also be used for
squamous gynecologic cancer; however, its yield appears
to be modest, as described in the previous paragraph. For
Flow cytometry
melanoma, a combination of a sensitive marker (S100) and
a specifc marker (Melan A, MART1, tyrosinase) can be Flow cytometry (FCM) is a technique of measuring the proper-
recommended. ties of cells in suspension after staining with a fuorescent dye.
Ultrastaging of the sentinel node 59

It is a fast technique in which thousands of cells can be (54%) lymph nodes that were negative on regular histology.
screened in minutes. The frst FCM approach for the detection Wascher et al.,74 using MAGE-A3 RT-PCR, found 28 of 73
of metastases in lymph nodes is with the same antibodies as (38%) histopathologically negative SNs to be RT-PCR positive.
for IHC, coupled with a fuorescent tag. However, such FCM Manzotti et al.75 found a high prevalence of positive RT-PCR in
is not very suitable for detecting rare events, so the additional histologically uninvolved SNs when considering single mark-
value to extensive histopathology is probably very limited. The ers, but when at least two of three markers (maspin, cytokera-
second approach is DNA FCM, in which a clone of cells with tin 19, and mammaglobin) were expressed, the concordance
abnormal DNA content or a high percentage of S-G2M-phase with either SN or axillary lymph node status was highest.75
cells may indicate metastatic cells. Joensuu et al.66 found the Some authors76 use RT-PCR routinely. For vulvar cancer, there
diagnostic accuracy of such DNA FCM analysis of fne-needle are at present no data on the value of RT-PCR. Van Trappen
aspirations of lymph nodes to be 92%, with a sensitivity of et al.77 applied a fully quantitative, real-time RT-PCR assay
91% and a specifcity of 95%. DNA FCM suggested the cor- to document absolute copy numbers of the epithelial marker
rect diagnosis in fve of the seven cytologically false-negative cytokeratin 19 in primary tumors, 156 lymph nodes from 32
cases and in nine of the twelve cytologically indeterminate or patients with cervical cancer (stages IA2, IB1, and IB2), and
suspicious cases. 32 lymph nodes from nine patients with benign disease. All
A more sophisticated approach is a DNA/cytokeratin primary tumors and histologically involved lymph nodes (six)
double-staining procedure, as described by Leers et al.67 In had increased expression of cytokeratin 19 mRNA, while
this study, the breast cancer paraffn pieces that were left over lower expression of cytokeratin 19 was detected in 66 (44%)
between step ribbons were used to prepare single-cell suspen- of 150 histologically uninvolved lymph nodes and in nodes
sions with preservation of the cytoplasm, so that epithelial from 16 of 32 patients with cervical cancer. Fifteen of these
cells could then be identifed by cytokeratin staining. 16 patients with evidence of micrometastases had the highest
The presence of 1% epithelial cells was very indicative of cytokeratin 19 transcription level in a frst lymph node drain-
metastases, but cell cycle parameters of the epithelial fraction age station. Transcription of cytokeratin 19 was found at a low
(DNA aneuploidy or a high percentage of S-phase) provided level in just 1 of 32 lymph nodes obtained from nine patients
further evidence. By this approach, metastases were found that with benign disease. The median copy number of cytokeratin
were not detected by H&E and IHC. 19 transcription was signifcantly higher in association with
adverse prognostic features. These results suggest that about
50% of early-stage cervical cancers shed tumor cells to the
pelvic lymph nodes, and the amount of cytokeratin 19 expres-
Molecular techniques sion was related to clinicopathologic features. However, fur-
An even more refned molecular biologic approach to the ther studies are required to document the clinical implications
detection of micrometastases is amplifcation by reverse- of molecular micrometastases.77
transcriptase polymerase chain reaction (RT-PCR) of messen- These studies underline the great promise of RT-PCR for
ger RNA (mRNA), which is expressed in the cancer cells of detecting SN metastases. However, several comments have
interest, but not by other lymph node cells. By dilution experi- to be made. First, a clear drawback of all of these studies is
ments,68,69 it has been estimated that a single cancer cell can that those pieces that were examined had not been subjected
be detected by RT-PCR among 106 –107 normal cells. This to the usual histopathologic investigation with H&E and IHC.
indicates that RT-PCR is by far the most sensitive method for If this had been done, more metastases might well have been
detection of metastases in SNs. detected with these conventional methods as well. Concluding
Different studies have evaluated the usefulness of RT-PCR that the molecular analysis is superior is therefore tricky. A
for detecting SN metastases. Schoenfeld et al.70 compared ker- better approach seems to be to subject those pieces to molecu-
atin 19 RT-PCR with histopathologic results (H&E and IHC) lar analysis that would normally be thrown away: the pieces
in axillary lymph nodes of breast cancer patients and found all between the step sections. However, this requires that RNA
18 histopathologically involved lymph nodes to be positive by be successfully extracted from paraffn-embedded tissue. This
RT-PCR. Of the 39 lymph nodes that were histologically nega- appears to be possible, as was shown by Palmieri et al. for mel-
tive, 14 were positive by RT-PCR. Noguchi et al.68,71 compared anoma,78 although the sensitivity of paraffn RT-PCR may be
MUC1 and keratin 19 RT-PCR and found all ten lymph nodes lower than that of frozen tissue. Second, contamination is a big
that were histopathologically positive to be also positive by potential problem, and histopathologic control of specimens
RT-PCR and that three (6%) and fve (9%) of histopathologi- homogenized for blind assays such as RT-PCR will always
cally negative (H&E only) lymph nodes expressed MUC1 and be necessary. A pitfall is epithelial or mesothelial inclusions
keratin 19, respectively, indicating the presence of metastases. and displaced benign cells.6 Third, healthy volunteers appear
Hoon et al.69 found occult metastases by beta–human chori- to express presumed specifc markers, such as CEA, CK-19,
onic gonadotropin (ß-HCG) RT-PCR in 25% of patients that GA733.2, and MUC-1,79 in their blood and lymph nodes,
were histopathologically lymph node negative, and Kataoka meaning that these markers can yield false-positive results.
et al.72 found conversion percentages of 25% for CEA and 21% Fourth, one must ask whether the high sensitivity of RT-PCR
for mammoglobin in an H&E-only study. In this last study, has clinical signifcance. As shown in several studies, second-
RT-PCR improved the prediction of axillary lymph node sta- echelon breast cancer metastases are hardly ever found when
tus to 98.5%. In a study on breast and gastrointestinal cancer, the true SN is negative by extensive histopathologic investiga-
Mori et al.73 detected metastases by CEA RT-PCR in 47/87 tion with step sections and IHC.28 It remains to be proven that
60 Clinical Lymphatic Mapping

RT-PCR may identify those few patients who escape extensive When we take the cumulative total of detected metastases
histopathologic investigation, but, especially for melanoma, at level 5 to be 100%, the percentage of SN-positive patients
there is promise. increased from 80%, 83%, 85%, 87% to 88% in the H&E
However, overall, RT-PCR may be a relatively quick and sections through levels 1–5, and with IHC from 86%, 90%,
cheap alternative,80 so further studies are necessary to develop 94%, 98% to 100%. With a similar protocol, an even higher
combinations of primers that are specifc enough to estab- conversion rate was found by Dowlatshahi et al.89 The clini-
lish the role of RT-PCR in detecting metastases in SNs and cal relevance of fnding micrometastases on the higher levels
to assess the clinical value of RT-PCR. Promising frst results is underlined by the fact that three of nine patients in whom
have been described.81 single-cell metastases were detected only at levels 3–5 had
One-step nucleic acid amplifcation (OSNA) is a rapid CK19 metastases in the subsequent axillary lymph node dissec-
mRNA PCR that can be applied intraoperatively.82 Fanfani et tion. Besides, we have seen several breast cancer cases with
al. studied OSNA for the detection of SN metastasis in women only a few metastatic cells in the SN detected by IHC, while
with endometrial cancer. OSNA detected a higher rate of second-echelon lymph nodes contained signifcant meta-
micrometastasis and a lower rate of macrometastasis and ITC static deposits.24,90 This is probably due to rerouting of the
when compared with standard ultrastaging, not allowing for lymph drainage because of lymph obstruction of the ‘real’
conclusions on the validity of this technique in endometrial SN by the heavy tumor load. Others have also reported rela-
cancer.83 tively high percentages of involved non-SNs in SN microme-
tastases.91 IHC is therefore essential when the initial H&E
SN (step) sections are all negative, and ribbons must be kept
when step-cutting.
The optimal standard protocol
Lamellate perpendicular to the long axis allows for maximum
visualization of the SN surface.11,41 Further, there is not much
consensus among different groups as to how many step sec- Summary remarks
tions are needed and what the step size should be.41 Breast
Ultrastaging, including step H&E-stained sections and IHC,
cancer studies have provided SN data that are useful to arrive
is mandatory for reliable detection of metastases in SNs of
at evidence-based guidelines in this respect.28,38,42,43 Turner et
gynecologic cancers. Intraoperative single H&E frozen-
al.38 examined 60 SNs by step H&E sections and cytokeratin
section and imprint cytologic analysis of SNs has been
IHC at ten levels separated by 40 µm, following the hilum
shown to be reasonably reliable for the detection of breast
approach. Levels 1 and 2 yielded additional micrometas-
cancer metastases, but a false-negative rate of 30%–40%
tases in nine SNs (15%), but in levels 3–10, only two (3%)
will have to be accepted. These are all standard techniques
further metastases were found. They therefore recommended
that are available in each pathology laboratory, so nothing
the study of only two levels separated by 40 µm. However,
special, except the dedication of the pathologist, is required.
their SN slices were 2–3 mm thick, so even with ten levels at
Although the workload for an SN is high for the pathologist,
40 µm, only 400 µm would be investigated, accounting for
it should be kept in mind that there will be fewer axillary
no more than 13%–20% of the SN slices. This seems to be
lymph node dissection specimens to handle, saving time.
insuffcient and may explain the disappointing yield of this
Altogether, the SN procedure may save costs.13 Further stud-
procedure. Rather than taking many sections at small inter-
ies are necessary to establish the role of FCM and sophis-
vals, it may be more effcient to take fewer sections at larger
ticated molecular biologic techniques such as RT-PCR in
step intervals. In the study of Cserni,4 SNs were serially sec-
detecting SN metastases. Table 9.3 summarizes our current
tioned and every 10th to 20th level was examined by H&E
recommendations for the SN evaluation and ultrastaging in
and/or IHC. A central cross-section through the SN would
gynecologic cancers.
have failed to detect metastases in 8/26 lymph nodes (31%),
leading to a false-negative SN status in 6/21 patients (29%).
The percentage of metastases found increased from 69% with
only a central cross-section to 77% with 5 further steps, to
81% with 10 steps, and to 96% with 15 steps. Only at 45 steps TABLE 9.3
was a 100% sensitivity found. Cserni stated that with a three- Recommendations for the SN Evaluation (IHC:
level approach at 25%, 50%, and 75% of the block, metastases immunohistochemistry) [see further 1,41]
would have been missed in 15% of patients. A previous non- Adenocarcinoma Single H&E frozen-section analysis and/or
SN study by Zhang et al.84 found almost all metastases with (cervix) imprint cytology. H&E step sections (fve with
such a three-level approach. Our own SN protocol includes 200–250 µm interval) with CAM5.2 IHC.
step sectioning at fve levels with an interval of 250 µm with Squamous cell cancer Single H&E frozen-section analysis with
H&E and IHC when the frst-level H&E section is nega- (vulva, cervix, vagina) imprint cytology. H&E step sections (fve
tive.1,2,10,12,42,43,85–88 Since we perform frozen-section analysis with 200–250 µm interval) with AE1/3 IHC.
routinely, leading to some loss of material, this ensures sam- Melanoma (vulva) Single H&E frozen-section analysis with
pling through the better part of the SN. In practice, this has imprint cytology. H&E step sections (two to
proven to provide an acceptable workload. The yield per level fve with 150 µm interval) with S100/Melan
A or IHC.
has been detailed, as shown in Table 9.1.42
Ultrastaging of the sentinel node 61

REFERENCES 21. Zurrida S, Mazzarol G, Galimberti V, et al. The problem of


the accuracy of intraoperative examination of axillary senti-
1. Van Diest PJ, Peterse HL, Borgstein PJ, et al. Pathologic
nel nodes in breast cancer. Ann Surg Oncol. 2001;8:817–20.
investigation of sentinel lymph nodes. Eur J Nuclear Med.
22. Richter T, Nährig J, Komminoth P, et al. Protocol for ultra-
1999;26:S43–A9.
rapid immunostaining of frozen sections. J Clin Pathol.
2. Van Diest PJ. Histopathologic workup of sentinel lymph
1999;52:461–3.
nodes: how much is enough? J Clin Pathol. 1999;52:871–3.
23. Levenback C, Coleman RL, Burke TW, et al. Lymphatic
3. Cserni G. How to improve low lymph node recovery rates
mapping and sentinel node identifcation in patients with
from axillary clearance specimens of breast cancer. A
cervix cancer undergoing radical hysterectomy and pelvic
short-term audit. J Clin Pathol. 1998;51:846–9.
lymphadenectomy. J Clin Oncol. 2002;20:688–93.
4. Cserni G. The reliability of sampling three to six nodes for
24. Rahusen FD, Van Diest PJ, Meijer S. Re: Chu et al; Do all
staging breast cancer. J Clin Pathol. 1999;52:681–3.
patients with sentinel node metastasis from breast carci-
5. Cserni G. Axillary staging of breast cancer and the sentinel
noma need complete axillary node dissection? Ann Surg.
node. J Clin Pathol. 2000;53:733–41.
2000;231:615–16.
6. van Deurzen CH, de Bruin PC, Koelemij R, et al. Isolated
25. Chu KU, Turner RR, Hansen NM, et al. Do all patients with
tumor cells in breast cancer sentinel lymph nodes: displace-
sentinel node metastasis from breast carcinoma need com-
ment or metastases? An immunohistochemical study. Hum
plete axillary node dissection? Ann Surg. 1999;229:536–41.
Pathol. 2009;40:778–82.
26. Giuliano AE. Sentinel lympadenectomy in primary breast
7. Liebens F, Carly B, Cusumano P, et al. Breast cancer seed-
carcinoma: an alternative to routine axillary dissection. J
ing associated with core needle biopsies: a systematic
Surg Oncol. 1996;62:75–6.
review. Maturitas. 2009;62:113–23.
27. Veronesi U, Paganelli G, Galimberti V, et al. Sentinel-node
8. Cserni G, Bianchi S, Boecker W, et al. European Working
biopsy to avoid axillary dissection in breast cancer with
Group for Breast Screening Pathology Improving the
clinically negative lymph-nodes. Lancet. 1997;349:1864–7.
reproducibility of diagnosing micrometastases and isolated
28. Turner RR, Ollila DW, Krasne DL, et al. Histopathologic
tumor cells. Cancer. 2005;103(2):358–67.
validation of the sentinel lymph node hypothesis for breast
9. Cserni G. Histopathologic examination of the sentinel
carcinoma. Ann Surg. 1997;226:271–8.
lymph nodes. Breast J. 2006;12(5 Suppl 2):S152–6.
29. Cibula D, Kocian R, Plaikner A et al. Sentinel lymph node
10. Van Diest PJ, Torrenga H, Meijer S, et al. Pathologic
mapping and intraoperative assessment in a prospective, inter-
analysis of sentinel lymph nodes. Semin Surg Oncol.
national, multicentre, observational trial of patients with cervi-
2001;20:238–45.
cal cancer: the SENTIX trial. Eur J Cancer. 2020;137:69–80.
11. Burg LC, Hengeveld EM, In‘t Hout J, et al. Ultrastaging
30. Roy M, Bouchard-Fortier G, Popa I, et al. Value of senti-
methods of sentinel lymph nodes in endometrial cancer –
nel node mapping in cancer of the cervix. Gynecol Oncol.
a systematic review. Int J Gynecol Cancer. 2020: Online
2011;122:269–74.
ahead of print.
31. Rychlik A, Angeles MA, Migliorelli F, et al. Frozen sec-
12. Van Diest PJ, Torrenga H, Borgstein PJ, et al. Reliability
tion examination of sentinel lymph nodes can be used as a
of intra-operative frozen section and imprint cytologi-
decisional tool in the surgical management of early cervical
cal investigation of sentinel lymph nodes in breast cancer.
cancer. Int J Gynecol Cancer. 2020;30(3):358–63.
Histopathology. 1999;35:14–18.
32. Balaya V, Guani B, Benoit L, et al. Diagnostic value of fro-
13. Flett MM, Going JJ, Stanton PD, et al. Sentinel node localiza-
zen section examination of sentinel lymph nodes in early-
tion in patients with breast cancer. Br J Surg. 1998;85:991–3.
stage cervical cancer at the time of ultrastaging. Gynecol
14. Galimberti V, Zurrida S, Intra M, et al. Sentinel node biopsy
Oncol. 2020;158(3):576–83.
interpretation: the Milan experience. Breast J. 2000;6:306–9.
33. Dostalek L, Slama J, Fisherova D, et al. Impact of sentinel
15. Viale G, Bosari S, Mazzarol G, et al. Intraoperative exami-
lymph node frozen section evaluation to avoid combined
nation of axillary sentinel lymph nodes in breast carcinoma
treatment in early-stage cervical cancer. Int J Gynecol
patients. Cancer. 1999;85:2433–8.
Cancer. 2020;30(6):744–8.
16. Rahusen FD, Pijpers R, Van Diest PJ, et al. The implemen-
34. Gentry JF. Pelvic lymph node metastases in prostatic car-
tation of the sentinel node biopsy as a routine procedure for
cinoma. The value of touch imprint cytology. Am J Surg
patients with breast cancer. Surgery. 2000;128:6–12.
Pathol. 1986;10:718–27.
17. Gulec SA, Su J, O’Leary JP, et al. Clinical utility of frozen
35. Hadjiminas DJ, Burke M. Intraoperative assessment of
section in sentinel node biopsy in breast cancer. Am Surg.
nodal status in the selection of patients with breast cancer
2001;67:529–32.
for axillary clearance. Br J Surg. 1994;81:1615–16.
18. Weiser MR, Montgomery LL, Susnik B, et al. Is routine
36. Fisher CJ, Boyle S, Burke M, et al. Intraoperative assess-
intraoperative frozen-section examination of sentinel
ment of nodal status in the selection of patients with breast
lymph nodes in breast cancer worthwhile? Ann Surg Oncol.
cancer for axillary clearance. Br J Surg. 1993;80:457–8.
2000;7:651–5.
37. Motomura K, Inaji H, Komoike Y, et al. Intraoperative
19. Veronesi U, Zurrida S, Galimberti V. Consequences of
sentinel lymph node examination by imprint cytology
sentinel lymph node in clinical decision making in breast
and frozen sectioning during breast surgery. Br J Surg.
cancer and prospects for future studies. Eur J Surg Oncol.
2000;87:597–601.
1998;24:93–5.
38. Turner RR, Ollila DW, Stern S, et al. Optimal histopatho-
20. Veronesi U, Zurrida S, Mazzarol G, et al. Extensive frozen
logic examination of the sentinel lymph node for breast car-
section examination of axillary sentinel nodes to determine
cinoma staging. Am J Surg Pathol. 1999;23:263–7.
selective axillary dissection. World J Surg. 2001;25:806–8.
62 Clinical Lymphatic Mapping

39. Ratanawichitrasin A, Biscotti CV, Levy L, et al. Touch squamous cell carcinoma of the vulva: a Gynecologic
imprint cytological analysis of sentinel lymph nodes for Oncology Group study. J Clin Oncol. 2012;30:3786–91.
detecting axillary metastases in patients with breast cancer. 57. Koskas M, Chereau E, Ballester M, et al. Accuracy of
Br J Surg. 1999;86:1346–8. a nomogram for prediction of lymph-node metastasis
40. Cserni G. The potential value of intraoperative imprint detected with conventional histopathology and ultrastaging
cytology of axillary sentinel lymph nodes in breast cancer in endometrial cancer. Br J Cancer. 2013;108:1267–72.
patients. Am Surg. 2001;67:86–91. 58. Kim CH, Soslow RA, Park KJ, et al. Pathologic ultrastag-
41. Euscher ED, Malpica A. Gynaecological malignancies and ing improves micrometastasis detection in sentinel lymph
sentinel lymph node mapping: an update. Histopathology. nodes during endometrial cancer staging. Int J Gynecol
2020;76:139–50. Cancer. 2013;23:964–70.
42. Torrenga H, Rahusen FD, Meijer S, et al. Sentinel node 59. Naoura I, Canlorbe G, Bendifallah S, et al. Relevance of
in breast cancer: detailed analysis of the yield from step sentinel lymph node procedure for patients with high-risk
sectioning and immunohistochemistry. J Clin Pathol. endometrial cancer. Gynecol Oncol. 2015;136:60–4.
2001;54:553–5. 60. Touhami O, Trinh XB, Gregoire J, et al. Predictors of non-
43. Torrenga H, Diest PJ van, Meijer S. Sentinel-node biopsy in sentinel lymph node (non-SLN) metastasis in patients with
breast cancer. Lancet. 2001;358:1814. sentinel lymph node (SLN) metastasis in endometrial can-
44. de Hullu JA, Hollema H, Piers DA, et al. Sentinel lymph cer. Gynecol Oncol. 2015;138:41–5.
node procedure is highly accurate in squamous cell carci- 61. Salvo G, Ramirez PT, Levenback CF, et al. Sensitivity and
noma of the vulva. J Clin Oncol. 2000;18:2811–16. negative predictive value for sentinel lymph node biopsy in
45. de Hullu JA, Doting E, Piers DA, et al. Sentinel lymph women with early-stage cervical cancer. Gynecol Oncol.
node identifcation with technetium-99m-labeled nano- 2017;145:96–101.
colloid in squamous cell cancer of the vulva. J Nucl Med. 62. Dundr P, Cibula D, Němejcová K, et al. Pathologic proto-
1998;39:1381–5. cols for sentinel lymph nodes ultrastaging in cervical can-
46. de Hullu JA, Piers DA, Hollema H, et al. Sentinel lymph cer. Arch Pathol Lab Med. 2020;144(8):1011–20.
node detection in locally recurrent carcinoma of the vulva. 63. Van den Brekel MWM, Stel HV, van der Valk P, et al.
Br J Obstet Gynaecol. 2001;108:766–8. Micrometastases from squamous cell carcinoma in
47. Levenback C. Intraoperative lymphatic mapping and sen- neck dissection specimens. Eur Arch Otorhinolaryngol.
tinel node identifcation: gynecologic applications. Recent 1992;249:349–53.
Results Cancer Res. 2000;157:150–8. 64. Motomura K, Inaji H, Komoike Y, et al. Gamma probe and
48. Euscher E, Sui DW, Soliman P, et al. Ultrastaging of sentinel ultrasonographically-guided fne-needle aspiration biopsy
lymph nodes in endometrial carcinoma according to use of of sentinel lymph nodes in breast cancer patients. Eur J
2 different methods. Int J Gynecol Pathol. 2018;37:242–51. Surg Oncol. 2001;27:141–5.
49. Terada KY, Shimizu DM, Wong JH. Sentinel node dissec- 65. Angelico G, Santoro A, Inzani F, et al. Ultrasound-guided
tion and ultrastaging in squamous cell cancer of the vulva. FNA cytology of groin lymph nodes improves the manage-
Gynecol Oncol. 2000;76:40–4. ment of squamous cell carcinoma of the vulva: results from
50. Molpus KL, Kelley MC, Johnson JE, et al. Sentinel lymph a comparative cytohistological study. Cancer Cytopathol.
node detection and micro-staging in vulvar carcinoma. J 2019;127:514–20.
Reprod Med. 2001;46:863–9. 66. Joensuu H, Klemi PJ, Eerola E. Flow cytometric DNA
51. Moore RG, Granai CO, Gajewski W, et al. Pathologic eval- analysis combined with fne needle aspiration biopsy in
uation of inguinal sentinel lymph nodes in vulvar cancer the diagnosis of palpable metastases. Analy Quant Cytol
patients: a comparison of immunohistochemical staining Histol. 1988;10:256–60.
versus ultrastaging with hematoxylin and eosin staining. 67. Leers M, Schoffelen R, Theunissen P, et al. Multiparameter
Gynecol Oncol. 2003;91(2):378–82. fow cytometry (MP-FCM) as a tool for the detection of
52. Van der Zee AG, Oonk MH, De Hullu JA, et al. Sentinel micrometastatic tumour cells in the sentinel lymph node
node dissection is safe in the treatment of early-stage vulvar procedure. J Clin Pathol. 2002;55:359–66.
cancer. J Clin Oncol. 2008;26;884–9. 68. Noguchi S, Aihara T, Motomura K, et al. Detection of
53. Euscher ED, Malpica A, Atkinson EN, et al. Ultrastaging breast cancer micrometastases in axillary lymph nodes by
improves detection of metastases in sentinel lymph nodes of means of reverse transcriptase-polymerase chain reaction.
uterine cervix squamous cell carcinoma. Am J Surg Pathol. Comparison between MUC1 mRNA and keratin 19 mRNA
2008;32:1336–43. amplifcation. Am J Pathol. 1996;148:649–56.
54. Devaja O, Mehra G, Coutts M, et al. A prospective study 69. Hoon DSB, Sarantou T, Doi F, et al. Detection of metastatic
of sentinel lymph node detection in vulval carcinoma: is it breast cancer by ß-hcg polymerase chain reaction. Int J
time for a change in clinical practice? Int J Gynecol Cancer. Cancer. 1996;69:369–74.
2011;21:559–64. 70. Schoenfeld A, Luqmani Y, Smith D, et al. Detection of breast
55. Ballester M, Dubernard G, Lecuru F, et al. Detection rate cancer micrometastases in axillary lymph nodes by using
and diagnostic accuracy of sentinel-node biopsy in early polymerase chain reaction. Cancer Res. 1994;54:2986–90.
stage endometrial cancer: a prospective multicentre study 71. Noguchi S, Tomohiko A, Motomura K, et al. Detection of
(SENTIENDO). Lancet Oncol. 2011;12:469–76. breast cancer micrometastases in axillary lymph nodes by
56. Levenback CF, Ali S, Coleman RL, et al. Lymphatic means of reverse transcriptase-polymerase chain reaction.
mapping and sentinel lymph node biopsy in women with Cancer. 1994;74:1595–600.
Ultrastaging of the sentinel node 63

72. Kataoka A, Mori M, Sadanaga N, et al. RT-PCR detection the detection of lymph node metastases in breast cancer. Int
of breast cancer cells in sentinel lymph nodes. Int J Oncol. J Cancer. 2008;122(11):2562–7.
2000;16:1147–52. 83. Fanfani F, Monterossi G, Di Meo ML, et al. Standard ultra-
73. Mori M, Mimori K, Inoue H, et al. Detection of cancer staging compared to one-step nucleic acid amplifcation for
micrometastases in lymph nodes by reverse transcriptase- the detection of sentinel lymph node metastasis in endome-
polymerase chain reaction. Cancer Res. 1995;55:3417–20. trial cancer patients: a retrospective cohort comparison. Int
74. Wascher RA, Bostick PJ, Huynh KT, et al. Detection of J Gynecol Cancer. 2020;30(3):372–7.
MAGE-A3 in breast cancer patients’ sentinel lymph nodes. 84. Zhang PJ, Reisner RM, Nangia R, et al. Effectiveness of
Br J Cancer. 2001;85:1340–6. multiple level sectioning in detecting axillary nodal micro-
75. Manzotti M, Dell’Orto P, Maisonneuve P, et al. Reverse metastasis in breast cancer. A retrospective study with
transcription-polymerase chain reaction assay for multiple immunohistochemical analysis. Arch Pathol Lab Med.
mRNA markers in the detection of breast cancer metastases 1998;122:687–90.
in sentinel lymph nodes. Int J Cancer. 2001;95:307–12. 85. Borgstein PJ, Pijpers R, Comans EF, et al. Sentinel lymph
76. Ishida M, Kitamura K, Kinoshita J, et al. Detection of node biopsy in breast cancer: guidelines and pitfalls of lym-
micrometastasis in the sentinel lymph nodes in breast can- phoscintigraphy and gamma probe detection. J Am Coll
cer. Surgery. 2002;131(1 Pt 2):S211–16. Surg. 1998;186:275–83.
77. Van Trappen PO, Gyselman VG, Lowe DG, et al. Molecular 86. Borgstein PJ, Meijer S, Pijpers R, et al. Functional lym-
quantifcation and mapping of lymph-node micrometasta- phatic anatomy for sentinel node biopsy in breast cancer:
ses in cervical cancer. Lancet. 2001;357:15–20. echoes from the past and the periareolar blue method. Ann
78. Palmieri G, Ascierto PA, Cossu A, et al. Detection of occult Surg. 2000;232:81–9.
melanoma cells in paraffn-embedded histologically nega- 87. Pijpers R, Meyer S, Hoekstra OS, et al. Impact of lympho-
tive sentinel lymph nodes using a reverse transcriptase poly- scintigraphy on sentinel node identifcation with techne-
merase chain reaction assay. J Clin Oncol. 2001;19:1437–43. tium-99m-colloidal albumin in breast cancer. J Nucl Med.
79. Bostick PJ, Chatterjee S, Chi DD, et al. Limitations of 1997;38:366–8.
specifc reverse-transcriptase polymerase chain reac- 88. Pijpers R, Borgstein PJ, Meijer S, et al. Sentinel node biopsy
tion markers in the detection of metastases in the lymph in melanoma patients: dynamic lymphoscintigraphy fol-
nodes and blood of breast cancer patients. J Clin Oncol. lowed by intraoperative gamma probe and vital blue guid-
1998;16:2632–40. ance. World J Surg. 1997;21:788–92.
80. van der Velde-Zimmermann D, Schipper ME, de Weger 89. Dowlatshahi K, Fan M, Bloom KJ, et al. Occult metas-
RA, et al. Sentinel node biopsies in melanoma patients: a tases in the sentinel lymph nodes of patients with early
protocol for accurate, effcient, and cost-effective analysis stage breast carcinoma: a preliminary study. Cancer.
by preselection for immunohistochemistry on the basis of 1999;86:990–6.
Tyr-PCR. Ann Surg Oncol. 2000;7:51–4. 90. Rahusen FD, Torrenga H, van Diest PJ, et al. Predictive
81. Bostick PJ, Huynh KT, Sarantou T, et al. Detection factors for metastatic involvement of nonsentinel nodes in
of metastases in sentinel lymph nodes of breast can- patients with breast cancer. Arch Surg. 2001;136:1059–63.
cer patients by multiple-marker RT-PCR. Int J Cancer. 91. Turner RR, Chu KU, Qi K, et al. Pathologic features asso-
1998;79:645–51. ciated with nonsentinel lymph node metastases in patients
82. Visser M, Jiwa M, Horstman A, et al. Intra-operative rapid with metastatic breast carcinoma in a sentinel lymph node.
diagnostic method based on CK19 mRNA expression for Cancer. 2000;89:574–81. Run ning Head Right- y of the vulva
10
Sentinel lymph node biopsy of the vulva

Maaike Oonk and Ate G.J. van der Zee

CONTENTS
Introduction .................................................................................................................................................................................... 65
Safety of the sentinel node procedure ............................................................................................................................................ 65
Can we extend the indication for sentinel node biopsy in vulvar cancer? ..................................................................................... 67
New directions in sentinel lymph node detection .......................................................................................................................... 68
Sentinel node metastasis ................................................................................................................................................................ 68
Conclusions .................................................................................................................................................................................... 69
References...................................................................................................................................................................................... 69

morbidity. The sentinel lymph node procedure was frst intro-


Introduction duced in vulvar cancer in 1994. Levenback et al. published the
frst feasibility study on sentinel lymph node biopsy with only
With an annual incidence of 2–3 per 100,000 women, vulvar isosulfan blue as a tracer and always followed by an inguino-
cancer is the fourth most common gynecologic malignancy. femoral lymphadenectomy.9 After that many small single-center
Squamous cell carcinoma is the most common histological studies were published in which the sentinel node procedure
type, comprising about 80% of all vulvar malignancies. Other was always followed by inguinofemoral lymphadenectomy to
types, like melanomas, adenocarcinomas, and basal cell can- investigate the feasibility and accuracy of this procedure. They
cers, are much less common. In this chapter we will focus on found the procedure was feasible, with a high detection rate,
squamous cell carcinoma of the vulva. For the last few decades especially with the combined technique (radioactive tracer and
the incidence of squamous cell carcinoma of the vulva has blue dye) (Figure 10.1). These studies also showed a high nega-
been increasing, with a more rapid rise in the rates seen in tive predictive value of a negative sentinel node.10,11
women aged <60 years at diagnosis.1
Over the last few decades, many modifcations have been
made in the standard treatment of vulvar cancer, all with the aim
to reduce treatment-related morbidity without compromising
Safety of the sentinel node procedure
survival rates. However, morbidity remained a serious problem
and was especially related to the inguinofemoral lymphadenec- The Groningen International Study on Sentinel Nodes in Vulvar
tomy. In 1977 Cabanas was the frst to introduce the sentinel cancer (GROINSS-V I) was the frst large study on the safety of
node technique in the treatment of patients with penile can- the sentinel node procedure in vulvar cancer and was published
cer.2 Vulvar cancer seemed an ideal candidate for sentinel node in 2008. In this multicenter international study only patients with
biopsy for a variety of reasons. First, most patients present a metastatic sentinel node underwent an inguinofemoral lymph-
with early-stage disease and only 30%–35% of them will have adenectomy. In patients with a negative sentinel node, it was
inguinofemoral lymph node involvement.3–5 Second, because observed whether omission of inguinofemoral lymphadenec-
of the absence of noninvasive imaging techniques that reliably tomy was safe. The eligibility criteria for this study were unifocal
exclude lymph node metastases,6 all patients with macroinva- squamous cell carcinoma of the vulva less than 4 cm in diameter,
sive disease require staging of the groin by sentinel node pro- without suspicious nodes at clinical examination. In 2.3% of the
cedure or lymphadenectomy. The morbidity of this procedure sentinel node–negative patients, groin recurrences were diag-
is signifcant, with frequent wound breakdown, lymphocysts, nosed during follow-up.3 In the same period, the Gynecological
lymphedema of the legs, and recurrent infections.7,8 And last, Oncology Group published their results of the GOG173 study,
since vulvar cancer is a cutaneous malignancy, the lesions are in which the sentinel node biopsy was routinely followed by
easily accessible for peritumoral tracer injection and also the an inguinofemoral lymphadenectomy. They found 2% of false
pattern of lymph drainage from the vulva is fairly predictable; negatives in patients with tumors <4 cm.4 After the publication
the frst draining lymph nodes are located in the groins. of these trials, the sentinel node procedure was included in many
Of all modifcations in standard treatment in the last decades, national and international treatment guidelines.
the introduction of the sentinel node procedure has prob- In 2015 Covens et al. published the results of their meta-
ably been the one with the most impact on treatment-related analysis which assessed the harms and benefts of the sentinel

DOI: 10.1201/9781003255536-10 65
66 Clinical Lymphatic Mapping

FIGURE 10.1 Sentinel node detection with the combined technique. (a) Preoperative lymposcintigram shows bilateral lymph drainage, with one
sentinel node located in each groin. (b) Just before the start of surgery, injection with blue dye to visualize the sentinel node and afferent lymph vessels.
(c) Using the gamma probe, the sentinel node is localized. (d) The blue afferent lymph vessels and radioactivity lead us to the sentinel node. (e) The
sentinel node is localized. (f) The sentinel node is sent for pathological evaluation with ultrastaging.
Sentinel lymph node biopsy of the vulva 67

node procedure. In this paper they showed the groin recurrence carcinoma smaller than 4 cm in maximum diameter, without
rates after a negative sentinel node biopsy were not signifcantly suspicious nodes at clinical examination and imaging. These
higher than after a negative inguinofemoral lymphadenec- criteria are based on the inclusion criteria of GROINSS-V I.
tomy. These were, respectively, 3% (95% confdence interval In the GOG173 study, tumors with a maximum diameter of
[CI] 2%–4%) and 1% (95% CI 0%–3%).12 In the AGO-CaRE-1 6 cm could be included. In this study the false-negative rate
study, 487 lymph node–negative patients who had undergone was much higher for patients with tumors ≥4 cm compared to
inguinofemoral lymphadenectomy were compared with 69 sen- those with tumors <4 cm (7.4% versus 2%).4 A multicenter
tinel lymph node–negative patients who had undergone sentinel German study in 127 women with primary T1–T3 vulvar
node biopsy only. Only those with tumors <4 cm were included. cancer published no separate analysis for tumors ≥4 cm.
The authors found similar results for both groups with regard However, three patients had false-negative sentinel nodes,
to recurrence rates and survival.13 A recent nationwide study in and two of them had primary tumors ≥4 cm.23 Nica et al.
Denmark in 286 early-stage vulvar cancer patients confrmed described 20 patients with a tumor ≥4 cm who had undergone
these results for the sentinel node procedure. Isolated groin sentinel node biopsy. Eleven of them had a negative sentinel
recurrences after a negative sentinel node were observed in node, and thus no adjuvant treatment. One of these patients
2.1% of the patients, all within 2 years after primary treatment.14 had an isolated groin recurrence (9%). Based on this fnd-
Standard sentinel node detection can be performed with a ing, the authors conclude that the sentinel node procedure
radioactive tracer (usually 99mTechnetium-labeled nanocolloid) in larger tumors might have a higher risk in terms of groin
and a blue dye. Blue dye enables the surgeon to directly visualize recurrences.24 Data on the sentinel node procedure in mul-
the lymph vessels and the sentinel node, and thereby it facilitates tifocal tumors are lacking. The GROINSS-V protocol was
the detection of the sentinel node. A meta-analysis of studies amended after 3 years of inclusion because of the high num-
on the sentinel node procedure in vulvar cancer showed that the ber of groin recurrences in patients with multifocal tumors
combination of technetium Tc99 and blue dye provides the best and a negative sentinel node (11.8% at the moment it was
detection rate: a detection rate of 97.7% for technetium Tc99 with decided to amend the protocol and multifocal disease was
blue dye, 94.0% for technetium Tc99 alone, and 68.7% for blue made an exclusion criterion).3 Other studies did not include
dye alone.15 Based on these results and the experience that the multifocal disease or did not describe these patients sepa-
learning curve is shorter with the combined technique, we advise rately.4,23 A study by Garganese et al. investigated the role
performing the sentinel node procedure with both a radioactive of sentinel node biopsy in patients without clinically suspect
tracer and blue dye. In a model-based economic evaluation of the groin nodes who were not eligible for the sentinel node pro-
sentinel node procedure using a radioactive tracer and blue dye cedure according to current treatment guidelines. They sug-
with ultrastaging, Sutton et al. concluded that this may be con- gest that sentinel lymph node biopsy can be accurate and safe
sidered the most cost-effective strategy, based on survival free even in clinically N0 patients who are currently excluded
of morbidity.16 Other studies confrmed these fndings and stated from this procedure, providing that a careful preoperative
that the key factor in this was the lower incidence of lymphedema selection is performed with a negative positron emission
in women who underwent sentinel node biopsy only.17,18 tomography–computed tomography (PET-CT).25 A Swedish
The morbidity of sentinel node biopsy alone is much study by Zach et al. is currently investigating the sentinel
lower compared with an inguinofemoral lymphadenectomy. node procedure in four groups of patients: frst, patients with
GROINSS-V showed that lymphedema and recurrent erysip- a primary tumor ≥4 cm; second, patients with multifocal dis-
elas occurred in, respectively, 1.9% and 0.4% after sentinel ease; third, patients with a local recurrence without previous
node biopsy alone, compared to 25.2% and 16.2%, respectively, inguinofemoral lymphadenectomy or radiation to the groins;
after inguinofemoral lymphadenectomy.3 Hospital stay is also and fourth, patients with a local recurrence with previous
shorter after sentinel node biopsy. Patients today usually can inguinofemoral lymphadenectomy and/or radiation to the
leave the hospital 1–2 days after the surgical intervention. groins. All patients in this study will undergo inguinofemo-
However, the morbidity for patients with a metastatic senti- ral lymphadenectomy after the sentinel node biopsy.26 In a
nel node is still high, as they all require additional inguinofem- retrospective analysis of 27 patients with local recurrence
oral lymphadenectomy. Different studies have reported on the who underwent a repeat sentinel node procedure, van Doorn
incidence of lower extremity lymphedema after vulvar cancer et al. showed the procedure is feasible in locally recurrent
surgery, and the range is wide: 14%–50%.19,20 Lower extremity disease. However, the detection rate is 78% lower compared
lymphedema has shown to be detrimental to quality of life, to the sentinel node procedure in the primary setting.27 This
daily functioning, and body image.21 Therefore, trials focusing group is currently prospectively investigating the sentinel
on prevention and management of this condition are needed. node procedure without subsequent inguinofemoral lymph-
adenectomy in case of a negative sentinel node in patients
with locally recurrent disease. In summary, several studies
Can we extend the indication for sentinel are investigating whether the inclusion criteria for sentinel
node biopsy can be extended so more vulvar cancer patients
node biopsy in vulvar cancer?
can be spared the negative consequences of an inguinofemo-
The criteria for application of the sentinel node procedure ral lymphadenectomy. Until now, we advise strictly adhering
in vulvar cancer have been the same since the introduc- to the criteria as formulated earlier in order to prevent (often
tion in our treatment guidelines:22 unifocal squamous cell fatal) groin recurrences.
68 Clinical Lymphatic Mapping

More recently, a superparamagnetic iron oxide has been


New directions in sentinel lymph introduced as an alternative tracer for sentinel lymph node
node detection detection. Superparamagnetic iron oxide particles have been
shown to be suitable for lymph node mapping using a mag-
Near-infrared fuorescence imaging has emerged as a new netic probe for nodal detection. The sentinel node can also
method for intraoperative visualization of sentinel nodes. be visualized since the particles are carboxydextran-coated,
Peritumoral injection of this tracer allows for real-time sen- giving the sentinel node a blackish-brown appearance. A
tinel node imaging. Near-infrared light is invisible to the meta-analysis and randomized controlled trial in breast can-
naked eye and therefore does not color the surgical feld when cer patients showed noninferiority to the standard technique
applied. Furthermore, it has a penetration depth of approxi- for sentinel node biopsy with technetium-labeled colloid and
mately 5–8 mm, allowing identifcation of structures even if blue dye.35,36 Therefore, it could be a viable alternative for a
they are not yet directly exposed to the surface.28 Several stud- radioactive tracer with blue dye or ICG. The advantages of this
ies have shown good feasibility of the method in identifying procedure are that the injection of the tracer can be done up
the sentinel node in vulvar cancer patients.29–31 to 7 days before surgery, allowing more fexibility in schedul-
A recent randomized controlled trial by Deken et al. com- ing surgery. Also, intraoperative injection can be performed,
pared standard sentinel node detection with a hybrid tracer allowing for a one-step procedure. Surgeons reported they
ICG-99Tc nanocolloid in 48 patients.32 They found that the felt comfortable with the magnetic technique after perform-
percentage of blue sentinel nodes was lower compared with the ing three to fve cases.36 Only one study investigated this tech-
percentage of fuorescent sentinel nodes (65.3% versus 92.5%, nique in vulvar cancer in a series of 20 patients. In this study
P < 0.001). The rate of successful sentinel node procedures the superparamagnetic iron oxide tracer was combined with
was not different between both groups (92.1% for the standard 99mTc, both injecting them a day before surgery. This study
group, 97.2% for blue dye, P = 0.33). There are several advan- showed that the use of superparamagnetic iron oxide tracer
tages of near-infrared fuorescence imaging over blue dye. was not inferior to the standard approach with a radioactive
First, the penetration depth of the fuorescence emission. Blue tracer.37 When this technique would show similar results in
dye cannot be seen through the skin or soft tissue, whereas a 1-day protocol, this could provide a great advantage, since
indocyanine green (ICG) has a penetration depth of up to 8 preoperative peritumoral tracer injection is a painful proce-
mm. Second, blue dye and has a lower sensitivity for sentinel dure for patients.
node detection. Furthermore, blue dye has a small risk of ana-
phylaxis, which is much lower for ICG.33 On the other hand,
near-infrared fuorescence imaging requires a fuorescent
camera system, training in near-infrared imaging, and conve- Sentinel node metastasis
nience with the technique. Also, there are some shortcomings Sentinel node biopsy allows the application of ultrastaging.
of this procedure. Because of its molecular size, ICG migrates Ultrastaging consists of multiple sectioning and immunohis-
through the lymph vessels without specifcity for the frst tochemistry. Different protocols for multiple sectioning have
draining lymph nodes compared to the higher-echelon nodes. been applied, but usually sentinel nodes are cut at 400- to
Because of this feature, it also induces spillage during surgi- 500-um intervals. At every level one slide is stained with
cal manipulation.34 There are only a few contraindications for standard hematoxylin-eosin staining and one with immu-
ICG: iodine allergy (since indocyanine green contains sodium nohistochemical staining for cytokeratin. Ultrastaging of
iodide), pregnancy or breastfeeding, liver disease, dialysis or sentinel nodes allows detection of smaller metastases, which
renal failure, and previous anaphylactic reaction.33 can easily be missed with standard hematoxylin-eosin stain-
The use of near-infrared imaging alone would have the ing (2- to 5-mm slides). In GROINSS-V I 41% of the meta-
potential advantage of becoming a one-step procedure, avoid- static sentinel nodes were detected only by ultrastaging.3 In
ing preoperative injection of a radioactive tracer and perform- the GOG173 study this percentage was 23%.4 An interesting
ing a lymphoscintigram. However, body weight is a limitation result is the publication of the interim fndings of an audit
for the use of ICG alone, since the penetrance is max 8 mm.29 on the outcomes of sentinel node biopsy in routine clinical
No other prospective studies have shown a single-tracer strat- practice in Australia and New Zealand. Two isolated groin
egy using ICG alone. The combination with 99mTc appears to recurrences were observed after a negative sentinel node,
be the optimal combination. The best of both was combined and in both cases, analysis showed the pathology protocol for
in the development of a hybrid tracer: ICG-99mTc-nanocolloid. sentinel node workup was not followed. This report shows
With this procedure, the sentinel node procedure can become that strict adherence to the pathology protocol is an essential
more accurate and independent of the use of blue dye.34 component in the utilization of the sentinel node procedure
In conclusion, the use of near-infrared imaging appears to in vulvar cancer.38
be of added value because of its high sensitivity, which outper- We know that the risk of non–sentinel node metastases
forms blue dye and is comparable to 99mTechnetium-labeled increases with the size of the sentinel node metastasis.
nanocolloid. Advantages might be that near-infrared imaging Analysis of metastatic sentinel nodes from GROINSS-V I
is not disturbed by the background signal, as sometimes is the showed that the risk of additional metastasis was 4.2% for
case with radiocolloid. Since the penetration depth is limited patients with isolated tumor cells in the sentinel node and
to approximately 8 mm, the concurrent use of a radioactive increased to 62.5% for patients with sentinel node metasta-
tracer is still needed. ses >10 mm. There was no threshold below which the risk
Sentinel lymph node biopsy of the vulva 69

of additional metastases was so low that inguinofemoral lymphadenectomy in vulvar cancer patients: a Dutch nation-
lymphadenectomy could be safely omitted.39 Therefore, all wide prospective study. Gynecol Oncol. 2017;146(3):580–7.
sentinel node metastases, even those with isolated tumor 8. Carlson JW, Kauderer J, Hutson A, et al. GOG 224 – the
cells, should be regarded as a metastatic sentinel, and lymphedema and gynecologic cancer (LEG) study: inci-
therefore have an indication for adjuvant treatment. At this dence and risk factors in newly diagnosed patients. Gynecol
moment, standard treatment for a metastatic sentinel node is Oncol. 2020;156(2):467–74.
an inguinofemoral lymphadenectomy of the affected groin. 9. Levenback C, Burke TW, Gershenson DM, et al.
A study by Nica et al. suggests an inguinofemoral lymph- Intraoperative lymphatic mapping for vulvar cancer. Obstet
adenectomy can be omitted and replaced by inguinofemoral Gynecol. 1994;84(2):163–7.
radiotherapy when the sentinel node contains metastasis ≤2 10. Hassanzade M, Attaran M, Treglia G, et al. Lymphatic
mm. However, these observations are based on retrospec- mapping and sentinel node biopsy in squamous cell carci-
tive analysis in a small number of patients.24 Therefore, noma of the vulva: a systematic review and meta-analsysi of
the literature. Gynecol Oncol. 2013;130(1):237–45.
we believe further prospective data should be awaited.
11. Lawrie TA, Patel A, Martin-Hirsch PP, et al. Sentinel node
GROINSS-V II investigated whether radiotherapy is a safe
assessment for diagnosis of groin lymph node involve-
alternative for patients with a micrometastasis in the senti-
ment in vulval cancer. Cochrane Database Syst Rev.
nel node. Patients with sentinel node metastasis ≤2 mm were
2014;2014(6):CD10409.
treated with inguinofemoral radiotherapy (50 Gy) instead of
12. Covens A, Vella ET, Kennedy EB, et al. Sentinel lymph
an inguinofemoral lymphadenectomy. Stopping rules were node biopsy in vulvar cancer: systematic review, meta-
constructed to monitor groin recurrence rate. This study analysis and guideline recommendations. Gynecol Oncol.
included patients from 2005 to 2016, and the results will 2015;137(2):351–61.
soon become available. GROINSS-V III will investigate 13. Klapdor R, Hillemans P, Wölber L, et al. Outcome after
whether chemoradiation is a safe alternative for an inguino- sentinel lymph node dissection in vulvar cancer: a sub-
femoral lymphadenectomy in patients with a macrometasta- group analysis of the AGO-CaRE-1 study. Ann Surg Oncol.
sis (>2 mm) in the sentinel node. 2017;24:1314–21.
14. Froeding LP, Høgdall C, Kristensen E, et al. Recurrence
and survival rates in node negative patients after sentinel
Conclusions node biopsy for early-stage vulva cancer – a nationwide
study. Gynecol Oncol. 2020;156:124–30.
Sentinel node biopsy is part of the standard of care today in 15. Meads C, Sutton AJ, Rosenthal AN, et al. Sentinel lymph
early-stage vulvar cancer treatment. The procedure can be per- node biopsy in vulval cancer: systematic review and meta-
formed with a radioactive tracer in combination with blue dye analysis. Br J Cancer. 2014;110(12):2837–46.
or ICG. The sentinel node procedure is cost-effective. Standard 16. Sutton AJ, Barton P, Sundar S, et al. Cost-effectiveness
treatment in case of a metastatic sentinel node (metastasis of of sentinel lymph node biopsy vs inguinofemoral lymph-
any size) is an inguinofemoral lymphadenectomy. Studies on adenectomy in women with vulval cancer. Br J Cancer.
radio(chemo)therapy instead of a lymphadenectomy might 2013;109(10):2533–47.
change future treatment guidelines. 17. Erickson BK, Divine LM, Leath CA, et al. Cost-
effectiveness analysis of sentinel lymph node biopsy in the
treatment of early-stage vulvar cancer. In J Gynecol Cancer.
REFERENCES 2014;24(8):1480–5.
1. Bray F, Laversanne M, Weiderpass E, et al. Geographic and 18. McCann GA, Cohn DE, Jewell EL, et al. Lymphatic mapping
temporal variations in the incidence of vulvar and vaginal and sentinel lymph node dissection compared to complete
cancer. In J Cancer. 2020;147(10):2764–71. lymphadenectomy in the management of early-stage vulvar can-
2. Cabanas RM. An approach for the treatment of penile car- cer: a cost-utility analysis. Gynecol Oncol. 2015;136(2):300–4.
cinoma. Cancer. 1977;39(2):456–66. 19. Wills A, Obermair A. A review of complications associ-
3. Van der Zee AG, Oonk MH, De Hullu JA, et al. Sentinel ated with the surgical treatment of vulvar cancer. Gynecol
node dissection is safe in the treatment of early-stage vulvar Oncol. 2013;131:467–79.
cancer. J Clin Oncol. 2008;26(6):884–9. 20. Huang J, Ye N, Wang Z, et al. Incidence of lower limb
4. Levenback CG, Ali S, Coleman RL, et al. Lymphatic map- lymphedema after vulvar cancer: a systemic review and
ping and sentinel lymph node biopsy in women with squa- meta-analysis. Medicine. 2017;96(46):e8722.
mous cell carcinoma of the vulva: a gynecologic oncology 21. Carter J, Huang HQ, Armer J, et al. GOG244 – the lymph-
group study. J Clin Oncol. 2012;30(31):3786–91. edema and gynecologic cancer (LeG) study: the impact of
5. Froeding LP, Høgdall C, Kristensen E, et al. Recurrence lower-extremity lymphedema on quality of life, psycho-
and survival rates in node negative patients after sentinel logical adjustment, physical ability, and function. Gynecol
node biopsy for early-stage vulva cancer – a nationwide Oncol. 2020; Epub ahead of print.
study. Gynecol Oncol. 2020;156(1):124–30. 22. Oonk MHM, Planchamp F, Baldwin P, et al. European soci-
6. Selman TJ, Luesley DM, Acheson N, et al. A systematic ety of gynaecological oncology guidelines for the manage-
review of the accuracy of diagnostic tests for inguinal lymph ment of patients with vulvar cancer. In J Gynecol Cancer.
node status in vulvar cancer. Gynecol Oncol. 2005:206–14. 2017;27:832–7.
7. Pouwer AW, Hinten F, van der Velden J, et al. Volume- 23. Hampl M, Hantschmann P, Michels P, et al. Validation
controlled versus short drainage after inguinofemoral of the accuracy of the sentinel lymph node procedure in
70 Clinical Lymphatic Mapping

patients with vulvar cancer: results of a multicenter study in 32. Deken MM, van Doorn HC, Verver D, et al. Near-infrared
Germany. Gynecol Oncol. 2008;111(2):282–8. fuorescence imaging compared to standard sentinel lymph
24. Nica A, Covens A, Vicus D, et al. Sentinel lymph nodes in node detection with blue dye in patients with vulvar can-
vulvar cancer: management dilemmas in patients with posi- cer – a randomized controlled trial. Gynecol Oncol. 2020;
tive nodes and larger tumors. Gynecol Oncol. 2019;94–100. Article in Press.
25. Garganese G, Collarino A, Fragomeni SM, et al. Groin senti- 33. Zapardiel I, Alvarez J, Barahona M, et al. Utility of intra-
nel node biopsy and 18F-FDG PET/CT-supported preoperative operative fuorescence imaging in Gynecologic surgery:
lymph node assessment in cN0 patients with vulvar cancer systematic review and consensus statement. Ann Surg
currently unft for minimally invasive inguinal surgery: the Oncol. 2020; Online Ahead of Print.
GroSNaPET study. Eur J Surg Oncol. 2017;43(9):1776–83. 34. KleinJan GH, Van Werkhoven E, van den Berg NS, et al.
26. Zach D, Kannisto P, Bohlin KS, et al. Can we extend the The best of both worlds: a hybrid approach for optimal pre-
indication for sentinel node biopsy in vulvar cancer? A and intraoperative identifcation of sentinel lymph nodes.
nationwide feasibility study from Sweden. Int J Gynecol Euro J Nucl Med Mol Imaging. 2018;45(11):1915–25.
Cancer. 2020;30:402–5. 35. Teshome M, Wei C, Hunt KK, et al. Use of a magnetic
27. Van Doorn HC, Van Beekhuizen HJ, Gaarenstroom KN, tracer for sentinel lymph node detection in early-stage
et al. Repeat sentinel lymph node procedure in patients breast cancer patients: a meta-analysis. Ann Surg Oncol.
with recurrent vulvar squamous cell carcinoma is feasible. 2016;23(5):1508–14.
Gynecol Oncol. 2016;140(3):415–19. 36. Alvarado MD, Mittendorf EA, Teshome M, et al.
28. Handgraaf HJ, Verbeek FP, Tummer QR, et al. Real-time SentimagIC: a non-inferiority trial comparing superpara-
near-infrared fuorescence guided surgery in gynecologic magnetic iron oxide versus technetium-99m and blue dye in
oncology: a review of the current state of the art. Gynecol the detection of axillary sentinel nodes in patients with early-
Oncol. 2014;135(3):606–13. stage breast cancer. Ann Surg Oncol. 2019;26(11):3510–16.
29. Crane LM, Themelis G, Arts HJ, et al. Intraoperative near- 37. Jedryka MA, Klimczak P, Kryszpin M, et al.
infrared fuorescence imaging for sentinel lymph node Superparamagnetic iron oxide: a novel tracer for senti-
detection in vulvar cancer: frst clinical results. Gynecol nel lymph node detection in vulvar cancer. Int J Gynecol
Oncol. 2011;120:291–5. Cancer. 2020;30(9):1280–4.
30. Schaafsma BE, Verbeek FP, Peters AA, et al. Near-infrared 38. Sykes P, Eva L, van der Griend R, et al. Pathological pro-
fuorescence sentinel lymph node biopsy in vulvar cancer: cess has a crucial role in sentinel node biopsy for vulvar
a randomised comparison of lymphatic tracers. BJOG. cancer. Gynecol Oncol. 2019;153:292–6.
2013;120:758–64. 39. Oonk MH, van Hemel BM, Hollema H, et al. Size of
31. Hutteman M, van der Vorst JR, Gaarenstroom KN, et al. sentinel-node metastasis and chances on non-sentinel-
Optimization of near-infrared fuorescent sentinel lymph node involvement and survival in early stage vulvar can-
node mapping for vulvar cancer. Am J Obstet Gynecol. cer: results from GROINSS-V, a multicentre observational
2012;206:89.e1–5. study. Lancet Oncol. 2010;11(7):646–52. Runa ping
11
Sentinel lymph node mapping in cervical cancer

David Cibula and Martina Borcinova

CONTENTS
Sentinel lymph node mapping in the pelvis................................................................................................................................... 71
Lymphatic drainage from the cervix .................................................................................................................................... 71
Lymphatic drainage in the pelvis ......................................................................................................................................... 71
Pelvic dissection and SLN removal technique ..................................................................................................................... 71
SLN localization in the pelvis .............................................................................................................................................. 72
SLN detection rate................................................................................................................................................................ 73
Sensitivity of SLN for pelvic lymph node staging ............................................................................................................... 74
SLN biopsy in clinical management .............................................................................................................................................. 74
Acceptance of SLN in current routine practice .................................................................................................................... 74
International recommendations ............................................................................................................................................ 74
SLN frozen section and intraoperative triage of patients ..................................................................................................... 75
Pathologic ultrastaging and prognostic role of micrometastases ......................................................................................... 75
SLN in fertility-sparing treatment ........................................................................................................................................ 77
SLN and lower leg lymphedema .......................................................................................................................................... 78
Ongoing clinical trials on the safety of SLN biopsy only .................................................................................................... 78
Summary ........................................................................................................................................................................................ 78
References...................................................................................................................................................................................... 79

vessels on the pelvic side wall, do not only carry lymph from
Sentinel lymph node mapping in the pelvis the central pelvic organs. Their main purpose is to drain the
lymph from the lower extremities cranially to the paracaval
Lymphatic drainage from the cervix and paraaortic lymphatic systems. These trunks receive multi-
The cervix is an optimal anatomical structure for SLN use. ple channels from the midline gynecologic organs. Two major
This organ is easily accessible transvaginally for a tracer appli- lymphatic trunks can be identifed on the lateral pelvic walls,
cation, and, in early stages, the metastatic spread is almost with multiple connections between them (https://surgery4u.eu/
always lymphogenic. It is very unlikely that, given the rich video-41).2 A ‘superfcial’ trunk enters the pelvis through the
network of lymph nodes (LNs) in the pelvis, lymph drainage femoral canal; it runs on the ventral walls of the external iliac
would bypass the pelvic basin completely so that tumor cells vessels, receives lymphatic channels from parametria, and
would be caught in the paraaortic region and all pelvic nodes continues on the ventral wall of the common iliac artery crani-
ally (Figure 11.1).
would remain negative. This is the main reason that pelvic LN
The ‘deep’ trunk enters the pelvis medially to the external
involvement is the most signifcant prognostic factor in early-
iliac vessels and creates a bundle of fatty lymphatic tissue
stage cervical cancer. And this is also why LN staging is such
surrounding the obturator nerve. From the obturator fossa, it
a signifcant step in disease management.
continues cranially to the space between the superior gluteal
As the cervix is the central pelvic organ, lymphatic drainage
vessels and the psoas muscle, where it splits into two parts.
is bilateral, and so each pelvic side must be staged separately.
The frst one creates a deep common iliac branch that runs
A negative sentinel lymph node (SLN) on one side of the pelvis
between the psoas muscle and the common iliac vein, while
does not guarantee LN negativity on the other side. If SLN is the deeper one, often overlooked, runs medially via a tun-
detected only unilaterally, systematic pelvic lymph node dis- nel below the common iliac vessels to the presacral region
section (PLND) must be performed on the other side.1 (Figure 11.2).

Lymphatic drainage in the pelvis Pelvic dissection and SLN removal technique
It is important to keep in mind that the lymphatic trunks, The goal of replacing systematic PLND by SLN biopsy
which are interrupted when LNs are harvested alongside large is to decrease postoperative morbidity, mainly the risk of

DOI: 10.1201/9781003255536-11 71
72 Clinical Lymphatic Mapping

developing lower leg lymphedema. The technique of pelvic


space dissection and SLN removal must therefore correspond
with this intention. If too many SLNs are removed and both
main lymphatic trunks running on the pelvic side wall are
disrupted, this will inevitably compromise drainage from the
lower extremities. Pelvic dissection should thus be as consider-
ate as possible and aim to preserve the lymphatic channels that
do not transport lymph to the SLN. When tracer-marked lym-
phatic channels are visible through the peritoneum, pelvic dis-
section can be targeted to one region only. If the channels are
not visible, however, regions with the most frequent SLN pres-
ence should be approached frst, namely the interiliac (obtura-
tor) and external iliac regions, by opening the paravesical and
lateral pararectal spaces. Other retroperitoneal spaces in the
level II should only be dissected if SLNs have not been found.

SLN localization in the pelvis


A familiarity with the regions in which SLNs are most fre-
quently found is crucial for their successful detection. For clin-
FIGURE 11.1 Superfcial lymphatic trunk on the right pelvic side wall. ical practice, it is useful to divide the pelvic lymphatic basin
EIA, external iliac artery; EIV, external iliac vein; IIV, internal iliac vein; into two levels, with a horizontal boundary at the level of the
IIA, internal iliac artery; CIA, common iliac artery; CIV, common iliac common iliac vessels’ bifurcation (Figure 11.3). It has been
vein; GN, genitofemoral nerve; PM, psoas muscle; A, aortic bifurcation; proven in large retrospective studies that the localization of all
SLT, superfcial lymphatic trunk; EI, external iliac region; SCI, superf- detected SLNs corresponds with the localization of positive
cial common iliac region. (From Ref. 2 with permission.)

FIGURE 11.2 Deep lymphatic trunk on the right pelvic side wall. EIV,
external iliac vein; EIA, external iliac artery; IIV, internal iliac vein; IIA,
internal iliac artery; CIA, common iliac artery; CIV, common iliac vein;
GN, genitofemoral nerve; PM, psoas muscle; A, lymphatic passage below
common iliac vessels; DLT, deep lymphatic trunk; OBT, obturator region; FIGURE 11.3 Anatomical distribution of SLN and positive SLN in the
II, internal iliac region; DCI, deep common iliac region. (From Ref. 2 pelvis in the SENTIX prospective trial (N = 395). SLN, sentinel lymph
with permission.) node. (From data in Ref. 5.)
Sentinel lymph node mapping in cervical cancer 73

SLNs. The majority of SLNs are typically found in level I, cau- patients in the SENTIX trial, paraaortic localization of SLN
dally to the common iliac vessel bifurcation. In level II, two has not been reported in any case.5
main anatomical regions are usually described on both sides
of the pelvis: the external iliac region around the external iliac
SLN detection rate
vessels and the interiliac region, which is anatomically identi-
cal with the obturator region. In the frst large retrospective Pelvic LN staging must be performed separately for both pel-
analysis on SLN mapping by Rob in 183 patients, 91% of SLNs vic side walls. SLN detection frequency is therefore reported
were detected in level I.3 In a combined analysis of two pro- as side-specifc, or separately for unilateral and bilateral detec-
spective French studies (SENTICOL I and II) in 405 patients,4 tion. The success rate for bilateral detection oscillates in the lit-
83% of SLNs were found in level I, and in a recent paper from erature between 55% and 100%, depending on the tracer used,
the SENTIX prospective study in 351 patients, it was 90%.5 size of the cohort, and experience of the surgeon – but mainly
Both spaces are well-defned by stable anatomical landmarks.6 on the year of publication.3–5,7,12–16 Lower detection frequency
SLNs are detected much less frequently in level II, between was reported in older studies in patients with larger tumors.12,13
the vena cava bifurcation cranially and the common iliac In a literature review published in 2013, the difference in side-
bifurcation caudally. On this level, typically three regions specifc detection was 25% between the groups with tumor
are distinguished: the common iliac region around the com- sizes of <2 cm (n = 768) and ≥2 cm (n = 724).17 However, with
mon iliac vessels on both sides of the pelvis and one central better standardization of SLN biopsy techniques, the differ-
presacral space between the common iliac vessels. These ences in detection between smaller and larger tumors have
regions also have clearly defned anatomical borders.6 In the progressively decreased.5,15 In a large retrospective study cov-
SENTIX trial positive SLNs in level II were detected only in ering 350 patients with stages pT1a–pT2, the detection rate
4% of patients, with 1.8% of cases with isolated positive SLNs was compared in three groups of different tumor sizes: <2 cm,
in level II only.5 All these results prove the fact that cervical 2–3.9 cm, and ≥4 cm.18 Unilateral and bilateral detection was
cancer in most patients progresses predictably, and so the most achieved in 93% and 80% of patients, respectively, without any
attention should always be paid to exploring the two regions major differences among groups. Successful SLN detection,
below the common iliac bifurcation, where the majority of however, requires adjustment of the technique of tracer appli-
SLNs and positive SLNs are located. cation in large tumors.18,19 Tracer cannot be applied superf-
Diverse data can be found in the literature about the pres- cially and submucosally; rather, it needs to be inserted into the
ence of SLN in so-called ‘atypical’ localizations. This is residual cervical stroma behind the tumor using a long needle
mainly because in the past some authors considered even (Figure 11.4).
level II or parametria ‘atypical’ regions.4,7,8 However, the pres-
ence of SLN in level II, including the presacral region, should
be considered typical for cervical cancer. Also, the parametria,
which are located between the cervix and pelvic side walls, are
a typical area for localization of cervical lymphatic drainage.
Back in 2000, Benedetti-Panici, who submitted isolated para-
metria for pathological ultrastaging, found LNs in the ventral
parametria in 12% and in lateral parametria in 38% of cases.9
It is likely that the detection of LNs in the parametria is seri-
ously underreported. Parametria are usually an integral part of
the surgical specimen after radical hysterectomy. Also, due to
the leak of color, fuorescent tracer, or radiation from the cer-
vix to the adjacent tissue, our ability to reliably detect subtle
parametrial LNs as SLNs is limited. This fact may gain impor-
tance in the future if simple hysterectomy is performed instead
of radical and, at the same time, systematic PLND is replaced
by SLN biopsy only. A combination of these recent trends may
increase the risk that positive parametrial LNs will be left in
situ and the patient will not receive adjuvant treatment if the
SLN is negative.
It seems, however, that another factor in the more frequent
atypical SLN localization in the past, mainly in the paraaortic
region, was the SLN biopsy learning curve. The more experi-
ence with SLN biopsy the surgeon has, the smaller the risk that
SLN is not identifed within the seven standard pelvic regions.
This historical trend is apparent in two consecutive studies
from France. While the paraaortic localization of SLN was
reported in 5% of patients in the earlier study, it was described
in only 1.5% of patients in a combined analysis of both tri- FIGURE 11.4 Tracer application into the residual cervical stroma in
als.10,11 In a recent paper from a large prospective cohort of 395 larger tumors.
74 Clinical Lymphatic Mapping

Overall, it appears that the main factor for successful bilat- negative despite the detection of MAC in non-SLN, but only
eral detection is the learning curve. A signifcantly higher MIC or ITC in SLN. A conventional SLN evaluation (with-
detection rate was reported in both multicenter prospective out ultrastaging) would fail to detect pelvic LN involvement
studies, SENTIX and SENTICOL, in centers with a higher in these patients, which would cause a drop of sensitivity
number of patients.4,5 More experience is likely the reason why to 80% and make SLN biopsy without additional PLND an
the more recent study SENTICOL II reached a better detec- unreliable and unsafe method of LN staging. Second, there
tion rate in comparison with the older study SENTICOL I.4 has always been a methodological bias when comparing SLN
The SENTIX study, which commenced in 2016, is the frst and non-SLN status. While SLNs are mostly assessed using
multicenter trial that recently reported a bilateral detection an ultrastaging protocol, other pelvic LNs are processed con-
rate higher than 90%.5 Data from 395 patients were analyzed. ventionally. So far, only a few studies on a limited number
The success rate of the detection was unaffected by tumor of patients assessed both SLNs and all other pelvic LNs by
size, tumor stage, or body mass index. On the other hand, ultrastaging.23–27 Although the risk of MIC in non-SLNs with
an interesting factor, which worsened the detection success negative SLNs is low, all available evidence comes from a
in both prospective studies, was the higher age of patients, cumulative group of 90 patients in which the methodology
determined as 60 years in the SENTIX study and ≥70 years of pathological evaluation was not identical and the risk of
in the SENTICOL study.4,5 The median number of removed LN involvement was low (12 cases). These limitations in our
SLNs in large studies consistently oscillates between two and knowledge justify the current ongoing trials, which both are
three.5,10,15 designed to address the safety of the replacement of PLND
by SLN biopsy only.
Sensitivity of SLN for pelvic lymph node staging
Sensitivity is the key parameter in assessing the reliability of
the SLN concept for pelvic LN staging. It indicates the portion SLN biopsy in clinical management
of patients in whom SLN status is negative and yet there is a
Acceptance of SLN in current routine practice
metastasis detected in other pelvic LNs. Therefore, sensitivity
can only be assessed when SLN is combined with systematic Even though there is robust evidence from retrospective stud-
PLND. ies about the high SLN detection rate and high sensitivity of
Similarly, the detection rates in earlier studies presented a SLN ultrastaging for pelvic LN staging, results from several
lower sensitivity of SLN biopsy in patients with larger tumors. surveys indicate that the acceptance of SLN biopsy in routine
In the frst multicenter study from Germany, the sensitivity clinical practice is still quite low.28
was 91% in tumors <2 cm, while only 73% in tumors ≥2 cm.13 For example, in a 2016 survey of 63 German institutions, SLN
A review of 15 publications with more than 50 cases from biopsy was reported to be performed in vulvar cancer in 73% of the
2013 showed that tumors smaller than 2 cm had a sensitivity institutions, but in cervical cancer only in 29%.29 And only 11%
of 100%, whereas in larger tumors it was 10% lower.20 With of the centers performed SLN biopsy without additional PLND.
growing experience and improved SLN biopsy technique stan- In an ESGO survey from 2018, which provided answers from 566
dards, the signifcance of tumor size decreases and the number respondents, SLN biopsy in cervical cancer was reported to be
of patients with falsely negative SLNs constantly stays very used in stage T1a1 by 17%, in stage T1a2 by 19%, and in stage
low. In the frst French prospective study, SENTICOL I, in T1b1 in tumors <2 cm by just 9% of the respondents.30
which SLN biopsy was followed by a systematic PLND, the There are probably multiple reasons for this apparent hesi-
sensitivity in patients with tumors <4 cm and bilateral SLN tation to avoid performing traditional systematic PLND and
detection reached 100%.7 In the largest retrospective study so rely on the SLN concept. First of all, LN involvement is the
far, in which there were 645 patients enrolled and disease stage most signifcant prognostic factor for an adjuvant treatment
varied from IA1 to IIB, the sensitivity reached 97%.21 decision in cervical cancer patients. While the prognosis in
Taking into consideration such a consistently reported low patients with positive LNs after a standard treatment includ-
SLN false-negative rate for pelvic LN staging, one could ing adjuvant radiotherapy is still very good, leaving positive
take the safety of SLN biopsy in cervical cancer for granted. LN without administering adjuvant treatment likely has fatal
There are, however, a few caveats in the available evidence. consequences for the patient. Second, the majority of the data
First, the sensitivity is very high, partially thanks to the fact on SLN biopsy in cervical cancer comes from retrospective
that the presence of any type of metastasis in SLN, includ- studies, which do have their limitations. This may be why
ing micrometastases or isolated tumor cells, is considered a many surgeons keep on performing systematic PLND – until
positive fnding. The classifcation of metastases was adopted the oncological safety of SLN biopsy without systemic PLND
from breast cancer, and it recognizes three groups based on has been proven in prospective studies.
size of metastasis as follows: macrometastases (MAC) >2 mm,
micrometastases (MIC) >0.2 mm up to 2 mm, and isolated
International recommendations
tumor cells (ITCs) up to 0.2 mm.
As an example, in a large retrospective study, metastatic Current international recommendations for the clinical man-
involvement was detected in non-SLN pelvic LNs in 23 agement of patients with cervical cancer have already opened
cases (3.6%) in whom only MIC or ITC were found in SLN the possibility of performing SLN biopsy only, without sys-
by ultrastaging.22 These cases were not considered false tematic PLND, but they vary in their indications.
Sentinel lymph node mapping in cervical cancer 75

European guidelines that were jointly developed by three MAC (4/9), all MIC (4/4), and all ITC (9/9).42 A Canadian
European medical societies (European Society Gynecologic study on 211 patients reported 10 of 13 cases with positive
Oncology-European Society for Radiotherapy and Oncology- SLN found falsely negative by frozen section, including 7
European Society of Pathology [ESGO-ESTRO-ESP]) con- MIC, 2 ITC cases, and 1 MAC with a size of 2.9 mm.43 In a
sider SLN biopsy alone suffcient only in stage IA;31 SLN large retrospective cohort of 309 cases, frozen section identi-
biopsy is suggested in patients with stage T1a1 LVSI–positive fed 67% of cases with MAC (16/24), but only 14% with MIC
or T1a2 LVSI–negative patients, and it is recommended in (2/14) and 0% with ITC (0/10).44 Similar results were con-
T1a2 LVSI–positive patients. On the other hand, in patients frmed recently from the SENTIX prospective study, which,
with stage T1b, SLN biopsy is highly recommended, though for the frst time in cervical cancer, included a central pathol-
it should be performed in combination with systematic PLND. ogy quality control algorithm.45 In the group of 395 patients,
The aim of SLN biopsy in these patients is not to avoid sys- MAC, MIC, and ITC were detected by frozen section in 72%
tematic staging, but to detect selected SLN for intraoperative (21/29), 10% (19/21), and 0% (0/12), respectively.5 The sensi-
assessment and increase the detection of smaller metasta- tivity of frozen section assessment for fnal SLN status reached
ses thanks to SLN ultrastaging. Published guidelines of the 76% for MAC and 46% for N1 (MAC and MIC together). In
European Society for Medical Oncology are in agreement summary, there is ample evidence that frozen section does not
with the European guidelines,32 as well as guidelines of the detect almost any MIC and ITC, but even more importantly, it
Spanish Society of Medical Oncology.33 also fails to detect about 30% of MAC, usually slightly above
In National Comprehensive Cancer Network (NCCN) 2 mm in size.
guidelines, SLN biopsy is considered an alternative to PLND The main purpose of SLN frozen section is to get an oppor-
in all tumors ≤4 cm except for stage IA1 without LVSI.1,34 tunity to change the management during the course of the sur-
These guidelines emphasize that the best mapping results are gery. If positive SLNs are found intraoperatively, any radical
obtained in tumors below 2 cm and that side-specifc LN stag- procedure, including systematic PLND and radical hysterec-
ing requires performance of a systematic PLND on the pelvic tomy, can be abandoned and the patient can be referred for
side where SLNs were not found. defnitive chemoradiation. The goal is to decrease extra mor-
Other societies worldwide also adopted the NCCN guide- bidity due to the combination of radical surgery and pelvic
lines, such as the International Gynecologic Cancer Society,35 radiotherapy. A one-step approach is even more important in
or reference both NCCN and European guidelines, such as fertility-sparing management (trachelectomy or conization).
Cancer Australia.36 Published guidelines of individual soci- This management is recommended by the European guide-
eties of Asian states are varying. In general, societies are in lines,46 which claim that one key objective in the management
agreement that SLN biopsy represents an option to PLND up to of early-stage disease should be avoiding combined treat-
stage T1a2.37,38 In contrast, the Korean Society of Gynecologic ment.47 A one-step protocol like this is obviously limited by
Oncology does not cover SLN biopsy in the guidelines and rec- the high false-negative rate of intraoperative SLN evaluation.44
ommend PLND as the standard of care for all disease stages.39 Both the clinician and the patient must be aware that about
50% of LN metastases will be diagnosed only after the surgery
by fnal ultrastaging, and therefore, a combined treatment can-
SLN frozen section and intraoperative
not be avoided in these cases.
triage of patients
One of the advantages of the SLN concept is that it provides
Pathologic ultrastaging and prognostic
the opportunity to identify a small number of LNs at the high-
role of micrometastases
est risk of metastatic involvement and send these for intra-
operative pathological assessment. The literature, however, is As discussed earlier, it is generally accepted that SLN biopsy
discrepant when it comes to the reliability of intraoperative increases the accuracy of pelvic LN staging.43,48,49 More fre-
SLN assessment.40 The frozen section technique is fairly uni- quent detection of positive LNs is a result of an intensive
form and usually involves the assessment of one slice from pathological assessment of a small number of SLNs. However,
each SLN or from each half of one SLN. A higher number of pathologic ultrastaging is a demanding and time-consuming
sections would prolong the surgery, and it could result in the technique, which cannot be applied to all pelvic LNs after sys-
loss of a substantial part of the SLN tissue for further ultra- tematic PLND, even though it increases the probability of fnd-
staging. Therefore, the key factor that infuences the sensitiv- ing metastases, especially smaller ones.
ity of the frozen section technique is the quality of the fnal Moreover, the role of SLN ultrastaging is still controversial.
SLN processing. The less intensive the ultrastaging protocol, The criticism stems mostly from experiences in other tumors,
the lower the chance for detecting smaller metastases. We predominantly breast cancer, in which the presence of MIC
can hypothesize that this phenomenon explains why some, was not confrmed to be a signifcant prognostic factor. It is,
mostly smaller, studies reported a fairly low false negativ- however, important to emphasize that experiences from one
ity of the frozen section technique for the fnal pelvic LN tumor cannot be fully extrapolated to a different one. While in
status.41 breast cancer there is an array of different prognostic factors,
More recent studies, however, consistently showed that fro- in cervical cancer, LN involvement is the decisive factor for
zen section assessment fails to detect small metastases. In the adjuvant treatment. Hematogenous and lymphogenous spread
French prospective study, SLN frozen section was performed is typical for breast cancer, while cervical cancer in the early
in 102 patients, and it failed to detect 70% of patients with stages spreads solely to pelvic lymph nodes.
76 Clinical Lymphatic Mapping

Critical arguments assume that ultrastaging detects only one showed a negative impact on the prognosis.23,53–58 In the
metastases smaller than 2 mm, MIC and ITC, whose prog- largest retrospective study published so far, data from 645
nostic signifcance has not yet been conclusively confrmed. cases were collected from seven institutions.21 All patients had
However, this is not entirely true. Let us use a comparison an SLN biopsy followed by PLND, and SLNs were processed
between frozen section and ultrastaging as an analogy. The by pathologic ultrastaging. Both MAC and MIC were associ-
intensity of pathological assessment by frozen section is fairly ated with similar and signifcantly decreased overall survival
similar to a standard assessment of pelvic LN. It has been (MAC: hazard ratio [HR] = 6.85; 95% confdence interval
well documented that ultrastaging is able to detect up to 30% [CI]: 2.59–18.05; MIC: HR = 6.86; 95% CI: 2.09–22.61). The
more MAC than frozen section. That is also a reason why SLN only retrospective study that failed to show MIC as a negative
biopsy is recommended as the method to increase the accuracy prognostic factor was a multi-institutional retrospective study,
of detection of positive pelvic LNs in the management of all which submitted tissue blocks for reevaluation in a group of
early stages in combination with PLND. 129 patients who were LN-negative at the time of primary
Nonetheless, a recent review article demonstrated how cur- treatment.57 Any immunoreactive tumor cells were classifed
rent approaches to ultrastaging protocols vary and how insuf- as MIC, and the results indicated that the presence of MIC was
fcient the protocol descriptions that have appeared in the not associated with a negative outcome. There were, however,
literature were.50 Among studies that reported results from only 11 recurrences in this group (8.5%), and patients with
SLN biopsy, 27% (27/97) did not give any details about the MIC were more likely to receive adjuvant radiotherapy than
ultrastaging protocol and, even more surprisingly, SLNs were those with negative LNs (39% vs. 18%). Recently, data from
completely processed in only 8 of them! Even if information two prospective French studies (SENTICOL I and II) were
about intervals between slices and the use of immunohisto- analyzed for the impact of MIC on prognosis.52 Positive LNs
chemistry (IHC) is given, the key parameter is the number were found in 41 out of 321 patients (13%), and 22 (54%) of
of levels (series), and this is mostly absent. It is important to them were low-volume metastases (MIC or ITC). The authors
emphasize that the number of levels is a key parameter for did not fnd any negative impact of the presence of MIC on
ultrastaging, since if only one or two levels are processed com- disease-free survival. It should be emphasized that the gen-
pletely, which is frequently the case, the remaining SLN tissue eral prognosis of patients was superior to any of the previous
is left in paraffn unprocessed. prospective trials, with only 6.5% of recurrences at 3 years,
Minimal requirements for the description of the protocol and neither low-volume disease nor the presence of MAC was
include the following items: 1) approach to gross process- associated with lower disease-free survival.
ing (slicing and thickness of slices), 2) number and distance Multiple reasons may explain discrepant results in the lit-
between levels (series), 3) number of sections (slides) per level, erature. First, the risk of recurrence is low in the early stages
and 4) use of IHC. A complete processing of all SLNs at a of cervical cancer, even in patients with positive pelvic LN,
few micrometer intervals would result in hundreds of slides so it requires a large cohort to demonstrate any signifcant
per node, which would not be sustainable in routine practice. impact on the prognosis. Second, and more importantly, in the
So, it is necessary to fnd a reasonable compromise between absence of any universal protocol for SLN ultrastaging, patho-
sensitivity, expense, and labor intensity. Recently a protocol logic processing is so different that such discrepancies inevi-
was suggested that is designed to pick up the majority of all tably affect the accuracy of detection of not only MIC but also
MICs (Figure 11.5).51 This protocol consists of slicing SLNs at small MAC. Third, the designs of the studies differed consid-
2-mm intervals and cutting the node into four sections (slides) erably. In some of them, SLNs were prospectively assessed by
at 200-μm levels (one hematoxylin-eosin [H&E], one IHC ultrastaging, while other pelvic LNs were processed by stan-
pan-cytokeratin antibody, and two unstained sections that can dard H&E evaluation. In others, tissue blocks from all pelvic
be used for assessing whether the tumor focus is becoming LNs were reevaluated retrospectively from patients who were
larger) (Figure 11.5b). It results in approximately 20 pairs of LN-negative at the time of the treatment. There are substantial
H&E and IHC-stained slides per SLN. Such a protocol is more differences in cohort sizes, disease stages, and the proportion
intensive than the ‘average’ protocol described in the literature of cases that received adjuvant therapy. And fnally, studies
(Figure 11.5a), and it represents a compromise that enables the differed substantially in the strategy of adjuvant treatment, and
detection of the majority of MAC and MIC. some of them did not even report what proportion of patients
It has been consistently shown that pathological ultrastag- with MAC or MIC received adjuvant radiotherapy.
ing detects about 5%–10% of patients with early-stage cervical Three ongoing international prospective trials planning
cancer with only MIC in their SLN.5,15,21,52 Numerous papers to accrue 600, 475, and 500 patients who will undergo SLN
presented data on the prevalence of MIC; very few of them, biopsy only should bring new evidence on the signifcance of
however, evaluated the impact of MIC on the prognosis, and MIC for the prognosis. Estimating a 10% rate of MIC in SLNs,
the data are not consistent. For the frst time, the potential these studies should result in cumulative data on about 150
signifcance of MIC was suggested by the French group.23 In cases with MIC. However, data on the oncological outcome
a case-control study, they compared a group of 26 recurred will not be available until 2022 and 2027. Furthermore, many
patients with the same number of matched controls without of the patients with MIC will likely receive adjuvant treatment,
recurrence. The relative risk of recurrence was 2.44 (p < 0.01) so any comparison with the LN-negative group will be biased.
for MIC and 2.64 (p < 0.01) for LVSI. Since the frst French Concerning ITC, available data are even more inconclu-
publication, another six retrospective studies have presented sive, and we must accept the fact that their impact on the
data about the risk of recurrence in patients with MIC; all but prognosis will likely never be proven. The main reason is that
Sentinel lymph node mapping in cervical cancer 77

FIGURE 11.5 Comparison between a commonly used ultrastaging protocol composed of two levels only (a) and a suggested protocol processing
SLN tissue completely (b). FFPE, formalin-fxed, paraffn-embedded; HE, hematoxylin-eosin stain; IHC, immunohistochemistry. (From Ref. 39 under
Creative Commons license.)

pathological processing will never be so intensive that it will


SLN in fertility-sparing treatment
be able to reliably detect all ITCs – ITC detection will always
be relative to the protocol for ultrastaging. The negative status of pelvic LNs is a key prerequisite for
In summary, although available data are inconsistent, for fertility-sparing management. Metastatic LN involvement
the safety of our patients, it is reasonable to consider the pres- shifts the patient to a high-risk group in which adjuvant treat-
ence of MIC as a negative prognostic factor. We should apply ment is indicated. With the current treatment options, we are
the same strategy to MIC patients as for patients with MAC, unable to offer a safe option for fertility-sparing treatment to
at least until the data from ongoing prospective trials proves LN-positive patients. The only available alternative is the pres-
that patients with MIC have an equal prognosis to LN-negative ervation of the ovaries for assisted reproductive techniques
patients, without adjuvant treatment. and surrogate pregnancy.
78 Clinical Lymphatic Mapping

Surgical LN staging should always be the frst step in the group as compared to no occurrence among 139 patients after
fertility-sparing algorithm. The advantages of SLN biopsy are SLN removal only.84 Finally, the SLN-only group of cervical
even more important in those cases than in nonfertility-sparing cancer patients in the prospective randomized SENTICOL II
management. Dispatching SLN for ultrastaging increases study also self-reported less severe symptoms when compared
the chances of detecting small metastases and improves the to the group who underwent full PLND.85
safety of fertility-sparing management. In cases with a posi- All available literature, mostly based on retrospective data,
tive result, intraoperative SLN assessment allows one to step consistently document that the risk of LLL is reduced to a min-
away from the planned fertility-sparing procedure and instead imum after RH in combination with SLN biopsy. Currently
perform paraaortic LN staging concomitantly with the trans- ongoing prospective trials should bring fnal evidence to con-
position of ovaries. frm these expectations.
Given the fact that fertility-sparing candidates are patients
with small tumors, the frequency of LN positivity in this
Ongoing clinical trials on the
population is fortunately low. Despite that, the result of frozen
section can be falsely negative, and the presence of a small- safety of SLN biopsy only
size metastasis can be reported as late as with defnite pathol- Three prospective trials are currently registered at clinicaltrials.
ogy.59 It is important to inform the patient about such an option gov with safety of SLN biopsy in cervical cancer treatment
before the surgery. as the primary endpoint and have been actively recruiting
patients. The frst study, SENTIX (NCT02494063), was reg-
istered in July 2015 and is a prospective observational single-
SLN and lower leg lymphedema
arm study with the aim of enrolling 600 cases with tumors
Lower leg lymphedema (LLL) is the most prevalent postopera- ≤4 cm, who will undergo SLN biopsy only.44 Enrollment is
tive morbidity following the treatment of gynecologic cancer. estimated to complete by the end of 2020, with the survival
Lymphedema is a manifestation of lymphatic system insuff- data maturing 2 years later. A Chinese trial was registered by
ciency and deranged lymph transport.29 It is characterized by Sun Yat-Sen University in December 2015 (NCT02642471).
the swelling of one or both lower limbs caused by excess accu- It is enrolling patients with tumors up to 3 cm and randomiz-
mulation of water, plasma proteins, extravascular blood cells, ing separately those with negative or positive SLN by frozen
and parenchymal/stromal cell products in the limb tissues.60 section to four arms, with and without additional PLND. An
LLL signifcantly decreases the quality of life of gyneco- update in 2020 reported that the target enrollment has been
logic cancer survivors, negatively affecting daily activities increased from 800 to 1080 cases, with the study comple-
as well as their social and sexual life.61 Since the worldwide tion date remaining December 2022. Another randomized
median of cervical cancer diagnosis is in the patients’ mid- trial, ARGACY/GINECO (NCT03386734), was registered
to-late 40s, minimizing the long-term risk of LLL in this in 2017 by a French group, with the aim of randomizing 950
relatively young population is of particular importance, and patients with tumors ≤4 cm into two arms: 1) SLN in combi-
it is the main reason for an effort to replace PLND by SLN nation with PLND and 2) SLN biopsy only, with estimated
biopsy only.62 completion in May 2026.86 These three trials should bring
The most signifcant risk factor for LLL development after practice-changing evidence about the safety of the SLN con-
cervical cancer treatment is the performance of PLND.63–66 cept in cervical cancer, if, hopefully, they deliver affrmative
Radical hysterectomy (RH) followed by PLND was described results.
to have an LLL incidence ranging between 20% and 56%.67–72
It was shown that the extent of PLND and the number of
removed LNs are factors directly related to the risk of LLL
Summary
development; however, the critical number of removed LNs
described in the literature varies between 10 and 31.72–77 Our knowledge about the SLN concept in cervical cancer has
Multiple randomized studies have demonstrated that the grown substantially since the frst edition of this book. The
implementation of SLN biopsy reduces the risk of LLL devel- technique of tracer application and SLN detection in the pelvis
opment after gynecologic cancer treatment, including the is currently well standardized, and we have robust data on the
treatment of endometrial or vulvar cancer.78–81 However, data SLN detection rate, as well as on the most frequent localiza-
on lymphedema in cervical cancer patients after SLN biopsy tion of SLN in the pelvis. We have gathered data from many
only are still scarce. retrospective studies showing that SLN biopsy signifcantly
In the prospective pilot paper of Niikura et al.,82 new symp- increases the sensitivity of pelvic LN staging thanks to inten-
tomatic LLL was identifed in 2 (8.7%) of the 23 cervical can- sive pathologic processing, which is able to detect more metas-
cer patients who underwent RH with SLN biopsy only and in tases of small dimensions, which would not be detected by a
5 (42%) of 12 patients who underwent RH with PLND. A ret- conventional pathological assessment. We have confrmed that
rospective review of 167 cervical cancer patients showed that SLN biopsy can be consistently used in all patients with early-
among 70 patients after RH with SLN biopsy only, none of stage disease, irrespective of tumor size or patient body mass
them developed LLL as compared to 13.4% of patients who index (BMI). We know that SLN frozen section can be used to
underwent RH with full PLND.83 Such a promising outcome adjust the management intraoperatively, but we have to accept
was confrmed by another retrospective study in which lymph- that approximately 50% of positive LNs will be detected as
edema was identifed in 22.4% (15/ 67) of patients in the PLND late as by fnal ultrastaging. Finally, available data confrm
Sentinel lymph node mapping in cervical cancer 79

that replacing PLND with SLN biopsy can nearly eradicate 14. Slama J, Dundr P, Dusek L, et al. Sentinel lymph node
the occurrence of LLL. status in patients with locally advanced cervical cancers
However, after 20 years of clinical research, we still have and impact of neoadjuvant chemotherapy. Gynecol Oncol.
some questions that remain unanswered. Discussions continue 2012;125(2):303–6.
about the signifcance of micrometastases for the prognosis 15. Salvo G, Ramirez PT, Levenback CF, et al. Sensitivity and
because the evidence is not conclusive and is based on very negative predictive value for sentinel lymph node biopsy in
small numbers of recurrences in these cases. Solving this puz- women with early-stage cervical cancer. Gynecol Oncol.
zle will also enable standardization of an international ultrast- 2017;145(1):96–101.
aging protocol. Foremost, we can expect to soon get evidence 16. Beavis AL, Salazar-Marioni S, Sinno AK, et al. Sentinel
from prospective studies about the safety of replacing tradi- lymph node detection rates using indocyanine green in
tional systematic PLND with SLN biopsy. women with early-stage cervical cancer. Gynecol Oncol.
2016;143(2):302–6.
17. Rob L, Robova H, Halaska MJ, et al. Current status of sen-
tinel lymph node mapping in the management of cervical
REFERENCES cancer. Expert Rev Anticancer Ther. 2013;13(7):861–70.
1. Wui-Jin K, Nadeem RA-R, Sarah B, et al. Cervical can- 18. Dostalek L, Zikan M, Fischerova D, et al. SLN biopsy in
cer, version 3.2019, NCCN clinical practice guidelines in cervical cancer patients with tumors larger than 2cm and
Oncology. J Natl Compr Canc Netw. 2019;17(1):64–84. 4cm. Gynecol Oncol. 2018;148(3):456–60.
2. Cibula D, Abu-Rustum NR. Pelvic lymphadenectomy in 19. Cibula D, Kuzel D, Slama J, et al. Sentinel node (SLN)
cervical cancer – surgical anatomy and proposal for a new biopsy in the management of locally advanced cervical can-
classifcation system. Gynecol Oncol. 2010;116(1):33–7. cer. Gynecol Oncol. 2009;115(1):46–50.
3. Rob L, Strnad P, Robova H, et al. Study of lymphatic map- 20. Rob L, Robova H, Chmel R, et al. Surgical options in early
ping and sentinel node identifcation in early stage cervical cervical cancer. Int J Hyperthermia. 2012;28(6):489–500.
cancer. Gynecol Oncol. 2005;98(2):281–8. 21. Cibula D, Abu-Rustum NR, Dusek L, et al. Bilateral ultra-
4. Balaya V, Bresset A, Guani B, et al. Risk factors for failure staging of sentinel lymph node in cervical cancer: lower-
of bilateral sentinel lymph node mapping in early-stage cer- ing the false-negative rate and improving the detection of
vical cancer. Gynecol Oncol. 2020;156(1):93–9. micrometastasis. Gynecol Oncol. 2012;127(3):462–6.
5. Cibula D, Kocian R, Plaikner A, et al. Sentinel lymph node 22. Cibula D, Abu-Rustum NR, Fischerova D, et al. Surgical
mapping and intraoperative assessment in a prospective, treatment of ‘intermediate risk’ lymph node negative cervi-
international, multicentre, observational trial of patients cal cancer patients without adjuvant radiotherapy: a retro-
with cervical cancer: the SENTIX trial. Eur J Cancer. spective cohort study and review of the literature. Gynecol
2020;137:69–80. Oncol. 2018;151(3):438–43.
6. Cibula D, Abu-Rustum NR. Classifcation system for pel- 23. Marchiole P, Buenerd A, Benchaib M, et al. Clinical sig-
vic lymphadenectomy in cervical cancer. Gynecol Oncol. nifcance of lympho vascular space involvement and lymph
2010;118(1):94–5. node micrometastases in early-stage cervical cancer: a
7. Lecuru F, Mathevet P, Querleu D, et al. Bilateral nega- retrospective case-control surgico-pathological study.
tive sentinel nodes accurately predict absence of lymph Gynecol Oncol. 2005;97(3):727–32.
node metastasis in early cervical cancer: results of the 24. Barranger E, Cortez A, Commo F, et al. Histopathological
SENTICOL study. J Clin Oncol. 2011;29(13):1686–91. validation of the sentinel node concept in cervical cancer.
8. Frumovitz M, Euscher ED, Deavers MT, et al. ‘Triple injec- Ann Oncol. 2004;15(6):870–4.
tion’ lymphatic mapping technique to determine if parame- 25. Popa I, Plante M, Renaud MC, et al. Negative sentinel
trial nodes are the true sentinel lymph nodes in women with lymph node accurately predicts negative status of pelvic
cervical cancer. Gynecol Oncol. 2012;127(3):467–71. lymph nodes in uterine cervix carcinoma. Gynecol Oncol.
9. Benedetti-Panici P, Maneschi F, D’Andrea G, et al. Early 2006;103(2):649–53.
cervical carcinoma. Cancer. 2000;88(10):2267–74. 26. Okamoto S, Niikura H, Nakabayashi K, et al. Detection of
10. Bats AS, Mathevet P, Buenerd A, et al. The sentinel node sentinel lymph node metastases in cervical cancer: assess-
technique detects unexpected drainage pathways and ment of KRT19 mRNA in the one-step nucleic acid amplif-
allows nodal ultrastaging in early cervical cancer: insights cation (OSNA) method. Gynecol Oncol. 2013;130(3):530–6.
from the multicenter prospective SENTICOL study. Ann 27. Cibula D, Zikan M, Slama J, et al. Risk of micrometasta-
Surg Oncol. 2013;20(2):413–22. ses in non-sentinel pelvic lymph nodes in cervical cancer.
11. Balaya V, Mathevet P, Magaud L, et al. Predictive factors Gynecol Oncol. 2016;143(1):83–6.
of unexpected lymphatic drainage pathways in early-stage 28. Vercellino GF, Erdemoglu E, Lichtenberg P, et al. A GCIG
cervical cancer. Gynecol Oncol. 2019;154(1):102–9. international survey: clinical practice patterns of sentinel
12. Wydra D, Sawicki S, Wojtylak S, et al. Sentinel node iden- lymph node biopsies in cervical cancer. Arch Gynecol
tifcation in cervical cancer patients undergoing transperi- Obstet. 2019;300(1):191–9.
toneal radical hysterectomy: a study of 100 cases. Int J 29. The diagnosis and treatment of peripheral lymphedema:
Gynecol Cancer. 2006;16(2):649–54. 2016 consensus document of the international society of
13. Altgassen C, Hertel H, Brandstadt A, et al. Multicenter lymphology. Lymphology. 2016;49(4):170–84.
validation study of the sentinel lymph node concept 30. Dostalek L, Avall-Lundqvist E, Creutzberg CL, et al. ESGO
in cervical cancer: AGO Study Group. J Clin Oncol. survey on current practice in the management of cervical
2008;26(18):2943–51. cancer. Int J Gynecol Cancer. 2018;28(6):1226–31.
80 Clinical Lymphatic Mapping

31. Cibula D, Potter R, Planchamp F, et al. The European 48. Bats AS, Clement D, Larousserie F, et al. Sentinel lymph
Society of Gynaecological Oncology/European Society node biopsy improves staging in early cervical cancer.
for Radiotherapy and Oncology/European Society of Gynecol Oncol. 2007;105(1):189–93.
Pathology guidelines for the management of patients with 49. Brar H, Hogen L, Covens A. Cost-effectiveness of sentinel
cervical cancer. Virchows Archiv. 2018;472(6):919–36. node biopsy and pathological ultrastaging in patients with
32. Marth C, Landoni F, Mahner S, et al. Cervical cancer: early-stage cervical cancer. Cancer. 2017;123(10):1751–9.
ESMO Clinical Practice Guidelines for diagnosis, treatment 50. Dundr P, Cibula D, Nemejcova K, et al. Pathologic proto-
and follow-up. Ann Oncol. 2017;28(Suppl 4):iv72–iv83. cols for sentinel lymph nodes ultrastaging in cervical can-
33. de Juan A, Redondo A, Rubio MJ, et al. SEOM clinical cer. Arch Pathol Lab Med. 2020;144(8):1011–20.
guidelines for cervical cancer (2019). Clin Transl Oncol. 51. Cibula D, McCluggage WG. Sentinel lymph node (SLN)
2020;22(2):270–8. concept in cervical cancer: current limitations and unan-
34. NCCN guidelines version 2.2020; 2020. Available from: swered questions. Gynecol Oncol. 2019;152(1):202–7.
www.nccn.org/professionals/physician_gls/pdf/cervical.pdf. 52. Guani B, Balaya V, Magaud L, et al. The clinical impact
35. International Gynecologic Cancer Society. Available from: of low-volume lymph nodal metastases in early-stage cer-
https://igcs.org/education-resources/clinical-resources/. vical cancer: the senticol 1 and senticol 2 trials. Cancers.
36. Cancer Australia. Available from: www.canceraustralia 2020;12(5).
.gov.au/affected-cancer/cancer-types/cervical-cancer/ 53. Fregnani JH, Latorre MR, Novik PR, et al. Assessment of
health-professionals. pelvic lymph node micrometastatic disease in stages IB
37. National Health Commission of the People’s Republic of and IIA of carcinoma of the uterine cervix. Int J Gynecol
C. Chinese guidelines for diagnosis and treatment of cer- Cancer. 2006;16(3):1188–94.
vical cancer 2018 (English version). Chin J Cancer Res. 54. Horn LC, Hentschel B, Fischer U, et al. Detection of micro-
2019;31(2):295–305. metastases in pelvic lymph nodes in patients with carci-
38. Ebina Y, Mikami M, Nagase S, et al. Japan Society of noma of the cervix uteri using step sectioning: frequency,
Gynecologic Oncology guidelines 2017 for the treatment of topographic distribution and prognostic impact. Gynecol
uterine cervical cancer. Int J Clin Oncol. 2019;24(1):1–19. Oncol. 2008;111(2):276–81.
39. Lim MC, Lee M, Shim SH, et al. Practice guidelines for 55. Cibula D, Abu-Rustum NR, Dusek L, et al. Prognostic signif-
management of cervical cancer in Korea: a Korean Society icance of low volume sentinel lymph node disease in early-
of Gynecologic Oncology Consensus Statement. J Gynecol stage cervical cancer. Gynecol Oncol. 2012;124(3):496–501.
Oncol. 2017;28(3):e22. 56. Colturato LF, Signorini Filho RC, Fernandes RC, et al.
40. Holman LL, Levenback CF, Frumovitz M. Sentinel lymph Lymph node micrometastases in initial stage cervical
node evaluation in women with cervical cancer. J Minim cancer and tumoral recurrence. Int J Gynaecol Obstet.
Invasive Gynecol. 2014;21(4):540–5. 2016;133(1):69–75.
41. Martinez A, Mery E, Filleron T, et al. Accuracy of intra- 57. Stany MP, Stone PJ, Felix JC, et al. Lymph node microme-
operative pathological examination of SLN in cervical can- tastases in early-stage cervical cancer are not predictive of
cer. Gynecol Oncol. 2013;130(3):525–9. survival. Int J Gynecol Pathol. 2015;34(4):379–84.
42. Bats AS, Buenerd A, Querleu D, et al. Diagnostic value of 58. Kocian R, Slama J, Fischerova D, et al. Micrometastases
intraoperative examination of sentinel lymph node in early in sentinel lymph nodes represent a signifcant negative
cervical cancer: a prospective, multicenter study. Gynecol prognostic factor in early-stage cervical cancer: a single-
Oncol. 2011;123(2):230–5. institutional retrospective cohort study. Cancers. 2020;12(6).
43. Roy M, Bouchard-Fortier G, Popa I, et al. Value of senti- 59. Sonoda K, Yahata H, Okugawa K, et al. Value of intra-
nel node mapping in cancer of the cervix. Gynecol Oncol. operative cytological and pathological sentinel lymph node
2011;122(2):269–74. diagnosis in fertility-sparing trachelectomy for early-stage
44. Dostalek L, Slama J, Fisherova D, et al. Impact of sentinel cervical cancer. Oncology. 2018;94(2):92–8.
lymph node frozen section evaluation to avoid combined 60. Grada AA, Phillips TJ. Lymphedema: pathophysiol-
treatment in early-stage cervical cancer. Int J Gynecol ogy and clinical manifestations. J Am Acad Dermatol.
Cancer. 2020;30(6):744–8. 2017;77(6):1009–20.
45. Nemejcova K, Kocian R, Kohler C, et al. Central pathol- 61. Dunberger G, Lindquist H, Waldenstrom AC, et al. Lower
ogy review in SENTIX, a prospective observational inter- limb lymphedema in gynecological cancer survivors –
national study on sentinel lymph node biopsy in patients effect on daily life functioning. Support Care Cancer.
with early-stage cervical cancer (ENGOT-CX2). Cancers. 2013;21(11):3063–70.
2020;12(5). 62. Torre LA, Islami F, Siegel RL, et al. Global cancer in
46. Cibula D, Potter R, Planchamp F, et al. The European women: burden and trends. Cancer Epidemiol Biomarkers
Society of Gynaecological Oncology/European Society Prev. 2017;26(4):444–57.
for Radiotherapy and Oncology/European Society of 63. Hareyama H, Hada K, Goto K, et al. Prevalence, classif-
Pathology guidelines for the management of patients with cation, and risk factors for postoperative lower extremity
cervical cancer. Radiother Oncol. 2018;127(3):404–16. lymphedema in women with gynecologic malignancies: a
47. Cibula D, Potter R, Planchamp F, et al. The European retrospective study. Int J Gynecol Cancer. 2015;25(4):751–7.
Society of Gynaecological Oncology/European Society 64. Todo Y, Yamamoto R, Minobe S, et al. Risk factors for
for Radiotherapy and Oncology/European Society of postoperative lower-extremity lymphedema in endometrial
Pathology guidelines for the management of patients with cancer survivors who had treatment including lymphad-
cervical cancer. Int J Gynecol Cancer. 2018;28(4):641–55. enectomy. Gynecol Oncol. 2010;119(1):60–4.
Sentinel lymph node mapping in cervical cancer 81

65. Roman MM, Barbieux R, Nogaret JM, et al. Use of lympho- radiotherapy for cervical cancer. Strahlenther Onkol.
scintigraphy to differentiate primary versus secondary lower 2008;184(4):206–11.
extremity lymphedema after surgical lymphadenectomy: a 76. Kunitake T, Kakuma T, Ushijima K. Risk factors for lower
retrospective analysis. World J Surg Oncol. 2018;16(1):75. limb lymphedema in gynecologic cancer patients after ini-
66. Halaska MJ, Novackova M, Mala I, et al. A prospective tial treatment. Int J Clin Oncol. 2020;25(5):963–71.
study of postoperative lymphedema after surgery for cervi- 77. Togami S, Kawamura T, Fukuda M, et al. Risk factors
cal cancer. Int J Gynecol Cancer. 2010;20(5):900–4. for lymphatic complications following lymphadenec-
67. Kim SI, Lim MC, Lee JS, et al. Comparison of lower tomy in patients with cervical cancer. Jpn J Clin Oncol.
extremity edema in locally advanced cervical cancer: pre- 2018;48(12):1036–40.
treatment laparoscopic surgical staging with tailored radio- 78. Huang J, Yu N, Wang X, et al. Incidence of lower limb
therapy versus primary radiotherapy. Ann Surg Oncol. lymphedema after vulvar cancer: a systematic review and
2016;23(1):203–10. meta-analysis. Medicine. 2017;96(46):e8722.
68. Kuroda K, Yamamoto Y, Yanagisawa M, et al. Risk factors 79. Geppert B, Lonnerfors C, Bollino M, et al. Sentinel
and a prediction model for lower limb lymphedema following lymph node biopsy in endometrial cancer: Feasibility,
lymphadenectomy in gynecologic cancer: a hospital-based ret- safety and lymphatic complications. Gynecol Oncol.
rospective cohort study. BMC Women’s Health. 2017;17(1):50. 2018;148(3):491–8.
69. Ohba Y, Todo Y, Kobayashi N, et al. Risk factors for lower- 80. Accorsi GS, Paiva LL, Schmidt R, et al. Sentinel lymph
limb lymphedema after surgery for cervical cancer. Int J node mapping vs systematic lymphadenectomy for endo-
Clin Oncol. 2011;16(3):238–43. metrial cancer: surgical morbidity and lymphatic complica-
70. Biglia N, Librino A, Ottino MC, et al. Lower limb lymph- tions. J Minim Invasive Gynecol. 2020;27(4):938–45.e2.
edema and neurological complications after lymphad- 81. Biglia N, Zanfagnin V, Daniele A, et al. Lower body lymph-
enectomy for gynecological cancer. Int J Gynecol Cancer. edema in patients with gynecologic cancer. Anticancer Res.
2015;25(3):521–5. 2017;37(8):4005–15.
71. Yoshihara M, Shimono R, Tsuru S, et al. Risk factors for 82. Niikura H, Okamoto S, Otsuki T, et al. Prospective study of
late-onset lower limb lymphedema after gynecological can- sentinel lymph node biopsy without further pelvic lymph-
cer treatment: a multi-institutional retrospective study. Eur adenectomy in patients with sentinel lymph node-negative
J Surg Oncol. 2020;46(7):1334–8. cervical cancer. Int J Gynecol Cancer. 2012;22(7):1244–50.
72. Mendivil AA, Rettenmaier MA, Abaid LN, et al. Lower- 83. Togami S, Kubo R, Kawamura T, et al. Comparison of lym-
extremity lymphedema following management for endome- phatic complications between sentinel node navigation sur-
trial and cervical cancer. Surg Oncol. 2016;25(3):200–4. gery and pelvic lymphadenectomy in patients with cervical
73. Abu-Rustum NR, Alektiar K, Iasonos A, et al. The inci- cancer. Jpn J Clin Oncol. 2020;50(5):543–7.
dence of symptomatic lower-extremity lymphedema fol- 84. Yahata H, Kobayashi H, Sonoda K, et al. Prognostic out-
lowing treatment of uterine corpus malignancies: a 12-year come and complications of sentinel lymph node navigation
experience at Memorial Sloan-Kettering Cancer Center. surgery for early-stage cervical cancer. Int J Clin Oncol.
Gynecol Oncol. 2006;103(2):714–18. 2018;23(6):1167–72.
74. Todo Y, Yamazaki H, Takeshita S, et al. Close relationship 85. Gianoni M, Mathevet P, Uzan C, et al. Does the sentinel
between removal of circumfex iliac nodes to distal exter- lymph node sampling alone improve quality of life in early
nal iliac nodes and postoperative lower-extremity lymph- cervical cancer management? Front Surg. 2020;7:31.
edema in uterine corpus malignant tumors. Gynecol Oncol. 86. Lecuru FR, McCormack M, Hillemanns P, et al. SENTICOL
2015;139(1):160–4. III: an international validation study of sentinel node biopsy
75. Fuller J, Guderian D, Kohler C, et al. Lymph edema in early cervical cancer. A GINECO, ENGOT, GCIG and
of the lower extremities after lymphadenectomy and multicenter study. Int J Gynecol Cancer. 2019;29(4):829–34. Run n ing
12
Sentinel lymph node mapping in breast cancer

Angelena Crown and Mary L. Gemignani

CONTENTS
Introduction.................................................................................................................................................................................... 83
Breast cancer.................................................................................................................................................................................. 83
Patterns of nodal metastasis ................................................................................................................................................. 83
Pathology.............................................................................................................................................................................. 83
Ductal carcinoma in situ .......................................................................................................................................... 83
Lobular carcinoma in situ ........................................................................................................................................ 84
Invasive ductal carcinoma........................................................................................................................................ 84
Infltrating lobular carcinoma............................................................................................................................................... 85
Staging for breast cancer................................................................................................................................................................ 85
The axilla ....................................................................................................................................................................................... 87
SLNB.................................................................................................................................................................................... 87
Pathologic analysis of the SLN...................................................................................................................................................... 89
Intraoperative techniques ............................................................................................................................................................... 90
Blue dye ......................................................................................................................................................................................... 90
Radioisotope/nuclear medicine...................................................................................................................................................... 90
Indocyanine green .......................................................................................................................................................................... 90
The SLN mapping procedure......................................................................................................................................................... 90
Management of the clinically negative axilla....................................................................................................................... 93
SLNB after NAC............................................................................................................................................................................ 93
Follow-up after SLNB ................................................................................................................................................................... 95
Conclusion ..................................................................................................................................................................................... 95
References...................................................................................................................................................................................... 95

Introduction Breast cancer


Breast cancer is the most common malignancy among women Patterns of nodal metastasis
in the United States. In 2019, an estimated 268,600 new cases
The ipsilateral axilla is the most common site of metastatic
of breast cancer were diagnosed in American women, compris-
spread of breast cancer. In contrast, metastasis to the internal
ing 15.2% of all new cancer cases.1 Women have a 12.8% (1 in
mammary nodal basin occurs less frequently and is an inde-
8) lifetime risk of developing breast cancer.1 Although breast
pendent predictor of poor outcomes.2 Spread to the internal
cancer poses a serious health concern, increased screening and
mammary nodes is most commonly observed in the setting of
early detection of breast cancer, coupled with advances in sys-
inner quadrant lesions and in the presence of axillary nodal
temic therapies, have led to dramatic improvements in relative
metastasis.
survival rates, which now approach 90% at 5 years.1
Despite an increasing role for genomics, axillary lymph
node status remains one of the most important prognos- Pathology
tic indicators in breast cancer. Sentinel lymph node biopsy
(SLNB) in breast cancer evolved out of efforts to minimize
Ductal carcinoma in situ
the morbidity associated with axillary lymph node dissection Ductal carcinoma in situ (DCIS) comprises a heterogeneous
(ALND) while still providing important staging information. group of lesions with distinct growth patterns and cytologic
Sentinel lymph node (SLN) mapping with the use of blue dye features. Classifcation is based on an architectural pattern
and/or radioisotope allows selective resection of representa- and includes comedo, cribriform, and micropapillary patterns
tive nodes to provide accurate axillary staging in patients (Figure 12.1). These noninvasive lesions range from low grade
with breast cancer. to high grade and are thought to be nonobligate precursors to

DOI: 10.1201/9781003255536-12 83
84 Clinical Lymphatic Mapping

invasive cancer with the potential to degenerate into invasive


carcinoma over time.

Lobular carcinoma in situ


Lobular carcinoma in situ (LCIS) is a noninvasive lesion char-
acterized by solid proliferation of small cells with round to
oval nuclei and distention of the terminal duct–lobular units.
It is a marker for increased breast cancer risk in either breast
and is associated with a 2% annual incidence of breast cancer
(Figure 12.2).3

Invasive ductal carcinoma


The category of invasive ductal carcinoma makes up the major-
ity of malignant mammary tumors (65%–80%) (Figure 12.3).
This category also includes a group of lesions termed ‘special
histologic subtypes,’ which is composed of tubular, medullary,
FIGURE 12.1 Ductal carcinoma in situ, intermediate grade (H&E, metaplastic, mucinous, papillary, and adenoid cystic carcino-
100×). (Courtesy of Dr. Dilip Giri from the Department of Pathology at mas, all of which are associated with favorable outcomes.4
Memorial Sloan Kettering Cancer Center.)

FIGURE 12.2 (a) Lobular carcinoma in situ, classical type, demonstrating the characteristic distended acini flled with small monomorphic cells with
round nuclei (H&E, 100×). (b) Arrow points to typical fried egg morphology of the neoplastic cells (H&E, 200×). (Courtesy of Dr. Dilip Giri from the
Department of Pathology at Memorial Sloan Kettering Cancer Center.)
Sentinel lymph node mapping 85

Infiltrating lobular carcinoma


Infltrating lobular carcinomas constitute 10%–14% of inva-
sive breast carcinomas (Figure 12.4). These tumors tend to
grow circumferentially around ducts and lobules, with a lin-
ear, ‘Indian fle,’ targetoid growth pattern.

Staging for breast cancer


The American Joint Committee on Cancer (AJCC) assigns
clinical and pathologic stages according to the tumor–node–
metastasis (TNM) system. The eighth edition of the AJCC
staging system for breast cancer incorporates SLNB results
into the anatomic stage and distinguishes micrometastases
from isolated tumor cells (ITCs) on the basis of size and his-
tologic evidence of malignant activity.5 In this most recent
edition, the AJCC assigns both anatomic stage groups and
FIGURE 12.3 Well to moderately differentiated invasive ductal carci-
prognostic stage groups that rely both on the anatomic
noma, tubular type. The tumor is exclusively composed of well-formed extent of disease and on biomarkers. The AJCC requires
tubular glands (arrows) (H&E, 100×). (Courtesy of Dr. Dilip Giri from the that cancer registries in the United States use the prognostic
Department of Pathology at Memorial Sloan Kettering Cancer Center.) stage groups for case reporting. These biomarkers include

FIGURE 12.4 (a and b) Invasive lobular carcinoma, classical type. Arrow denotes characteristic Indian-fle linear arrangement (H&E, 200×).
(Courtesy of Dr. Dilip Giri from the Department of Pathology at Memorial Sloan Kettering Cancer Center.)
86 Clinical Lymphatic Mapping

expression of estrogen receptor (ER) and progesterone


receptor (PR), both of which are considered positive if ≥1%
of tumor cells stain for the receptor on immunohistochemi-
cal (IHC) evaluation regardless of staining intensity (Figure
12.5). Human epidermal growth factor receptor 2 (HER2) is
evaluated by immunohistochemistry or, in equivocal cases,
by fuorescent in situ hybridization (Figure 12.6). Histologic
grade is calculated as the sum of values assigned to mor-
phologic features, including tubule formation, mitotic count,
and nuclear pleomorphism. The Oncotype DX (Genomic
Health, Redwood City, CA) genetic assay assigns recurrence
scores (RSs) that are incorporated into staging for patients
with ER/PR-positive, HER2-negative tumors that are >5
mm and ≤50 mm in size.

FIGURE 12.5 (a) Well to moderately differentiated invasive ductal car-


cinoma with well-formed glandular structures (H&E, 100×). (b) Tumor
is strongly positive for ER with >95% of tumor cells exhibiting strong FIGURE 12.6 (a) Moderately differentiated invasive ductal carcinoma
nuclear staining (6F11, Leica, 100×). (c) Tumor is strongly positive for with micropapillary features (H&E, 100×). (b) The tumor is HER2+ with
PR with >95% of tumor cells exhibiting strong nuclear staining (Clone 3+ staining in 100% of tumor cells (4B5, Ventana, 100×). (Courtesy of
16, Leica, 100×). (Courtesy of Dr. Dilip Giri from the Department of Dr. Dilip Giri from the Department of Pathology at Memorial Sloan
Pathology at Memorial Sloan Kettering Cancer Center.) Kettering Cancer Center.)
Sentinel lymph node mapping 87

node metastasis may be present in >10% of patients and that


The axilla the incidence of lymph node metastasis increases with larger
tumor size.6
The axilla is a pyramidal space between the arm and thoracic A complete level I and II ALND provides excellent regional
wall. It contains the axillary vessels and their branches, the control, with axillary recurrence occurring in <1% of patients,
brachial plexus and its branches, and lymph nodes embedded but comes at the expense of signifcant morbidity, with 10%–
in fatty tissue. The primary route of lymphatic drainage of the 15% of patients developing lymphedema.7–9 Additionally, arm
breast is through the ipsilateral axillary lymph nodes (Figure numbness and tingling, pain, shoulder abduction defcits, and
12.7a). The boundaries of a level I and II ALND include the weakness are well-documented sequelae of this procedure.9
axillary vein superiorly, the serratus medially, and the latis-
simus dorsi laterally. The inferior extent is defned by the con-
vergence of the latissimus dorsi and serratus anterior muscles. SLNB
Care is taken to avoid injury to the long thoracic neurovascular
bundle, which travels medially and inserts into the serratus Metastatic involvement of lymph nodes is thought to occur in
anterior at approximately T4, and to the thoracodorsal neuro- a stepwise manner. The SLN concept was frst introduced by
vascular bundle, which innervates the latissimus dorsi. Cabanas in penile carcinoma.10 He postulated that the SLN,
At the time of Halsted’s radical mastectomy, complete the frst lymph node in a regional lymphatic basin to receive
ALND, including levels I–III, was routine, given the impor- lymph fow from a primary tumor, would accurately refect the
tance of nodal status for prognostication. The status of the status of the remainder of the nodes in that regional lymphatic
axilla remains one of the most important prognostic factors basin (Figure 12.7b). Rosen demonstrated the incidence of
today. Although valuable, clinical examination of the axilla “skip metastasis” (nodal disease present in level 3 but in nei-
is neither sensitive nor specifc, and a negative clinical exam ther level 1 nor level 2) to be <2%.11 Given the predictable pat-
does not guarantee absence of metastatic disease. Multiple tern of metastasis, SLNB emerged as an alternative to ALND
studies have shown that even in tumors <1 cm in size, lymph for axillary staging.

FIGURE 12.7 (a) Axillary lymph nodes. The axillary and substernal lymph nodes that drain the breast are shown. The lymph nodes are also divided
into levels by location relative to the pectoralis minor. Level I (low) lymph nodes lie lateral to the lateral border of the pectoralis minor muscle. Level
II (mid) nodes lie behind the pectoralis minor muscle, and level 3 (high) nodes are medial (deep) to the medial border of this muscle. (From Gemignani
ML, Sentinel lymph node mapping in breast cancer and melanoma, Operative Techniques in Gynecologic Surgery [2001] 6:16–20, with permission.)
88 Clinical Lymphatic Mapping

FIGURE 12.7 (b) The sentinel lymph node is the frst lymph node that receives direct drainage from the primary tumor. Because it is the frst site of
drainage, it should be the frst site of lymphatic spread. A tumor-free sentinel lymph node implies the absence of lymph node metastases in the entire
lymphatic basin.

Initial studies in melanoma by Morton and colleagues dem- TABLE 12.1


onstrated the feasibility of the SLNB concept in patients with Selected Large Validation Studies of SLNB in Breast Cancer
cutaneous melanoma.12 This initial study reported success-
False negative
ful identifcation of the SLN in 82% of the 237 patients who
Trial N Treatment arms rate
received intradermal injections of blue dye. SLNB accurately
predicted the regional nodal status in 99% of successful proce- Louisville 1436 Group 1: SLNB with blue 8.3%
Sentinel Node dye -> ALND (range,
dures and in 95% of cases with positive nodes.
Trial17 Group 2: SLNB with colloid 4.3%–14.3%)
The validity of the SLN concept hinges upon the accurate (2001) -> ALND
prediction of the status of the regional lymphatic basin based Group 3: SLNB with dual
on the SLN status. Studies have shown that successful iden- mapping -> ALND
tifcation of the SLN occurs 92%–98% of the time, with the Milan16 516 Group 1: SLNB + ALND 8.8%
combination of blue dye and radioisotope improving the rate (2003) Group 2: SLNB with ALND
of detection of SLNs. The positive predictive value of the tech- only if SLN+
nique approaches 100%, with a negative predictive value close ALMANAC14 1031 SLNB with blue dye + 6.7%
to 95% when both blue dye and radioisotope are used. The (2006) colloid followed by ALND
false-negative rate (FNR) is approximately 5%–10% in most NSABP B-3215 5611 Group 1: SLNB + ALND 9.8%
studies (Table 12.1).13–17 (2007) Group 2: SLNB with ALND
The positive predictive value of SLNB increases with the only if SLN+
number of sentinel nodes retrieved, with positive predictive SNAC13 1088 Group 1: SLNB with ALND 5.5%
(2009) only if SLN+
values ranging from 75% for a single sentinel node to 98% for
Group 2: SLNB + ALND
three sentinel nodes.18 Resection of non-SLNs does not appear
to add value to the SLNB procedure. Turner et al. performed Note: These trials all demonstrated false-negative rates between 5% and 10%.
IHC staining of all lymph nodes removed, sentinel and nonsen- Abbreviations: SLNB, sentinel lymph node biopsy; ALND, axillary
tinel, in a series of women undergoing standard ALND with lymph node dissection; SLN, sentinel lymph node; ALMANAC, Axillary
clinically negative nodes (cN0).19 A total of 157 SLNs were Lymphatic Mapping Against Nodal Axillary Clearance; NSABP,
examined; 10 (6%) demonstrated IHC positivity compared National Sentinel Adjuvant Breast and Bowel Project; SNAC, Sentinel
with only 1 of 1087 non-SLNs (0.09%). They concluded that Node Biopsy vs. Axillary Clearance
the probability of non-SLNs containing metastases was <0.1%
when both hematoxylin-eosin (H&E) and IHC stains of the
SLN were negative, indicating that resection of SLNs does not the management of any cN0 patients with invasive breast can-
signifcantly increase the positive predictive value of SLNB. cer. SLNB should be performed by a team that can proceed to
SLNB can be performed in conjunction with either mastec- ALND in the event that the SLN procedure is unsuccessful.
tomy or breast-conserving surgery (BCS) and is standard in It is not routinely indicated for DCIS and should be reserved
Sentinel lymph node mapping 89

for cases in which there is a suspicion of microinvasion, as nodal metastases and is possible because only a few SLNs are
the rate of positive nodes approaches 6%, or in the setting of obtained at the time of the procedure. Neither serial sectioning
mastectomy, where occult invasive disease is found in 20% of nor IHC would be practical or cost-effective in a conventional
mastectomy specimens.20–22 ALND, which yields an average of 20 nodes.33
Because of the paramount importance of nodal status in IHC stains used in breast cancer are monoclonal antibod-
the prognosis of breast cancer, fndings on SLNB inform sys- ies against the cytokeratins AE1/AE3 and CAM 5.2. Several
temic therapy recommendations. Nodal metastasis is currently studies have demonstrated an increase in the diagnostic yield
an indication for adjuvant chemotherapy in all breast cancer of occult nodal metastases with the use of IHC analysis in
subtypes.23 The RxPONDER trial is evaluating whether low- patients thought to have negative nodes through standard
volume nodal disease in the setting of a low RS should remain histologic analysis using H&E stains. Use of IHC stains can
an indication for adjuvant chemotherapy in women with cN0 increase the yield of detection of tumor cells by 10%–20%
ER/PR+, HER2− breast cancer.24 over standard histopathologic analysis.34,35
Use of SNLB in elderly patients is more nuanced. Hughes According to AJCC staging, metastases <2 mm in size are
et al. reported a 12-year follow-up on 636 women age ≥70 classifed as micrometastases.5 The question of whether the
years with cT1N0M0 tumors who were randomized to presence of isolated micrometastases on SLNB necessitates
lumpectomy and tamoxifen or to lumpectomy, tamoxifen, full ALND was initially controversial; however, two prospec-
and adjuvant radiation therapy (RT) and found that omission tive national trials found that micrometastases are associated
of RT was not associated with a deleterious effect on sur- with minimal changes in prognosis. The American College of
vival.25 Breast cancer–specifc survival at 10 years was 97% Surgeons Oncology Group (ACOSOG) Z0010 trial evaluated
in the tamoxifen group compared to 98% in the tamoxifen the impact of occult lymph node metastases on survival in
plus radiation group.25 The majority of patients in the study, women undergoing breast-conserving therapy (BCT); it found
64%, did not undergo axillary surgery.26 After 12 years of that 10.5% of women who had negative SLNs on H&E had
follow-up, only six patients (1% of those who did not undergo positive fndings on IHC.34 There was no difference in overall
axillary surgery) in the entire study had an axillary recur- survival (OS) or disease-free survival (DFS) in women who
rence – all in patients who did not undergo ALND and did were found to have micrometastases compared to women
not receive RT.25 Given these reassuring fndings, this study whose lymph nodes were negative on both H&E and IHC.
provided level 1 evidence for selective use of axillary surgery The National Surgical Adjuvant Breast and Bowel Project
in patients age ≥70 years. Indeed, the Society of Surgical (NSABP) B-32 study randomized women with cT1–2N0
Oncology issued a “Choosing Wisely” recommendation that undergoing BCT or mastectomy with negative SLNs on H&E
SLNB be omitted in cN0 women age ≥70 years with early- to SLNB alone or full ALND. The study found that occult
stage ER/PR+, HER2– breast cancers; however, this recom- metastases were present in 15.9% of women and that the pres-
mendation allows for individual consideration of axillary ence of occult nodal metastases was associated with a 1.2%
staging in cases in which a positive result would affect sys- reduction in OS, a 2.8% decrease in DFS, and a 2.8% reduction
temic therapy recommendations.27 in distant DFS at 60 months.7
Historically, SLN mapping was thought be compromised Galimberti and colleagues reported results from
in cases of disrupted or aberrant lymphatic drainage pat- International Breast Cancer Study Group 23–01, a random-
terns, which may be caused by large tumors, previous RT, ized control trial that confrmed the oncologic safety of the
multicentric tumors, or the presence of a large cavity pro- omission of ALND in patients with cT1–2N0 tumors undergo-
duced from a prior excision. With modern mapping tech- ing BCT or mastectomy with fndings of micrometastases on
niques, these conditions do not constitute contraindications SNLB.36 In this trial, women with early-stage breast cancer
to SLNB, and we routinely perform SLNB in these cases. with no palpable adenopathy who were found to have micro-
Pregnancy was also considered a relative contraindication metastases on SLNB were randomized to completion ALND
to SLNB due to concerns for radiation exposure to the fetus or no further axillary surgery. After a median follow-up of
from the radioisotope and for allergic reactions to blue dye; 9.7 years, they found no difference in DFS between groups:
however, modern series quantifying radiation dose to the 76.8% in the no further axillary surgery group and 74.9% in
fetus have demonstrated that fetal exposure is negligible and the completion axillary dissection group.37 However, lymph-
have suggested that technetium colloid use for SLNB is safe edema rates were 13% in the completion ALND group, com-
during pregnancy.28–30 pared to 5% in the no further axillary surgery group, again
highlighting the increased morbidity associated with conven-
tional ALND.
These prospective trials support the current practice of
Pathologic analysis of the SLN omitting ALND in patients with isolated micrometastases.
In contrast to ALND, SLNB has the advantage of facilitating Additionally, together with the landmark ACOSOG Z0011
the use of enhanced pathologic techniques, such as IHC, for trial that confrmed the oncologic safety of omitting ALND
the evaluation of the SLNs. Overall, greater scrutiny is paid to in patients with fewer than three nodes with macrometastases,
SLNs through serial sectioning and IHC staining. Serial sec- these trials have led to more selective use of IHC staining, as
tioning improves the detection of metastases and increases the the presence of isolated micrometastases has been shown to
yield (up to 30%) in H&E analysis over single sectioning.31,32 have minimal prognostic value outside of the neoadjuvant che-
The addition of IHC staining further increases the detection of motherapy setting.38
90 Clinical Lymphatic Mapping

Intraoperative techniques Radioisotope/nuclear medicine


Frozen-section H&E analysis allows visualization of the In the United States, the majority of the published experience
nodal architecture and can be used to inform operative deci- has been with technetium-99m sulfur colloid (99mTc-SC),
sion making. It has been reported to have an FNR of 6%–24% while in Europe, 99mTc colloidal albumin is used. Filtration of
compared with fnal analysis on paraffn and is more reliable the colloid produces uniformity of particle size and is standard
for detection of macrometastatic disease.31,32 Disadvantages of in lymphatic mapping in melanoma, whereas the unfltered
this technique include frozen-tissue artifact, the consumption colloid is suffcient in breast cancer.45 In melanoma, lympho-
of tissue, and intraoperative delay. scintigraphy reveals anomalous drainage patterns and iden-
The use of imprint cytology has also been reported in SLN tifes the lymphatic basin to be investigated. Because the
mapping. Imprint cytology uses touch preparations, which can ipsilateral axilla is the primary focus of SLNB in breast can-
provide excellent cytologic detail and allow rapid analysis, as cer, a negative lymphoscintigram does not preclude successful
well as increased tissue preservation for use in paraffn sec- intraoperative localization of the SLN.
tions; however, there are inherent limitations to imprint cytol- A number of studies have found that SLNs identifed by
ogy, including the smaller number of cells examined compared intraparenchymal, subdermal, intradermal, or subareolar
with frozen-section analysis and an increased incidence of injection stage the axilla with comparable accuracy.46 It is
indeterminate results.39 thought that the entire breast and overlying skin function as
In the era of ACOSOG Z0011, we reserve frozen-section one, with drainage to an identifable SLN in most patients.47
nodal analysis for women undergoing mastectomy and for This may explain why such a wide variation in isotope tech-
women who received neoadjuvant chemotherapy (NAC), niques, including dosage, medium location, and timing of
as the chance of fnding more than two positive nodes in injection, produces similar SLN identifcation rates.
women with cT1–2N0 disease undergoing up-front surgery
is very low.
Indocyanine green
A novel tracer, indocyanine green (ICG), was developed in
Blue dye
2005.48 Advantages of ICG include delineating lymphatic
Isosulfan blue dye (Lymphazurin, Hirsch Industries, channels and real-time fow of lymph. Initial studies reported
Richmond, VA) is a monosodium salt of a 2,5-disulfonated tri- an SLN detection rate of 94%, with subsequent studies report-
phenyl methane dye.40 It binds weakly to albumin and is taken ing SLN detection rates approaching 100%, providing increas-
up selectively by the lymphatics. Incidence of severe allergic ing support for the use of ICG.49–51 In a study of 821 women
reaction is <1%. Allergic reactions include urticaria, rashes, with cN0 breast cancer who underwent SLNB, Sugie et al.
and ‘blue hives’; however, anaphylactic reactions with cardio- compared SLNB using ICG to SLNB using traditional radio-
vascular collapse have also been reported. isotope.52 Rates of SLN detection were equivalent (both 97%),
A study from Memorial Sloan Kettering Cancer Center and use of both methods together led to a SLN detection rate
(MSKCC) reported an allergic reaction rate of 1.6% among of 99.8%. ICG detected more SLNs (2.3 nodes vs. 1.7 nodes
2392 patients who received isosulfan blue dye as part of for radioisotope) and increased the detection of positive nodes
the SLNB procedure.41 The majority of the reactions (69%) (7.2% of positive nodes were fuorescent but not radioactive).
were blue hives and urticaria. The incidence of hypotensive No ICG hypersensitivity reactions were observed. The use of
reaction was 0.5%. Other severe allergic reactions included ICG requires a special camera to allow visualization and is
bronchospasm and respiratory compromise; these events less readily accessible than current techniques used for SLNB
required short-term pressor support but did not require (blue dye and radioisotope). Further study defning the role of
emergency intubation. No cross-sensitivity between sulfa ICG in SLNB in breast cancer is necessary.
allergy and isosulfan blue dye was observed. A subse-
quent prospective series reported by King and colleagues
from MSKCC demonstrated a similar rate of allergic reac-
tions, 1.8%, among 1728 patients.42 Hypotensive reactions
The SLN mapping procedure
comprised 0.2% of events, with 0.1% of patients requiring Our current approach at MSKCC includes both isotope and
pressor support. Additionally, a nonsignifcant trend toward blue dye for lymphatic mapping for the majority of up-front
fewer allergic reactions with smaller volumes of dye was cases and for all cases following NAC.53 The morning of the
noted. operation, the patient receives 0.1 mCi of unfltered 99mTc-SC
Patent blue-V dye is a triphenyl methane dye similar to iso- (CIS-U.S. Inc., Bedford, MA) intradermally overlying the site of
sulfan blue that is used in Europe. Patent blue-V dye has been the tumor. Alternatively, mapping may be performed the night
used in lymphangiography longer than isosulfan blue dye. The before surgery; this has not been demonstrated to have a clini-
frequency of allergic reactions to patent blue dye has been esti- cally meaningful impact on successful SLN identifcation.54 A
mated to be 0.2%–2.7%.43,44 lymphoscintigram is obtained preoperatively (Figure 12.7c).
Sentinel lymph node mapping 91

FIGURE 12.7 (c) Intraoperative identifcation of the sentinel lymph node. The use of lymphoscintigraphy to identify the sentinel lymph node in
melanoma is well documented. The value of the preoperative lymphoscintigram in sentinel lymph node mapping in breast cancer is controversial.
Injection of radioisotope in conjunction with blue dye has been shown to increase the success of fnding the sentinel lymph node and thus decrease the
learning curve associated with the procedure.

FIGURE 12.8 (a) Injection of the blue dye at the time of surgery is
helpful in the sentinel lymph node procedure. Adverse reactions occur
in approximately 1% of sentinel lymph node biopsies. The use of the blue
dye in conjunction with radioisotope increases the success rate of lym-
phatic mapping compared to either technique alone.
FIGURE 12.8 (c) Injection of the blue dye can also be performed intra-
dermally, but will require a smaller volume.

One to two hours after the isotope injection, the patient is


taken to the operating room. Under either local or general
anesthesia, the breast and axilla are prepped and draped in
the usual manner, an intraparenchymal injection of 2–8 mL
of isosulfan blue dye is injected into the subareolar plexus,
and the breast is massaged for 5 minutes (Figure 12.8a–e).
Within 5–10 minutes, the axilla is explored through a small
transverse incision (Figure 12.8f). Identifcation and removal
of all hot and blue nodes are done with the aid of a hand-
held gamma probe and identifcation of the blue lymphatic
FIGURE 12.8 (b) The gamma probe (C-Trak, Care Wise, Inc., Morgan
channels (Figure 12.9a–d). In cases of mastectomy or follow-
Hill, CA) can be used to identify intraoperatively the ‘hot’ sentinel lymph ing NAC, the SLNs are submitted for frozen-section analy-
node during dissection. The gamma probe provides an auditory signal that sis, and if metastases are identifed, an immediate ALND is
can help direct the dissection toward the location of the sentinel lymph node. performed.
92 Clinical Lymphatic Mapping

FIGURE 12.8 (d) The radioactive counts help to determine the amount FIGURE 12.9 (a) The gamma probe is dressed with a sterile cover and
of radioisotope that is present in the injection site. is used to help localize the direction to proceed for identifcation of the
sentinel lymph node.

FIGURE 12.9 (b) Identifcation of the ‘blue’ sentinel lymph node is noted.
FIGURE 12.8 (e) The area of maximum radioactivity is noted. Surgical
exploration will begin directly over this area to have the greatest potential
in identifcation of the sentinel lymph node.

FIGURE 12.9 (c) The use of both radioisotope and blue dye will ensure
success for the SLN procedure. An average of two nodes is usually found
during this procedure. However, any blue or hot node will need to be
removed to decrease the false-negative procedure rate. Nodes that emit
FIGURE 12.8 (f) Careful dissection is performed so as not to disrupt 10% or more of the strongest signal that was detected in a sentinel node
the blue lymphatics, which will lead to the sentinel lymph node. require resection as sentinel nodes.
Sentinel lymph node mapping 93

radiation tangents cover low-level axillary nodes, it is thought


that some axillary treatment occurs during WBRT. Unless
they met criteria for postmastectomy RT, most women under-
going mastectomy would not receive RT and therefore risk
undertreatment of their axillae. Additionally, although nodal
tangents were explicitly disallowed in the Z0011 protocol, post
hoc review of available radiation plans revealed that approxi-
mately 19% of patients received axillary RT.61 These fndings
raised questions regarding the optimal radiation treatment
approach to patients with low-volume axillary disease treated
with SNLB alone.
The AMAROS study provided additional data to guide
axillary management in women with low-volume axillary
disease.62 In this prospective randomized study, women with
cT1-2N0 undergoing mastectomy or BCT with positive fnd-
ings on SLNB were randomized to ALND or axillary RT.
FIGURE 12.9 (d) After removal of the blue nodes, the gamma probe Locoregional recurrence, OS, and DFS were similar between
is introduced into the axilla to check for any residual radioactivity groups. Additional fndings of the study included lower rates
hotspots, which may indicate an additional sentinel lymph node. A four- of lymphedema and shoulder dysfunction in the axillary RT
fold decrease in counts is considered a successful radiolocalization. This group. This study provided the data to support axillary RT as
reduced count is recorded. a potential alternative to ALND in patients undergoing mas-
tectomy that were found to have positive SLNs.
The National Cancer Institute of Canada MA.20 study
It is important to perform careful intraoperative palpation evaluated whether the addition of regional nodal irradiation
of the axilla, as uptake of both isotope and blue dye can be (RNI) to WBRT would improve survival outcomes in patients
impaired by gross tumor involvement of nodes. Cody and col- with node-positive or high-risk node-negative breast cancers.63
leagues reported that clinically suspicious nodes were present Addition of RNI did not result in any difference in OS, but
in more than 50% of false-negative SLNB procedures.53,55 In DFS was 5% higher in the RNI group: 82% compared to 77%
these cases, gross tumor involvement of the nodes may com- in the WBRT-only group. Benefts were seen in both loco-
promise the identifcation of the true sentinel node, and these regional recurrence and distant recurrence with the addition
palpable nodes should be resected as sentinel nodes. of RNI. Despite nearly ubiquitous use of ALND (96%), the
results of this trial may apply to Z0011 patients who are treated
with SNLB for low-volume axillary disease. This reasoning
Management of the clinically negative axilla
is predicated on the assumption that SNLB likely resects the
Until the ACOSOG Z0011 trial, standard of care mandated majority of the disease burden in the axilla, as well as the
completion ALND in the setting of a positive SLN. The Z0011 observation of similar locoregional recurrence rates regardless
trial originated from the observation that the SLN is the only of whether an ALND was performed in Z0011.
positive lymph node in the majority of women and from a
desire to spare women the morbidity of an operation that, in the
setting of a single positive node, would be unlikely to provide
SLNB after NAC
signifcant oncologic beneft.56–59 This landmark randomized
study assigned women undergoing BCS and whole-breast RT Although adjuvant chemotherapy was frmly established as
(WBRT) with cT1-T2N0M0 breast cancer with one or two pos- the backbone of systemic therapy for breast cancer treatment,
itive SLNs to one of two treatment arms: completion ALND or there was interest in elucidating the ideal timing of chemo-
no ALND.38 Of note, 27% of women in the ALND group were therapy. NAC, which is delivered prior to surgery, may allow
found to have additional positive nodes in their axillary speci- locally advanced primary tumors to become amenable to
mens. Women who underwent SLNB alone presumably had a breast conservation, as well as facilitate downstaging of the
similar rate of residual disease; despite no targeted therapy to axilla. The NSABP explored the role of chemotherapy in the
the axilla, there was no difference in locoregional recurrence, neoadjuvant setting in two large randomized trials, B-18 and
OS, or DFS between groups after a median follow-up of 9.3 B-27, and found no difference in OS or DFS between women
years.60 Results of this trial were practice changing and led to who received NAC and those who received adjuvant chemo-
the routine omission of ALND for women with cT1-2N0 breast therapy.64,65 Pathologic complete response (pCR) was a predic-
cancer undergoing BCT who were found to have one to two tor of OS and DFS in both studies. In B-27, pCR following
positive SLNs. NAC was a prognostic factor for survival independent of the
Of note, women undergoing mastectomy were not included response in the breast. These fndings led to interest in defn-
in the Z0011 trial; consequently, results of the trial cannot be ing a role for SLNB in women undergoing NAC.
extrapolated to them. Targeted treatment of the axilla is still Given concern for potential fbrosis of lymphatic channels,
warranted in women who elect mastectomy and are found the accuracy of SLNB following NAC was evaluated in mul-
to have one to two positive SLNs. Because standard breast tiple studies. The GANEA study prospectively evaluated the
94 Clinical Lymphatic Mapping

feasibility of SLNB after NAC; in cN0 women, the identifca- Building on the fndings in Z1071, Caudle and the University
tion rate was 95% with an FNR of 9% – comparable to rates of Texas MD Anderson Cancer Center group described a
that have been reported for cN0 women undergoing up-front novel technique, the TArgeted lymph node Dissection (TAD),
surgery.66 With the ability to perform an accurate SLNB after in which the biopsy-proven positive node was clipped prior
NAC confrmed, the next series of questions focused on opti- to NAC and then targeted at the time of surgery in addition
mal timing of axillary staging for women receiving NAC. to a standard SNLB.72 Multiple groups reported outcomes of
The SENTInel NeoAdjuvant (SENTINA) study demon- this technique and found that the combination of SLNB and
strated that SLNB prior to NAC and after NAC (repeat SLNB) excision of the biopsy-proven node reduced the FNR to below
was associated with an identifcation rate of 61% and an FNR 10% and had the potential to tailor axillary management.
of 52%, indicating that repeat SLNB was not an appropriate Additionally, they demonstrated that the clipped node was not
option.67 Knowing that axillary staging can only be performed an SLN in up to 24% of cases, suggesting that SLNB alone
once and that confrmation of nodal pCR could obviate the may miss the initially positive node in up to a quarter of cases.
need for ALND, performing SLNB after NAC became the pre- Frozen-section analysis is of paramount importance in the
ferred approach. Hunt et al. evaluated the oncologic safety of setting of NAC, as metastases of any size, including ITCs
this approach with a series of 3746 women with cT1-3N0 who and micrometastases, indicate residual disease and warrant
underwent SLNB at the University of Texas MD Anderson ALND (Figure 12.10a–c). Indeed, it was with the inclusion
Cancer Center. Patients who were found to be pN0 follow-
ing NAC and underwent SLNB alone had a 4-year regional
recurrence rate of 1.2%, similar to the 0.9% observed among
patients who had negative SLNs and underwent SLNB alone
in the up-front surgery setting.68
Having verifed the oncologic safety of SLNB following
NAC in cN0 patients, attention then turned to evaluating the
feasibility and accuracy of SLNB in clinically node-positive
(cN+) patients who convert to cN0 following NAC. The
ACOSOG Z1071 trial evaluated results of SLNB in 649 biopsy-
proven cN+ women; 83% converted to cN0 after NAC.69 The
SLN identifcation rate was 93%, and the FNR in women who
had two or more SLNs resected was 13%. This FNR failed
to meet the predefned threshold of 10% to consider SLNB
appropriate in this population. A similar study, Sentinel Node
Biopsy Following Neoadjuvant Chemotherapy (SN FNAC),
also examined the technical success and accuracy of SLNB
in biopsy-proven cN+ women. This study reported comparable
results, with an SLN identifcation rate of 88% and an FNR of
8% when ITCs were considered node positive and 13% when
ITCs were considered node negative.70
Although the FNRs in these studies failed to meet the pre-
determined threshold to indicate oncologic appropriateness
of SLN in cN+ women undergoing NAC, unplanned explor-
atory analyses in Z1071 suggested strategies to help reduce the
unacceptably high FNR. These strategies included use of dual
tracer (11% false negative vs. 20% FNR with single agent) and
removal of three or more SLNs (FNR 9% vs. 21% with two
SLNs). With FNRs so close to the predetermined threshold
and a signal in Z1071 suggesting that the FNR could reach
acceptable range with some technical modifcations, the next
series of studies focused on refning the SLN technique and
generating clinical criteria to ensure oncologic safety in this
population.
In an early study assessing the feasibility and clinical impact
of fnding three or more SLNs after NAC, Mamtani et al.
reported on a prospective cohort of 132 women who presented
with cN+ disease and converted to cN0 after NAC. They found
that the probability of fnding three or more SLNs was 86%
FIGURE 12.10 (a) Frozen section of a sentinel lymph node involved
and that 47% of women in this population were spared ALND with metastatic carcinoma (H&E, 200×). Arrows demonstrate clusters of
after NAC, suggesting that fnding three or more SLNs was tumor cells in the background of lymphoid tissue (star). (b) Lymph node
both feasible in most women and that the results affected sur- with treatment effect following NAC with accumulation of macrophages
gical management in almost half of these women.71 (H&E, 50×).
Sentinel lymph node mapping 95

The long-term morbidities of SLNB require ongoing long-


term follow-up. A prospective trial comparing sensory mor-
bidity after SLNB and ALND found that sensory defcits
occurred twice as frequently in women who underwent ALND
as they did in women who underwent SLNB.77 In a study of 187
women who underwent BCS with either SLNB or ALND, the
rates of subjective arm numbness, chest or axillary numbness,
arm or hand swelling, and objective neurosensory changes in
the axilla and arm were signifcantly higher in women who
underwent ALND.78
Studies demonstrate a lower rate of lymphedema in women
who undergo SLNB compared to those who undergo ALND.
McLaughlin et al. reported a series of 936 women who under-
went axillary surgery at MSKCC; women who required ALND
had a 16% rate of lymphedema at 5 years follow-up compared
to 5% in women who required SLNB alone.79 Risk factors
associated with the development of lymphedema included
higher body mass index and infection or injury of the ipsilat-
FIGURE 12.10 (c) Arrow denotes pale-appearing macrophages embedded eral arm. Studies exploring the incidence and management of
in a background of fbrosis (H&E, 200×). (Courtesy of Dr. Dilip Giri from
the Department of Pathology at Memorial Sloan Kettering Cancer Center.)
lymphedema are ongoing, with additional work focusing on
prevention and early detection.

of micrometastases that the SN FNAC study was able to


achieve an FNR <10%. To elucidate the relationship between
low-volume axillary disease on SLNB and additional disease Conclusion
within the axilla, Moo and colleagues at MSKCC evaluated
SLNB allows accurate staging in patients with breast cancer
702 women who underwent SLNB after NAC. They found that
and identifcation of patients who require ALND to optimize
23% of women with micrometastases and 63% of women with
locoregional control. SLNB also helps guide systemic therapy
macrometastases on frozen section had one or more additional
recommendations and affords decreased morbidity by avoid-
positive lymph nodes on ALND.73 Additionally, 17% of women
ing ALND in appropriate patients.
with ITCs had one or more additional positive nodes on ALND.
Together, these results indicate that low-volume SLN disease
does not identify women who are at low risk for additional axil-
lary metastases and confrm the role for ALND in this setting.
REFERENCES
Although there is a large volume of data to guide manage- 1. Surveillance Epidemiology and End Results (SEER)
ment of the axilla after NAC, there is a paucity of data defn- Program, SEER Stat Fact Sheet: Female Breast Cancer
ing best practices for axillary management after neoadjuvant [cited 2020 Mar 17]. Available from: http://seer.cancer.gov/
endocrine therapy, and this will be an area of intense research statfacts/html/breast.html.
activity in the coming years. 2. Shen J, et al. Intramammary lymph node metastases are
an independent predictor of poor outcome in patients with
breast carcinoma. Cancer. 2004;101(6):1330–7.
3. King TA, et al. Lobular carcinoma in situ: a 29-year lon-
Follow-up after SLNB gitudinal experience evaluating clinicopathologic fea-
tures and breast cancer risk. J Clin Oncol. 2015;33(33):
Local recurrence after SLNB for melanoma has been
3945–52.
reported.74 Recent reports have found that the rates of isolated 4. Tadros AB, Wen HY, Morrow M. Breast cancers of spe-
axillary relapse after a negative SLNB are comparable to a cial histologic subtypes are biologically diverse. Ann Surg
conventional ALND; indeed, long-term follow up of the B-32 Oncol. 2018;25(11):3158–64.
trial and Milan trials demonstrated similar rates of regional 5. Amin MB, et al. The eighth edition AJCC cancer staging
nodal recurrence – all under 1%.7,8 Madsen et al. reported the manual: continuing to build a bridge from a population-
MSKCC experience: among 1529 women with negative SLNs based to a more ‘personalized’ approach to cancer staging.
at the time of SLNB, cumulative incidence of axillary recur- CA Cancer J Clin. 2017;67(2):93–9.
rence as a frst breast event was 1% at 15 years.75 6. Bevilacqua JLB, et al. Doctor, what are my chances of hav-
Importantly, there are no long-term data assessing the onco- ing a positive sentinel node? A validated nomogram for risk
logic safety of SLNB alone for patients who convert from cN+ estimation. J Clin Oncol. 2007;25(24):3670–9.
to pN0. Galimberti and colleagues reported zero axillary fail- 7. Krag DN, et al. Sentinel-lymph-node resection compared
ure events at 61 months among 70 patients who converted from with conventional axillary-lymph-node dissection in clini-
cN+ to pN0 and were treated with SLNB alone.76 Reports on cally node-negative patients with breast cancer: overall sur-
oncologic safety and rates of regional recurrence are highly vival fndings from the NSABP B-32 randomised phase 3
anticipated as the data from Z1071 mature. trial. Lancet Oncol. 2010;11(10):927–33.
96 Clinical Lymphatic Mapping

8. Veronesi U, et al. Sentinel lymph node biopsy in breast can- 25. Hughes KS, et al. Lumpectomy plus tamoxifen with or
cer: ten-year results of a randomized controlled study. Ann without irradiation in women age 70 years or older with
Surg. 2010;251(4):595–600. early breast cancer: long-term follow-up of CALGB 9343. J
9. Ashikaga T, et al. Morbidity results from the NSABP B-32 Clin Oncol. 2013;31(19):2382–7.
trial comparing sentinel lymph node dissection versus axil- 26. Hughes KS, et al. Lumpectomy plus tamoxifen with or
lary dissection. J Surg Oncol. 2010;102(2):111–18. without irradiation in women 70 years of age or older
10. Cabanas RM. An approach for the treatment of penile car- with early breast cancer. N Engl J Med. 2004;351(10):
cinoma. Cancer. 1977;39(2):456–66. 971–7.
11. Rosen PP, et al. Discontinuous or ‘skip’ metastases in breast 27. Oncology, S.o.S. Don’t routinely use sentinel node biopsy
carcinoma. Analysis of 1228 axillary dissections. Ann in clinically node negative women ≥70 years of age with
Surg. 1983;197(3):276–83. early stage hormone receptor positive, HER2 negative inva-
12. Morton DL, et al. Technical details of intraoperative lym- sive breast cancer. Choosing Wisely; 2016 [2020 Feb 29].
phatic mapping for early stage melanoma. Arch Surg. Available from: www.choosingwisely.org/clinician-lists/
1992;127(4):392–9. sso-sentinel-node-biopsy-in-node-negative-women-70-and-
13. Gill G, S.T.G.o.t.R.A.C.o. Surgeons, and N.C.T. Centre, over/.
Sentinel-lymph-node-based management or routine axil- 28. Gentilini O, et al. Safety of sentinel node biopsy in pregnant
lary clearance? One-year outcomes of sentinel node biopsy patients with breast cancer. Ann Oncol. 2004;15(9):1348–51.
versus axillary clearance (SNAC): a randomized controlled 29. Pandit-Taskar N, et al. Organ and fetal absorbed dose esti-
surgical trial. Ann Surg Oncol. 2009;16(2):266–75. mates from 99mTc-sulfur colloid lymphoscintigraphy and
14. Goyal A, et al. Factors affecting failed localisation and sentinel node localization in breast cancer patients. J Nucl
false-negative rates of sentinel node biopsy in breast Med. 2006;47(7):1202–8.
cancer – results of the ALMANAC validation phase. Breast 30. Spanheimer PM, et al. Measurement of uterine radiation
Cancer Res Treat. 2006;99(2):203–8. exposure from lymphoscintigraphy indicates safety of
15. Krag DN, et al. Technical outcomes of sentinel-lymph-node sentinel lymph node biopsy during pregnancy. Ann Surg
resection and conventional axillary-lymph-node dissec- Oncol. 2009;16(5):1143–7.
tion in patients with clinically node-negative breast cancer: 31. Turner RR, Giuliano AE. Intraoperative pathologic
results from the NSABP B-32 randomised phase III trial. examination of the sentinel lymph node. Ann Surg Oncol.
Lancet Oncol. 2007;8(10):881–8. 1998;5(8):670–2.
16. Veronesi U, et al. A randomized comparison of sentinel- 32. Turner RR, et al. Intraoperative examination of the sentinel
node biopsy with routine axillary dissection in breast can- lymph node for breast carcinoma staging. Am J Clin Pathol.
cer. N Engl J Med. 2003;349(6):546–53. 1999;112(5):627–34.
17. Wong SL, et al. Sentinel lymph node biopsy for breast can- 33. Boughey JC, et al. Number of lymph nodes identifed at
cer: impact of the number of sentinel nodes removed on the axillary dissection: effect of neoadjuvant chemotherapy
false-negative rate. J Am Coll Surg. 2001;192(6):684–9; and other factors. Cancer. 2010;116(14):3322–9.
discussion 689–91. 34. Giuliano AE, et al. Association of occult metastases in sen-
18. McCarter MD, et al. The breast cancer patient with mul- tinel lymph nodes and bone marrow with survival among
tiple sentinel nodes: when to stop? J Am Coll Surg. women with early-stage invasive breast cancer. JAMA.
2001;192(6):692–7. 2011;306(4):385–93.
19. Turner RR, et al. Histopathologic validation of the senti- 35. Weaver DL, et al. Effect of occult metastases on sur-
nel lymph node hypothesis for breast carcinoma. Ann Surg. vival in node-negative breast cancer. N Engl J Med.
1997;226(3):271–6; discussion 276–8. 2011;364(5):412–21.
20. Flanagan MR, et al. Is sentinel lymph node biopsy required 36. Galimberti V, et al. Axillary dissection versus no axillary
for a core biopsy diagnosis of ductal carcinoma in situ with dissection in patients with sentinel-node micrometastases
microinvasion? Ann Surg Oncol. 2019;26(9):2738–46. (IBCSG 23–01): a phase 3 randomised controlled trial.
21. Pilewskie M, et al. Is sentinel lymph node biopsy indicated Lancet Oncol. 2013;14(4):297–305.
at completion mastectomy for ductal carcinoma in situ? 37. Galimberti V, et al. Axillary dissection versus no axil-
Ann Surg Oncol. 2016;23(7):2229–34. lary dissection in patients with breast cancer and sentinel-
22. Mittendorf EA, et al. Core biopsy diagnosis of ductal carci- node micrometastases (IBCSG 23–01): 10-year follow-up
noma in situ: an indication for sentinel lymph node biopsy. of a randomised, controlled phase 3 trial. Lancet Oncol.
Curr Surg. 2005;62(2):253–7. 2018;19(10):1385–93.
23. National Comprehensive Cancer Network, Breast Cancer 38. Giuliano AE, et al. Axillary dissection vs no axillary dis-
(Version 3.2019), in NCCN Clinical Practice Guidelines in section in women with invasive breast cancer and senti-
Oncology; 2019. nel node metastasis: a randomized clinical trial. JAMA.
24. Southwest Oncology Group, A phase III, randomized 2011;305(6):569–75.
clinical trial of standard adjuvant endocrine therapy +/− 39. Tew K, et al. Meta-analysis of sentinel node imprint cytol-
chemotherapy in patients with 1–3 positive nodes, hormone- ogy in breast cancer. Br J Surg. 2005;92(9):1068–80.
responsive and HER2-negative breast cancer according 40. Hirsch JI, et al. Use of isosulfan blue for identifcation of
to Recurrence Score [cited 2020 Feb 1]. Available from: lymphatic vessels: experimental and clinical evaluation.
www.swog.org/clinical-trials/s1007. AJR Am J Roentgenol. 1982;139(6):1061–4.
Sentinel lymph node mapping 97

41. Montgomery LL, et al. Isosulfan blue dye reactions dur- 60. Giuliano AE, et al. Effect of axillary dissection vs no axil-
ing sentinel lymph node mapping for breast cancer. Anesth lary dissection on 10-year overall survival among women
Analg. 2002;95(2):385–8. with invasive breast cancer and sentinel node metastasis:
42. King TA, et al. A prospective analysis of the effect of blue- the ACOSOG Z0011 (Alliance) randomized clinical trial.
dye volume on sentinel lymph node mapping success and JAMA. 2017;318(10):918–26.
incidence of allergic reaction in patients with breast cancer. 61. Jagsi R, et al. Radiation feld design in the ACOSOG Z0011
Ann Surg Oncol. 2004;11(5):535–41. (Alliance) Trial. J Clin Oncol. 2014;32(32):3600–6.
43. Kalimo K, Saarni H. Immediate reactions to patent blue 62. Donker M, et al. Radiotherapy or surgery of the axilla after a
dye. Contact Dermatitis. 1981;7(3):171–2. positive sentinel node in breast cancer (EORTC 10981–22023
44. Mortazavi SH, Burrows BD. Allergic reaction to patient blue AMAROS): a randomised, multicentre, open-label, phase 3
dye in lymphangiography. Clin Radiol. 1971;22(3):389–90. non-inferiority trial. Lancet Oncol. 2014;15(12):1303–10.
45. Linehan DC, et al. Sentinel lymph node biopsy in breast 63. Whelan TJ, et al. Regional nodal irradiation in early-stage
cancer: unfltered radioisotope is superior to fltered. J Am breast cancer. N Engl J Med. 2015;373(4):307–16.
Coll Surg. 1999;188(4):377–81. 64. Bear HD, et al. Sequential preoperative or postoperative
46. Martin RC, et al. Highest isotope count does not predict docetaxel added to preoperative doxorubicin plus cyclo-
sentinel node positivity in all breast cancer patients. Ann phosphamide for operable breast cancer: National Surgical
Surg Oncol. 2001;8(7):592–7. Adjuvant Breast and Bowel Project Protocol B-27. J Clin
47. Borgstein PJ, et al. Functional lymphatic anatomy for senti- Oncol. 2006;24(13):2019–27.
nel node biopsy in breast cancer: echoes from the past and 65. Fisher B, et al. Effect of preoperative chemotherapy on
the periareolar blue method. Ann Surg. 2000;232(1):81–9. local-regional disease in women with operable breast can-
48. Kitai T, et al. Fluorescence navigation with indocyanine cer: fndings from National Surgical Adjuvant Breast and
green for detecting sentinel lymph nodes in breast cancer. Bowel Project B-18. J Clin Oncol. 1997;15(7):2483–93.
Breast Cancer. 2005;12(3):211–15. 66. Classe JM, et al. Sentinel lymph node biopsy after neo-
49. Sugie T, et al. Comparison of the indocyanine green fuo- adjuvant chemotherapy for advanced breast cancer: results
rescence and blue dye methods in detection of sentinel of Ganglion Sentinelle et Chimiotherapie Neoadjuvante,
lymph nodes in early-stage breast cancer. Ann Surg Oncol. a French prospective multicentric study. J Clin Oncol.
2013;20(7):2213–18. 2009;27(5):726–32.
50. Tagaya N, et al. A novel approach for sentinel lymph node 67. Kuehn T, et al. Sentinel-lymph-node biopsy in patients with
identifcation using fuorescence imaging and image over- breast cancer before and after neoadjuvant chemotherapy
lay navigation surgery in patients with breast cancer. World (SENTINA): a prospective, multicentre cohort study.
J Surg. 2011;35(1):154–8. Lancet Oncol. 2013;14(7):609–18.
51. Tagaya N, et al. Intraoperative identifcation of senti- 68. Hunt KK, et al. Sentinel lymph node surgery after neoadjuvant
nel lymph nodes by near-infrared fuorescence imaging chemotherapy is accurate and reduces the need for axillary dis-
in patients with breast cancer. Am J Surg. 2008;195(6): section in breast cancer patients. Ann Surg. 2009;250(4):558–66.
850–3. 69. Boughey JC, et al. Sentinel lymph node surgery after
52. Sugie T, et al. Evaluation of the clinical utility of the ICG neoadjuvant chemotherapy in patients with node-positive
fuorescence method compared with the radioisotope breast cancer: the ACOSOG Z1071 (Alliance) clinical trial.
method for sentinel lymph node biopsy in breast cancer. JAMA. 2013;310(14):1455–61.
Ann Surg Oncol. 2016;23(1):44–50. 70. Boileau JF, et al. Sentinel node biopsy after neoadjuvant
53. Cody HS. Sentinel lymph node mapping in breast cancer. chemotherapy in biopsy-proven node-positive breast cancer:
Oncology (Williston Park). 1999;13(1):25–34; discussion the SN FNAC study. J Clin Oncol. 2015;33(3):258–64.
35–6, 39, 43. 71. Mamtani A, et al. How often does neoadjuvant chemother-
54. Dodia N, El-Sharief D, Kirwan CC. The use of isotope apy avoid axillary dissection in patients with histologically
injections in sentinel node biopsy for breast cancer: are confrmed nodal metastases? Results of a prospective study.
the 1- and 2-day protocols equally effective? Springerplus. Ann Surg Oncol. 2016;23(11):3467–74.
2015;4:495. 72. Caudle AS, et al. Improved axillary evaluation following
55. Hill AD, et al. Lessons learned from 500 cases of lymphatic neoadjuvant therapy for patients with node-positive breast
mapping for breast cancer. Ann Surg. 1999;229(4):528–35. cancer using selective evaluation of clipped nodes: imple-
56. Albertini JJ, et al. Lymphatic mapping and sentinel mentation of targeted axillary dissection. J Clin Oncol.
node biopsy in the patient with breast cancer. JAMA. 2016;34(10):1072–8.
1996;276(22):1818–22. 73. Moo T-A, et al. Is low-volume disease in the sentinel node
57. Giuliano AE, et al. Lymphatic mapping and sentinel lymph- after neoadjuvant chemotherapy an indication for axillary
adenectomy for breast cancer. Ann Surg. 1994;220(3):391– dissection? Ann Surg Oncol. 2018;25(6):1488–94.
8; discussion 398–401. 74. Gershenwald JE, et al. Patterns of recurrence following a
58. Krag D, et al. The sentinel node in breast cancer – a multi- negative sentinel lymph node biopsy in 243 patients with
center validation study. N Engl J Med. 1998;339(14):941–6. stage I or II melanoma. J Clin Oncol. 1998;16(6):2253–60.
59. O’Hea BJ, et al. Sentinel lymph node biopsy in breast can- 75. Matsen C, et al. Late axillary recurrence after negative sen-
cer: initial experience at Memorial Sloan-Kettering Cancer tinel lymph node biopsy is uncommon. Ann Surg Oncol.
Center. J Am Coll Surg. 1998;186(4):423–7. 2016;23(8):2456–61.
98 Clinical Lymphatic Mapping

76. Galimberti V, et al. Sentinel node biopsy after neoadjuvant 78. Crane-Okada R, et al. Long-term morbidity of sentinel node
treatment in breast cancer: fve-year follow-up of patients biopsy versus complete axillary dissection for unilateral
with clinically node-negative or node-positive disease breast cancer. Ann Surg Oncol. 2008;15(7):1996–2005.
before treatment. Eur J Surg Oncol. 2016;42(3):361–8. 79. McLaughlin SA, et al. Prevalence of lymphedema in
77. Temple LK, et al. Sensory morbidity after sentinel lymph women with breast cancer 5 years after sentinel lymph node
node biopsy and axillary dissection: a prospective study of biopsy or axillary dissection: objective measurements. J
233 women. Ann Surg Oncol. 2002;9(7):654–62. Clin Oncol. 2008;26(32):5213–19.
13
Sentinel lymph node biopsy of the endometrium

Jennifer J. Mueller and Nadeem R. Abu-Rustum

CONTENTS
Epidemiology of the disease .......................................................................................................................................................... 99
Historical perspective on lymph node status in endometrial cancer .............................................................................................. 99
Early sentinel lymph node mapping feasibility studies in endometrial cancer .............................................................................. 99
SLN mapping patterns in endometrial cancer .............................................................................................................................. 100
SLN mapping: techniques and examples ..................................................................................................................................... 102
SLN detection using an algorithm ............................................................................................................................................... 103
SLN detection: false-negative rates.............................................................................................................................................. 103
SLN mapping versus lymphadenectomy: the great debate .......................................................................................................... 104
SLN mapping and oncologic outcomes ....................................................................................................................................... 104
Endometrioid histology ...................................................................................................................................................... 104
High-risk histology............................................................................................................................................................. 105
Future directions .......................................................................................................................................................................... 106
References.................................................................................................................................................................................... 106

Epidemiology of the disease with increasing FIGO grade and depth of myometrial inva-
sion, which has been reproduced in modern patient cohorts.6–8
Endometrial cancer is the fourth most common malignancy This helped catalyze a change in endometrial cancer manage-
among women in the United States. As the annual incidence ment, with patients stratifed based on clinical and pathologic
rate continues to rise from year to year, an estimated 65,620 risk factors for recurrence to help determine who would most
new cases will be diagnosed in 2020 and 12,590 women will beneft from radiation therapy. Prospective trials conducted
die from this disease.1 Endometrial cancer most often presents by the GOG and the Post-Operative Radiation Therapy in
at an early stage, and in the absence of concerns such as medi- Endometrial Cancer (PORTEC) groups9–11 examined the
cal frailty or fertility preservation, is managed surgically with oncologic beneft of adjuvant radiation therapy in patients with
hysterectomy, bilateral oophorectomy, and lymph node evalu- high-to-intermediate-risk endometrial cancer, and although
ation.2 Compared to patients with extrauterine disease spread, these studies did not show an improvement in survival, they
particularly nodal disease, the vast majority of women with did provide justifcation for postoperative radiation therapy to
early-stage disease have an excellent prognosis.3 reduce the risk of recurrence.
Around that time, surgical strategies began to incorporate
lymph node dissection (pelvic and paraaortic) to tailor therapy
Historical perspective on lymph node based on uterine risk factors and nodal positivity. Two land-
status in endometrial cancer mark prospective trials examined the oncologic impact of
lymphadenectomy as part of surgical staging in patients with
The historical management of apparent early-stage endome-
early-stage endometrial cancer. These randomized trials dem-
trial cancer was discussed more thoroughly in an earlier pub-
onstrated lymphadenectomy was signifcantly associated with
lished edition of this chapter4 and will be briefy reviewed here.
prognosis (e.g., detecting node-positive disease) but not with
Prior to the 1980s, the approach to endometrial cancer therapy
survival.12,13 Furthermore, lymphadenectomy carries added
included primary radiation followed by surgery. This was per-
surgical morbidity, including leg lymphedema, higher rates of
formed to sterilize extrauterine spread of disease and spare
intraoperative blood loss, and vascular and nerve injury.14–17
extensive surgery, including lymph node evaluation. In 1987,
Creasman et al.5 published the fndings of a clinicopathologic
study (Gynecologic Oncology Group [GOG] 33) that examined Early sentinel lymph node mapping
occult nodal involvement in surgically staged early endome- feasibility studies in endometrial cancer
trial cancer and related nodal involvement to pathologic uter-
ine factors, including International Federation of Gynecology The concept of sentinel lymph node (SLN) mapping was
and Obstetrics (FIGO) grade and myometrial invasion. The described in the literature as early as the 1970s,18 although
study’s results revealed that lymph node involvement increases 2 decades would pass before its application in endometrial

DOI: 10.1201/9781003255536-13 99
100 Clinical Lymphatic Mapping

cancer was frst reported.19 Endometrial tumors present many to receive either ICG followed by blue dye or blue dye followed
challenges for SLN mapping, including a tumor not clinically by ICG. Higher rates of bilateral mapping were seen with ICG
or radiographically visible to inject and complex, bilateral lym- (77%–80%) over blue dye (29%–32%) in both groups.28
phatic drainage pathways. Early reports and studies of SLN
mapping in endometrial cancer consisted of small numbers
of patients and looked at various injection sites (e.g., myome-
trium, uterine serosa, cervix, and endometrium) or combina-
SLN mapping patterns in endometrial cancer
tions of these sites to determine optimal injection location. The lymphatic drainage of the uterus is reviewed in detail in
The ultimate goal of these initial studies was to determine an Chapter 2. The uterus follows three general lymphatic drain-
injection method that would incorporate both the ovarian and age pathways.37,38 The frst drainage pathway is the main target
pelvic drainage pathways. Investigators who have incorporated for pelvic SLN mapping in endometrial cancer. This pathway
a fundal and cervical injection to optimize dye uptake in the drains the cervical plus mid/lower uterine lymphatics and
pelvic and ovarian lymphatic drainage pathways have reported empties to pelvic nodal basins along the iliac and obtura-
mapping success rates in the range of 80%.20 Hysteroscopic tor vessels. Lymphatic trunks will condense in the paracer-
endometrial injection, with the advantage that a peritumoral vix and cross medial to lateral over the cranial part of the
injection maps to the most relevant drainage pathways of the obliterated umbilical ligament (upper or ventral paracervical
primary tumor site, including the paraaortic (ovarian) drain- pathway). Less commonly, these trunks condense in the para-
age route, has been proposed as an alternative approach. This cervix and traverse dorsally in the meso-ureter to drain the
approach, however, has not been broadly adopted, possibly presacral common iliac nodes (lower or dorsal paracervical
due to its associated technical challenges and poor reproduc- pathway). The second drainage pathway is the fundal portion
ibility.21 The Memorial Sloan Kettering Cancer Center (MSK) of the uterus, including the serosa, which joins with the ovar-
group pioneered and standardized the cervical injection tech- ian vessels and drains to paraaortic nodal basins. The third
nique for SLN mapping in endometrial cancer.2 Although ini- (and least dominant) pathway is the mid-uterine lymphatics,
tially met with resistance from gynecologic surgeons, many which travel within the round ligament to the inguinal nodal
institutions eventually realized the value of this simple injec- basins. Following a cervical injection of dye, the most com-
tion approach, and adoption rates have increased.22,23 Rossi et mon SLN locations in the pelvis are found along external
al. compared a cervical versus hysteroscopic injection using iliac, internal iliac, and obturator vessels and, less frequently,
indocyanine green (ICG) in patients with endometrial cancer along the common iliac vessels and presacral region of the
and demonstrated higher SLN detection rates with a cervi- pelvis. A subset of patients (10%–15%) will map to the para-
cal injection, as well as comparable drainage patterns, which aortic nodal regions. When these nodes are encountered, they
included aortic regions, suggesting cervical injection alone is are frequently found above the level of the inferior mesenteric
suffcient for lymphatic mapping.24 artery (IMA).39,40
Cervical injection has been widely adopted for SLN map- Understanding the two well-established mapping patterns
ping in endometrial cancer and has been shown in two meta- of paracervical lymphatic channel fow leading to SLNs41 can
analysis studies to enhance pelvic SLN detection compared help surgeons reliably locate true SLNs and reduce the risk
with alternative sites of injection.25,26 In one study, SLNs of false-negative nodes. The most common pelvic mapping
detected with cervical injection were three times more likely pattern occurs when lymphatic channels travel through the
to harbor metastatic disease than non-SLNs, even without paracervix and cross over the obliterated umbilical ligament
ultrastaging, demonstrating increased intraoperative surgical to an SLN located medial to the external iliac vessels, ven-
precision.21,27 The technique consists of injecting 1 cc of dye tral to the internal iliac vessels, or in the superior portion of
superfcially (1–2 mm) and deep (1–2 cm) at the 3 and 9 o’clock the obturator space. This is the upper or ventral paracervical
positions of the cervix for a total of 4 cc of dye. This is the most pathway. SLNs are frequently found along the dorsal cranial
common approach in current practice2 and was also used in the surface of external iliac vessels, but the obturator space is
Fluorescence Imaging for Lymphatic Mapping (FILM) ran- also a common location. If the retroperitoneal spaces are not
domized trial of blue dye versus ICG.28 These early studies are opened and explored, the surgeon may remove a secondary
discussed in greater detail in excellent literature reviews.20,29,30 echelon node and mistake it for the true SLN. The less com-
A variety of tracers and dyes, including technetium-99 and mon pelvic mapping pattern will demonstrate dye uptake
colored dyes such as vital blue (isosulfan blue, methylene blue, in lymphatic channels that do not cross over the obliterated
Lymphazurin) and ICG, have been studied in SLN mapping umbilical ligament and instead course cephalad within the
for endometrial cancer. Bilateral SLN detection rates range dorsal meso-ureter to drain to a common iliac or presacral
from 31% to 100%, depending on the study and injection agent node. This is the lower or dorsal paracervical pathway. These
used.31 Clinical practice has shifted to the use of ICG with lymph nodes can be more challenging to identify, especially
near-infrared (NIR) imaging due to its consistent and supe- in obese patients. Awareness of this less common map-
rior bilateral mapping success rates, including in morbidly ping pattern can enable the surgeon to locate atypical SLN
obese patients, and excellent safety profle.32–36 The random- locations and reduce rates of failed mapping (Figure 13.1).
ized, controlled FILM study compared both blue dye and ICG Uncommon or atypical SLN mapping examples in endome-
within each of the 180 patients with clinical stage I endome- trial cancer are depicted in Figure 13.2, as variability can
trial or early-stage cervical cancer undergoing laparoscopic exist within a patient. Figure 13.3 provides technical pointers
staging that included SLN mapping. Patients were randomized for pelvic SLN mapping success.
Biopsy of the endometrium 101

FIGURE 13.1 Pelvic lymphatic mapping patterns in endometrial cancer using a cervical injection technique. (a) This illustrates the most common
lymphatic drainage pattern following an intracervical injection of colored dye. Channels condense in the paracervix and cross medial to lateral over
the obliterated umbilical ligament, draining to external iliac and obturator nodal basins. (c) A patient with endometrial cancer has mapped via the
more common mapping pattern. Channels have crossed over the umbilical ligament (arrow) and lead to a right external iliac sentinel lymph node. (b)
This illustrates the less common lymphatic drainage pattern following an intracervical injection of colored dye. Channels condense in the paracervix
and travel cranially in the meso-ureter to drain common iliac and presacral nodal basins. (d) A patient with endometrial cancer has mapped via the
less common mapping pattern. Channels are seen traveling cephalad in the meso-ureter (arrow) and leading to a right presacral common iliac sentinel
lymph node. In both patient examples (c and d), the sentinel node (star in each image) appears bright white in a black background using the ‘spy’ mode
(Stryker, Kalamazoo, MI) perfusion assessment mode (side panel, middle images), fuoresces bright green using indocyanine green with near-infrared
imaging (side panel, lower images), and bright red with heat map assessment using color-segmented fuorescence in which gray-blue is least intense
and red is most intense (main images).
102 Clinical Lymphatic Mapping

FIGURE 13.2 Uncommon and atypical sentinel lymph node mapping examples in endometrial cancer following a cervical injection of dye. Top row:
(a) Indocyanine green (ICG) uptake within a left external iliac lymph node (double star) initially labeled as the sentinel node was actually a secondary
echelon node. (b) Retroperitoneal spaces opened and explored, and the true sentinel lymph node was located in the obturator space, which is bright
green using near-infrared imaging (star). (c) Lymphatics are seen crossing over the obliterated umbilical ligament (arrows in a and b) and leading to
a node in the obturator space (star). Efferent channels are seen traveling from this obturator sentinel lymph node to the previously seen external iliac
node (a). Surgeons may miss the true sentinel lymph node if the retroperitoneal spaces are not opened and explored. (d) Example of an empty nodal
packet. Dilated lymphatic channels mapping to the left pelvic sidewall consolidated to create a more intense signal (medial to the umbilical ligament)
on both perfusion and heat-based mapping (seen as red using color-segmented fuorescence mode and bright white using ‘spy’ mode). No nodal tis-
sue was palpated upon removal. Palpating excised tissue bedside during surgery can improve sentinel node retrieval and reduce false-negative cases.
Middle row: (a) Example of methylene blue dye uptake in a single lymphatic channel (arrow) in the left pelvis, which is coursing cranially parallel
with the iliac vessels to the presacral region and leads to a common iliac sentinel lymph node. (b and c) Cervical injection site locations for sentinel
lymph node mapping in endometrial cancer. Our preference is to inject dye 1 cc superfcially and 1 cc deep at the 3 and 9 o’clock positions for a total
of 4 cc of dye. (d) ICG uptake in the right pelvis within the dominant lymphatic channel traveling within the meso-ureter to a subaortic sentinel lymph
node (star). Lower row: (a) ICG fuorescing lymphatic channel in the left pelvis crosses the obliterated umbilical ligament and travels along the iliac
vessels past the common iliac vessels (arrow), with no sentinel nodes seen in the pelvis. This channel led to a left paraaortic sentinel lymph node. (b)
A lymphatic channel with methylene blue uptake (arrow) is seen terminating adjacent to a grossly enlarged precaval lymph node (star). Nodes flled
with tumor may block uptake of dye coming from lymphatic channels. Suspicious or enlarged lymph nodes must be removed regardless of mapping. (c)
A right pelvic primary lymphatic channel seen crossing over the obliterated umbilical ligament (common mapping pattern) and bifurcated, leading to
two true sentinel lymph nodes (both were removed and underwent pathologic ultrastaging). (d) A right-sided internal iliac sentinel lymph node mapped
with isosulfan blue. (*Several panels courtesy of Drs. Mario M. Leitao Jr and Nadeem R. Abu-Rustum.)

• Inject dye slowly into the cervix using a good injection technique
• Perform sentinel lymph node mapping first
• You can reinject the cervix if there is no dye uptake
• Keep your surgical field clean, avoid disrupting lymphatic channels
• Take it slow and be precise with your movements
• Find the obliterated umbilical ligament then trace it cranially
• Follow the lymphatic channels as they cross the umbilical ligament (this will lead to
the sentinel node in most cases)
• Do not remove the first green node you see
• Open the retroperitoneal spaces and trace the channels
• Remember the less common mapping pattern

FIGURE 13.3 Tips for fnding the pelvic sentinel lymph node

the MSK group estimated that 30 cases were needed to achieve


SLN mapping: techniques and examples good detection rates and low false-negative rates.42 Another
single-institution surgical group used a logistic model to assess
Although a learning curve is associated with SLN mapping the probability of successful SLN mapping and noted 40 cases
in endometrial cancer, there is no consensus on the number were needed to consistently achieve successful bilateral SLN
of cases needed to declare mastery. In their initial experience, mapping.43 The Society of Gynecologic Oncology (SGO)
Biopsy of the endometrium 103

recommends a minimum of 20 SLN mapping procedures with has provided a valuable framework for surgeons to move
concurrent lymphadenectomy or a <5% false-negative rate.44 beyond simply removing the SLN to considering important
Given the potential for variability, it is important for each sur- factors such as clinically enlarged nodes, peritoneal spread
geon to analyze the pathologic and clinical outcomes of all of disease, and management standards for failed mapping
their SLN mapping cases in order to reduce the risk of false- in one or both hemipelvis. Beginning in 2014, the National
negative nodes and empty nodal packets. Comprehensive Cancer Network (NCCN) guidelines incor-
SLN mapping can be performed via laparotomy or a mini- porated the MSK surgical staging algorithm for endometrial
mally invasive approach. If blue dye is used, special equip- cancer as an option for surgical staging (Figure 13.4).
ment is not required. There are multiple laparoscopic imaging
systems (e.g., PINPOINT from Stryker, Kalamazoo, MI and
Visera System from Olympus Medical Systems, Hamburg,
Germany), as well as a robotic platform (DaVinci Surgical
SLN detection: false-negative rates
System from Intuitive Surgical, Sunnyvale, CA), that incor- Acceptable false-negative rates for SLN mapping in endome-
porate ICG with NIR imaging. There are also handheld and trial cancer are derived from profciency metrics in SLN map-
mountable NIR imaging systems (e.g., Stryker and Olympus ping for early-stage breast cancer. Based upon these metrics,
Medical Systems). Some imaging systems include a trimodal surgeons should meet a <5% false-negative rate threshold to
view of NIR, white light (perfusion-based assessment), and be considered profcient in the technique.47 False-negative
color-segmented fuorescence (a heat map showing intensity rates for SLN mapping in endometrial cancer range widely
of ICG uptake). The addition of color-segmented fuorescence in the literature, refecting surgeon experience, tracer selec-
may enable surgeons to better differentiate lymphatic channels tion, body mass index (BMI) of the patient population, and
from SLNs and SLNs from secondary echelon nodes.45 whether a surgical staging algorithm was used. The contempo-
rary literature reports false-negative rates of less than 5% and
negative predictive values of 97%–100%.39,44,46 Holloway et
al. retrospectively compared patients with endometrial cancer
SLN detection using an algorithm
(n = 780) who had undergone lymphadenectomy alone com-
Early reports of SLN mapping in endometrial cancer focused pared with SLN mapping plus lymphadenectomy. The two
on the feasibility of the technique. Later work sought to deter- groups were similar across all comparisons, except patients
mine whether SLN mapping was a reasonable alternative to who had undergone SLN mapping demonstrated a signifcant
lymphadenectomy in apparent early-stage disease. increase in the detection of lymph node metastasis, which was
Numerous publications have demonstrated the technical fea- associated with an increase in the use of adjuvant therapy. In
sibility of SLN mapping in endometrial cancer; however, the SLN-positive cases, the SLN was the only positive node 50%
high false-negative rates reported in these early papers raised of the time and the SLN false-negative rate was 2.8%.48 The
concern. MSK incorporated a surgical staging algorithm for FIRES trial was a single-arm prospective cohort study of 340
endometrial cancer and applied it retrospectively to a cohort patients undergoing surgical staging with SLN mapping fol-
of women staged with SLN plus lymphadenectomy. The appli- lowed by lymphadenectomy for apparent early-stage endome-
cation of the algorithm reduced the false-negative rate in the trial cancer (any grade or histology). Investigators performed
cohort from 15% to 2%.46 The MSK surgical staging algorithm site visits to observe surgeons performing SLN mapping as

FIGURE 13.4 Memorial Sloan Kettering Cancer Center surgical algorithm for endometrial cancer. (Adopted by the National Comprehensive Cancer
Center; from Ref. 46, with permission.)
104 Clinical Lymphatic Mapping

a quality metric prior to patient enrollment. The investiga­ negative pelvic nodes in a cohort of patients with isolated
tors reported a high rate of successful mapping to at least one positive paraaortic nodes led to an approximate 30% detec­
hemipelvis (86%), a low false-negative rate (3%), and a high tion rate of pelvic nodal metastasis, previously undetected,
negative predictive value (99.6%).39 These results support SLN which reduced the rate of isolated paraaortic nodal metastases
mapping in endometrial cancer as an acceptable alternative to to 1.5% in this series.53 Mueller et al.8 from MSK published a
lymphadenectomy.2,44,49 clinical and pathologic study of 1044 patients with endome­
trial cancer staged with bilateral SLN mapping over a 10-year
period and reported that 6% (61 of 1044 patients) had isolated
tumor cells (ITCs) that would be missed with routine hema­
SLN mapping versus lymphadenectomy: toxylin and eosin pathologic nodal processing (Table 13.1).8
the great debate Kim et al. from MSK reported on a cohort of 508 patients with
Lymphadenectomy is associated with increased periopera­ clinical stage I endometrial cancer retrospectively subjected to
tive risk, including bleeding, operative injury, lymphocele pathologic SLN ultrastaging and detected an additional 4.5%
development, and chronic leg lymphedema, which can have incidence of nodal metastases with the use of ultrastaging.54
a dramatic impact on quality of life. Prospective trials con­ However, this remains an area of debate in the field, particu­
ducted in patients with apparent early-stage endometrial can­ larly in high-risk endometrial histology. For now, we accept
cer undergoing comprehensive nodal staging demonstrated the small percentage of patients (1%–3%) with potentially
that nodal evaluation is prognostic but not associated with a missed isolated paraaortic nodal metastasis using SLN map­
survival advantage,12,13 providing the impetus for the explo­ ping in endometrial cancer and leave the decision of whether to
ration of SLN techniques in endometrial cancer. SLN map­ proceed with a paraaortic lymphadenectomy to the surgeon’s
ping is associated with decreased morbidity and increased discretion after careful consideration of the surgical staging
precision in detecting nodal disease and provides valuable algorithm (Figure 13.4). It is important to note that this is not
prognostic information. Leitao et al. used a mailed survey to a unique, recent clinical scenario and that paraaortic metas­
describe the first patient-reported outcomes on postoperative tases were unfortunately missed even in the pre-SLN era. In
leg lymphedema in surgically staged patients with endome­ fact, the findings of an SGO survey by Soliman et al. showed
trial cancer, with encouraging results in support of SLN versus that only 11% of gynecologic oncologists routinely performed
lymphadenectomy.16 a paraaortic dissection to renal vessels as part of staging.55
SLN mapping in endometrial cancer has concerned some in
the field, given the risk of missing a positive paraaortic SLN.50
Work by the Mayo Clinic showed that in women with apparent
early-stage endometrial cancer found to have a positive pel­ SLN mapping and oncologic outcomes
vic node, approximately 50% will have a positive paraaortic Endometrioid histology
node, most often located above the IMA.51 Conversely, in the
setting of negative pelvic nodes, an isolated positive paraaor­ There are limited publications comparing oncologic outcomes
tic node will be found in 2%–3% of patients.51,52 The value of between patients who have undergone SLN mapping and
paraaortic lymphadenectomy to capture this small percentage those who have undergone lymphadenectomy. The literature
of patients is unclear. Pathologic ultrastaging of pelvic lymph regarding oncologic safety is limited to observational data,
nodes was not performed in these frequently cited studies; and there have not been any high-level prospective trials com­
thus, micrometastatic disease may have gone undetected. In a paring the two techniques.56,57 As a reminder, pelvic lymph­
recent paper from the Mayo Clinic, ultrastaging of presumably adenectomy for staging did not become a standard of care

TABLE 13.1
Incidence of Sentinel Lymph Node Metastases Reported by Grade and Depth of Myometrial Invasion in Endometrioid Endometrial
Carcinoma (n = 959)
Grade 1 Grade 2 Grade 3
n (%) n (%) n (%)
Depth of invasion ITC Micro/macro ITC Micro/macro ITC Micro/macro
None 2/449 (<1) 0/449 (0) 0/61 (0) 0/61 (0) 0/20 (0) 1/20 (5)
Inner half 20/202 (10) 9/202 (4.5) 4/76 (5) 3/76 (4) 2/31 (6) 1/31 (3)
Outer half 19/62 (31) 6/62 (10) 7/41 (17) 8/41 (20) 3/17 (18) 4/17 (24)
Total 41/713 (6) 15/713 (2) 11/178 11/178 (6) 5/68 (7) 6/68 (9)
(6)

Notes: Depth of invasion: inner half defined as <50% myoinvasion; outer half defined as ≥50% myoinvasion; Grade: Refers to the International Federation
of Gynecology and Obstetrics (FIGO) grading system for endometrial carcinoma.

Abbreviation: ITC, isolated tumor cells.

Source: Reprinted from Ref. 8, with permission from Elsevier.


Biopsy of the endometrium 105

based on prospective randomized trials showing an oncologic node-positivity rate (the only positive node in 40% of cases),
beneft. For SLN mapping, the best available data we have for and the false-negative rate was 4.3%. The authors concluded
now come from the collaborative work of the Mayo Clinic and SLN mapping was a safe alternative to lymphadenectomy,
MSK, who have reported on the staging, treatment, and onco- with an acceptable false-negative rate in this patient popu-
logic outcomes of two different low-grade endometrial cancer lation. The prospective FIRES trial included a substantial
populations. The Mayo Clinic used data from a period when number (n = 100) of patients with endometrial cancer of high-
surgical practice was to perform a comprehensive pelvic and grade histology and reported an overall false-negative rate
aortic node dissection for high-risk uterine factors; MSK used of 3%.39 The SENTOR trial62 was a prospective study of 156
data from a period when surgical practice was to perform SLN patients with clinical stage I endometrial cancer with inter-
mapping. In the pooled results of 1135 women with endome- mediate- and high-risk histologic subtypes that examined
trioid endometrial cancer with <50% myoinvasion, metasta- the sensitivity of the SLN algorithm in this high-risk popula-
ses to pelvic nodes were detected in 5.1% of MSK patients tion. Patients underwent minimally invasive staging surgery,
compared with 2.6% of Mayo patients (p = 0.03). A similar including SLN mapping, followed by pelvic lymphadenec-
number of positive paraaortic nodal metastases were detected tomy in patients with FIGO grade 2 histology and pelvic and
in both groups despite fewer paraaortic nodes removed in paraaortic lymphadenectomy for all other subtypes, with a
the MSK-staged patients. The 3-year disease-free survival median of 16 pelvic and 5 paraaortic lymph nodes removed.
rates were not signifcantly different (94.9% [95% confdence An SLN was detected in 97% of patients (78% bilateral detec-
interval (CI), 92.4–97.5] for SLN-mapped patients compared tion rate), the false-negative rate was 4% (95% CI, 0.1–1–19),
with 96.8% [95% CI, 95.2–98.5] for lymphadenectomy-staged and the negative predictive value was 99% (95% CI, 96–100).
patients).58 Of the 27 node-positive patients in the study, 52% had metas-
This same collaboration between the Mayo Clinic and MSK tases in the SLN alone. The authors concluded that SLN map-
studied 176 patients with endometrioid endometrial cancer with ping in intermediate- and high-grade endometrial cancer has
outer-half myoinvasion. On multivariable analysis, stage  III excellent diagnostic accuracy and may be a suitable replace-
disease and receipt of adjuvant treatment were associated with ment for lymphadenectomy in low- and high-grade endome-
overall survival in the combined cohort. When analyzing node- trial cancer. No oncologic outcomes were reported in these
negative patients, there was no association between overall sur- prospective trials.
vival and the method of nodal assessment if patients received A group from Johns Hopkins University published a retro-
adjuvant therapy. The adjusted hazard ratios for progression- spective study of 52 patients with clinical stage I high-grade
free survival, nonvaginal progression-free survival, and overall endometrial cancer undergoing surgical staging with SLN
survival were 0.69 (95% CI, 0.23–2.03), 0.67 (95% CI, 0.19– mapping using a staging algorithm, followed by lymphad-
2.33), and 0.81 (95% CI, 0.16–4.22), respectively.59 enectomy.63 The authors reported a false-negative rate of 22%
A Canadian research group retrospectively compared 275 (two of nine node-positive patients) but also reported that
patients with surgically staged endometrial cancer who had SLN mapping did not affect adjuvant therapy choice or risk of
undergone SLN mapping with 197 patients who had undergone recurrence in the node-positive patients.
lymphadenectomy. They observed no difference in survival The MSK and Mayo groups published their retrospective
between the groups but reported a pelvic sidewall recurrence experience on staging, treatment, and oncologic outcomes in
rate of 30% in SLN-staged patients compared with 71% in high-risk endometrial cancer histologies with SLN mapping
lymphadenectomy-staged patients. The authors concluded that (MSK) compared with lymphadenectomy (Mayo Clinic).
the SLN mapping approach was the most accurate method in Ducie et al. published on patients with deeply invasive FIGO
detecting nodes at greatest risk for disease.56 grade 3 plus any serous or clear cell histology with any myo-
invasion, with approximately 200 patients from each institu-
tion.64 The authors reported no difference in FIGO stage IIIC
High-risk histology
detection (21.7% SLN versus 19.4% lymphadenectomy; p =
Some question the oncologic safety of SLN mapping in high- 0.68) and similar rates of paraaortic lymph node metastases
risk endometrial cancers, given the increased risk of recur- in their high-risk cohort (8.3% SLN versus 12.6% lymph-
rence and death compared with low-risk disease, even when adenectomy; p = 0.29). Schiavone et al. published the MSK
uterine confned. There is concern that the false-negative rates experience of 248 patients with serous endometrial cancer; 95
in these high-risk cancers may be higher than reported in the were staged with lymphadenectomy and 153 with SLN map-
literature, as most studies report from experience with low- ping.65 Similar rates of FIGO stage IIIC detection, receipt of
grade histologic subtypes. There is also concern that endome- adjuvant therapy, and oncologic outcomes were noted among
trial cancers of high-risk histology may harbor occult aortic the two groups. The 2-year progression-free survival rate was
nodal metastasis with a higher frequency than cancers of low- 77% (CI 68%–83%) for SLN-staged patients and 71% (95%
risk histology.60 CI, 61%–79%) for lymphadenectomy-staged patients. In 2019,
Researchers at the MD Anderson Cancer Center conducted Basaran et al. published results from a retrospective cohort
a prospective study of 101 patients with clinical stage I high- of 245 patients with serous endometrial carcinoma surgically
grade endometrial subtypes who underwent staging surgery, staged according to the ‘ideal use’ of the MSK surgical stag-
including SLN mapping using a surgical staging algorithm, ing algorithm (bilateral SLN retrieval without lymphadenec-
followed by pelvic and aortic lymphadenectomy.61 At least tomy, n = 79) versus lymphadenectomy alone (n = 166).66 The
one SLN was removed in 89% of patients. There was a 23% 2-year overall survival rates were 96.6% for the SLN group
106 Clinical Lymphatic Mapping

FIGURE 13.5 Representative example of clinical positron lymphography (PLG) with positron emission tomography/computed tomography (PET/
CT) and intracervical injection of 18F-fuoro-2-deoxy-D-glucose (18F-FDG) in a patient with uterine carcinosarcoma. (a and b) PLG demonstrated three
sentinel lymph nodes (SLNs) in the right pelvis, (b) one of which demonstrated a prolonged and delayed uptake pattern, as shown in the time-activity
curves (c). (d) Intraoperative measurements of counts per minute (CPM) of the excised lymph nodes demonstrated higher uptake in the suspicious
node even 5 hours after tracer injection. (e) 3D surface reconstruction of the PET/CT at 25 minutes showing the higher uptake in the suspicious node
(blue circle) and the same node shown on the axial CT (f). (g) Intraoperative view of the node stained by vital blue dye. Histology – low (h) and
high magnifcation (i and j) confrming the presence of metastatic carcinoma within the suspicious node. (This research was originally published in
JNM. Mueller JJ, Dauer LT, Murali R, Iasonos A, Pandit-Taskar N, Abu-Rustum N, Grimm J. Positron Lymphography via Intracervical 18 F-FDG
Injection for Pre-Surgical Lymphatic Mapping in Cervical and Endometrial Malignancies. J Nucl Med. 2020 Jan 10:jnumed.119.230714. doi: 10.2967/
jnumed.119.230714. Online ahead of print.)

versus 89.6% for the lymphadenectomy group in stage I and II widely available radiotracer 18F-fuorodeoxyglucose (18F-
patients (p = 0.8) and 73.6% for the SLN group versus 77.3% FDG) to locally inject the cervix, followed by dynamic posi-
for the lymphadenectomy group in stage III patients (p = 0.8). tron emission tomography/computed tomography imaging
In a study of 136 patients with endometrial carcinosarcoma (Figure 13.5). This technique has been shown to not only map
staged with SLN mapping or lymphadenectomy,67 Schiavone to the SLN on imaging but has also been able to delineate an
et al. reported that node-positive disease detection was similar SLN that harbors metastasis. Mueller et al. from MSK pub-
between the two groups, and the 2-year progression-free sur- lished the frst in-human results from 20 patients with endome-
vival was 23 months for each group. trial and cervical cancers using this technique and detected all
No prospective trials have examined oncologic outcomes cases (3 patients) with pathologically confrmed SLN metasta-
in patients staged with SLN mapping. Some have raised con- ses, including 2 patients with micrometastatic disease.69
cern for nodal recurrences either in the aortic region or pel- Use of novel imaging modalities may enhance metastatic
vis due to false-negative SLNs at initial surgery. Aloisi et al. node detection. MSK is currently looking at clinical applica-
published on patterns of frst recurrence in a cohort of 207 tions of Cerenkov luminescence imaging. Cerenkov light is
stage IIIC1 patients staged with SLN mapping alone (no aortic emitted by many different radiopharmaceuticals, including 18F-
nodal dissection) and reported a 30% recurrence rate. Of these FDG. When these charged particles travel through a nondialec-
frst recurrences, only 8% were pure node, of which 4% were tric medium (such as water in a cell), they emit light within the
isolated aortic disease.68 There are limited data on this topic. visible blue spectrum, which can be captured with Cerenkov
At this time, SLN mapping using the MSK NCCN surgical imaging systems. Grimm and colleagues published their work
staging algorithm for endometrial cancer is deemed a reason- using integrated positron lymphography with Cerenkov imag-
able alternative to lymphadenectomy, with predominantly ret- ing in a melanoma murine model and showed a clear delin-
rospective, observational research to support this practice.44 eation of normal versus malignant nodal tissue confrmed on
histopathology.70 Technological advances such as these may
help add even more value to SLN mapping for our patients.
Future directions
Investigators are focused on improving SLN mapping tech- REFERENCES
niques using novel approaches to radiotracers and imaging 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA
modalities. For example, positron lymphography uses the Cancer J Clin. 2020;70(1):7–30.
Biopsy of the endometrium 107

2. Koh WJ, Abu-Rustum NR, Bean S, et al. Uterine Neoplasms, prevalence of lower-extremity lymphedema after sentinel
Version 1.2018, NCCN clinical practice guidelines in oncol- lymph node mapping versus lymphadenectomy. Gynecol
ogy. J Natl Compr Canc Netw. 2018;16(2):170–99. Oncol. 2020;156(1):147–53.
3. Surveillance, Epidemiology, and End Results Program. 17. Accorsi GS, Paiva LL, Schmidt R, et al. Sentinel lymph
Cancer Stat Facts: Uterine Cancer [cited 2020 May 22]. node mapping vs systematic lymphadenectomy for endo-
Available from: https://seer.cancer.gov/statfacts/html/corp. metrial cancer: surgical morbidity and lymphatic complica-
html. tions. J Minim Invasive Gynecol. 2020;27(4):938–45.
4. Levenback C, van der Zee AGJ, Coleman RL. Clinical 18. Cabanas RM. An approach for the treatment of penile car-
Lymphatic Mapping of Gynecologic Cancers. London: cinoma. Cancer. 1977;39(2):456–66.
CRC Press; 2004. 19. Burke TW, Levenback C, Tornos C, et al. Intraabdominal
5. Creasman WT, Morrow CP, Bundy BN, et al. Surgical patho- lymphatic mapping to direct selective pelvic and para-
logic spread patterns of endometrial cancer. A Gynecologic aortic lymphadenectomy in women with high-risk endo-
Oncology Group Study. Cancer. 1987;60(8 Suppl):2035–41. metrial cancer: results of a pilot study. Gynecol Oncol.
6. Chi DS, Barakat RR, Palayekar MJ, et al. The incidence of 1996;62(2):169–73.
pelvic lymph node metastasis by FIGO staging for patients 20. Abu-Rustum NR, Khoury-Collado F, Gemignani ML.
with adequately surgically staged endometrial adenocar- Techniques of sentinel lymph node identifcation for
cinoma of endometrioid histology. Int J Gynecol Cancer. early-stage cervical and uterine cancer. Gynecol Oncol.
2008;18(2):269–73. 2008;111(2 Suppl):S44–50.
7. Creasman WT, Ali S, Mutch DG, et al. Surgical- 21. Buda A, Lissoni A, Milani R. Sentinel lymph node detec-
pathological fndings in type 1 and 2 endometrial cancer: tion in endometrial cancer: hysteroscopic peritumoral ver-
an NRG Oncology/Gynecologic Oncology Group study on sus cervical injection. J Gynecol Oncol. 2016;27(1):e11.
GOG-210 protocol. Gynecol Oncol. 2017;145(3):519–25. 22. Renz M, Diver E, English D, et al. Sentinel lymph node
8. Mueller JJ, Pedra Nobre S, Braxton K, et al. Incidence of biopsies in endometrial cancer: practice patterns among
pelvic lymph node metastasis using modern FIGO staging gynecologic oncologists in the United States. J Minim
and sentinel lymph node mapping with ultrastaging in sur- Invasive Gynecol. 2020;27(2):482–8.
gically staged patients with endometrioid and serous endo- 23. Casarin J, Multinu F, Abu-Rustum NR, et al. Factors
metrial carcinoma. Gynecol Oncol. 2020;157(3):619–23. infuencing the adoption of the sentinel lymph node tech-
9. Creutzberg CL, van Putten WL, Koper PC, et al. Surgery nique for endometrial cancer staging: an international
and postoperative radiotherapy versus surgery alone for survey of gynecologic oncologists. Int J Gynecol Cancer.
patients with stage-1 endometrial carcinoma: multicentre 2019;29(1):60–7.
randomised trial. PORTEC Study Group. Post Operative 24. Rossi EC, Jackson A, Ivanova A, et al. Detection of sentinel
Radiation Therapy in Endometrial Carcinoma. Lancet. nodes for endometrial cancer with robotic assisted fuores-
2000;355(9213):1404–11. cence imaging: cervical versus hysteroscopic injection. Int
10. Nout RA, Smit V, Putter H, et al. Vaginal brachytherapy J Gynecol Cancer. 2013;23(9):1704–11.
versus pelvic external beam radiotherapy for patients with 25. Bodurtha Smith AJ, Fader An, Tanner EJ. Sentinel
endometrial cancer of high-intermediate risk (PORTEC-2): lymph node assessment in endometrial cancer: a sys-
an open-label, non-inferiority, randomised trial. Lancet. tematic review and metaanalysis. AM J Obstet Gynecol.
2010;375:816–23. 2017;216(5):459–76.
11. Keys HM, Roberts JA, Brunetto VL, et al. A phase III 26. Kang S, Yoo HJ, Hwang JH, et al. Sentinel lymph node
trial of surgery with or without adjunctive external pelvic biopsy in endometrial cancer: meta analysis of 26 studies.
radiation therapy in intermediate risk endometrial adeno- Gynecol Oncol. 2011;123(3):522–7.
carcinoma: a Gynecologic Oncology Group study. Gynecol 27. Khoury-Collado F, Murray MP, Hensley ML, et al. Sentinel
Oncol. 2004;92:744–51. lymph node mapping for endometrial cancer improves the
12. Benedetti Panici P, Basile S, Maneschi F, et al. Systematic detection of metastatic disease to regional lymph nodes.
pelvic lymphadenectomy vs. no lymphadenectomy in early- Gynecol Oncol. 2011;122(2):251–4.
stage endometrial carcinoma: randomized clinical trial. J 28. Frumovitz M, Plante M, Lee PS, et al. Near-infrared fuo-
Natl Cancer Inst. 2008;100(23):1707–16. rescence for detection of sentinel lymph nodes in women
13. Barton DP, Naik R, Herod J. Effcacy of systematic pel- with cervical and uterine cancers (FILM): a randomised,
vic lymphadenectomy in endometrial cancer (MRC phase 3, multicentre, non-inferiority trial. Lancet Oncol.
ASTEC Trial): a randomized study. Int J Gynecol Cancer. 2018;19(10):1394–403.
2009;19(8):1465. 29. Khoury-Collado F, Abu-Rustum NR. Lymphatic map-
14. Abu-Rustum NR, Alektiar K, Iasonos A, et al. The inci- ping in endometrial cancer: a literature review of current
dence of symptomatic lower-extremity lymphedema fol- techniques and results. Int J Gynecol Cancer. 2008;18(6):
lowing treatment of uterine corpus malignancies: a 12-year 1163–8.
experience at Memorial Sloan-Kettering cancer center. 30. Papadia A, Gasparri ML, Buda A, et al. Sentinel lymph
Gynecol Oncol. 2006;103:714–18. node mapping in endometrial cancer: comparison of
15. Dessources K, Aviki E, Leitao Jr MM. Lower extremity fuorescence dye with traditional radiocolloid and blue. J
lymphedema in patients with gynecologic malignancies. Int Cancer Res Clin Oncol. 2017;143(10):2039–48.
J Gyecol Cancer. 2020;30(2):252–60. 31. Rossi EC. Current state of sentinel lymph nodes for
16. Leitao MM Jr, Zhou QC, Gomez-Hidalgo NR, et al. Patient- women with endometrial cancer. Int J Gynecol Cancer.
reported outcomes after surgery for endometrial carcinoma: 2019;29(3):613–21.
108 Clinical Lymphatic Mapping

32. Eriksson AG, Beavis A, Soslow RA, et al. A comparison in endometrial cancer staging: beyond removal of blue
of the detection of sentinel lymph nodes using indocyanine nodes. Gynecol Oncol. 2012;125(3):531–5.
green and near-infrared fuorescence imaging versus blue 47. Lyman GH, Giuliano AE, Somerfeld MR, et al. American
dye during robotic surgery in uterine cancer. Int J Gynecol Society of Clinical Oncology guideline recommendations
Cancer. 2017;27(4):743–7. for sentinel lymph node biopsy in early-stage breast cancer.
33. Eriksson AG, Montovano M, Beavis A, et al. Impact of obe- J Clin Oncol. 2005;23(30):7703–20.
sity on sentinel lymph node mapping in patients with newly 48. Holloway RW, Gupta S, Stavitzski NM, et al. Sentinel
diagnosed uterine cancer undergoing robotic surgery. Ann lymph node mapping with staging lymphadenectomy for
Surg Oncol. 2016;23(8):2522–8. patients with endometrial cancer increases the detection of
34. Rozenholc A, Samouelian V, Warkus T, et al. Green versus metastasis. Gynecol Oncol. 2016;141(2):206–10.
blue: randomized controlled trial comparing indocyanine 49. SGO Clinical Practice Statement: The Role of Sentinel
green with methylene blue for sentinel lymph node detection Lymph Node Mapping in Endometrial Cancer; 2015 [cited
in endometrial cancer. Gynecol Oncol. 2019;153(3):500–4. 2020 Feb 10]. Available from: www.sgo.org/clinical-prac-
35. Plante M, Touhami O, Trinh XB, et al. Sentinel node tice/guidelines/the-role-of-sentinel-lymph-node-mapping-
mapping with indocyanine green and endoscopic near- in-endometrial-cancer/.
infrared fuorescence imaging in endometrial cancer. A 50. Frumovitz M, Coleman RC, Soliman PT, et al. A case for
pilot study and review of the literature. Gynecol Oncol. caution in the pursuit of the sentinel node in women with
2015;137(3):443–7. endometrial carcinoma. Gynecol Oncol. 2014;132(2):275–9.
36. Buda A, Bussi B, Di Martino G, et al. Sentinel lymph node 51. Kumar S, Podratz KC, Bakkum-Gamez JN, et al.
mapping with near-infrared fuorescent imaging using Prospective assessment of the prevalence of pelvic, para-
indocyanine green: a new tool for laparoscopic platform aortic and high paraaortic lymph node metastasis in endo-
in patients with endometrial and cervical cancer. J Minim metrial cancer. Gynecol Oncol. 2014;132(1):38–43.
Invasive Gynecol. 2016;23(2):265–9. 52. James JA, Rakowski JA, Jeppson CN, et al. Robotic transperi-
37. Henriksen E, Murrieta T. Adenocarcinoma of the cor- toneal infra-renal aortic lymphadenectomy in early-stage
pus uteri; a clinicopathological study. West J Surg Obstet endometrial cancer. Gynecol Oncol. 201;136(2):285–92.
Gynecol. 1950;58(7):331–43. 53. Multinu F, Casarin J, Cappuccio S, et al. Ultrastaging of
38. Masubuchi S, Fujimoto I, Masubuchi K. Lymph node negative pelvic lymph nodes to decrease the true prevalence
metastasis and prognosis of endometrial carcinoma. of isolated paraaortic dissemination in endometrial cancer.
Gynecol Oncol. 1979;7(1):36–46. Gynecol Oncol. 2019:154(1):60–4.
39. Rossi EC, Kowalski LD, Scalici J, et al. A comparison of 54. Kim CH, Soslow RA, Park KJ, et al. Pathologic ultrastag-
sentinel lymph node biopsy to lymphadenectomy for endo- ing improves micrometastasis detection in sentinel lymph
metrial cancer staging (FIRES trial): a multicentre, pro- nodes during endometrial cancer staging. Int J Gynecol
spective, cohort study. Lancet Oncol. 2017;18(3):384–92. Cancer. 2013;23(5):964–70.
40. Jewell EL, Huang JJ, Abu-Rustum NR, et al. Detection of 55. Soliman PT, Frumovitz M, Spannuth W, et al.
sentinel lymph nodes in minimally invasive surgery using Lymphadenectomy during endometrial cancer staging:
indocyanine green and near-infrared fuorescence imag- practice patterns among gynecologic oncologists. Gynecol
ing for uterine and cervical malignancies. Gynecol Oncol. Oncol. 2010;119(2):291–4.
2014;133(2):274–7. 56. How J, Gauthier C, Abitbol J, et al. Impact of sentinel
41. Abu-Rustum NR. Sentinel lymph node mapping for endo- lymph node mapping on recurrence patterns in endometrial
metrial cancer: a modern approach to surgical staging. J cancer. Gynecol Oncol. 2017;144(3):503–9.
Natl Compr Canc Netw. 2014;12(2):288–97. 57. Buda A, Restaino S, Di Martino G, et al. The impact of
42. Khoury-Collado F, Glaser GE, Zivanovic O, et al the type of nodal assessment on prognosis in patients with
Improving sentinel lymph node detection rates in endome- high-intermediate and high-risk ESMO/ESGO/ESTRO
trial cancer: how many cases are needed? Gynecol Oncol. group endometrial cancer. A multicenter Italian study. Eur
2009;115(3):453–5. J Surg Oncol. 2018;44(10):1562–7.
43. Tucker K, Staley SA, Gehrig PA, et al. Defning the learn- 58. Eriksson AGZ, Ducie J, Ali N, et al. Comparison of a
ing curve for successful staging with sentinel lymph node sentinel lymph node and a selective lymphadenectomy
biopsy for endometrial cancer among surgeons at an aca- algorithm in patients with endometrioid endometrial car-
demic institution. Int J Gynecol Cancer. 2020;30(3):346–51. cinoma and limited myometrial invasion. Gynecol Oncol.
44. Holloway RW, Abu-Rustum NR, Backes FJ, et al. Sentinel 2016;140(3):394–9.
lymph node mapping and staging in endometrial can- 59. Schlappe BA, Weaver AL, Ducie JA, et al. Multicenter
cer: a Society of Gynecologic Oncology literature review study comparing oncologic outcomes between two nodal
with consensus recommendations. Gynecol Oncol. assessment methods in patients with deeply invasive endo-
2017;146(2):405–15. metrioid endometrial carcinoma: a sentinel lymph node
45. Lopez Labrousse MI, Frumovitz M, Guadalupe Patrono M, algorithm versus a comprehensive pelvic and paraaortic
et al. Sentinel lymph node mapping in minimally invasive lymphadenectomy. Gynecol Oncol. 2018;151(2):235–42.
surgery: role of imaging with color-segmented fuorescence 60. Mariani A, Dowdy SC, Cliby WA, et al Prospective assess-
(CSF). Gynecol Oncol. 2017;146(3):676–7. ment of lymphatic dissemination in endometrial can-
46. Barlin JN, Khoury-Collado F, Kim CH, et al. The impor- cer: a paradigm shift in surgical staging. Gynecol Oncol.
tance of applying a sentinel lymph node mapping algorithm 2008;109(1):11–18.
Biopsy of the endometrium 109

61. Soliman PT, Westin SN, Dioun S, et al. A prospective vali- 66. Basaran D, Bruce S, Aviki EM, et al. Sentinel lymph node
dation study of sentinel lymph mapping for high-risk endo- mapping alone compared to more extensive lymphadenec-
metrial cancer. Gynecol Oncol. 2017;146(2):234–9. tomy in patients with uterine serous carcinoma. Gynecol
62. Cusimano MC, Vicus D, Pulman K, et al. Assessment Oncol. 2020;156(1):70–6.
of sentinel lymph node biopsy vs lymphadenectomy for 67. Schiavone MB, Zivanovic O, Zhou Q, et al. Survival of
intermediate- and high-grade endometrial cancer staging. patients with uterine carcinosarcoma undergoing sentinel
JAMA Surg. 2021;156(2):157–64. https://doi.org/10.1001/ lymph node mapping. Ann Surg Oncol. 2016;23(1):196–202.
jamasurg.2020.5060. 68. Aloisi A, Casanova JM, Tseng JH, et al. Patterns of
63. Tanner EJ, Ojalvo L, Stone RL, et al. The utility of senti- FIRST recurrence of stage IIIC1 endometrial cancer with
nel lymph mapping in high-grade endometrial cancer. Int J no PARAAORTIC nodal assessment. Gynecol Oncol.
Gynecol Cancer. 2017;27(7):1416–21. 2018;151(3):395–400.
64. Ducie JA, Eriksson AGZ, Ali N, et al. Comparison of a sen- 69. Mueller JJ, Dauer LT, Murali R, et al. Positron lymphog-
tinel lymph node mapping algorithm and comprehensive raphy via intracervical 18F-FDG injection for pre-surgical
lymphadenectomy in the detection of stage IIIC endome- lymphatic mapping in cervical and endometrial malig-
trial carcinoma at higher risk for nodal disease. Gynecol nancies. J Nucl Med. 2020;61(8);1123–30. https://doi.
Oncol. 2017;147(3):541–8. org/10.2967/jnumed.119.230714.
65. Schiavone MB, Scelzo C, Straight C, et al. Survival 70. Lockau H, Neuschmelting V, Ogirala A, et al. Dynamic
of patients with serous uterine carcinoma undergo- 18F-FDG PET lymphography for in vivo identifcation of
ing sentinel lymph node mapping. Ann Surg Oncol. lymph node metastases in murine melanoma. J Nucl Med.
2017;24(7):1965–71. 2018;59(2):210–15.
14
Impact of sentinel lymph node mapping on quality of life

Mariana Antonella Nigra, Pedro T. Ramirez, and Larissa Meyer

CONTENTS
Quality of life in gynecologic cancer ........................................................................................................................................... 111
Sentinel lymph node mapping and lymphedema ......................................................................................................................... 112
Vulvar cancer: quality of life after sentinel lymph node mapping ............................................................................................... 112
Endometrial cancer: quality of life after sentinel lymph node mapping ...................................................................................... 113
Cervical cancer: quality of life after sentinel lymph node mapping ............................................................................................ 113
Patient-reported outcomes in gynecologic cancer ....................................................................................................................... 114
Conclusion ................................................................................................................................................................................... 115
References.................................................................................................................................................................................... 115

Quality of life in gynecologic cancer fve functional scales (physical, role, cognitive, emotional,
and social), three symptom scales (fatigue, pain, and nausea/
Early detection and novel therapies in the management of vomiting), a global health status, and a number of single items
patients with gynecologic cancers have led to signifcant assessing additional symptoms commonly reported by cancer
improvements in survival. As a result, there are an increas- patients (dyspnea, loss of appetite, insomnia, constipation, and
ing number of cancer survivors for whom quality of life (QoL) diarrhea) and perceived fnancial impact of the disease.7
is a major determinant on return to normal daily activities.1 QoL assessments are a key factor in assessing cancer bur-
QoL is defned as overall general well-being, which comprises den and the effect of treatment.4 The literature has extensively
objective descriptions and subjective evaluations of physical, described the toxicities and side effects related to such treat-
material, social, and emotional well-being, together with the ments, as is the case of lower limb lymphedema. This is a
extent of personal development, all weighted by a personal set condition, often irreversible, that affects many patients after
of values.2 The principle of QoL has been studied since the lymphadenectomy for gynecologic cancer, and it has been con-
1960s, and it has been routinely associated with the discussion sistently shown to affect multiple QoL metrics.8 The overall
of numerous items ranging from the relative ‘liveability’ and incidence of treatment-related lymphedema is approximately
effects of social policy, to the benefts of different models of 25% in endometrial or cervical cancer, but it may be as high
human service providers, to the individualized outcomes of a as 70% in vulvar cancer if a lymphadenectomy is performed.9
vast number of medical and psychotherapeutic practices.3 The clinical presentations of lower limb lymphedema are usu-
Currently, it is possible to quantify QoL to establish numeri- ally chronic and progressive, leading to a reduction in QoL.10
cal parameters for the evaluation of medical interventions. In Ryan et al. showed in a retrospective study that among patients
order to measure QoL, several questionnaires have been cre- with vulva, endometrial, and cervical cancer, 17% had lower
ated to obtain data on physical functioning, role limitations extremity lymphedema and 51% reported alterations in their
due to physical problems, social functioning, bodily pain, gen- daily activities.11 A cohort study evaluated QoL of women
eral mental health, role limitations due to emotional problems, treated with either pelvic lymphadenectomy and pelvic radia-
and vital and general health perceptions.4 A frst-generation tion therapy or pelvic radiation alone for gynecologic cancer
core questionnaire, the European Organization for Research (endometrial, ovarian, cervical, vulvar).12 The authors used a
and Treatment of Cancer (EORTC) QLQ-C36, was developed questionnaire addressing physical symptoms that originated
in 1987.5 This 36-item questionnaire was designed to be cancer in the pelvis, demographic, psychological, and QoL factors. A
specifc and applicable across cultural settings. The only scale total of 616 gynecologic cancer survivors answered the ques-
that required substantial revision, due to inadequate reliabil- tionnaire, and 36% reported lower limb lymphedema. Overall
ity, was the eight-item emotional functioning scale. In the next QoL was signifcantly lower among cancer survivors with
generation of the instrument, this scale was replaced by a four- lower limb lymphedema. They were also less satisfed with
item emotional functioning scale previously used in EORTC their sleep, more concerned with the recurrence of cancer, and
clinical trials. A second-generation core questionnaire, the more likely to interpret the body’s symptoms as recurrence.
frst version of the 30-item EORTC QLQ-C30, was subse- Cancer survivors reported that lymphedema of the lower
quently developed.6 The QLQ-C30 version 1.0 incorporates extremities prevented them from physical activity (45%) and

DOI: 10.1201/9781003255536-14 111


112 Clinical Lymphatic Mapping

housework (29%) and affected their ability to participate in cancer of the vulva and studied the safety and clinical util-
social activities (27%) or meet friends (20%). The authors thus ity of this procedure.21 When the sentinel node was found to
showed a negative impact of lymphedema on QoL and activi- be negative in the pathological ultrastaging, inguinofemoral
ties of daily living. lymphadenectomy was omitted. The patients were observed
Aesthetic and functional changes secondary to surgical with follow-up for 2 years at intervals of every 2 months.
treatment can lead to depression, anxiety, and a negative body Short-term morbidity decreased in patients after sentinel node
image. In its severe stages, lymphedema can affect a patient’s dissection compared to patients with a positive sentinel node
ability to perform the functions of daily living.13 In a retro- who underwent inguinofemoral lymphadenectomy (groin
spective study, 152 patients who underwent lymphadenec- wound rupture: 11.7% vs. 34.0%, respectively, and cellulite:
tomy for endometrial, cervical, or ovarian cancer reported the 4.5% vs. 21.3%, respectively). Long-term morbidity was also
incidence of lower limb lymphedema and neurological com- observed less frequently after sentinel node removal compared
plications collected by questionnaire for three months after to sentinel node removal and inguinofemoral lymphadenec-
surgery.14 The incidence of lower limb lymphedema and neu- tomy (recurrent erysipelas: 0.4% vs. 16.2%, respectively, and
rological complications after lymphadenectomy was 36%. lymphedema of legs: 1.9% vs. 25.2%, respectively). However,
an evaluation of QoL in the GROINSS V study was not
reported in the initial publication. Oonk et al. performed a ret-
rospective study in patients who participated in GROINSS-V
Sentinel lymph node mapping and to compare QoL in vulvar cancer patients who were treated
lymphedema with SLN mapping only to those who underwent inguino-
femoral lymphadenectomy.22 Patients were invited to fll out
For the purpose of reducing the morbidity associated with
three questionnaires: the EORTC QLQ-C30, a vulvar-specifc
lymphadenectomy, less invasive operative techniques, such as
questionnaire, and a questionnaire regarding the opinion of
lymphatic mapping and sentinel lymph node (SLN) mapping,
patients on new treatment options 33 weeks after surgery. With
have been developed.15 A prospective study of patients with
a response rate of 85%, 35 patients after SLN mapping and
stage I–IIIA melanoma of the extremities, trunk, or neck who
27 patients after inguinofemoral lymphadenectomy flled out
underwent SLN mapping evaluated the development of lymph-
the questionnaires. No difference in overall QoL was observed
edema and QoL 6 months after surgery using a functional
between the two groups. Reasons for not fnding an improve-
evaluation questionnaire for breast cancer therapy (FACT-B).16
ment in QoL was the limitation of the small study population
A total of 14 (2%) of 694 patients had lymphedema. Stage I–II
that underpowered the result. The major difference was the
patients with complete QoL data demonstrated that weighted
increase in complaints of lymphedema of the lower extremities
QoL was strongly associated with perceived changes rather
after inguinofemoral lymphadenectomy.
than actual changes in limb size. Subsequently, the sentinel
Conversely, other publications have shown a change in QoL,
node was implemented in other cancers, such as breast cancer,
such as a retrospective study, where the QoL was measured
where a signifcant reduction in the prevalence of lymphedema
in women after a complete lymphadenectomy or SLN map-
(<10%) was shown when patients underwent SLN mapping
ping for early-stage vulvar cancer.23 The severity of lymph-
without lymphadenectomy.17 Furthermore, its implementation
edema symptoms was recorded. The QoL-adjusted survival
in gynecologic cancer patients has been described in several
was measured using the Utility-Based Questionnaire-Cancer,
studies.9,18,22,25
a cancer-specifc validated QoL instrument. Sixty-three per-
In gynecologic cancers, QoL has been evaluated as it per-
cent of women reported lymphedema after complete lymph-
tains to the presence of lymphedema. A study of 789 women
adenectomy (48%) or sentinel node biopsy (15%). The activity
treated with either pelvic radiotherapy alone or with surgery
most affected in all women was physical function, especially
and diagnosed with endometrial cancer, cervical cancer, vagi-
the ability to perform vigorous activities such as running and
nal cancer, or ovarian cancer evaluated the impact of lower
climbing stairs. Social life and self-care activities were the
limb lymphedema on overall QoL, sleep, sexuality, and daily
least affected. Thirty-six women (60%) identifed themselves
life activities reported by gynecologic cancer survivors.18
as being sexually active. Of these women, 18 (50%) reported
that their health did not affect their sex life, whereas the same
number of women reported that it did. When comparing those
Vulvar cancer: quality of life after who reported lymphedema or leg pain with those who did not
experience these symptoms, QoL scores were signifcantly
sentinel lymph node mapping lower in the symptomatic group. In women reporting lymph-
Treatment for vulvar cancer has been associated with sig- edema, the greatest negative effects were on sex life, physical
nifcant QoL disruption, including sexual dysfunction, vulvar function, and social activities. The women who experienced
pain or numbness, and lymphedema.19 Patients with vulvar leg pain had higher levels of emotional distress and loss of
cancer after inguinofemoral lymphadenectomy had shown a ability to perform self-care activities.
rate of lower extremity lymphedema of 10% in retrospective Furthermore, in a retrospective study of 21 patients with
reports and 73% in studies assessing patient-reported symp- vulvar carcinoma FIGO stage I–IIIA who underwent vulvec-
toms.20 GROINSS V is a multicenter observational study that tomy or wide excision with sentinel node biopsy or inguinal
evaluated the detection of sentinel nodes using a radioactive lymphadenectomy, the authors investigated factors infuenc-
marker and blue dye in patients with T1–T2 squamous cell ing QoL and sexual function.24 All patients were interviewed
Impact of sentinel lymph node mapping 113

according to the Female Sexual Function Index questionnaire with a primary aim to measure the incidence of lower limb
(FSFI) and the Short Form 12 questionnaire (SF12). This ques- lymphedema.29 Secondary objectives included observation
tionnaire uses 12 items to describe a summary scale for QoL. of changes in lower extremity function and QoL with the
It is divided into eight subscales, providing two partial scores: FACT-G questionnaire and the Lower Extremity Functional
the Mental Component Summary and the Physical Component Scale (LEFS). The FACT-G was used to evaluate the impact of
Summary. In total, 10 patients had undergone an inguinal staging surgery and lymphedema on QoL. The LEFS is a vali-
lymphadenectomy, 5 patients a sentinel node biopsy, and 6 dated survey that consists of 20 questions that assess the effect
without lymph node evaluation. Of the patients who underwent of a lower limb condition on a person’s ability to perform
a lymphadenectomy, 90% scored low on the FSFI, but only everyday tasks. The rate of lower limb lymphedema was 25%
45% scored low without this procedure. Lymphadenectomy at 4–6 weeks, 19% at 6–9 months, and 27% at 12–18 months
was the only factor infuencing sexual function. Patients who postoperatively. The presence of lower limb lymphedema
did not undergo lymphadenectomy or sentinel node biopsy was associated with a signifcant worsening from baseline
scored better in terms of sexual function. LEFS scores at 4–6 weeks (−27.0% vs. −3.7%, p = 0.02) and
6–9 months (−13.0% vs. 0%, p = 0.01). Lower limb lymph-
edema was not associated with a change in patient-reported
global QoL.
Endometrial cancer: quality of life Another retrospective study in patients who underwent pri-
after sentinel lymph node mapping mary surgery for endometrial cancer evaluated the impact of
The implementation of the sentinel node in endometrial can- lymphadenectomy on QoL. Women were mailed a survey that
cer has been evaluated extensively, including in two prospec- included a validated 13-item lower limb lymphedema screening
tive studies. The SENTI-ENDO study demonstrated that SLN questionnaire and QoL questionnaire QLQ-C30 and EORTC
biopsy adequately predicts lymph node status, especially in QLQ-EN24 for a minimum of 44 months after surgery.30
patients with stage I endometrial cancer and increased detec- This measured fve functional scales, three symptom scales,
tion of lymph node involvement when ultrastaging was per- global health status, and a number of single items assessing
formed.25 In another key study, the FIRES trial evaluated additional symptoms commonly reported by cancer patients.
the use of the indocyanine green–labeled sentinel nodes, and Of 1275 potential participants, 599 were evaluable (180 SLN,
86% of patients had successful mapping, and nodal metasta- 352 lymphadenectomy, and 67 hysterectomies alone). Self-
ses were detected in 97% of endometrial cancer metastasis.26 reported lower limb lymphedema prevalence was 27% in the
In that study, a signifcant decrease in morbidity associated SLN group (49/180) and 41% in the lymphadenectomy group
with complete lymphadenectomy was observed. In addition, (144/352). Lymphadenectomy retained an independent associ-
a prospective study compared the rate of lymphatic complica- ation with an increased prevalence of lower limb lymphedema
tions in 188 patients with endometrial cancer undergoing SLN over SLN (p < 0.003). Patients with self-reported lower limb
biopsy versus a full pelvic and infrarenal paraaortic lymphad- lymphedema had signifcantly worse QoL compared to those
enectomy. SLN biopsy alone resulted in a lower incidence of without self-reported lower limb lymphedema.
leg lymphedema than lymphadenectomy (1.3% vs. 18.1%, p =
0.0003).27 However, none of these prospective trials evaluated
patient QoL. Cervical cancer: quality of life after
A number of investigators have explored QoL as it pertains
sentinel lymph node mapping
to the use of SLN mapping. Rowlands et al. assessed and com-
pared QoL 3–5 years after surgery for endometrial cancer in SLN biopsy in cervical cancer was validated in the SENTICOL I
women with lower limb lymphedema, lower limb swelling, or study, a retrospective study that determined sensitivity when
without.28 The patients underwent either pelvic lymphadenec- this technique was implemented.30 However, this study did
tomy or SLN. The Medical Outcomes Study Short Form-12 not evaluate morbidity of patients undergoing surgery or their
(SF-12) Health Survey was used to assess physical and mental subsequent QoL. With the need to learn about the impact of
QoL. The scale is made up of eight subscales (physical func- different treatment approaches on the QoL of patients, Park et
tioning, bodily pain, general health, physical and emotional al. aimed to identify problems related to long-term QoL and
role limitations, vitality, social functioning, and mental health), sexual function in cervical cancer survivors.31 A total of 860
which are combined to form two summary scores: physical women with a history of cervical cancer (stage I–IVa) and 494
and mental QoL. In total, 639 women were categorized as controls were selected randomly. Subjects flled out a question-
women with lower limb lymphedema (n = 68), women with naire that included the European Organization for Research
lower limb swelling (n = 177), or women without (n = 394). and Treatment of Cancer (EORTC) QLQ-C30, its Cervical
Women with lower limb lymphedema had clinically lower Cancer Module, and additional sexual function items. Cervical
physical QoL than women without; however, their mental QoL cancer survivors had clinically signifcant worse problems
was within the normal range. Women with lower limb swelling with social functioning, constipation, diarrhea, and diffcul-
had signifcantly lower physical and mental QoL than women ties with their fnances than controls (p < 0.01). Survivors also
without lower limb lymphedema or lower limb swelling. reported more severe lymphedema and menopausal symptoms
A prospective study was performed in 97 women who and worse body image, sexual and/or vaginal functioning, and
underwent minimally invasive staging for endometrial cancer sexual worry (p < 0.01). Anxiety about sexual performance
114 Clinical Lymphatic Mapping

was more problematic in survivors than in controls (p < 0.01), PROs have the potential to improve the quality and patient-
as was dyspareunia for women who received radiotherapy centeredness of medical care in a variety of ways. They can
(p < 0.01). be used at the patient level to improve interactions between
In another study, the QoL of patients was evaluated regard- patients and clinicians and can also be used in research to
less of the approach of lymph node evaluation. Le Borgne et al. identify benefts and harms of interventions. Finally, they
prospectively evaluated long-term QoL in cervical cancer sur- have a role in policy making and population surveillance,
vivors 5, 10, and 15 years after diagnosis.32 Five QoL question- including contributing to guideline development, inform-
naires (SF-36, EORTC QLQ-C30, the cervical cancer-specifc ing coverage and reimbursement decisions, evaluating care
module [EORTC QLQ-CX24], the MFI fatigue questionnaire, quality, and identifying the impacts of policy options. This
and the STAI for anxiety) and a life condition questionnaire initiative of the Patient-Reported Outcomes Measurement
were used. A total of 173 patients (42% treated with surgery Information System (PROMIS) establishes a national
alone and 58% with a combination of treatments) and 594 con- resource for precise and effcient measurement of PROs.
trols participated in the study. Compared to controls, patients The main goal of the PROMIS initiative is to develop and
had an overall good QoL, except the psycho-emotional decline evaluate to the clinical research community a set of publicly
of survivors after 15-years was worse (p < 0.01). Worsening of available, effcient, and fexible PRO measurements, includ-
some symptoms was observed over time; 15-year survivors, ing QoL.38 The PROMIS Network has developed a compre-
in particular, reported signifcantly more lymphedema than hensive repository of electronic patient-reported outcomes
5-year survivors (p = 0.009) and 10-year cervical cancer sur- measures (ePROs) of major symptom domains that have been
vivors (p = 0.002). Compared with cervical cancer survivors validated in cancer patients.
treated by surgery alone, QoL of cervical cancer survivors With the growing focus on patient-centered care, PROs are
who underwent radiotherapy was signifcantly more affected becoming an important component to clinical trials and quality
in terms of cervical cancer–specifc problems, such as sexual metrics. Tools have been studied to measure PROs in gyne-
dysfunction (p = 0.002), voiding and abdominal symptoms cologic cancer patients. A prospective study performed over 6
(p = 0.01), and lymphedema (p = 0.01). months in a gynecologic oncology outpatient clinic evaluated
Currently, the SENTIX trial is designed to determine the feasibility and acceptability of PROMIS and assessed if it
the oncological safety and postoperative morbidity of SLN can identify severely symptomatic patients and increase refer-
biopsy, only without subsequent pelvic lymph node dissec- ral to supportive services.39 Of the 336 patients who completed
tion in women with early-stage cervical cancer. However, the study, 35% had a current cancer and 19% had experienced
this study does not include a formal evaluation of QoL.33 The at least one disease recurrence. Sixty-nine percent demon-
SENTICOL III study is currently underway as a prospective, strated moderate to severe physical dysfunction (60%), pain
multicenter, randomized trial examining the oncologic out- (36%), fatigue (28%), anxiety (9%), depression (8%), and sexual
comes of sentinel lymph node mapping vs. complete lymphad- dysfunction (32%). Thirty-nine (12%) severely symptomatic
enectomy in patients with cervical cancer. One of its objectives patients were referred to services such as psychiatry, pallia-
will be the evaluation of QoL using the questionnaires QLQ- tive care, pain management, social work, or integrative oncol-
C30 and QLQ-CX24, measuring three specifc dimensions: ogy care. Most survey respondents identifed ePROs as helpful
pain, global health score, and physical functioning scores at 3 (78%) and easy to complete (92%). Concluding that outpatient
years. However, one of the potential limitations of this study is PROMIS administration is feasible and acceptable to gyneco-
that patients will be aware as to which surgery they had based logic oncology patients and can help identify severely symp-
on lymph node count in the fnal pathology report and may be tomatic patients for referral to ancillary support services.
biased to report higher rates of lymphedema and worse QoL if The PRO measure has been implemented in different felds of
knowing that they were randomized to the full lymphadenec- oncological gynecology. In a study by Meyer et al., the authors
tomy group.34 Therefore, at this time there is a paucity of data evaluated patient symptom burden in 29 patients.40 Women
evaluating QoL of patients after the implementation of sentinel were undergoing laparotomy in the gynecologic oncology
node mapping in cervical cancer. department. Symptoms were assessed using the MD Anderson
Symptom Inventory (MDASI-OC), a validated 27-item tool
used in patients with ovarian cancer. The MDASI-OC was
administered as a preoperative reference daily while admitted
Patient-reported outcomes in to the hospital after surgery, twice a week in the frst week after
gynecologic cancer discharge, and then weekly for up to 8 weeks after the opera-
tion. Seventy-fve percent of patients had a diagnosis of ovarian
Patient-reported outcomes (PROs) are defned by the Food cancer. Of these patients, half underwent primary cytoreduction
and Drug Administration (FDA) and National Quality Forum surgery and the other half received neoadjuvant chemotherapy
(NQF) as “a report that comes directly from the patient about prior to cytoreductive interval surgery. In the postoperative hos-
the status of a patient’s health condition without amend- pital setting, the fve symptoms with the highest overall burden
ment or interpretation of the patient’s response by a clini- were fatigue, pain, abdominal pain, dry mouth, and drowsiness.
cian or anyone else.”35,36 PRO instruments aim to measure The study concluded that PROs are a crucial component of
one or more aspects of PROs, which can include topics such patient-centered research and longitudinal compilation and that
as symptom burden to more complex, multidimensional con- analysis of symptom burden may allow for more meaningful
cepts, such as QoL.37 comparisons of surgical technique and preoperative care.
Impact of sentinel lymph node mapping 115

Other tools have been used in studies to measure PROs. In 6. Aaronson NK, Ahmedzai S, Bergman B, et al. The
a systematic review aiming to summarize the existing litera- European Organization for Research and Treatment of
ture related to PROs among endometrial cancer survivors, the Cancer QLQ-C30: a quality-of-life instrument for use in
investigators included articles according to exposures or spe- international clinical trials in oncology. J Nat Cancer Inst.
cifc PROs.41 Of 1722 unique studies, 102 full-text articles were 1993;85(5):365–76.
reviewed, of which a total of 27 studies fulflled the inclusion 7. The EORTC QLQ-C30 Manuals, Reference Values and
criteria. The most commonly used PRO questionnaires were Bibliography. EORTC Quality of Life Group. Belgium:
the European Organization for Research and Treatment of Brussels EORTC; 2002.
Cancer Quality of Life Questionnaire-Core 30 (EORTC QLQ- 8. Dunberger G, Lindquist H, Waldenström A, et al. Lower
C30) (n = 9), Short Form 36 Questionnaire (SF-36, n = 8), the limb lymphedema in gynecological cancer survivors –
Functional Assessment of Cancer Therapy-General (FACT-G, effect on daily life functioning. Support Care Cancer.
n = 5), and the Female Sexual Function Index (FSFI, n = 4). 2013;21(11):3063–70.
9. Dessources K, Aviki E, Leitao MM Jr. Lower extremity
Obesity was associated with lower QoL and physical function-
lymphedema in patients with gynecologic malignancies. Int
ing. Treatment type affected several outcomes. Laparoscopy
J Gynecol Cancer. 2020;30(2):252–60.
generally resulted in better QoL outcomes when compared to
10. Rowlands IJ, Beesley VL, Janda M, et al. Quality of life
laparotomy. Sexual function outcomes were dependent on age,
of women with lower limb swelling or lymphedema 3–5
time since diagnosis, and having consulted a physician before
years following endometrial cancer. Gynecol Oncol.
engaging in sexual activities. In addition, a physical activity 2014;133(2):314–18.
intervention was associated with improved sexual interest but 11. Ryan M, Stainton MC, Slaytor EK, et al. A etiology and
not sexual function. This review provides insight into the expe- prevalence of lower limb lymphedema following treatment
rience of endometrial cancer survivors. Factors that contribute for gynaecological cancer. Aust N Z J Obstet Gynecol.
to QoL, such as pain, fatigue, and emotional and social func- 2003;43(2):148–51.
tioning, should be monitored following an endometrial cancer 12. Dunberger G, Lindquist H, Waldenström A, et al. Lower
diagnosis. To our knowledge, there are no studies evaluating limb lymphedema in gynecological cancer survivors –
PROs after the implementation of SLN mapping in gyneco- effect on daily life functioning. Support Care Cancer.
logic cancer. 2013;21(11):3063–70.
13. Fu MR, Ridner SH, Hu SH, et al. Psychosocial impact of
lymphedema: a systematic review of literature from 2004
to 2011. Psychooncology. 2013;22(7):1466–84.
Conclusion
14. Biglia N, Librino A, Ottino M, et al. Lower limb lymph-
The evaluation of QoL and PROs in patients with cancer has edema and neurological complications after lymphad-
become a topic of increasing relevance both in the clinical enectomy for gynecological cancer. Int J GynecolCancer.
management of patients and in research. The increase in the 2015;25(3):521–5.
survival of patients secondary to new therapies requires the 15. Morton RL, Wen DR, Wong JH, et al. Technical details
evaluation of its impact on patient recovery and daily activi- of intraoperative lymphatic mapping for early stage mela-
ties. With the integration of SLN mapping, the most frequent noma. Arch Surg. 1992;127(4):392–9.
and disabling complications, such as lymphedema of the lower 16. Morton RL, Tran A, Vessey J, et al. Quality of life fol-
limbs, have decreased signifcantly, allowing an improvement lowing sentinel node biopsy for primary cutaneous mela-
in the functional capacity of patients. Although there are data noma: health economic implications. Ann Surg Onco.
on QoL in survivors of gynecologic cancers, there is very 2017;24(8):2071–9.
limited information regarding assessment of QoL specifc 17. Cormier JN, Askew RL, Mungovan KS, et al. Lymphedema
beyond breast cancer: a systematic review and meta-
to patients who have undergone SLN mapping alone. Future
analysis of cancer-related secondary lymphedema. Cancer.
studies should aim to prospectively evaluate QoL and PROs in
2010;116(22):5138–49.
patients undergoing SLN mapping.
18. Dunberger G, Lindquist H, Waldenström A, et al. Lower
limb lymphedema in gynecological cancer survivors –
effect on daily life functioning. Support Care Cancer.
REFERENCES 2013;21(11):3063–70.
1. Beesley VL, Eakin EG, Janda M, et al. Gynecological can- 19. Wenzel L. Patient-reported outcomes in sentinel lymph
cer survivors health behaviors and their associations with node procedure versus inguinofemoral lymphadenectomy –
quality of life. Cancer Causes Control. 2008;19(7):775–82. what is the next step? Gynecol Oncol. 2009;113(3):299–300.
2. Antaki C, Rapley M. Quality of life talk: the liberal paradox 20. Berger J, Scott E, Sukumvanich P, et al. The effect of groin
of psychological testing. Discourse Soc. 1996;7(3):293–316. treatment modality and sequence on clinically signifcant
3. Rapley M. Quality of Life Research: A Critical Introduction. chronic lymphedema in patients with vulvar carcinoma. Int
London: Springer; 2004. pp. 1021–1024. J Gynecol Cancer. 2015;25(1):119–24.
4. Mooney A. Quality of life: questionnaires and questions. J 21. Van der Zee AG, Oonk MH, De Hullu JA, et al. Sentinel
Health Commun. 2006;11(3):327–41. node dissection is safe in the treatment of early stage vulvar
5. Aaronson NK, Bullinger M, Ahmedzai S. A modular cancer. J Clin Oncol. 2008;26(6):884–9.
approach to quality-of-life assessment in cancer clinical tri- 22. Oonk MH, Van Os MA, De Bock GH, et al. A compari-
als. Recent Results Cancer Res. 1988;111:231–49. son of quality of life between vulvar cancer patients after
116 Clinical Lymphatic Mapping

sentinel lymph node procedure only and inguinofemoral 32. Park SY, Bae DS, Nam JH, et al. Quality of life and
lymphadenectomy. Gynecol Oncol. 2009;113(3):301–5. sexual problems in disease-free survivors of cervical
23. Farrell R, Gebski V, Hacker NF. Quality of life after com- cancer compared with the general population. Cancer.
plete lymphadenectomy for vulvar cancer: do women pre- 2007;110(12):2716–25.
fer sentinel lymph node biopsy? Int J Gynecol Cancer. 33. Cibula D, Dusek J, Jarkovsky J, et al. A prospective mul-
2014;24(4):813–19. ticenter trial on sentinel lymph node biopsy in patients
24. Forner DM, Dakhil R, Lampe B. Quality of life and sexual with early-stage cervical cancer (SENTIX). Int J Gynecol
function after surgery in early stage vulvar cancer. Eur J Cancer. 2019;29(1):212–15.
Surg Oncol. 2015;41(1):40–5. 34. Lecuru FR, McCormack M, Hillemanns P, at el.
25. Daraï E, Dubernard G, Bats AS, et al. Sentinel node biopsy SENTICOL III: an international validation study of sen-
for the management of early stage endometrial cancer: long- tinel node biopsy in early cervical cancer. A GINECO,
term results of the SENTI-ENDO study. Gynecol Oncol. ENGOT, GCIG and multicenter study. Int J Gynecol
2015;136(1):54–9. Cancer. 2019;29(4):829–34.
26. Rossi EC, Kowalski LD, Scalici J, et al. A comparison of 35. U.S. Food and Drug Administration. Guidance for Industry.
sentinel lymph node biopsy to lymphadenectomy for endo- Patient reported outcome measures: use in medical product
metrial cancer staging (FIRES trial): a multicentre, pro- development to support labeling claims. Federal Register.
spective, cohort study. Lancet Oncol. 2017;18(3):384–92. 2009;74:65132–3.
27. Geppert B, Lönnerfors C, Bollino M, et al. Sentinel lymph 36. National Quality Forum. Patient-Reported Outcomes [2012
node biopsy in endometrial cancer-feasibility, safety and lym- Dec 10]. Available from: www.qualityforum.org/Projects/
phatic complications. Gynecol Oncol. 2018;148(3):491–8. n-r/ Patient-Repor ted_Outcomes/ Patient-Repor ted _
28. Rowlands IJ, Beesley VL, Janda M. Quality of life of Outcomes.aspx.
women with lower limb swelling or lymphedema 3–5 37. Snyder CF, Jensen RE, Segal JB, et al. Patient-reported out-
years following endometrial cancer. Gynecol Oncol. comes (PROs). Med Care. 2013;51:S73–9.
2014;133(2):314–18. 38. Ahmed S, Berzon RA, Revicki DA, et al. The use of patient-
29. Watson CH, Lopez-Acevedo M, Broadwater G, et al. A reported outcomes (PRO) within comparative effectiveness
pilot study of lower extremity lymphedema, lower extrem- research: implications for clinical practice and health care
ity function, and quality of life in women after minimally policy. Med Care. 2012;50(12):1060–70.
invasive endometrial cancer staging surgery. Gynecol 39. Gressel GM, Dioun SM, Richley M, et al. Utilizing the
Oncol. 2019;153(2):399–404. Patient Reported Outcomes Measurement Information
30. Leitao MM Jr, Gomez-Hidalgo NR, Abu-Rustum NR, et System (PROMIS®) to increase referral to ancillary sup-
al. Patient-reported outcomes after surgery for endometrial port services for severely symptomatic patients with gyne-
carcinoma: prevalence of lower-extremity lymphedema cologic cancer. Gynecol Oncol. 2019;152(3):509–13.
after sentinel lymph node mapping versus lymphadenec- 40. Meyer LA, Nick AM, Shi Q, et al. Perioperative trajectory
tomy. Gynecol Oncol. 2020;156(1):147–53. of patient reported symptoms: a pilot study in gynecologic
31. Lécuru F, Mathevet P, Querleu D, et al. Bilateral nega- oncology patients. Gynecol Oncol. 2015;136(3):440–5.
tive sentinel nodes accurately predict absence of lymph 41. Shisler R, Sinnott JA, Wang V, et al. Life after endometrial
node metastasis in early cervical cancer: results of the cancer: a systematic review of patient-reported outcomes.
SENTICOL study. J Clin Oncol. 2011;29(13):1686–91. Gynecol Oncol. 2018;148(2):403–13.
15
Radiation oncology considerations

Gwendolyn Joyce McGinnis and Anuja Jhingran

CONTENTS
Introduction .................................................................................................................................................................................. 117
Radiation therapy targets ............................................................................................................................................................. 117
Nodal target delineation ..................................................................................................................................................... 117
Pelvic nodes........................................................................................................................................................................ 117
Paraaortic nodes ................................................................................................................................................................. 118
Inguinal nodes .................................................................................................................................................................... 118
Radiation therapy techniques....................................................................................................................................................... 118
3D conformal radiation therapy and intensity-modulated radiation therapy ..................................................................... 118
Image-guided radiotherapy ................................................................................................................................................ 120
Literature review by disease site .................................................................................................................................................. 120
Vulva .................................................................................................................................................................................. 120
Endometrium...................................................................................................................................................................... 121
Early stage ............................................................................................................................................................. 121
Advanced stage ...................................................................................................................................................... 123
Conclusion ................................................................................................................................................................................... 124
References.................................................................................................................................................................................... 124

Introduction endometrial, and cervical cancers. We will also discuss the


data available to inform decision making regarding adjuvant
Radiation therapy has been used to treat cancer for over a cen- radiation for these disease sites. While adjuvant radiation fol-
tury. In recent years, radiation has become a more sophisti- lowing sentinel lymph node biopsy is important to understand
cated tool through technological advancements that allow for in the context of breast cancer, it is outside the scope for this
improved effectiveness and minimized side effects. Radiation chapter. When considering the use of radiation therapy in the
therapy is employed in the treatment of approximately 60% of treatment of gynecologic cancer patients after surgical inter-
cancer patients1 – often to complement surgical or systemic vention, it is frst important to understand the most common
approaches to minimize the chance of microscopic residual techniques used to plan and deliver radiation therapy for these
disease. As surgical approaches become more nuanced, so disease sites.
must our understanding of how to best incorporate radiation
therapy to maximize oncologic outcomes while minimizing
short- and long-term toxicity from treatment.
Radiation therapy is often employed in the adjuvant setting Radiation therapy targets
with the goal of eradicating microscopic disease. For this rea- Nodal target delineation
son, increased utilization of sentinel lymph node biopsy will
introduce new adjuvant treatment decisions in the manage- Deciding on target volumes of interest when planning radia-
ment of many cancers. In some disease sites, like the breast tion treatment is often a nuanced decision. Regional lymph
or vulva, there are relatively robust data to support the role of nodes are often included as targets of interest when treating
adjuvant radiation, depending on fndings at the time of sen- gynecologic cancers with radiation therapy. Which lymph
tinel lymph node biopsy. However, in other cases, like uterine node basins to include depends on the primary site and likeli-
or cervical cancer, there are less high-level data on which to hood of regional spread in the context of individual patients.
base treatment recommendations and, instead, expert opinion
prevails. Pelvic nodes
In this chapter, we will offer a brief overview of modern
external beam radiation techniques and targets relevant to Pelvic nodes, including obturator, internal iliac, external iliac,
patients undergoing postoperative radiation after sentinel presacral, and common iliac basins, are commonly included in
lymph node biopsy for gynecologic cancers, including vulvar, radiation felds, given the propensity for gynecologic cancers

DOI: 10.1201/9781003255536-15 117


118 Clinical Lymphatic Mapping

to spread to lymph nodes in these regions. Radiation felds are


designed to encompass all sites of primary drainage, including Radiation therapy techniques
lymph nodes found at the junction between the external iliac,
internal iliac, and common iliac vessels to the level of the com- 3D conformal radiation therapy and
mon iliac bifurcations. Presacral nodes are usually covered in intensity-modulated radiation therapy
patients with cervical cancer and upper vaginal involvement Conformal radiation therapy refers to radiotherapy treat-
but are not routinely covered in patients with lower vaginal ment designed to deliver high doses to areas of interest while
cancer, vulvar cancer unless the apex of the vagina is involved, minimizing doses to normal structures. Within the category
or endometrial cancer unless there is cervical stromal involve- of conformal techniques, both 3D conformal radiation ther-
ment. Perirectal lymph nodes are included for posterior apy (3D-CRT) and intensity-modulated radiation therapy
involvement, including the rectovaginal septum, cul-de-sac, (IMRT) have been used in the treatment of gynecologic can-
or rectum. Typical pelvic felds have a superior border at the cers. 3D-CRT became a standard for treating pelvic felds in
level of the aortic bifurcation, or approximately at the L4/L5 gynecologic cancers by utilizing four beams (anterior, poste-
interspace. rior, right lateral, and left lateral) to create a four-feld ‘box’ to
cover volumes at risk within the pelvis. While this technique
Paraaortic nodes adequately covers the pelvic targets as described earlier, it fre-
quently includes a substantial volume of bowel, which contrib-
Paraaortic nodes are often treated in cases of uterine fundus, utes to acute and chronic diarrhea seen with radiation therapy.
fallopian tube, or ovarian involvement. They are also included In IMRT, the dose is shaped around target structures, with
if there are any grossly involved paraaortic lymph nodes. The multiple converging beams or arcs utilizing dose-calculation
term ‘extended felds’ is used when paraaortic lymph nodes algorithms. Precise delineation of volumes at risk and consid-
are included. Radiation felds are extended approximately to eration of internal organ motion within the pelvis are critical.
the level of T12, or 1.5–2 cm above the level of the most supe- IMRT allows for a signifcantly more nuanced treatment plan
rior grossly involved lymph node. Lower paraaortic nodes are design, which has repeatedly demonstrated improved dosim-
covered prophylactically when there is positive common iliac etry2–5 as well as decreased patient-reported toxicity.6,7 RTOG
nodes up to the level of L2. 1203 was a randomized trial of 278 women with cervical or
endometrial cancer undergoing either standard pelvic radia-
Inguinal nodes tion or IMRT.8 Pelvic IMRT was found to be associated with
much lower rates of gastrointestinal (GI) and urinary toxicity.
Inguinal lymph nodes are routinely treated in cases of vulvar Figures 15.1 and 15.2 show two different plans for a patient
cancer. They are usually included in the treatment of cervi- with vulvar cancer comparing the 3D conformal technique
cal, vaginal, or endometrial cancer when the tumor involves to IMRT. Figures 15.3 and 15.4 show typical IMRT felds for
the distal one-third of the vagina or there are grossly involved patients treated with pelvic felds, as well as patients treated
inguinal lymph nodes. with paraaortic felds.

FIGURE 15.1 A volumetric modulated arc (VMAT) IMRT plan prescribed to 45 Gy (blue isodose line) with a 50 Gy simultaneous integrated boost
(SIB; red isodose line) (a) and a four-feld 3D CRT plan prescribed to 50 Gy with hotspots blocked (b) that could be used to treat a patient with vulvar
cancer. Target volumes include primary (flled red), nodal clinical target volume (flled pink), and nodal planning target volume (flled aquamarine).
Radiation oncology considerations 119

FIGURE 15.2 A comparison of the dose volume histogram (DVH) for IMRT (solid lines) as compared to 3D-CRT (dashed lines). Note lower doses
delivered to bladder, rectum, sigmoid, bowel, and femoral heads without sacrifcing dose to target volumes.

FIGURE 15.3 An example of the target volumes and dose delivered for a postoperative patient prescribed to 45 Gy (red isodose line) to the pelvis.
Target contours include vaginal internal target volume (flled maroon) and nodal clinical target volume (flled blue).
120 Clinical Lymphatic Mapping

FIGURE 15.4 An example of the target volumes and dose delivered for a postoperative patient dispositioned to extended feld coverage. Dose is
prescribed to 45 Gy (blue isodose line) to the pelvis and extended nodal regions with a simultaneous integrated boost to the involved lymph node (flled
pink) to 50 Gy (red isodose line). In addition to the grossly involved lymph node, target contours include vaginal internal target volume (flled red) and
nodal clinical target volume (flled yellow).

Image-guided radiotherapy resulted in signifcantly lower late grade 2 or higher bowel tox-
icity (2.0% versus 8.7%), likely driven by signifcantly lower
An essential element of high-quality radiation treatment is doses delivered to the bowel.
accurate patient positioning and treatment delivery. A num-
ber of factors introduce uncertainty in this process, includ-
ing imprecise patient positioning, inherent organ motion, and
technical variation. In order to compensate for this uncertainty Literature review by disease site
inherent in the patient setup and treatment process, radiation
Vulva
oncologists use an additional margin when designing target
volumes in excess of the volumes of clinical interest. However, Lymph node status is the most important predictor for overall
the use of real-time image guidance can allow for the use of survival in patients with vulvar cancer.10–12 Thorough lymph
smaller margins, as the treating radiation oncologist can be node evaluation is necessary for patient selection for radia-
certain of the position of the treatment target and organs at risk tion therapy. Sentinel node biopsy in addition to radical local
at the time of treatment. Image guidance may include x-rays excision has become the standard of care in the treatment
aligning to bone or cone beam computed tomography (CT) of early-stage vulvar cancer.13 The sentinel node has gained
scans aligning to soft tissue, or even just looking at positions popularity because it has been shown to have low groin
of target organs or a combination of both. recurrence and low morbidity and prevents overtreatment.14
The use of image-guided IMRT to patients receiving adju- Adjuvant radiation therapy may play a role in the manage-
vant cervical radiation was investigated in the PARCER trial, ment of patients with vulvar cancer in several scenarios,
and preliminary results have been presented in abstract form.9 including prophylactic irradiation and management of micro-
This study of 300 patients found that image-guided IMRT metastatic disease.
Radiation oncology considerations 121

Patient selection for defnitive radiation treatment to the is treatment-related morbidity. Chemoradiation will be 48–50
groin in place of groin lymphadenectomy remains an area of Gy to the groin clinical target volume (CTV) with simultane-
controversy.15 Past efforts have aimed to investigate differ- ously integrated boost (SIB) to 56 Gy delivered with weekly
ences between groin dissection versus groin radiation. GOG cisplatin. The outcome of this study will be very useful to see if
37 found that radiation after radical vulvectomy and inguinal radiation therapy can replace lymph node dissection in patients
lymphadenectomy signifcantly reduced local relapses and with micrometatasis. Figure 15.5 shows a CT scan of a patient
cancer-related deaths.16,17 Retrospective data suggested that who had sentinel lymph node biopsy that was negative; how-
inguinofemoral irradiation could be a viable alternative to ever, a node was present on the preoperative scan that was not
lymphadenectomy for squamous cell carcinoma of the vulva. a sentinel node and was not removed and later was found to be
18 However, GOG 88 randomized patients undergoing radical positive—one of the major concerns about just using sentinel
vulvectomy for T1–T3/N0–N1 (FIGO, 1971) vulvar cancer to node data. Postoperative scans should usually be done to make
bilateral groin dissection or bilateral groin radiation therapy.19 sure that all suspicious nodes are removed, even with SLND.
The study was closed early after interim analysis revealed
signifcantly higher rates of groin failure in patients receiving
Endometrium
radiation (18.5% versus 0%).19 However, subsequent analyses
have demonstrated that the groin radiation felds prescribed to Surgical staging is a cornerstone in the management of endo-
3 cm did not adequately treat groin lymph nodes, which can metrial cancer and often historically included systematic
range in depth from 2 to 18.5 cm.20 It is also notable that 20% lymphadenectomy. Much of the beneft of lymph node dis-
of patients in the surgical arm were found to have involved section appears to come from surgical staging rather than
groin nodes and subsequently received adjuvant groin and pel- therapeutic effect.27,28 It is known that metastatic lymph node
vic radiation. Subsequent retrospective data suggest that pro- involvement is a signifcant prognostic factor.29,30 Detection of
phylactic inguinofemoral irradiation may be an alternative to lymph node involvement often substantially changes adjuvant
primary lymphadenectomy with similar oncologic outcomes.21 treatment recommendations, including the utilization of post-
The data for the role of adjuvant radiation following the operative radiation therapy. This is especially important for
discovery of metastatic groin nodes are more robust. It has patients with grade 2–3 endometrioid endometrial, clear cell,
been established that patients with increased nodal burden or papillary serous tumors. However, the optimal management
have increased risk of cancer recurrence. Patients with more of patients with grade 1 endometrioid endometrial cancer is a
than two involved lymph nodes have signifcantly higher risk subject up for greater debate. In recent years, sentinel lymph
of recurrence, and overall survival declines with each positive node biopsy has become a favored alternative to lymph node
node.18 The impact of lymph node involvement differs signif- dissection.31
cantly by adjuvant treatment. In patients without adjuvant radi- Current guidelines provide a framework for conceptualiz-
ation, the number of involved lymph nodes has been highly ing adjuvant treatment of endometrial cancer, including the
correlated with prognosis; however, this difference is dimin- infuence of lymph node involvement.32–34 Women with low-
ished in the setting of adjuvant radiation.22 AGO-CaRE-1 was risk disease defned as surgical specimens with no or mini-
a multicenter retrospective cohort study that showed a beneft mal grade 1–2 residual disease (<50% myometrial invasion)
in progression-free survival (PFS) and a trend in overall sur- without additional risk factors (age over 60 or lymphovascu-
vival (OS) with adjuvant radiation in node-positive patients. lar space involvement [LVSI]). Women who are found to have
This beneft was seen regardless of the number of positive grade 1–2 disease with more substantial myometrial invasion
nodes, grade, or depth of invasion.23 There was a subgroup (>50%) or women with less invasive (<50% myometrial inva-
analysis of patients undergoing sentinel lymph node dissection sion) but high-grade disease are typically recommended for
(SLND).24 vaginal brachytherapy. Pelvic radiation is typically reserved
At present, there are no established guidelines for the man- for women with high-grade disease with >50% myometrial
agement of SLN micrometastasis (≤2 mm deposit) or isolated invasion, deep cervical stromal invasion, or positive nodes.
tumor cells (ITCs) (≤0.2 mm). In the GROINSS-V study, the Lymph node involvement is a relatively common fnding
rate of positive non-SLNs was found to be low in patients with upon surgical staging, though the involvement is commonly
ITCs (4.2%) and in patients with micrometastasis (11.1%); low volume (qualifying as either ITCs or micrometastatic dis-
however, the authors concluded that no cutoff size was found ease).35,36 Many patients who have low-volume lymph node
below which the chance of non-SLN metastasis was close to involvement have otherwise favorable disease.36 Therefore,
zero, emphasizing the role of adjuvant treatment.25 Current adjuvant treatment recommendations after discovery of low-
National Cancer Comprehensive Network (NCCN) guidelines volume lymph node involvement can require nuanced decision
recommend adjuvant radiation with or without concurrent che- making. Likely, there is no role for additional surgery after
motherapy for a single positive sentinel lymph node measuring positive sentinel lymph node biopsy. Rather, some combina-
2 mm or less.26 tion of radiation and chemotherapy should be utilized.
GROINSS-VIII is a prospective phase II trial designed
to test the effcacy and safety of adjuvant chemoradiation in
Early stage
patients with a groin metastasis over 2 mm in a sentinel node
in patients with unifocal, squamous cell carcinomas of the Multiple large trials, including PORTEC 137 and GOG 9938,
vulva with a primary tumor less than 4 cm. The primary out- seem to suggest that adjuvant radiation does not improve
come is the groin recurrence rate, and the secondary outcome overall survival in early-stage, low-risk disease. However,
122 Clinical Lymphatic Mapping

FIGURE 15.5 An example of an inguinal node recurrence after partial radical vulvectomy and negative sentinel node biopsies. The patient was a
70-year-old woman who underwent initial radical partial vulvectomy with sentinel lymph node biopsy. Bilateral sentinel lymph nodes were mapped
and three additional groin nodes were dissected; all were negative for metastatic disease. However, a left groin lymph node, which measured 1.5 × 0.8
cm on initial scan (a) and subsequently enlarged to 1.5 × 1.2 cm (b), was not removed. The lymph node was appreciated on postoperative restaging
imaging (c and d) at 5 months, and fne needle aspiration (FNA) confrmed metastatic squamous cell carcinoma. The patient went on to undergo salvage
left inguinal lymphadenectomy showing 5 mm focus of squamous cell carcinoma, followed by adjuvant external beam radiation therapy (EBRT) to the
left inguinal operative bed and distal left pelvic lymph nodes. She had no evidence of disease and minimal toxicity at 5 years after initial diagnosis.
(From Eiffel P, Klopp AH, Gynecologic Radiation Oncology: A Practical Guide, Wolters Kluwer Health, Inc., 2016, with permission.)

in patients with high risk disease or those with metastatic of adjuvant treatment. A total of 31 patients with ITCs alone
involvement of the lymph nodes, data show a beneft to adju- were included. Eleven (35%) patients with ITCs received
vant therapy. adjuvant chemotherapy ± whole pelvic radiation therapy
A large retrospective review of patients who underwent (WPRT), 10 (32%) received WPRT, and 10 (32%) received
sentinel node biopsy showed patients with low-volume lymph either no adjuvant treatment or vault brachytherapy (VBT)
node disease frequently received adjuvant treatment and had only. At 3 years, PFS in ITC patients (95.5%) was similar to
improved oncologic outcomes compared to patients with mac- node-negative (87.6%) and micrometastatic (85.5%) disease,
rometastatic lymph node disease.39 In this study, 844 patients but signifcantly better than patients with macrometastases
were included, 44 of whom had low-volume metastatic lymph (58.5%). Only 1 of the 31 patients with ITCs had a recurrence
node disease (LVM; defned as ITCs or micrometastatic dis- (FIGO stage IB, carcinosarcoma histology) despite adjuvant
ease) and 47 had macrometastatic lymph node involvement treatments.
(MM); 753 patients were lymph node negative. In terms of Another multi-institutional retrospective study of 175
adjuvant therapy, 82% of patients with LVM, 89% of patients patients with early-stage endometrial cancer (9% had stage IA,
with MM, and 14% of patients without lymph node involve- 39% stage IB, and 12% stage II disease) found to have ITCs
ment received adjuvant therapy. Recurrence-free survival was on SLNB confrmed similar fndings.40 Completion lymphad-
90% for N0, 86% for N0(i+), 86% for N1(mic), and 71% for enectomy was performed in 49% of patients. Many patients
N1+ patients. (43%) received either no adjuvant treatment or VBT alone,
It appears that the risk of recurrence with ITCs alone is low, 12% had EBRT, and 45% had chemotherapy with or without
regardless of adjuvant therapy. In a retrospective review of radiation therapy. Nine recurrences were noted (fve distant,
519 patients undergoing surgical management for endometrial three retroperitoneal, one vaginal). Chemotherapy, external
cancer, including sentinel lymph node mapping, outcomes beam radiation, and type of lymph node dissection were not
for patients with ITCs alone appeared excellent regardless associated with recurrence-free survival.
Radiation oncology considerations 123

Among higher-risk patients, another retrospective review undergoing a primary surgical approach if pathologic fnd-
found a higher risk of recurrence with any lymph node dis- ings indicate a high risk of disease recurrence. Many of these
ease, including ITC or micrometastatic disease.41 A total of factors focus on the nature of the primary disease, including
61 patients were included, with 9 patients with LVM and 52 clinical tumor size, LVSI, and depth of tumor invasion into the
without lymph node disease. Presence of ITC/MM was found cervical stroma.51,52 However, lymph node involvement is also
to be an independent risk factor for recurrence, and 8-year OS a strong prognostic indicator in cervical cancer and therefore
and RFS were lower in the ITC/MM group when compared to an item of importance when considering adjuvant treatment
the N0 group. recommendations.53,54 OS has repeatedly been found to be sig-
nifcantly shorter in patients with lymph node involvement.55
According to current guidelines by the NCCN, full pelvic
Advanced stage
lymphadenectomy remains the standard of care for early-stage
Patients with advanced disease are at a higher risk of local cervical cancer patients except for those with IA1 disease with-
recurrence42 and the use of adjuvant radiation therapy is more out LVSI.56 However, sentinel lymph node biopsy is becom-
established.43,44 Patients are also at increased risk of distant ing increasingly popular and is the recommended approach
failure, which is why chemotherapy is a mainstay of adjuvant for pelvic lymph node staging by the European Society of
treatment. Gynecological Oncology.57,58 Patients may beneft from a more
Several studies have compared radiation alone versus che- minimal surgical approach, given morbidity after pelvic and/
motherapy. GOG 122 demonstrated that OS was improved in or paraaortic lymphadenectomy.59–61 Toxicity after lymphad-
patients receiving chemotherapy versus whole-abdomen radia- enectomy is further increased in the setting of adjuvant radia-
tion therapy.45 Another randomized phase III trial of advanced tion.62 Sentinel lymph node biopsy appears to be a reasonable
endometrial cancer comparing adjuvant chemotherapy versus alternative, with high-quality data showing bilateral detection
pelvic radiation therapy showed PFS and OS were higher with rates in more than 90% of patients, even with small cervical
chemotherapy versus pelvic radiation.46 However, in Maggi cancers.63,64
et al., there was no signifcant difference in survival between Although sentinel lymph node biopsy is becoming a more
high-risk endometrial cancer patients treated with chemother- popular approach in early-stage cervical cancer, there are lim-
apy as compared to pelvic radiation.47 ited data to guide optimal adjuvant therapy based on the patho-
Subsequent trials examined the combination of chemother- logical fndings in sentinel lymph nodes. Nevertheless, these
apy and radiation. PORTEC 3 was a randomized trial between fndings are currently being used to shape treatment decisions.
radiation alone versus chemoradiation and showed no differ- In a 2017 survey of the European Society of Gynaecological
ence in OS with the addition of chemotherapy, but did show Oncology, macrometastases, micrometastases, and ITCs in
an improvement in RFS (75% vs. 68%), especially in stage lymph nodes were considered indications for adjuvant treat-
III patients.48 The NSGO/EORTC/MaNGO trial compared ment by 96%, 93%, and 68% of respondents, respectively.57
sequential chemotherapy with external beam radiation versus Currently, postoperative chemoradiation therapy is recom-
radiation alone and found an improvement in PFS.49 In GOG mended for all patients at high risk for recurrence.65,66 Patients
258, 707 patients with stage III or IVA endometrial carcinoma are considered high risk if surgical pathology demonstrates
were randomized to receive either chemoradiotherapy or che- positive margins, positive pelvic or paraaortic lymph nodes,
motherapy alone. Patients with stage I or II clear-cell or serous and positive parametrium.67,68
endometrial carcinoma and positive peritoneal washings were Data for adjuvant management of early-stage cervical can-
also included. Patients underwent hysterectomy and bilateral cer patients at high risk for recurrence include SWOG 8797/
salpingo-oophorectomy with optional pelvic and paraaortic RTOG 9112/GOG 109. This was a prospective randomized
lymph node dissection. Patients were randomized to either trial that investigated the role of adding concurrent chemo-
cisplatin plus EBRT followed by carboplatin/paclitaxel or car- therapy to adjuvant pelvic radiation in women with FIGO
boplatin/paclitaxel only. EBRT was delivered to the pelvis, stage IA2, IB, or IIA cervical cancer at high risk of recur-
with or without paraaortic felds, with a planned total dose of rence after radical hysterectomy and pelvic lymph node dis-
45 Gy in 25 fractions. IMRT and VBT were allowed only in section based on pathological features. A total of 127 patients
the chemoradiation group. Recurrence-free survival at 5 years with pelvic nodal metastases, positive parametrial invasion, or
was almost identical: 59% in the chemoradiation group versus positive surgical margins were randomized to radiation versus
58% in radiation alone. There were no signifcant subgroup radiation with the addition of chemotherapy, delivered every
differences. Chemoradiation was associated with a lower rate 3 weeks as four cycles of cisplatin (CDDP) at 70 mg/m2 intra-
of vaginal recurrences at 5 years (2% vs. 7%) and paraaortic venous (IV) infusion and 5-fuorouracil (5-FU) at 4000 mg/
node recurrence at 5 years (11% vs. 20%). However, distant m2 over a 4-day continuous infusion beginning on day 1 of
recurrence was more common in the chemoradiation group radiation. Postoperative radiation was delivered as 49.3 Gy
(27% vs. 21%). In quality-of-life analysis, chemoradiation in 1.7-Gy fractions to at least the whole pelvis, with elective
demonstrated poorer outcomes, though this did not exceed the paraaortic extended nodal feld irradiation to 45 Gy in 30 frac-
‘clinically signifcant’ threshold.50 tions if common iliac nodes were found to be involved. On
long-term update, both PFS and OS were improved with the
Cervix addition of chemotherapy.69
Surgical management is the mainstay of treatment of early- Another study of high-risk patients was a retrospective
stage cervical cancer. Adjuvant therapy is indicated for patients review of patients with FIGO stage IB–IIB cervical cancer
124 Clinical Lymphatic Mapping

with at least two positive pelvic lymph nodes and/or metastatic


common iliac nodes who received adjuvant chemotherapy in Conclusion
addition to postoperative radiation and showed consistent fnd-
ings.70 Of the 25 patients included, 64% were disease free at Radiation therapy plays an important role in the treatment of
30 months from treatment and no paraaortic node recurrences gynecologic cancers, both as a defnitive treatment and as an
were observed. adjuvant to surgery. Continuous improvements in radiation
Finally, a retrospective review examined adjuvant che- planning and techniques coupled with surgical strategies like
motherapy alone for patients with FIGO stage IB and IIA sentinel node biopsy appear to result in better local tumor con-
cervical cancer with features increasing risk of recurrence, trol and reduced acute and long-term toxicity.
including deep cervical stromal invasion (> 50%; DCSI), posi- The integration of sentinel node biopsy into the routine care
tive surgical margin, parametrial involvement, and/or pelvic of women with gynecologic cancer raises new questions for
nodal metastases following radical hysterectomy and pel- study. Do all micrometastases and ITCs require adjuvant radi-
vic lymphadenectomy. A total of 65 patients were included. ation therapy? Should patients with regional macrometastases
Chemotherapy consisted of bleomycin (5 mg IV continuous undergo lymphadenectomy prior to regional radiation therapy?
infusion for 7 days), vincristine (0.7 mg/m2 IV bolus on day When should radiation felds be extended? What is the role for
7), mitomycin C (7 mg/m2 IV bolus on day 7), and CDDP (10 new strategies like targeted therapy and immunotherapy with
mg IV continuous infusion for 7 days). Patients with DCSI as existing combinations? Radiation oncologists have decades-
their only risk factor received only four cycles of chemother- long experience with the treatment of women with gyneco-
apy, while others received fve cycles. Results showed 5-year logic cancers and must remain as fully engaged members of
disease-free survival of 100% for patients with squamous cell multidisciplinary teams in the continuous effort to develop
cancers (SCCs) with DCSI alone, 71.4% for adenosquamous safer and more effective combinations.
histology and DCSI alone, 89.3% for SCC and at least one
other risk factor, and 71.4% for adenosquamous histology with
at least one other risk factor.71 A phase III trial is undergo- REFERENCES
ing evaluating the advantage of additional chemotherapy to 1. Slotman BJ, Cottier B, Bentzen SM, et al. Overview of
chemoradiation for these high-risk patients, and the results national guidelines for infrastructure and staffng of radio-
should be ready by 2022. therapy. ESTRO-QUARTS: work package 1. Radiother
Patients with intermediate risk factors for recurrence, Oncol. 2005;75(3):349–54.
including large tumor size, DCSI, and LVSI, also beneft 2. Luo H-C, Lin G-S, Liao S-G, et al. Cervical cancer treated
from adjuvant therapy. This was demonstrated in GOG 92, a with reduced-volume intensity-modulated radiation therapy
phase III randomized trial of 277 patients with FIGO stage IB base on Sedlis criteria (NCCN VS RTOG). Br J Radiol.
cervical cancer with the risk factors previously mentioned.52,72 2018;91(1081):20170398.
Patients were randomized to postoperative radiation therapy 3. Du X-L, Tao J, Sheng X-G, et al. Intensity-modulated radia-
versus no further treatment. Radiation was delivered as tion therapy for advanced cervical cancer: a comparison of
46–50.4 Gy at 1.8–2.0 Gy per fraction over 4.5–6 weeks to dosimetric and clinical outcomes with conventional radio-
the pelvis. Results showed that local (13.9% vs. 20.7%) and therapy. Gynecol Oncol. 2012;125(1):151–7.
distant (2.9% vs. 8.6%) relapse rates were lower in the radia- 4. Gandhi AK, Sharma DN, Rath GK, et al. Early clinical out-
tion therapy arm. On long-term follow-up, the radiation arm comes and toxicity of intensity modulated versus conven-
demonstrated longer PFS; however, no difference was seen in tional pelvic radiation therapy for locally advanced cervix
carcinoma: a prospective randomized study. Int J Radiat
OS. After controlling for prognostic factors, patients in the
Oncol Biol Phys. 2013;87(3):542–8.
radiation therapy arm were 44% less likely to fail. This fnd-
5. Naik A, Gurjar OP, Gupta KL, et al. Comparison of dosi-
ing was especially dramatic in patients with nonsquamous
metric parameters and acute toxicity of intensity-modulated
histologies (8.8% vs. 44% experiencing recurrences). A 2012
and three-dimensional radiotherapy in patients with cer-
meta-analysis, which also includes the data from GOG 92,
vix carcinoma: a randomized prospective study. Cancer/
also demonstrated the beneft of radiation for intermediate Radiothérapie. 2016;20(5):370–6.
risk factors.72 6. Mell LK, Sirák I, Wei L, et al. Bone marrow-sparing inten-
Additionally, GOG 0263 is a phase III prospective trial cur- sity modulated radiation therapy with concurrent cisplatin
rently accruing and designed to determine if postoperative for stage IB-IVA cervical cancer: an international multi-
adjuvant chemoradiation can signifcantly improve recurrence- center Phase II Clinical Trial (INTERTECC-2). Int J Radiat
free survival when compared to radiation alone in stage I–IIA Oncol Biol Phys. 2017;97(3):536–45.
cervical cancer patients with intermediate risk factors after 7. Wang X, Shen Y, Zhao Y, et al. Adjuvant intensity-
treatment with radical hysterectomy. However, the bigger modulated radiotherapy (IMRT) with concurrent paclitaxel
question is that with no beneft in OS with adjuvant radiation and cisplatin in cervical cancer patients with high risk fac-
therapy, whether there is a need anyhow for adjuvant therapy tors: a phase II trial. Eur J Surg Oncol. 2015;41(8):1082–8.
with modern imaging and surgery. Therefore, a new phase III 8. Klopp AH, Yeung AR, Deshmukh S, et al. Patient-
will start enrolling in 2022 randomizing patients with inter- reported toxicity during pelvic intensity-modulated radia-
mediate risk factors to surgery plus or minus VBT versus sur- tion therapy: NRG oncology – RTOG 1203. J Clin Oncol.
gery plus pelvic radiation therapy. 2018;36(24):2538.
Radiation oncology considerations 125

9. Chopra S, Dora T, Gupta S, et al. Phase III randomized trial 24. Klapdor R, Hillemanns P, Wölber L, et al. Outcome after
of postoperative adjuvant conventional radiation (3DCRT) sentinel lymph node dissection in vulvar cancer: a sub-
versus image guided intensity modulated radiotherapy group analysis of the AGO-CaRE-1 study. Ann Surg Oncol.
(IG-IMRT) in cervical cancer (PARCER): fnal analysis. 2017;24(5):1314–21.
Int J Radiat Oncol Biol Phys. 2020;108(3):S1–S2. 25. Oonk MH, van Hemel BM, Hollema H, et al. Size of
10. Canlorbe G, Rouzier R, Bendifallah S, et al. Impact sur la sentinel-node metastasis and chances of non-sentinel-
survie de la procédure du ganglion sentinelle dans le cancer node involvement and survival in early stage vulvar can-
de la vulve : analyse de la SEER database. Gynécol Obstét cer: results from GROINSS-V, a multicentre observational
Fertil. 2012;40(11):647–51. study. Lancet Oncol. 2010;11(7):646–52.
11. Burger MP, Hollema H, Emanuels AG, et al. The importance 26. NCCN Clinical Practice Guidelines in Oncology: Vulvar
of the groin node status for the survival of T1 and T2 vulval Cancer (Squamous Cell Carcinoma); 2020. Available from:
carcinoma patients. Gynecol Oncol. 1995;57(3):327–34. www.nccn.org/professionals/physician_gls/pdf/vulvar_
12. Farias-Eisner R, Cirisano FD, Grouse D, et al. Conservative blocks.pdf.
and individualized surgery for early squamous carcinoma 27. Kitchener H, Swart AM, Qian Q, et al. Effcacy of sys-
of the vulva: the treatment of choice for stage I and II (T1–2 tematic pelvic lymphadenectomy in endometrial can-
N0–1 M0) disease. Gynecol Oncol. 1994;53(1):55–8. cer (MRC ASTEC trial): a randomised study. Lancet.
13. Covens A, Vella ET, Kennedy EB, et al. Sentinel lymph 2008;373(9658):125–36.
node biopsy in vulvar cancer: systematic review, meta- 28. Panici PB, Basile S, Maneschi F, et al. Systematic pelvic
analysis and guideline recommendations. Gynecol Oncol. lymphadenectomy vs no lymphadenectomy in early-stage
2015;137(2):351–61. endometrial carcinoma: randomized clinical trial. J Natl
14. Van der Zee AGJ, Oonk MH, De Hullu JA, et al. Sentinel Cancer Inst. 2008;100(23):1707–16.
node dissection is safe in the treatment of early-stage 29. DiSaia PJ, Creasman WT, Boronow RC, et al. Risk factors
vulvar cancer. J Clin Oncol Off J Am Soc Clin Oncol. and recurrent patterns in Stage I endometrial cancer. Am J
2008;26(6):8884–9. Obstet Gynecol. 1985;151(8):1009–15.
15. Jolly S, Soni P, Gaffney DK, et al. ACR Appropriateness 30. Barlin JN, Zhou Q, St Clair CM, et al. Classifcation and
Criteria® adjuvant therapy in vulvar cancer. Oncology. regression tree (CART) analysis of endometrial carci-
2015;29(11). Available from: www.cancernetwork.com/ noma: seeing the forest for the trees. Gynecol Oncol.
oncology-journal/acr-appropriateness-criteria-adjuvant- 2013;130(3):452–6.
therapy-vulvar-cancer/page/0/1. 31. Soliman PT, Westin SN, Dioun S, et al. A prospective vali-
16. Homesley HD, Bundy BN, Sedlis A, et al. Radiation therapy dation study of sentinel lymph node mapping for high-risk
versus pelvic node resection for carcinoma of the vulva with endometrial cancer. Gynecol Oncol. 2017;146(2):234–9.
positive groin nodes. Obstet Gynecol. 1986;68(6):733–40. 32. Meyer LA, Bohlke K, Powell MA, et al. Postoperative radi-
17. Kunos C, Simpkins F, Gibbons H, et al. Radiation therapy ation therapy for endometrial cancer: American Society of
compared with pelvic node resection for node-positive vul- Clinical Oncology clinical practice guideline endorsement
var cancer: a randomized controlled trial. Obstet Gynecol. of the American Society for Radiation Oncology evidence-
2009;114(3):537–46. based guideline. J Clin Oncol. 2015;33(26):2908–13.
18. Petereit DG, Mehta MP, Buchler DA, et al. Inguinofemoral 33. Klopp A, Smith BD, Alektiar K, et al. The role of post-
radiation of N0,N1 vulvar cancer may be equivalent to operative radiation therapy for endometrial cancer: execu-
lymphadenectomy if proper radiation technique is used. Int tive summary of an American Society for Radiation
J Radiat Oncol Biol Phys. 1993;27(4):963–7. Oncology evidence-based guideline. Pract Radiat Oncol.
19. Stehman FB, Bundy BN, Thomas G, et al. Groin dissec- 2014;4(3):137–44.
tion versus groin radiation in carcinoma of the vulva: a 34. Jones E, Beriwal S, Beyer D, et al. An analysis of appropriate
Gynecologic Oncology Group study. Int J Radiat Oncol delivery of postoperative radiation therapy for endometrial
Biol Phys. 1992;24(2):389–96. cancer using the RAND/UCLA appropriateness method:
20. Koh WJ, Chiu M, Stelzer KJ, et al. Femoral vessel depth executive summary. Adv Radiat Oncol. 2016;1(1):26–34.
and the implications for groin node radiation. Int J Radiat 35. Plante M, Stanleigh J, Renaud M-C, et al. Isolated tumor
Oncol Biol Phys. 1993;27(4):969–74. cells identifed by sentinel lymph node mapping in endo-
21. Hallak S, Ladi L, Sorbe B. Prophylactic inguinal- metrial cancer: does adjuvant treatment matter? Gynecol
femoral irradiation as an alternative to primary lymphad- Oncol. 2017;146(2):240–6.
enectomy in treatment of vulvar carcinoma. Int J Oncol. 36. Clinton LK, Kondo J, Carney ME, et al. Low-volume lymph
2007;31(5):1077–85. node metastases in endometrial carcinoma. Int J Gynecol
22. Woelber L, Eulenburg C, Choschzick M, et al. Prognostic Cancer. 2017;27(6):1165–70.
role of lymph node metastases in vulvar cancer and impli- 37. Creutzberg CL, van Putten WLJ, Koper PCM, et al. Surgery
cations for adjuvant treatment. Int J Gynecol Cancer. and postoperative radiotherapy versus surgery alone for
2012;22(3):503–8. patients with stage-1 endometrial carcinoma: multicentre
23. Mahner S, Jueckstock J, Hilpert F, et al. Adjuvant ther- randomised trial. Lancet. 2000;355(9213):1404–11.
apy in lymph node – positive vulvar cancer: the AGO- 38. Keys HM, Roberts JA, Brunetto VL, et al. A phase III
CaRE-1 study. J Natl Cancer Inst. 2015;107(3). https://doi. trial of surgery with or without adjunctive external pel-
org/10.1093/jnci/dju426. vic radiation therapy in intermediate risk endometrial
126 Clinical Lymphatic Mapping

adenocarcinoma: a Gynecologic Oncology Group study. a Gynecologic Oncology Group Study. Gynecol Oncol.
Gynecol Oncol. 2004;92(3):744–51. 1999;73(2):177–83.
39. St. Clair CM, Eriksson AGZ, Ducie JA, et al. Low-volume 53. Creasman WT, Kohler MF. Is lymph vascular space
lymph node metastasis discovered during sentinel lymph involvement an independent prognostic factor in early cer-
node mapping for endometrial carcinoma. Ann Surg Oncol. vical cancer? Gynecol Oncol. 2004;92(2):525–9.
2016;23(5):1653–9. 54. Biewenga P, van der Velden J, Mol BWJ, et al. Prognostic
40. Backes FJ, Felix AS, Grégoire J, et al. Sentinel lymph node model for survival in patients with early stage cervical can-
(SLN) isolated tumor cells (ITCs) in otherwise stage I/II cer. Cancer. 2011;117(4):768–76.
endometrioid endometrial cancer: to treat or not to treat? 55. Fuller AF Jr, Elliott N, Kosloff C, et al. Determinants of
Gynecol Oncol. 2018;149:7–8. increased risk for recurrence in patients undergoing radical
41. Todo Y, Kato H, Okamoto K, et al. Isolated tumor cells and hysterectomy for stage IB and IIA carcinoma of the cervix.
micrometastases in regional lymph nodes in stage I to II Gynecol Oncol. 1989;33(1):34–9.
endometrial cancer. J Gynecol Oncol. 2016;27(1):e1. 56. NCCN clinical practice guidelines in Oncology (NCCN
42. Mundt AJ, McBride R, Rotmensch J, et al. Signifcant pel- Guidelines) cervical cancer. National Comprehensive
vic recurrence in high-risk pathologic stage I – IV endome- Cancer Network. Published 2020 [cited 2020 Oct 8].
trial carcinoma patients after adjuvant chemotherapy alone: Available from: www.nccn.org/professionals/physician_
implications for adjuvant radiation therapy. Int J Radiat gls/pdf/cervical_blocks.pdf.
Oncol Biol Phys. 2001;50(5):1145–53. 57. Dostalek L, Åvall-Lundqvist E, Creutzberg CL, et al. ESGO
43. Stewart KD, Martinez AA, Weiner S, et al. Ten-year out- survey on current practice in the management of cervical
come including patterns of failure and toxicity for adjuvant cancer. Int J Gynecol Cancer. 2018;28(6):1226–31.
whole abdominopelvic irradiation in high-risk and poor 58. Cibula D, Pötter R, Planchamp F, et al. The European
histologic feature patients with endometrial carcinoma. Int Society of Gynaecological Oncology/European Society for
J Radiat Oncol Biol Phys. 2002;54(2):527–35. Radiotherapy and Oncology/European Society of pathol-
44. Mundt AJ, Murphy KT, Rotmensch J, et al. Surgery and ogy guidelines for the management of patients with cervical
postoperative radiation therapy in FIGO Stage IIIC cancer. Virchows Arch. 2018;472(6):919–36.
endometrial carcinoma. Int J Radiat Oncol Biol Phys. 59. Beesley V, Janda M, Eakin E, et al. Lymphedema after
2001;50(5):1154–60. gynecological cancer treatment: prevalence, correlates, and
45. Randall ME, Filiaci VL, Muss H, et al. Randomized phase supportive care needs. Cancer. 2007;109(12):2607–14.
III trial of whole-abdominal irradiation versus doxorubicin 60. Mathevet P, Lecuru F. Effect of sentinel lymph-node
and cisplatin chemotherapy in advanced endometrial carci- biopsy alone on the morbidity of the surgical treatment
noma: a Gynecologic Oncology Group Study. J Clin Oncol. of early cervical cancer: results from the prospective
2006;24(1):36–44. randomized study Senticol2. J Clin Orthod. 2015;33
46. Susumu N, Sagae S, Udagawa Y, et al. Randomized phase III (15_suppl):5521.
trial of pelvic radiotherapy versus cisplatin-based combined 61. Niikura H, Okamoto S, Otsuki T, et al. Prospective study of
chemotherapy in patients with intermediate- and high-risk sentinel lymph node biopsy without further pelvic lymph-
endometrial cancer: a Japanese Gynecologic Oncology adenectomy in patients with sentinel lymph node – negative
Group study. Gynecol Oncol. 2008;108(1):226–33. cervical cancer. Int J Gynecol Cancer. 2012;22(7):1244–
47. Maggi R, Lissoni A, Spina F, et al. Adjuvant chemotherapy 50. Published online 2012. https://ijgc.bmj.com/content/
vs radiotherapy in high-risk endometrial carcinoma: results 22/7/1244.abstract.
of a randomised trial. Br J Cancer. 2006;95(3):266–71. 62. Landoni F, Maneo A, Colombo A, et al. Randomised study
48. de Boer SM, Powell ME, Mileshkin L, et al. Adjuvant of radical surgery versus radiotherapy for stage Ib-IIa cervi-
chemoradiotherapy versus radiotherapy alone for women cal cancer. Lancet. 1997;350(9077):535–40.
with high-risk endometrial cancer (PORTEC-3): fnal 63. Cibula D, Kocian R, Plaikner A, et al. Sentinel lymph node
results of an international, open-label, multicentre, ran- mapping and intraoperative assessment in a prospective,
domised, phase 3 trial. Lancet Oncol. 2018;19(3):295–309. international, multicentre, observational trial of patients
49. Hogberg T, Signorelli M, de Oliveira CF, et al. Sequential with cervical cancer: the SENTIX trial. Eur J Cancer.
adjuvant chemotherapy and radiotherapy in endometrial 2020;137:69–80.
cancer – results from two randomised studies. Eur J Cancer. 64. Altgassen C, Hertel H, Brandstädt A, et al. Multicenter
2010;46(13):2422–31. validation study of the sentinel lymph node concept
50. Matei D, Filiaci V, Randall ME, et al. Adjuvant chemother- in cervical cancer: AGO Study Group. J Clin Oncol.
apy plus radiation for locally advanced endometrial cancer. 2008;26(18):2943–51.
N Engl J Med. 2019;380(24):2317–26. 65. Wolfson AH, Varia MA, Moore D, et al. ACR
51. Delgado G, Bundy B, Zaino R, et al. Prospective surgical- Appropriateness Criteria® role of adjuvant therapy in the
pathological study of disease-free interval in patients management of early stage cervical cancer. Gynecol Oncol.
with stage IB squamous cell carcinoma of the cervix: 2012;125(1):256–62.
a Gynecologic Oncology Group study. Gynecol Oncol. 66. Chino J, Annunziata CM, Beriwal S, et al. Radiation
1990;38(3):352–7. therapy for cervical cancer: executive summary of an
52. Sedlis A, Bundy BN, Rotman MZ, et al. A randomized ASTRO clinical practice guideline. Pract Radiat Oncol.
trial of pelvic radiation therapy versus no further therapy 2020;10(4):220–34.
in selected patients with stage IB carcinoma of the cervix 67. Peters WAI, Liu PY, Barrett RJII, et al. Concurrent chemo-
after radical hysterectomy and pelvic lymphadenectomy: therapy and pelvic radiation therapy compared with pelvic
Radiation oncology considerations 127

radiation therapy alone as adjuvant therapy after radical 70. Cheng X, Cai SM, Li ZT, et al. Concurrent chemotherapy
surgery in high-risk early-stage cancer of the cervix. Obstet and adjuvant extended feld irradiation after radical surgery
Gynecol Surv. 2000;55(8):491. for cervical cancer patients with lymph node metastases. Int
68. Rotman M, Sedlis A, Piedmonte MR, et al. A phase III ran- J Gynecol Cancer. 2008;18(4):779–84.
domized trial of postoperative pelvic irradiation in stage IB 71. Takeshima N, Umayahara K, Fujiwara K, et al. Treatment
cervical carcinoma with poor prognostic features: follow- results of adjuvant chemotherapy after radical hysterectomy
up of a gynecologic oncology group study. Int J Radiat for intermediate- and high-risk stage IB – IIA cervical can-
Oncol Biol Phys. 2006;65(1):169–76. cer. Gynecol Oncol. 2006;103(2):618–22.
69. Monk BJ, Wang J, IMS, et al. Rethinking the use of radiation 72. Rogers L, Siu SSN, Luesley D, et al. Radiotherapy and
and chemotherapy after radical hysterectomy: a clinical- chemoradiation after surgery for early cervical cancer.
pathologic analysis of Gynecologic Oncology Group/ Cochrane Database Syst Rev. 2012;5. Available from:
Southwest Oncology Group/Radiation Therapy Oncology www.cochranelibrary.com/cdsr/doi/10.1002/14651858.
Group trial. Gynecol Oncol. 2005;96(3):721–8. CD007583.pub3/abstract.
16
Special considerations

Michael Frumovitz

CONTENTS
Introduction .................................................................................................................................................................................. 129
Vaginal cancer .............................................................................................................................................................................. 129
Ovarian cancer ............................................................................................................................................................................. 130
Vulvar melanoma ......................................................................................................................................................................... 131
References.................................................................................................................................................................................... 133

Introduction lymphatic system of the female reproductive tract, Plentl and


Friedman admit that the lymphatic drainage of the vagina is
As lymphatic mapping and sentinel lymph node biopsy have complex and uncertain and that “unusual variations” are fre-
become part of the standard of care in the treatment of women quently reported.1
with vulvar cancer and are options frequently utilized in the As reliable pathways for lymphatic drainage of the vagina
surgical treatment of cervical and uterine cancers, gyneco- are lacking, lymphatic mapping may be an essential part of
logic oncologists are exploring their utility and validity in therapeutic planning for treating women with vaginal cancer.
other malignancies of the female reproductive tract. Although Typically vaginal cancers located in the upper and middle
the diagnosis is exceedingly rare, women with vaginal can- vagina are treated with radiation therapy. With the variable
cer may beneft from lymphatic mapping even when radiation drainage patterns of the vagina, lymphatic mapping may be
therapy is being considered instead of surgery. For patients very useful in these patients prior to radiation planning to
with clinically early ovarian cancer, lymphatic mapping and assure adequate coverage of potential metastatic sites in drain-
sentinel lymph node biopsy may allow for less extensive sur- ing basins. For women with vaginal cancers in the lower third
gery while still obtaining important prognostic information. or introitus and felt to be surgically resectable, lymphatic
The application of lymphatic mapping and sentinel lymph mapping may help surgeons plan the surgical approach and
nodes will be reviewed in the frst part of this chapter. Finally, whether to sample inguinofemoral or pelvic nodes (or possibly
vulvar melanoma presents an interesting intersection of muco- rectal nodes) as part of their surgery.
sal squamous cancer and cutaneous melanoma. The manage- Multiple case reports have detailed the location of sentinel
ment of sentinel nodes in cutaneous melanoma has evolved nodes after lymphatic mapping of a primary vaginal cancer.
signifcantly over the last decade, and how those lessons may These publications typically report traditional pathways of
be applied to vulvar melanoma will be explored at the end of lymphatic drainage of the vagina, with the upper vagina drain-
this chapter. ing to the pelvis and the lower vagina draining to the groin.2–6
In a study of seven patients by Hertel and colleagues, lymphatic
drainage also seemed to follow the more traditional routes.7
Patients with disease exclusively in the upper or middle third
Vaginal cancer
had drainage to the pelvis, while the one patient with disease
Many clinicians assume that lymphatic drainage from the limited to only the lower third had drainage to the groin. One
upper vagina follows cervical routes into the pelvis, while patient had disease along the entire vagina, and lymphatic
drainage from the lower vagina and introitus follow vulvar mapping revealed sentinel nodes in both the groin and pel-
routes to the inguinofemoral triangle. The middle portion vis. The last patient had disease in the lower and middle third
of the vagina may therefore drain to either basin. These pat- and had lymphatic drainage to the pelvis only with no map-
terns, however, are inconsistent, and lymphatic drainage of ping in the groin. Other prospective studies have confrmed
the vagina may actually follow ventral and dorsal routes more the unpredictable lymphatic pathways draining the vagina. In
commonly than drainage based on the regions of the proximal, the largest study of lymphatic mapping in women with vaginal
mid, and distal vagina (or upper, middle, and lower thirds).1 cancer, Frumovitz et al. identifed at least one sentinel node in
In this ventral/dorsal model, lymphatics from the ventral por- 11 (79%) of 14 women who underwent lymphatic mapping.8
tion of the vagina drain to the pelvis, while the dorsum of the When tumors were located in the lower third or at the introitus,
vagina may drain to rectal nodes. In their defnitive text on the sentinel nodes were detected in the groin 50% of the time, in

DOI: 10.1201/9781003255536-16 129


130 Clinical Lymphatic Mapping

TABLE 16.1 disease spread to lymph nodes may affect recommendations


Lymph Node Basins Draining the Vagina by Tumor Location * for observation or adjuvant chemotherapy in certain histologic
subtypes. Similar to performing complete lymphadenectomy
Location of sentinel node(s)
in other gynecologic cancers, removing all at-risk nodes in
Tumor location (by thirds) Groin Pelvis Groin and pelvis women with gross ovary-confned disease adds surgical mor-
Lower/introitus (n = 11) 64% 22% 22% bidity and operative time and is overtreatment for the 85% of
Middle (n = 1) 0% 100% 0% women who will ultimately have node-negative disease. For
Upper (n = 8) 25% 75% 0%
these reasons, investigators have started to explore the role of
lymphatic mapping and sentinel nodes in women with early-
Note: *Tumors that crossed one or more regions were excluded. stage ovarian cancer.
In order to apply mapping principles to ovarian cancer, early
investigators frst had to work out the location and timing of
the pelvis 17% of the time, and in both the groin and pelvis injections. Injection of mapping substances directly into the
33% of the time. Tumors in the upper third drained to the groin ovarian cortex was fraught with risk of rupture and upstaging
50% of the time and to the pelvis in the other half of cases. of disease, so this approach was quickly abandoned. Some of
The one case with disease in the middle third drained to the the initial studies injected dyes into the mesovarium, myome-
pelvis. When assessing tumor location by anterior or posterior, trium, and uterine cervix, but ultimately leading researchers
anterior lesions drained to the groin in 25% of cases and to the adopted injection in the subcuticular tissue of the infundibu-
pelvis in 75% of cases. Posterior lesions drained to the groin lopelvic and utero-ovarian ligaments as the areas most likely
in 50% of cases, to the pelvis in 17% of cases, and to both to accurately map the ovary.12 Utilizing this technique, at least
the groin and pelvis in 33%. Other authors have also reported one sentinel node will be identifed in approximately 94% of
cases of vaginal cancer located in the lower third of the vagina cases. Sentinel nodes will be found in the aortocaval region
mapping to both groin and pelvic basins.9 Table 16.1 summa- alone 66% of the time, in the pelvis only 12% of the time, and
rizes the location of sentinel nodes based on tumor location in in both regions 22% of the time13 (Figure 16.1). Surprisingly,
the vagina. sentinel nodes along the vena cava and aorta were more fre-
quently detected below the inferior mesenteric artery than
above it (57% vs. 43%).14–18 Although injecting the infundib-
ulopelvic and utero-ovarian ligaments seems to be the best
Ovarian cancer option for lymphatic mapping ovarian cancer, one of the limi-
The main lymphatic drainage of the ovary follows the ovar- tations of this technique in ovarian cancer is the lack of drain-
ian vessels to the aortocaval region to the vena cava and aorta ing basins detected in the contralateral pelvis even though
as high as the cisterna chyli.1 However, nodal basins drain- metastatic disease to lymph nodes can be found in those nodes
ing the ovary may also be found in multiple locations in the in 3.2% of cases.12
deep pelvis and along the iliac vessels. The right ovary fre- The timing of injections is still not entirely worked out. As
quently drains to the right and anterior to the vena cava above the decision to perform lymph node sampling is typically made
the inferior mesenteric artery, while the lymphatic basins of after a frozen section has established malignancy, there are two
the left ovary are more often found overlying the aorta and a possibilities of when to inject the dyes. One option is to inject
bit more cephalad. Occasionally the lymphatic channels found all cases with a suspicion of malignancy at the beginning of the
in the infundibulopelvic ligament will terminate lower along case and then explore for sentinel nodes only after the frozen
the vena cava and aorta (below the inferior mesenteric artery) section returns cancer. One problem with this approach is that
and even below the bifurcation of the aorta along the sacral blue dye and indocyanine green only stay in the sentinel node
promontory. There are also lymphatic channels that fow down for a limited time, so they may not be found if the frozen sec-
the posterolateral aspects of the uterus to join the cervical tion takes a long time. Radiocolloid will stay in the sentinel
channels leading deep into the pelvis to the obturator space node for hours, so this tracer may be more appropriate for this
and external iliac vessels. In addition, the ovary has lymphatic approach. Another potential problem with this approach is that
drainage directly to the pelvis through channels in the folds it may be diffcult to safely access the infundibulopelvic and/or
of the broad ligament terminating near the bifurcation of the utero-ovarian ligaments for injection if the pelvic tumor is large
internal and external iliac vessels.1 These channels may also or fxed. The other technique is to remove the adnexal mass, and
continue into the inguinal triangle. Although rare, women with once a frozen section reveals malignancy, then inject the rem-
ovarian cancer do occasionally present with metastatic disease nants of the infundibulopelvic and utero-ovarian ligaments. This
in the groin nodes as disease progresses past the pelvic nodes timing protocol will have a sentinel node detection rate in 90%–
through these channels. 100% of cases.17,18 Frequently a diagnosis of cancer is made post-
Complete lymphadenectomy in patients with stage III/ operatively requiring a staging surgery as a second procedure.
IV ovarian cancer has been shown to be unnecessary,10 but Attempts have been made in these circumstances to perform
its role in clinically early-stage disease remains unclear. In lymphatic mapping by injecting the infundibulopelvic and utero-
women with disease grossly limited to the ovary, lymphad- ovarian ligament stumps/remnants, but detection rates go down
enectomy will confrm metastatic disease to lymph nodes in signifcantly when this is done. In one study, the sentinel node
as many as 15% of cases.11 Although the therapeutic value of detection rate in these delayed staging cases was 42% compared
resecting these lymph nodes is likely minimal, the presence of to 89% when performed during the initial surgery (p = 0.003).17
Special considerations 131

FIGURE 16.1 Sentinel lymph nodes of the right ovary identifed by injection of blue dye in the hilum of the ovary. The nodes are anterior to the
inferior vena cava (IVC) and between the IVC and aorta. Left ovarian sentinel nodes are typically at level of the left renal vein.

There are very few prospective validation studies for senti- (ESMO) and the National Comprehensive Cancer Network
nel nodes in women with ovarian cancer. The Sentinel Nodes (NCCN) recommend a wide excision with 0.5-cm margins for
in Early Stage Ovarian Cancer (SELLY) trial performed lym- in situ lesions, 1 cm for tumors <2 mm thick, and obtaining a
phatic mapping and sentinel lymph node biopsy followed by margin of 2 cm for lesions ≥ 2 mm thick22 (Figure 16.3). For
complete lymphadenectomy in women with histologically tumors >1 mm thick, sentinel lymph node biopsies are also
confrmed ovarian cancer. The authors reported on the frst
31 patients in a preliminary report.17 The overall detection rate
was 68% (21 of 31 patients). Four patients had metastatic dis-
ease to lymph nodes, and the sentinel node identifed all of
them (sensitivity 100%, false-negative rate 0%). In addition,
the negative predictive value was 100%.17 The SELLY trial
continues to accrue and ultimately will enroll 176 patients,
which will provide more robust data on the validity of the
technique.19

Vulvar melanoma
Although vulvar melanoma is the most common nonsqua-
mous malignancy of the vulva, it is an exceedingly rare dis-
ease, accounting for only 5% of vulvar cancers. In the United
States there are only 0.136 cases per 100,000 women.20 Vulvar
melanoma is an aggressive disease with a 5-year survival rate
of approximately 33%.21 The disease occurs most frequently in
Caucasian women at an average age of 60, and older age has
been shown to be an independent prognostic factor for sur-
vival. Tumors are most frequently found on the labia minora
and clitoris (Figure 16.2).
Much of the treatment recommendations for vulvar mela-
noma have been extrapolated from the literature for the more
common cutaneous melanoma. Surgical resection is the pre-
ferred primary management for patients with vulvar melanoma.
In their guidelines, the European Society for Medical Oncology FIGURE 16.2 Vulvar melanoma involving the clitoris.
132 Clinical Lymphatic Mapping

p = 0.78). This study established sentinel lymph node biopsy


as the standard of care in patients with cutaneous melanoma.
As immediate regional lymphadenectomy after detection
of a positive sentinel node was part of the MSLT-I protocol,
this, too, became part of the standard of care for cutaneous
melanoma. However, at that time there were no prospective
studies showing that completion lymphadenectomy improved
oncologic outcomes, although it was well known that it greatly
increased adverse events and morbidity.
The second Multicenter Selective Lymphadenectomy Trial
(MSLT-II) evaluated the utility of completion lymphadenec-
tomy in patients with cutaneous melanoma who were found
to have a positive sentinel node. Patients with metastatic dis-
ease to sentinel nodes were randomly assigned to completion
regional lymphadenectomy or an observation group entailing
clinical exam and nodal ultrasonography assessment with
completion lymph node dissection if the patient developed a
regional lymph node recurrence. As expected, regional lymph
node recurrence was higher in the observation group as com-
pared to the completion group (23% vs. 8%, p < 0.001), but
this did not translate into a survival difference. At a median
follow-up of 43 months, the 3-year melanoma-specifc sur-
vival rate was equivalent between the two groups (86% for
both groups, p = 0.42).25 The DeCOG-SLT study also found
completion lymphadenectomy did not improve survival over
observation with exam and ultrasound after a positive senti-
FIGURE 16.3 Surgical resection of clitoral melanoma with adequate nel node was detected in patients with cutaneous melanoma.26
margins. Although the recurrence rate was lower than predicted in both
groups, leading to a study that was ultimately underpowered,
the study showed no difference in survival between the two
recommended. In addition, sentinel lymph node biopsy is rec- groups (3-year survival 82% in the observation group com-
ommended for lesions 0.75–1 mm thick that have additional pared to 81% in the completion group, p = 0.87). Again, nodal
risk factors such as ulceration or high mitotic rate. recurrences were higher in the observation group (16%) than
With efforts led by Dr. Donald Morton in the early 1990s, they were in the completion group (11%).
surgeons began to investigate the safety of sentinel lymph node Based on these studies, the American Society of Clinical
biopsy alone as a replacement for complete regional lymph- Oncology (ASCO) and the Society for Surgical Oncology
adenectomy in patients with cutaneous melanoma.23 The (SSO) recommend wide excision and sentinel lymph node
Multicenter Selective Lymphadenectomy Trial-I (MSLT-I) biopsy alone for patients with intermediate-thickness cutane-
randomized 2001 patients with cutaneous melanoma with a ous melanoma (1–4 mm) and for those with thin lesions (<1
Breslow thickness >1 mm to either wide excision and observa- mm thick) with risk factors such as ulceration.27 For patients
tion with regional lymphadenectomy only at the time of recur- with thick melanomas (>4 mm), sentinel lymph node biopsy
rence (observation group) or wide excision and sentinel lymph may be considered for prognostic purposes and to aid in guid-
node biopsy followed by observation if the sentinel node was ing adjuvant therapy, even though MSLT-1 and other studies
negative (biopsy group). In those patients with positive sen- have not shown a survival beneft in these patients. Based on
tinel nodes, complete regional lymphadenectomy was per- the MSLT-II and DeCOG-SLT studies, the NCCN states that
formed in a second surgery (biopsy group).24 For patients with observation after a positive sentinel node is preferred, although
intermediate-thickness melanomas (1.2–3.5 mm), there was no completion lymphadenectomy may be considered.28
difference in melanoma-specifc survival for the entire biopsy Lymphatic mapping and sentinel lymph node biopsy for
group compared to the observation group (81% vs. 78% at 10 vulvar melanoma follow the same techniques described ear-
years, p = 0.18). However, there was a signifcant improvement lier in this book (see Chapter 5). Wechter et al. reported on
in melanoma-specifc survival in those patients who had a pos- ten patients with vulvar melanoma who underwent sentinel
itive sentinel lymph node compared to those in the observation lymph node biopsy.29 Two patients had positive sentinel nodes,
group who developed clinical disease in a regional lymph node but there was no description of long-term follow-up in these
basin (62% vs. 42%, p = 0.006). For patients with thick mela- patients. In another retrospective study, De Hullu et al. treated
nomas (>3.5 mm), similarly there was no difference in survival 33 patients with vulvar melanoma in whom 9 underwent
for the entire group, including for those patients with positive lymphatic mapping and sentinel lymph node biopsy.30 Two
nodes (33%) who had sentinel lymph node biopsy as compared (22%) of the 9 patients who had a sentinel lymph node biopsy
to those who had regional nodes observed and only managed at recurred in the groin compared to 0 of the 24 who had full
the time of clinical presentation for the disease (48% vs. 46%, inguinofemoral lymphadenectomies. However, both of these
Special considerations 133

patients had lesions >4 mm thick, which made them exceed- with indocyanine green in a minimally invasive setting: a
ingly high risk for recurrence. Other than an additional few feasible study. J Minim Invasive Gynecol. 2017;24:165–70.
case reports, there are no other publications on outcomes for 16. Nyberg RH, Korkola P, Maenpaa JU. Sentinel node and
women with vulvar melanoma after sentinel node biopsy. ovarian tumors: a series of 20 patients. Int J Gynecol
Cancer. 2017;27:684–9.
17. Uccella S, Nero C, Vizza E, et al. Sentinel-node biopsy in
REFERENCES early-stage ovarian cancer: preliminary results of a pro-
1. Plentl AA, Friedman EA. Lymphatic System of the Female spective multicentre study (SELLY). Am J Obstet Gynecol.
Genitalia. Philadelphia: W.B. Saunders Company; 1971. 2019;221:324e1–e10.
2. Ishida H, Nagai T, Sato S, et al. Concomitant sentinel lymph 18. Lago V, Bello P, Montero B, et al. Sentinel lymph node
node biopsy leading to abbreviated systematic lymphad- technique in early-stage ovarian cancer: a phase II clinical
enectomy in a patient with primary malignant melanoma of trial. Int J Gynecol Cancer. 2020;30(9):1390–6.
the vagina. Springerplus. 2015;4;102. 19. Scambia G, Nero C, Uccella S, et al. Sentinel-node biopsy
3. Nakagawa S, Koga K, Kugu K, et al. The evaluation of in early stage ovarian cancer: a prospective multicentre
the sentinel node successfully conducted in a case of malig- study (SELLY). Int J Gynecol Cancer. 2019;29:1437–9.
nant melanoma of the vagina. Gynecol Oncol. 2002;86; 20. Boer FL, Ten Eikelder MLG, Kapiteijn EH, et al. Vulvar
387–9. malignant melanoma: pathogenesis, clinical behaviour and
4. Dhar KK, Das N, Brinkman DA, et al. Utility of sentinel management: review of the literature. Cancer Treat Rev.
node biopsy in vulvar and vaginal melanoma: report of two 2019;73:91–103.
cases and review of the literature. Int J Gynecol Cancer. 21. Weinstock MA. Malignant melanoma of the vulva and
2007;17:720–3. vagina in the United States: patterns of incidence and pop-
5. Abramova L, Parekh J, Irvin WP Jr, et al. Sentinel node ulation-based estimates of survival, Am J Obstet Gynecol.
biopsy in vulvar and vaginal melanoma: presentation 1994;171:1225–30.
of six cases and a literature review. Ann Surg Oncol. 22. Dummer R, Hauschild A, Lindenblatt N, et al. Cutaneous
2002;9:840–6. melanoma: ESMO Clinical Practice Guidelines for diag-
6. Rodier JF, Janser JC, David E, et al. Radiopharmaceutical- nosis, treatment and follow-up. Ann Oncol. 2015;26(Suppl
guided surgery in primary malignant melanoma of the 5):126–32.
vagina. Gynecol Oncol. 1999;75:308–9. 23. Morton DL, Cochran AJ, Thompson JF, et al. Sentinel node
7. Hertel H, Soergel P, Muecke J, et al. Is there a place for biopsy for early-stage melanoma: accuracy and morbidity
sentinel technique in treatment of vaginal cancer?: feasibil- in MSLT-I, an international multicenter trial. Ann Surg.
ity, clinical experience, and results. Int J Gynecol Cancer. 2005;242:302–11; discussion 311–13.
2013;23:1692–8. 24. Morton DL, Thompson JF, Cochran AJ, et al. Final trial
8. Frumovitz M, Gayed IW, Jhingran A, et al. Lymphatic report of sentinel-node biopsy versus nodal observation in
mapping and sentinel lymph node detection in women with melanoma. N Engl J Med. 2014;370:599–609.
vaginal cancer. Gynecol Oncol. 2008;108:478–81. 25. Faries MB, Thompson JF, Cochran AJ, et al. Completion
9. van Dam P, Sonnemans H, van Dam PJ, et al. Sentinel dissection or observation for sentinel-node metastasis in
node detection in patients with vaginal carcinoma. Gynecol melanoma. N Engl J Med. 2017;376:2211–22.
Oncol. 2004;92:89–92. 26. Leiter U, Stadler R, Mauch C, et al. Complete lymph node
10. Harter P, Sehouli J, Lorusso D, et al. A randomized trial of dissection versus no dissection in patients with sentinel
lymphadenectomy in patients with advanced ovarian neo- lymph node biopsy positive melanoma (DeCOG-SLT):
plasms. N Engl J Med. 2019;380:822–32. a multicentre, randomised, phase 3 trial. Lancet Oncol.
11. Piver MS, Barlow JJ, Lele SB. Incidence of subclinical 2016;17:757–67.
metastasis in stage I and II ovarian carcinoma. Obstet 27. Wong SL, Balch CM, Hurley P, et al. Sentinel lymph
Gynecol. 1978;52:100–4. node biopsy for melanoma: American Society of Clinical
12. Uccella S, Zorzato PC, Lanzo G, et al. The role of sentinel Oncology and Society of Surgical Oncology joint clinical
node in early ovarian cancer: a systematic review. Minerva practice guideline. J Clin Oncol. 2012;30:2912–18.
Med. 2019;110:358–66. 28. Network NCC. Cutaneous Melanoma (Version 3.2020)
13. Dell’Orto F, Laven P, Delle Marchette M, et al. Feasibility [2020 Jun 10]. Available from: www.nccn.org/professionals/
of sentinel lymph node mapping of the ovary: a systematic physician_gls/pdf/cutaneous_melanoma.pdf.
review. Int J Gynecol Cancer. 2019;29:1209–15. 29. Wechter ME, Gruber SB, Haefner HK, et al. Vulvar mela-
14. Kleppe M, Brans B, Van Gorp T, et al. The detection of noma: a report of 20 cases and review of the literature. J Am
sentinel nodes in ovarian cancer: a feasibility study. J Nucl Acad Dermatol. 2004;50:554–62.
Med. 2014;55:1799–804. 30. de Hullu JA, Hollema H, Hoekstra HJ, et al. Vulvar mel-
15. Buda A, Passoni P, Corrado G, et al. Near-infrared anoma: is there a role for sentinel lymph node biopsy?
fuorescence-guided sentinel node mapping of the ovary Cancer. 2002;94:486–91.
17
Clinical trial design

Scott Jordan and Brian M. Slomovitz

CONTENTS
Introduction .................................................................................................................................................................................. 135
Endometrial cancer ...................................................................................................................................................................... 135
Cervical cancer............................................................................................................................................................................. 136
Vulvar cancer ............................................................................................................................................................................... 138
Summary ...................................................................................................................................................................................... 139
References.................................................................................................................................................................................... 139

Introduction Endometrial cancer


Since frst being described for use in penile cancer in 1977, The frst report of sentinel node injection in endometrial can-
sentinel lymph node (SLN) evaluation has slowly become the cer was published in 1996 by Burke et al., a single-institution
standard-of-care surgical approach to staging across multiple pilot study with 15 patients who underwent myometrial injec-
cancer sites.1 Acceptance among gynecologic oncologists has tion of isosulfan blue dye at the time of abdominal hysterec-
been dependent on clinical trials demonstrating SLN evalua- tomy for endometrial cancer.2 Statistical endpoints were not
tion to have a low false-negative rate, noninferior survival out- defned, and neither sensitivity nor negative predictive value
comes, and an improvement in long- and short-term surgical (NPV) were reported. Multiple small prospective case series
morbidity. followed, investigating the feasibility of sentinel node identi-
In order to accomplish this, a stepwise progression from fcation and the sensitivity and NPV for cancer detection.3–16
reports of a novel surgical approach to large randomized con- Many of these pilot studies also reported the most common
trolled trials is required. The approach to this has differed locations for mapping as part of feasibility.
among disease sites due to unique challenges present in each Early studies were divided in surgical methodology between
cancer, but the basic principles are similar. Initially, small uterine injection of tracer (either into the fundus or directly
case series are published, demonstrating the technical ability into the tumor via hysteroscopy) and cervical injection. Due
of radiocolloid or dye to identify sentinel nodes in patients who to the relative ease of cervical injection compared to hystero-
are planned for complete lymphadenectomy. Larger retrospec- scopic or fundal injection, small trials were designed compar-
tive series and smaller prospective studies describe institutional ing the two injection sites with the goal of comparing node
experience with technique, further refning a standard proto- detection rates. In 2008, Perrone et al. compared sentinel node
col for use in practice-changing trials. The primary objective detection rates between two cohorts. The frst received techni-
of traditional phase III trials is to establish prospectively the cium-99 (Tc-99m) injected preoperatively into the cervix, and
sensitivity of SLNevaluation, often using the patient herself as the second cohort received hysteroscopically Tc-99m directly
the control group, by comparing excised sentinel nodes to the into the tumor. Detection rates between the cohorts did not
standard of complete lymphadenectomy. A second objective differ signifcantly.17 In 2013, Rossi et al. compared cervical
is to establish the safety of the technique by reporting short- versus hysteroscopic injection of indocyanine green (ICG) dye
and long-term procedural morbidity, recurrence rate, and prior to robotic sentinel lymph node biopsy, demonstrating a
recurrence-free survival data in patients undergoing sentinel signifcantly higher detection rate in the cervical group.18 A
node biopsy compared with full lymphadenectomy. Because study incorporating single photon emission computed tomog-
of the rarity of some of the gynecologic cancers, the variety of raphy (SPECT) rather than surgical detection methods con-
endpoints that we deem to be most clinically appropriate, and frmed cervical injection to yield an improved detection rate
the strong views of experts in our feld, carrying out phase III compared with hysteroscopic peritumoral injection.19 Two
trials is not always possible, and we need to use novel study large meta-analyses have confrmed these results.20,21
designs to demonstrate meaningful data that will change the While initial studies utilized either Tc-99m, blue dye, or a
standard of care. combination of the two, the addition of near-infrared imaging

DOI: 10.1201/9781003255536-17 135


136 Clinical Lymphatic Mapping

technology has allowed the inclusion of ICG tracer to be com-


pared as well. Sinno et al. compared detection rates between Cervical cancer
ICG and isosulfan blue dye cohorts of patients undergoing
robotic-assisted SLN biopsy, demonstrating signifcantly SLN mapping in cervical cancer was frst reported in 1999, in
higher rates of sentinel node identifcation in the ICG cohort a prospective series by Echt et al., demonstrating a subopti-
compared with isosulfan blue dye.22 A comparison by How et mal identifcation rate of 15.4% in 13 cervical cancer patients
al. utilizing all three tracers injected into the cervical stroma of injected with lymphazurine.32 In the following years, similar
the same patient demonstrated poorer performance of isosul- to endometrial cancer, many small case series were published
fan blue dye compared with either ICG or Tc-99m, which per- demonstrating the sensitivity of SLN mapping in cervical can-
formed similarly.23 In a randomized controlled trial of sentinel cer, including sensitivity and NPV. These small studies were
node detection in endometrial and cervical cancer powered for included in meta-analyses by van de Land et al. in 2007 (sen-
noninferiority, ICG demonstrated a signifcantly higher detec- sitivity 92%) and by Frumovitz et al. in 2008 (sensitivity 92%,
tion rate compared with isosulfan blue, leading the authors to NPV 97%).33,34
suggest ICG to in fact be the superior agent.24 The frst multicenter study was published in 2008 by
Two large phase III, prospective, multisite studies demon- Altagassen et al. (Table 17.1). It included 590 prospectively
strated acceptable sensitivity and NPV values (Table 17.1). The registered patients, with endpoints of detection rate (88.6%),
SENTI-ENDO study, frst published in 2011, evaluated sensitiv- sensitivity (77.4%), and NPV (94.3%) calculated after SLN
ity and NPV in SLN evaluation using each hemipelvis as a unit excision followed by systematic pelvic and paraaortic lymph
of analysis, as well as by using the patient as a unit of analysis. node dissection. Despite a low sensitivity in the total cohort,
Tc-99m and isosulfan blue cervical injections were utilized for a post hoc analysis demonstrated higher sensitivity and NPV
identifcation. Completion pelvic lymphadenectomies were per- in patients with tumors under 2 cm, suggesting that patients
formed following sentinel lymph node excision for calculation with smaller tumors are the most appropriate for SLN biopsy.35
of NPV and sensitivity. The study demonstrated 100% NPV and The multicenter SENTICOL study was similarly designed, but
100% sensitivity in each hemipelvis, with a 97% NPV and 84% explicitly included up to stage IB1 disease. The protocol also
sensitivity in each patient.25 A survival analysis was published included ultrastaging of sentinel nodes, which was not done in
in 2015, showing no difference in recurrence-free survival the previous study. Sensitivity of this study was higher at 92%,
between patients with and without detected SLN or between and NPV was 98.2%.36
those with and without positive SLN. Adjuvant therapy was also In an effort to further increase sensitivity and NPV while
given more frequently in patients with positive SLN, demon- minimizing surgical morbidity, Cormier et al. published a
strating the impact on therapeutic decision making.26 prospective study evaluating the sensitivity and NPV of SLN
The FIRES trial, published in 2017, was similarly designed, mapping within a single institution compared with the theo-
utilizing ICG as a tracer. The primary endpoints were sensi- retical sensitivity and specifcity had the proposed algorithm
tivity and NPV, and with 356 patients enrolled, it is the larg- been applied. This algorithm included excision of all mapped
est prospective endometrial SLN study to date. Sensitivity SLNs (with ultrastaging), removal of all suspicious nodes,
was reported as 97.2% with an NPV of 99.6%.27 The fndings side-specifc pelvic lymph node dissection (LND) perfor-
of these large prospective studies corroborate the published mance in a nonmapping hemipelvis, and en block paramet-
results of the three largest meta-analyses on endometrial SLN rectomy being performed with resection of the primary tumor.
biopsy.20,21,28 This analysis demonstrated an increase in patient-specifc
Initially, due to controversy surrounding the utility of com- specifcity of 87.5% and NPV of 96.8% to 100% and 100%,
prehensive universal lymphadenectomy in the surgical man- respectively, while avoiding bilateral complete LND in all but
agement of early-stage endometrial cancer, SLN evaluation 6.6% of patients.37
presented an attractive alternative due to decreased surgical Unique to the surgical management of cervical cancer is the
intervention but equivalent detection rates.29 With increasing potential for abandoning the surgical approach in the setting of
adoption of the technique, retrospective and prospective data positive lymph nodes in order to reduce treatment-related mor-
were published demonstrating the utility of SLN in high-risk bidity and provide optimal anatomy to permit maximum radi-
histologies. Schiavone et al. published the results of an analysis ation dose delivery. Thus, additional studies were designed to
of a prospectively maintained database including 136 patients determine the utility of SLN evaluation intraoperatively. Fader
with carcinosarcoma. No difference in progression-free or et al. published a prospective series evaluating 38 patients’
overall survival was seen between patients who underwent SLN excision specimens intraoperatively via frozen section
complete lymphadenectomy or SLN biopsy alone.30 The rate and postoperatively via ultrastaging. Comparison of these two
of positive nodes was also equivalent between the two groups. methods demonstrated frozen section to correctly identify
Soliman et al. published a prospective trial of 123 patients with lymph node metastases in only 33% of cases.38 Retrospective
high-grade endometrial cancers, demonstrating 95% sensitiv- evaluation of the SENTICOL study, which only performed
ity and a false-negative rate of 4.3%.31 intraoperative examination in the case of macroscopic nodal
Given the prevalence of endometrial cancer, the stepwise enlargement, similarly demonstrated sensitivity of intraop-
demonstration of safety and effcacy, and the confrmation erative evaluation to be 21%.39 A larger retrospective trial in
in phase III prospective trials, SLN for endometrial cancer 2013, including 225 patients, compared results of intraopera-
is considered the standard of care for patients with presumed tive SLN frozen section to postoperative ultrastaging, again
early-stage endometrial cancer. showing a poor sensitivity result of only 56%.40
Clinical trial design 137

TABLE 17.1
Study Details
Trial name Disease site Population Study design Endpoints Enrollment Result
How 2012 23 Endometrium All histologies Cervical Tc-99 and patent blue Detection rate, 100 Detection 92%,
dye injection, robotic SLN sensitivity, specifcity, sensitivity 89%,
excision, PLND +/− PALND NPV NPV 99%
Rossi (FIRES) Endometrium Clinical stage I, Cervical ICG injection, robotic 1) Sensitivity of sentinel 340 patients Sensitivity to detect
201727 all histologies SLN excision, followed by lymph node specimen in metastatic disease:
complete PLND +/− PALND detecting metastatic 97.2%. NPV 99.6%
disease, 2) NPV for
lymph node involvement
Ballester Endometrium Clinical stage Cervical Tc-99 and patent blue Negative predictive 125 patients NPV for each
(SENTI-ENDO) I–II, all injection, open or laparoscopic value in 1) each hemipelvis: 100%;
201125 histologies SLN excision, followed by hemipelvis and 2) each NPV for each patient:
PLND +/− PALND patient 97%
Darai (SENTI- Endometrium Clinical stage Cervical Tc-99 and patent blue Recurrence-free survival 125 patients No difference in RFS
ENDO) 201526 I–II, all injection, open or laparoscopic between patients with
histologies SLN excision, followed by and without detected
PLND +/− PALND SLN, or between
patients with and
without LN
metastases
Tanner 201746 Endometrium Early stage, Cervical ICG injection, Side-specifc PLND rate 113 patients Side-specifc PLND:
grade 1–2 laparoscopic SLN excision, and nodal metastases 5.3%, overall PLND
endometrioid followed by TLH/BSO; in identifcation rate rate: 7.1%; nodal
nonbilaterally mapping metastases rate: 8%
patients, only those that met
high-risk criteria underwent
completion PLND on the
nonmapping side
Paley 201647 Endometrium Early stage, all ICG injection, SLN excision, Evaluate feasibility and 85 patients SLN sensitivity of
histologies PPALND in serous, clear cell, accuracy of SLN in 100%, specifcity of
carcinosarcoma, tumor size >2 women at high risk for 100%
cm, >50% myometrial wall LN involvement
invasion, grade 3 histology,
grossly suspicious nonsentinel
nodes, or positive SLN on
touch prep
Soliman 201731 Endometrium Stage I: Injection with ICG, T99, or False-negative rate, 101 Hemipelvis:
High-risk blue dye, SLN excision, sensitivity, and negative False-negative rate:
histologies or followed by PLND and predictive value by 7.1%, NPV 98%,
deep PALND patient and by sensitivity 92.9%,
myometrial hemipelvis Patient: False-
invasion; negative rate: 5%,
stage II: any NPV 98.6%,
histology Sensitivity 95%
Sawicki 201548 Endometrium Stage I–II, all Injection Tc99 to cervix and Compare SLN detection 188 Detection rate in
histologies methylene blue to fundus rates after cervical and Tc-99/methylene
versus methylene blue injected subserosal blue group: 95.1%,
into cervix and fundus, open administration of two methylene blue only
SLN biopsy, followed by tracers, sensitivity of group: 87.7%,
PLND + PALND in high-risk SLN biopsy sensitivity: 87.5%,
tumors NPV 97.7%
Lecuru Cervix Stage IA1 with T99 and patent blue injection, Determine sensitivity 139 patients Sensitivity 92%,
(SENTICOL) LVSI-IB1 SLN excision, PLND and NPV of SLN biopsy NPV 98.2%
201136 +/− PALND
Altgassen 200835 Cervix All stages T99 or blue dye injection, SLN Sensitivity and NPV in 507 patients Sensitivity 77.4%,
planned for excision, PLND +/− PALND total population NPV 94.3%
surgical staging

(Continued )
138 Clinical Lymphatic Mapping

TABLE 17.1
(Continued)
Trial name Disease site Population Study design Endpoints Enrollment Result
Cormier 2011 37 Cervix Stage IAI with Blue dye +/− Tc-99m Sensitivity and NPV 122 Patient specifc:
LVI to IIA injection, SLN excision, rates by patient and by Sensitivity 87.5%,
PLND +/− PALND hemipelvis; NPV 96.8%; side
retrospective evaluation specifc: sensitivity
of SLN excision 92.6%, NPV 98.9%,
algorithm algorithm: Sensitivity
100%, NPV 100%
Frumovitz 201824 Cervix and Clinical stage I Isosulfan blue + ICG dye Effcacy of 163 patients 97% of excised
uterus cervical or injection, SLN identifcation intraoperative ICG lymph nodes
uterine cancer, and excision versus isosulfan blue identifed with ICG,
all histologies (noninferiority) 47% of excised
lymph nodes
identifed with
blue dye
Van der Zee Vulva T1 or T2, less T99 and isosulfan injection, Evaluate safety of 403 In SLN group, 2-year
(GROINS VI) than 4 cm, groin SLN excision, if omitting inguinofemoral recurrence rate of
200842 squamous cell positive, inguinofemoral lymphadenectomy in 2.3%, 3-year survival
carcinoma of lymphadenectomy was patients with negative 97%, improved
the vulva, with performed; if more than one sentinel lymph node, short- and long-term
>1 mm DOI nodal metastases was evaluate short- and operative morbidity
and clinically identifed, postoperative RT long-term morbidity of
nonsuspicious was recommended sentinel node removal
LN and complete
inguinofemoral
lymphadenectomy
Levenback Vulva Squamous cell Isosulfan blue +/− T99 Establish negative 452 patients NPV 96.3% by
(GOG 173) carcinoma, >1 injection, groin SLN excision, predictive value and patient, 97.4% by
201244 mm invasion, followed by inguinofemoral sensitivity of groin SLN groin, sensitivity
tumor size lymphadenectomy evaluation 91.7% by patient,
between 2 and 92.1% by groin
6 cm clinically
nonsuspicious
LN
GROINS VII45 Vulva T1 or T2, less Isosulfan blue + T99 injection,
than 4 cm, groin SLN excision
squamous cell
carcinoma of
the vulva, with
>1 mm DOI
and clinically
nonsuspicious
LN

Similar to studies in uterine cancer, the optimal radiotracer of the approach and should be performed regardless of open or
was also investigated. In a meta-analysis combining six studies minimally invasive surgery.
that compared indocyanine green (ICG) dye to blue dye, Tc99-m,
or a combination, ICG was demonstrated to be superior to blue
dye but equivalent to the combination of Tc-99m and blue dye.41
The FILM study by Frumovitz et al. comparing blue dye to ICG
Vulvar cancer
uptake rates in patients who received both tracers was powered SLN mapping in vulvar cancer was the frst gynecologic
for noninferiority, but 97% of excised nodes were ICG positive, malignancy studied. This was due to the predictable lym-
compared to just 47% of nodes with blue dye positivity.24 phatic drainage of the vulva, the ease of tracer injection, and
Similar to endometrial cancer, SLN detection in cervical the morbidity associated with complete groin lymphadenec-
cancer patients has been studied prospectively and may be tomy. Given the rarity of this disease, traditional clinical trial
incorporated into the standard of care. One issue indirectly design (phase I, II, III) is not possible. Instead, large coopera-
related to this procedure relates to performing it during an tive group observational trials were designed to best answer
exploratory laparotomy, not during minimally invasive sur- the question of whether the sentinel node procedure could be
gery. The principles of SLN detection are the same regardless incorporated into the standard of care.
Clinical trial design 139

The GROINSS-V1 study (GROnigen INternational Study on case of sentinel node metastasis >2 mm. The protocol was
Sentinel nodes in Vulvar cancer) was an observational study of amended, with only patients with metastasis ≤2 mm receiving
276 women with early-stage vulvar cancer who had a primary radiotherapy and those >2 mm undergoing complete lymph-
tumor smaller than 4 cm and no metastases detected on SLN adenectomy. Final analysis after ≥2 years revealed an accept-
biopsy. The primary objective of this observational study was able groin recurrence rate; six isolated groin recurrences in
to investigate the safety of omitting inguinofemoral lymphade- 160 patients with a sentinel node micrometastasis (3.8%).
nectomy in patients with a negative sentinel node. For patients Two developed recurrence in the contralateral (sentinel node–
with unifocal disease, these investigators found a groin recur- negative) groin and two had refused radiotherapy.45
rence rate of 2.3% over a median follow-up time of 35 months. With the completion of GROINSSV1, GOG173, and
The false-negative rate was 5.9%, and the false-negative pre- GROINSSV2/GOG270, the standard of care has been changed
dictive value was 2.9%.42 to incorporate SLN biopsy in all patients with unifocal vulvar
At the same time, GOG protocol 173, a multi-institutional cancer 4 cm or less. For patients with negative nodes, obser-
observational trial of 452 women with early-stage vulvar vation is safe. For patients with positive nodes less than 2
cancer, was performed. Unlike the GROINS-V1 trial, in mm and without extracapsular spread, radiotherapy to 50 Gy
this study, all of the patients underwent an SLN biopsy fol- yields an acceptable treatment option without full lymph node
lowed by a complete inguinofemoral lymph node dissection dissection.
(IFLD). The primary objective of this study was to estimate GROINSV3/CV2020 is a NCI cancer therapy evaluation
the negative predictive value of a sentinel groin lymph node program (CTEP)-approved study to investigate prospectively
(i.e., the probability of all unilateral groin lymph nodes from if increasing radiotherapy to 56 Gy and adding chemosen-
a dissection being negative when the unilateral sentinel node sitization can adequately treat patients with greater than 2
is negative). The study also utilized the false-negative predic- mm of lymph node involvement or with extracapsular spread.
tive value, incorporating the false-negative identifcation rate This is a prospective phase II trial only enrolling patients
in affected groins and the true negative rate in unaffected dis- after SLN biopsy. Similar to the other GROINS studies,
sected groins, calculated by 1 – the NPV. The study results early stopping rules are incorporated to prevent an ineffec-
yielded a false-negative predictive value of 3.7%. There was tive treatment.
an improved false-negative SLN biopsy rate (2.0 vs. 7.4%) in Based on three large prospective studies, the SLN procedure
patients with tumors 2–3.9 cm compared to tumors 4–6 cm, has become the standard of care for women with early-stage
respectively.43,44 vulvar cancer.
The results of these two large prospective trials were enough
to incorporate the sentinel node procedure into the standard of
care for women with apparent early-stage squamous cell vul-
var cancer with unifocal lesions that were less than 4 cm. In Summary
these patients, a negative sentinel lymph node was enough to Prospective phase II treatment trials and randomized trials
abandon a complete lymphadenectomy and to avoid radiother- have been performed to demonstrate the safety and effcacy of
apy. However, further research needed to be done to improve the SLN procedure in patients with endometrial, cervical, and
the care for women with a positive sentinel node. vulvar cancer. Future trials will continue to investigate areas
One limitation to both of these initial trials was the time of high unmet need and further evaluate newer modalities for
it took to enroll patients. To overcome this barrier, the third these advanced procedures.
large observational trial for early-stage vulvar cancer brought
together two large cooperative groups to perform one study.
GROINSS-V II/GOG 270 was a prospective multicenter REFERENCES
phase II trial, including patients with early-stage squamous
1. Cabanas RM. An approach for the treatment of penile
cell carcinoma (SCC) of the vulva (diameter <4 cm) with-
carcinoma. Cancer. 1977;39(2):456–66. https://doi.org/10.
out suspicious lymph nodes at imaging, who had primary
1002/1097-0142(197702)39:23.0.CO;2-I.
surgical treatment with a sentinel node procedure. In case
2. Burke TW, Levenback C, Tornos C, et al. Intraabdominal
of a metastatic sentinel node (metastasis of any size), radio- lymphatic mapping to direct selective pelvic and para-
therapy was given to the groin(s) (50 Gy). In case of a nega- aortic lymphadenectomy in women with high- risk endo-
tive sentinel node, patients were followed up for ≥2 years. metrial cancer: results of a pilot study. Gynecol Oncol.
Stopping rules were defned for both groups to monitor groin 1996;62(2):169–73. http://doi.org/10.1006/gyno.1996.0211.
recurrence rate. These rules pertained to the occurrence of 3. Allameh T, Hashemi V, Mohammadizadeh F, et al. Sentinel
groin recurrences in those patients with metastatic SLN(s). lymph node mapping in early stage of endometrial and cer-
The rules were based on grouped sequential analysis, with vical cancers. J Res Med Sci. 2015;20(2):169–73. https://
alpha and beta spending properties. The maximum num- pubmed.ncbi.nlm.nih.gov/25983771. Available from: www.
ber of patients needed was 150: when the upper and lower ncbi.nlm.nih.gov/pmc/articles/PMC4400713/.
boundary of the stopping rule would not be passed, the study 4. Altgassen C, Pagenstecher J, Hornung D, et al. A new
would close when 150 patients with a metastatic SLN had approach to label sentinel nodes in endometrial cancer.
completed 2 years of follow-up.45 Gynecol Oncol. 2007;105(2):457–61. Available from:
After activation of the stopping-rule in June 2010, interim www.sciencedirect.com.ezproxy.fu.edu/science/article/pii/
analysis showed an increased risk for groin recurrence in S0090825807000054.
140 Clinical Lymphatic Mapping

5. Bats A, Clément D, Larousserie F, et al. Does sentinel node endometrial cancer. Eur J Gynaecol Oncol. 2015;36(6):
biopsy improve the management of endometrial cancer? 643–6. http://europepmc.org/abstract/MED/26775344.
data from 43 patients. J Surg Oncol. 2008;97(2):141–5. 17. Perrone AM, Casadio P, Formelli G, et al. Cervical and hys-
https://doi.org/10.1002/jso.20857. teroscopic injection for identifcation of sentinel lymph node
6. Delaloye J, Pampallona S, Chardonnens E, et al. in endometrial cancer. Gynecol Oncol. 2008;111(1):62–7.
Intraoperative lymphatic mapping and sentinel node biopsy Available from: www.sciencedirect.com.ezproxy.fu.edu/
using hysteroscopy in patients with endometrial cancer. science/article/pii/S0090825808004277.
Gynecol Oncol. 2007;106(1):89–93. Available from: www. 18. Rossi EC, Jackson A, Ivanova A, et al. Detection of sen-
sciencedirect.com.ezproxy.fiu.edu/science/article/pii/ tinel nodes for endometrial cancer with robotic assisted
S0090825807001825. fuorescence imaging: cervical versus hysteroscopic injec-
7. Favero G, Pfffer T, Ribeiro A, et al. Laparoscopic senti- tion. Int J Gynecol Cancer. 2013;23(9):1704–11. https://doi.
nel lymph node detection after hysteroscopic injection of org/10.1097/IGC.0b013e3182a616f6.
technetium-99 in patients with endometrial cancer. Int J 19. Sahbai S, Taran F, Fiz F, et al. Pericervical injection of
Gynecol Cancer. 2015;25(3):423–30. https://doi.org/10.1097/ 99mTc-nanocolloid is superior to peritumoral injection for
IGC.0000000000000387. sentinel lymph node detection of endometrial cancer in
8. Gien LT, Kwon JS, Carey MS. Sentinel node mapping SPECT/CT. Clin Nucl Med. 2016;41(12). Available from:
with isosulfan blue dye in endometrial cancer. J Obstet https://journals.lww.com/nuclearmed/Fulltext/2016/12000/
Gynaecol Can. 2005;27(12):1107–12. Available from: Pericervical_Injection_of_99mTc_Nanocolloid_Is.3.aspx.
www.sciencedirect.com.ezproxy.fu.edu/science/article/pii/ 20. Kang S, Yoo HJ, Hwang JH, et al. Sentinel lymph node
S1701216316303930. biopsy in endometrial cancer: meta-analysis of 26 studies.
9. Kataoka F, Susumu N, Yamagami W, et al. The importance Gynecol Oncol. 2011;123(3):522–7. https://doi.org/10.1016/j.
of para-aortic lymph nodes in sentinel lymph node mapping ygyno.2011.08.034.
for endometrial cancer by using hysteroscopic radio-isotope 21. Bodurtha Smith AJ, Fader AN, Tanner EJ. Sentinel lymph
tracer injection combined with subserosal dye injection: node assessment in endometrial cancer: a systematic review
prospective study. Gynecol Oncol. 2016;140(3):400–4. and meta-analysis. Obstet Gynecol. 2017;216(5):459–76.
Available from: www.sciencedirect.com.ezproxy.fu.edu/ e10. Available from: www.sciencedirect.com.ezproxy.fu.
science/article/pii/S0090825815302250. edu/science/article/pii/S0002937816320579. https://doi.
10. Kuru Oğ, Topuz S, Sen S, et al. Sentinel lymph node org/10.1016/j.ajog.2016.11.1033.
biopsy in endometrial cancer: description of the tech- 22. Sinno AK, Fader AN, Roche KL, et al. A comparison
nique and preliminary results. J Turk Ger Gynecol Assoc. of colorimetric versus fuorometric sentinel lymph node
2011;12(4):204–8. Available from: www.ncbi.nlm.nih. mapping during robotic surgery for endometrial can-
gov/pmc/articles/PMC3939250/. https://doi.org/10.5152/ cer. Gynecol Oncol. 2014;134(2):281–6. https://doi.org/
jtgga.2011.52. S0090-8258(14)00982-2.
11. Laios A, Volpi D, Tullis IDC, et al. A prospective pilot 23. How J, Gotlieb WH, Press JZ, et al. Comparing indo-
study of detection of sentinel lymph nodes in gynaecologi- cyanine green, technetium, and blue dye for sentinel
cal cancers using a novel near infrared fuorescence imag- lymph node mapping in endometrial cancer. Gynecol
ing system. BMC Res Notes. 2015;8(1):608. https://doi. Oncol. 2015;137(3):436–42. Available from: www.
org/10.1186/s13104-015-1576-z. sciencedirect.com.ezproxy.fiu.edu/science/article/pii/
12. Lopes LAF, Nicolau SM, Baracat FF, et al. Sentinel S0090825815007945. https://doi.org/10.1016/j.ygyno.2015
lymph node in endometrial cancer. Int J Gynecol .04.004.
Cancer. 2007;17(5):1113–17. https://doi.org/10.1111/j.1525- 24. Frumovitz M, Plante M, Lee PS, et al. Near-infrared
1438.2007.00909.x. fuorescence for detection of sentinel lymph nodes in
13. López-De la Manzanara Cano C, Cordero García JM, women with cervical and uterine cancers (FILM): a ran-
Martín-Francisco C, et al. Sentinel lymph node detection domised, phase 3, multicentre, non-inferiority trial.
using 99mTc combined with methylene blue cervical injec- Lancet Oncol. 2018;19(10):1394–403. Available from:
tion for endometrial cancer surgical management: a pro- www.sciencedirect.com.ezproxy.fiu.edu/science/article/
spective study. Int J Gynecol Cancer. 2014;24(6):1048–53. pii/S1470204518304480. https://doi.org/10.1016/S1470
https://doi.org/10.1097/IGC.0000000000000158. -2045(18)30448-0.
14. Mücke J, Klapdor R, Schneider M, et al. Isthmocervical 25. Ballester M, Dubernard G, Lécuru F, et al. Detection rate
labelling and SPECT/CT for optimized sentinel detection and diagnostic accuracy of sentinel-node biopsy in early
in endometrial cancer: technique, experience and results. stage endometrial cancer: a prospective multicentre study
Gynecol Oncol. 2014;134(2):287–92. Available from: (SENTI-ENDO). Lancet Oncol. 2011;12(5):469–76. https://
www.sciencedirect.com.ezproxy.fu.edu/science/article/pii/ doi.org/10.1016/S1470-2045(11)70070-5.
S0090825814009561. 26. Daraï E, Dubernard G, Bats A, et al. Sentinel node biopsy
15. Solima E, Martinelli F, Ditto A, et al. Diagnostic accu- for the management of early stage endometrial cancer: long-
racy of sentinel node in endometrial cancer by using hys- term results of the SENTI-ENDO study. Gynecol Oncol.
teroscopic injection of radiolabeled tracer. Gynecol Oncol. 2015;136(1):54–9. Available from: www.sciencedirect.com.
2012;126(3):419–23. Available from: www.sciencedirect. ezproxy.fiu.edu/science/article/pii/S0090825814013389.
com.ezproxy.fu.edu/science/article/pii/S0090825812003988. https://doi.org/10.1016/j.ygyno.2014.09.011.
16. Valha P, Kucera E, Sak P, et al. Intraoperative subserosal 27. Rossi EC, Kowalski LD, Scalici J, et al. A comparison
approach to label sentinel nodes in intermediate and high-risk of sentinel lymph node biopsy to lymphadenectomy for
Clinical trial design 141

endometrial cancer staging (FIRES trial): a multicentre, 39. Bats A, Buénerd A, Querleu D, et al. Diagnostic value
prospective, cohort study. Lancet Oncol. 2017;18(3):384– of intraoperative examination of sentinel lymph node in
92. Available from: www.sciencedirect.com.ezproxy.fu. early cervical cancer: a prospective, multicenter study.
edu/science/article/pii/S1470204517300682. https://doi.org/ Gynecol Oncol. 2011;123(2):230–5. Available from:
10.1016/S1470-2045(17)30068-2. www.sciencedirect.com.ezproxy.fiu.edu/science/article/
28. Ansari M, Rad MA, Hassanzadeh M, et al. Sentinel node pii/S0090825811006950. https://doi.org/10.1016/j.ygyno.2011
biopsy in endometrial cancer: systematic review and .08.010.
meta-analysis of the literature. Eur J Gynaecol Oncol. 40. Slama J, Dundr P, Dusek L, et al. High false negative rate of
2013;34(5):387–401. frozen section examination of sentinel lymph nodes in patients
29. NCCN.org. NCCN Clinical Practice Guidelines in with cervical cancer. Gynecol Oncol. 2013;129(2):384–8.
Oncology (NCCN Guidelines ®) Uterine Neoplasms Available from: www.sciencedirect.com.ezproxy.fu.edu/
Continue NCCN Guidelines for Patients ®; 2021. Available science/article/pii/S0090825813000723. https://doi.org/10
from: www.nccn.org/patients. .1016/j.ygy2013.02.001.
30. Schiavone MB, Zivanovic O, Zhou Q, et al. Survival of 41. Ruscito I, Gasparri ML, Braicu EI, et al. Sentinel node
patients with uterine carcinosarcoma undergoing sentinel mapping in cervical and endometrial cancer: indocyanine
lymph node mapping. Ann Surg Oncol. 2016;23(1):196– green versus other conventional dyes-A meta-analysis. Ann
202. https://doi.org/10.1245/s10434-015-4612-2. Surg Oncol. 2016;23(11):3749–56. https://doi.org/10.1245/
31. Soliman PT, Westin SN, Dioun S, et al. A prospective vali- s10434-016-5236-x.
dation study of sentinel lymph node mapping for high-risk 42. Van der Zee AGJ, Oonk MH, De Hullu JA, et al. Sentinel
endometrial cancer. Gynecol Oncol. 2017;146(2):234–9. node dissection is safe in the treatment of early-stage vul-
https://doi.org/10.1016/j.ygyno.2017.05.016. var cancer. JCO. 2008;26(6):884–9. https://doi.org/10.1200/
32. Echt ML, Finan MA, Hoffman MS, et al. Detection of sen- JCO.2007.14.0566.
tinel lymph nodes with lymphazurin in cervical, uterine, 43. Coleman RL, Ali S, Levenback CF, et al. Is bilateral lymph-
and vulvar malignancies. South Med J. 1999;92(2):204–8. adenectomy for midline squamous carcinoma of the vulva
https://doi.org/10.1097/00007611-199902000-00008. always necessary? An analysis from gynecologic oncol-
33. van de Lande J, Torrenga B, Raijmakers PGHM, et al. ogy group (GOG) 173. Gynecol Oncol. 2013;128(2):155–9.
Sentinel lymph node detection in early stage uterine cer- https://doi.org/10.1016/j.ygyno.2012.11.034.
vix carcinoma: a systematic review. Gynecol Oncol. 44. Levenback CF, Ali S, Coleman RL, et al. Lymphatic map-
2007;106(3):604–13. Available from: www.sciencedirect ping and sentinel lymph node biopsy in women with squa-
.com/science/article/pii/S0090825807003381. https://doi mous cell carcinoma of the vulva: a gynecologic oncology
.org/10.1016/j.ygyno.2007.05.010. group study. J Clin Oncol. 2012;30(31):3786–91. Available
34. Frumovitz M, Ramirez PT, Levenback CF. Lymphatic from: www.ncbi.nlm.nih.gov/pmc/articles/PMC3478573/.
mapping and sentinel lymph node detection in women with https://doi.org/10.1200/JCO.2011.41.2528.
cervical cancer. Gynecol Oncol. 2008;110(3, Suppl 2):S17– 45. Slomovitz B, Oonk M, Monk BJ, et al. Validation of sen-
S20. Available from: www.sciencedirect.com.ezproxy.fu tinel lymph biopsy in patients with early stage vulvar
.edu/science/article/pii/S0090825808002059. https://doi cancer: a prospective trial of 1552 women (GROINSS-V
.org/10.1016/j.ygyno.2008.03.012. II/GOG270). Gynecol Oncol. 2020;159:2–3. https://doi
35. Altgassen C, Hertel H, Brandstädt A, et al. Multicenter vali- .org/10.1016/j.ygyno.2020.06.004.
dation study of the sentinel lymph node concept in cervi- 46. Tanner E, Puechl A, Levinson K, et al. Use of a novel sen-
cal cancer: AGO study group. JCO. 2008;26(18):2943–51. tinel lymph node mapping algorithm reduces the need for
https://doi.org/10.1200/JCO.2007.13.8933. pelvic lymphadenectomy in low-grade endometrial can-
36. Lécuru F, Mathevet P, Querleu D, et al. Bilateral nega- cer. Gynecol Oncol. 2017;147(3):535–40. Available from:
tive sentinel nodes accurately predict absence of lymph www.sciencedirect.com.ezproxy.fiu.edu/science/article/
node metastasis in early cervical cancer: results of the pii/S0090825817314233. https://doi.org/10.1016/j.ygyno
SENTICOL study. JCO. 2011;29(13):1686–91. https://doi. .2017.10.020.
org/10.1200/JCO.2010.32.0432. 47. Paley PJ, Veljovich DS, Press JZ, et al. A prospective inves-
37. Cormier B, Diaz JP, Shih K, et al. Establishing a sentinel tigation of fuorescence imaging to detect sentinel lymph
lymph node mapping algorithm for the treatment of early nodes at robotic-assisted endometrial cancer staging.
cervical cancer. Gynecol Oncol. 2011;122(2):275–80. Obstet Gynecol. 2016;215(1):117.e1–.e7. Available from:
https://doi.org/10.1016/j.ygyno.2011.04.023. www.sciencedirect.com.ezproxy.fiu.edu/science/article/
38. Fader AN, Edwards RP, Cost M, et al. Sentinel lymph node pii/S0002937815026502. https://doi.org/10.1016/j.ajog.2015.
biopsy in early-stage cervical cancer: utility of intraop- 12.046.
erative versus postoperative assessment. Gynecol Oncol. 48. Sawicki S, Lass P, Wydra D. Sentinel lymph node biopsy
2008;111(1):13–17. Available from: www.sciencedirect.com in endometrial Cancer – Comparison of 2 detection meth-
.ezproxy.fu.edu/science/article/pii/S009082580800437X. ods. Int J Gynecol Cancer. 2015;25(6):1044–50. https://doi
https://doi.org/10.1016/j.ygyno.2008.06.009. .org/10.1097/IGC.0000000000000447.
Index

Note: Page numbers in italics indicate a fgure and page numbers in bold indicate a table on the corresponding page.

99mTc colloidal albumin, 90 sentinel lymph node (SLN), 41, 41–42 posterior trunk, 25, 26
99mTc-labeled radiopharmaceuticals, 44 unidirectional lymphatics fow, 39 topographic anatomy, 23, 23–24
99mTechnetium-labeled nanocolloid, 68 breast cancer, 7 tracer injection, 31
[99mTechnetiumTc] tilmanocept, 50 AMAROS study, 93 chemoradiation, 75, 121, 123, see also
axilla, 87, 87–89, 88, 88 radiation oncology
A blue dye, 90, 91 circumfex vasculature, 17
clinically negative axilla, management, 93 clinical management, SLN biopsy
abdomen and pelvis, lymphatics, 1, 2 ductal carcinoma in situ (DCIS), 83, 84 acceptance in current routine practice, 74
adenocarcinoma metastases, 26, 57 follow-up after SLNB, 95 in fertility-sparing treatment, 77–78
adjuvant chemotherapy, 41, 89, 93, gamma probe, 91 frozen section and intraoperative triage of
122–124, 130 imprint cytology, 56 patients, 75
adjuvant radiation therapy (RT), 89, 99, 120, indocyanine green, 90 international recommendations, 74–75
123–124 infltrating lobular carcinoma, 85, 85 lower leg lymphedema (LLL), 78
AJCC Cancer Staging Manual for breast intraoperative techniques, 90, 91 micrometastases, pathologic ultrastaging
cancer, 7 invasive ductal carcinoma, 84, 85–86 and prognostic, 75–77, 77
Alcian blue, 57 lobular carcinoma in situ (LCIS), 84, 84 ongoing clinical trials, 78
American College of Surgeons Oncology nodal metastasis, patterns, 83 clinical target volume (CTV), 121
Group 2001 Trial, 41 paraffn pieces, 59 clinical trial design
American Joint Committee on Cancer (AJCC), pathology, 83, 84 cervical cancer, 136–138
7, 85–86 radioisotope/nuclear medicine, 90, 92 endometrial cancer, 135–136
American Society of Clinical Oncology sentinel lymph node mapping, 83–95 study details, 137–138
(ASCO), 132 SLN, pathologic analysis, 89 vulvar cancer, 138–139
anaphylaxis, 44, 49, 68 SLNB after NAC, 93–95, 94–95 clitoral lymphatics to pelvis, 18
anastomosis of Poirier channel, 30 SLN mapping procedure, 90–93, 91–93 colloids and lymphoscintigraphy, 44–45
anterior trunk, 25–26 staging, 85–86, 86 color-segmented fuorescence, 103
antigen-presenting cells (APCs), 11 breast-conserving surgery (BCS), 88 complete lymphadenectomy, 130–131, 135
ARGACY/GINECO randomized trial, 78 contralateral lymph node metastases, 36
ascites, 9 C contrast-enhanced ultrasound, 51
automated immunostainers, 57–58 Cooper’s ligaments, 39
axillary sheath, breast and axilla, 39 Cancer Australia, 75 counts per minute (CPM), 106
Cancer Care Ontario group, 46 cribriform fascia, 17, 20
cancer-specifc validated QoL instrument, 112 cribriform lamina, 17
B
carbon nanoparticles, 50–51 cutaneous lymphatics, 3–4
BCS, see breast-conserving surgery (BCS) cardinal ligaments, 24 cutaneous melanoma, 6
beta-human chorionic gonadotropin (β-HCG) caveats, 57, 74 cytokeratin 19 transcription, 59
RT-PCR, 59 Cerenkov luminescence imaging, endometrial cytoreductive interval surgery, 114
bilateral efferent drainage, 18 cancers, 106
bilateral inguinal femoral lymphadenectomy, cervical cancers, 6, 26, 31 D
3, 5–6 biopsy in clinical management, 74–78
bilateral lymph drainage, 66 clinical trial design, 136–138 dendritic cells, 11
bilateral pelvic lymphadenectomy, 7 natural history of, 26, 26 direct lymphatic drainage, 41
bilateral sentinel lymph nodes, 122 in pelvis, 71–74 disease-free survival (DFS) in women, 89
blind FNAB, 58 quality of life, sentinel lymph node distal interiliac nodal basins, 25–26
blue dye agents/tracer, 4, 44, 47 mapping on, 113–114 DNA aneuploidy, 59
breast cancer, 90, 91 sentinel lymph node mapping in, 71–78 DNA/cytokeratin double-staining procedure,
ICG dose on SLN mapping, 49 cervical injection, 100 59
mapping tracers, 44 of dye, 102 DNA FCM, 59
breast and axilla technique, 101 dose volume histogram (DVH), 119
axillary sheath, 39 cervix/cervical
internal mammary artery medially, 39 anterior trunk, 25–26 E
internal mammary nodes, 40–41 cervical lymphatics, topographic anatomy
lymphatic system, 39 of, 24–26 edema, 9–10
lymph nodes draining, classifcation, 41 cervix cancer, natural history of, 26, 26 electronic patient-reported outcomes measures
nodes level, 41 lateral trunk, 24–25, 25 (ePROs), 114
pectoralis major muscle, 39 lymphatic drainage, 1, 2, 4 empty lymph node packet syndrome, 45–46
primary blood supply, 40 lymphatics, topographic anatomy of, 24–26 en bloc tumor resections, 19
primary lymph nodes of, 40 lymphatics of, 23–26 endometrial adenocarcinomas, 11
Rotter’s interpectoral group, 40 midbody uterine drainage pathway, 30 endometrial cancers, 10, 31, 60, 101

143
144 Index

Cerenkov luminescence imaging, 106 fne-needle aspiration cytology, 58 hot node, 44


cervical injection of dye, 102 FIRES trial, 136 human epidermal growth factor receptor 2
clinical trial design, 135–136 fow cytometry (FCM), 58–59 (HER2), 86
detection using an algorithm, 103 Fluorescence Imaging for Lymphatic Mapping human papillomavirus (HPV) infection, 26
early studies in, 99–100 (FILM) randomized trial, 100 hybrid tracer, 68
endometrioid histology, 104–105 FNAB, see fne-needle aspiration biopsy
false-negative rates, 103–104 (FNAB) I
high-risk histology, 105–106 Food and Drug Administration (FDA), 47
lymphadenectomy vs., 104 frozen section, sentinel node, 55–56, 75 ICG dye, see indocyanine green (ICG) dye
lymph node status in, 99 functional evaluation questionnaire for breast iliac lymph nodes, 26
patterns in, 100, 101–102 cancer therapy (FACT-B), 112 image-guided radiotherapy, 120
positron lymphography, 106 fundus lymphatic channels, 29 imaging-enhanced modalities, 51
quality of life, sentinel lymph node immunohistochemical (IHC) evaluation,
mapping on, 113 57–58, 86
G
survivors, 115 imprint cytology, 56
technique by disease site, 47–48 Gamma photon emission energy, 44 India ink, 4, 51
techniques and examples, 102–103 gamma probe, 41, 44–47, 51, 58, 66, 91–92 indocyanine green (ICG) dye, 45–46, 68, 90, 100,
ultrastaging protocols, 57 generator-produced radioisotopes, 44 113, 135, see also blue dye agents/tracer
endometrial lymphatic anatomy, 29 glands sentinel, 9 blue dye, 49
endometrioid endometrial carcinoma, 104 groin anatomy, 16 cervical cancer, 48–49
endometrioid histology, 104–105 cribriform fascia, 20 indocyanine green, 49–50
endometrium cross-sectional anatomy, 20 ovarian cancer, 49
endometrial cancer, early studies in, 99–100 lymphatic anatomy, 19 safety, 49–50
endometrial cancer, lymph node status in, 99 operative and cross-sectional anatomy, 21 on SLN mapping, 48–50
epidemiology, 99 sentinel lymph nodes and inguinal femoral technetium, 49
Memorial Sloan Kettering Cancer Center anatomy, 20 inferior mesenteric artery (IMA), 100
surgical algorithm, 103 superfcial inguinal nodes, 20 infundibulopelvic ligament, 4, 49
pelvic lymphatic mapping patterns, 101 vulvar and inguinal lymphatic anatomy, 21 inguinal lymphatics, topography of, 15–17, 17
radiation oncology, 121–124 GROINSS-VII/GOG 270, 139 inguinal lymph nodes, radiation
sentinel lymph node biopsy of, 99–106 GROINSS-VIII, 121 oncology, 118
superfcial lymphatics, 29 GROINSV3/CV2020, 139 inguinal nodes
energy discrimination, 45 Groningen International Study on Sentinel and pelvic nodes, 3
epithelial marker cytokeratin, 19, 59 Nodes in Vulvar cancer (GROINSS-V I), radiation oncology, 118
epithelial ovarian cancer (EOC), 36 65, 67, 69, 121, 139 recurrence, 122
estrogen receptor (ER), 86 Gynecological Oncology Group, 65 inguinofemoral lymphadenectomy, 12–13,
European Organization for Research and gynecologic cancers/malignancies, 12 65, 112
Treatment of Cancer (EORTC), 113 management, 7 inguinofemoral lymph node dissection
European Organization for Research and quality of life, sentinel lymph node (IFLD), 139
Treatment of Cancer Quality of Life mapping on, 111–112 internal mammary artery medially, 39
Questionnaire-Core 30 (EORTC SN evaluation and ultrastaging in, 60, 60 internal mammary nodes, 40–41
QLQ-C30), 115 gynecologic oncology, 1, 11–12 International Atomic Energy Agency
European Society for Medical Oncology Gynecologic Oncology Group (GOG), 6, 20 guidelines, 49
(ESMO), 131 gynecologic tumors, 1, see also gynecologic International Breast Cancer Study Group
European Society of Gynecological cancers/malignancies 23–01, 89
Oncology, 123 International Commission on Radiological
exocervix, 23 H Protection, 49
extravisceral trunks, 24 International Gynecologic Cancer Society, 75
Halstedian principles, 5 International Sentinel Node Society, 7
hematogenous spread, breast cancer, 75 intraoperative identifcation techniques
F
hematoxylin-eosin (H&E) stain, 88 carbon nanoparticles, 50–51
FACT-B, see functional evaluation hepatobiliary insuffciency, 49 India ink, 51
questionnaire for breast cancer therapy high-reliability organizations (HROs), 7 [99mTechnetiumTc] tilmanocept, 50
(FACT-B) history superparamagnetic iron, 51
false-negative rate (FNR), 88, 103–104 abdomen and pelvis, lymphatics, 1, 2 intraoperative lymphatic mapping, 6
false-negative sentinel node, 7 cervix, lymphatic drainage, 1, 2, 4 intraoperative near-infrared fuorescence
FCM, see fow cytometry (FCM) inguinal nodes and pelvic nodes, 3 imaging, 46
Federation of Gynecologists and Obstetricians lymphatic anatomy, Cadaver studies of, 1 intraoperative probes, 45
(FIGO), 35, 99 lymphatic mapping concept, 6, 6–7 intravenous and submucosal
female genitalia, 16 penile lymphography, Cabanas’s technique administration, 45
female genitalia, external, 16 for, 5 in vivo injection of dyes, 3–4
Female Sexual Function Index questionnaire safety and surgical innovation, 7 isolated micrometastases, 89
(FSFI), 113 sentinel node identifcation, early efforts isolated tumor cells (ITCs), 85, 104, 123
femoral (deep) inguinal lymphatics, 17–18 at, 4–6 ITCs, see isolated tumor cells (ITCs)
FIGO, see Federation of Gynecologists and solid tumors, Halsted model for surgical
Obstetricians (FIGO) management, 3 K
fltered lymph, 45 in vivo injection of dyes, 3–4
fne-needle aspiration biopsy (FNAB), 58 vulvar lymphatics, 1, 2 Korean Society of Gynecologic Oncology, 75
Index 145

L empty lymph node packet syndrome, optimal standard protocol, sentinel node, 60
45–46 ovarian cancer, 35, 130–131, 131
labial-crural fold, 5 indocyanine green, 45–46 ovarian drainage pathway, 30
lateral trunk, 24–25, 25 MD Anderson Symptom Inventory ovary
Lower Extremity Functional Scale (LEFS), 113 (MDASI-OC), 114 and fallopian tube, lymphatic drainage, 36
lower leg lymphedema (LLL), 78 Medical Outcomes Study Short Form-12 lymphatic anatomy, 35
lymphadenectomy, 12, 99, 113 (SF-12) Health Survey, 113 ovarian cancer, sentinel node detection
bilateral inguinal femoral, 3, 5–6 melanoma, 7 in, 36
bilateral pelvic, 7 Memorial Sloan Kettering Cancer Center pelvic and aortic lymph node metastasis
complete, 130–131, 135 (MSKCC), 4–5, 90 distribution, 35–36
vs. endometrial cancers, 104 group, 100 studies, 35
inguinofemoral, 12–13, 65, 112 surgical algorithm, 103 overall survival (OS), 89
paraaortic, 104 metastable isotope, 44
pelvic, 1 metastatic carcinoma, sentinel lymph node
regional, 6–7 P
involved in, 94–95
lymph and lymphatic vessels, 10 metastatic lymph nodes, 31 paraaortic lymphadenectomy, 104
lymphangiogenesis, 10–11 methylene blue, 44 paraaortic nodes, 118
lymphangiography, 17 microanatomy and physiology, lymphatic paraffn-embedded tissue, 59
lymphangions, 10 anatomy parametrium, 48, 51, 71, 73, 123–124
lymphatic anatomy, Cadaver studies of, 1 lymph and lymphatic vessels, 10 parotid cancer, 4
lymphatic capillaries, blind terminal sacs lymph nodes, 10–11 patent blue-V dye, 90
of,  10 physiology, 10 pathologic complete response (pCR), 93
lymphatic channels, 29 regional lymph nodes, immunology, 11–12 Patient-Reported Outcomes Measurement
lymphatic drainage, 17–18 studies, 9 Information System (PROMIS), 114
fuid, 10 surgical anatomy, 12–13 patient-reported outcomes (PROs), 114–115
patterns, 4 micrometastases (MIC), 74–77, 123 pectoralis major muscle, breast and axilla, 39
routes, 29–30, 30 minimally invasive radical hysterectomy, 48 pelvic lymphadenectomy, 1
lymphatic dysfunction, 9 molecular techniques, sentinel node, 59–60 pelvic lymphatic mapping patterns, 101
lymphatic mapping monoclonal antibodies, breast cancer, 89 pelvic nodes, radiation oncology, 117–118
concept, 6, 6–7 morbidity, 112 pelvis, sentinel lymph node mapping in
studies in women, 18 Morton, D., 6 anatomical distribution, 72
for tumors, 1 Multicenter Selective Lymphadenectomy atypical localizations, 73
lymphatic spread of breast cancer, 9 Trial-I (MSLT-I), 132 cervix, lymphatic drainage from, 71
lymphatic system Multicenter Selective Lymphadenectomy deep lymphatic trunk, 72
breast and axilla, 39 Trial-II (MSLT-II), 132 detection rate, 73–74
studies of, 9 localization, 72–73
surgical anatomy, 12–13 lymphatic drainage, 71
lymphatic trunks, 12 N
pelvic dissection and SLN removal
lymphatic vessel endothelial hyaluronan National Cancer Comprehensive Network technique, 71–72
receptor 1 (LYVE1), 10 (NCCN), 75, 121, 131 pelvic lymph node staging, sensitivity
lymphedema, 10, 12–13, 112 National Cancer Institute of Canada MA.20 of, 74
lymph node dissection (LND), 136 study, 93 residual cervical stroma, tracer
lymph nodes, 10–11 National Council on Radiation Protection application, 73
anatomy, 11 (NCRP) recommendations, 49 SENTIX study, 74
draining, breast and axilla, 41 National Quality Forum (NQF), 114 superfcial lymphatic trunk, 72
involvement, 121 National Surgical Adjuvant Breast and Bowel penile carcinoma, 4–5, 87
metastases, 11 Project (NSABP) B-32 study, 89 penile lymphography, 4, 5
metastasis, 57–58 National Surgical Quality Improvement periodic acid-Schiff (PAS), 57
sampling, 130 Database (NSQIP), 7 perirectal lymph nodes, radiation oncology, 118
status in endometrial cancers, 99 negative predictive value (NPV), 135 PORTEC 3, 123
lymphocytes, 11 neoadjuvant chemotherapy (NAC), 90 positron emission tomography (PET), 50
from lymph nodes, 11 nodal metastases, 31, 89 positron lymphography, endometrial cancers, 106
tumor antigens, 11 nodal target delineation, radiation oncology, 117 posterior trunk, 25, 26
lymphogenous spread, breast cancer, 75 node of Cloquet, 3 postoperative chemoradiation therapy, 123
lymphoscintigram, 9, 90 nodes level, breast and axilla, 41 Post-Operative Radiation Therapy in
lymphoscintigraphy, 6, 10, 44 nonkeratinizing squamous epithelium, 23 Endometrial Cancer (PORTEC)
Lymphoseek, 50 non-sentinel node metastases, 68–69 groups, 99
lymphovascular space involvement (LVSI), 121 NSGO/EORTC/MaNGO trial, 123 pregnancy, mapping in, 50
lymphovascular systems, 45 nuclear medicine, 44–45, 51, 90 preoperative lymphoscintigraphy, 45–46
primary blood supply, breast and axilla, 40
M O primary lobular breast cancer, 57
primary lymph nodes of breast and axilla, 40
macrometastases (MAC), 60, 74–77, 123 obesity, mapping in, 50 progesterone receptor (PR), 86
MAGE-A3 RT-PCR, 59 oncotic proteins, 44 prospero homeobox protein (PROX1), 10
mapping tracers Oncotype DX genetic assay, 86 prostate, lymph node metastases of, 56
blue dye agents, 44 one-step nucleic acid amplifcation psycho-emotional decline, 113
colloids and lymphoscintigraphy, 44–45 (OSNA), 60 pulse oximetry, 49
146 Index

Q sentinel node, 1, 4, 7 cervical cancers, 31


biopsy, 1, 112 cervical/midbody uterine drainage
quality of life, sentinel lymph node mapping on fne-needle aspiration cytology, 58 pathway, 30
cervical cancer, 113–114 fow cytometry (FCM), 58–59 cervical tracer injection, 31
endometrial cancer, 113 frozen section, 55–56 endometrial cancers, 31
gynecologic cancer, 111–112 identifcation, 4–7 endometrial lymphatic anatomy, 29
lymphedema, 112 immunohistochemistry, 57–58 endometrium, superfcial lymphatics, 29
patient-reported outcomes (PROs), imprint cytology, 56 fundus lymphatic channels, 29
114–115 molecular techniques, 59–60 lymphatic channels, 29
vulvar cancer, 112–113 optimal standard protocol, 60 lymphatic drainage routes, 29–30, 30
recommendations for the SN Evaluation, 60 metastatic lymph nodes, 31
R serial sectioning, 57 nodal metastases, 31
ultrastaging of, 56–60 ovarian drainage pathway, 30
radiation oncology Sentinel Node Biopsy Following Neoadjuvant sentinel lymph node (SLN) mapping, 31, 32
dose volume histogram (DVH), 119 Chemotherapy (SN FNAC), 93 tumor metastases, 30–31
endometrium, 121–124 Sentinel Nodes in Early Stage Ovarian Cancer uterine corpus, lymphatic drainage
image-guided radiotherapy, 120 (SELLY) trial, 131 patterns, 29
inguinal lymph nodes, 118 serial sectioning, sentinel node, 57 utero-ovarian ligament, 49
inguinal nodes, 118 shielding, 45 Utility-Based Questionnaire-Cancer, 112
nodal target delineation, 117 Short Form 12 questionnaire (SF12), 113
paraaortic nodes, 118 simultaneously integrated boost (SIB), 121
pelvic nodes, 117–118 V
single photon emission computed tomography
perirectal lymph nodes, 118 (SPECT), 44, 135 vaginal cancer, 129, 129–130
protection, 49 sinus-lining (dendritic) cells, 57–58 vaginal epithelium, 23
radiation therapy techniques, 118–120, skip metastasis, 87 vascular endothelial growth factor receptor-3
119–120 sky-blue dye, 4 (VEGFR3), 10
therapy techniques, 118–120, 119–120 Society for Surgical Oncology (SSO), 132 vascular endothelial growth factor (VEGF), 11
volumetric modulated arc (VMAT) IMRT Society of Gynecologic Oncology (SGO), vault brachytherapy (VBT), 122
plan, 118, 119 102–103 vesicocervicovaginal space, 48
vulva, 120–121 solid tumors, Halsted model for surgical volumetric modulated arc (VMAT) IMRT
radical hysterectomy (RH), 78 management, 3 plan, 118, 119
radical vulvectomy, 3, 5–6 spatial resolution, 44–45 vulvar cancer, 7
radiocolloid tracers, 46 squamous cell carcinomas, 1, 11, 65 clinical trial design, 138–139
radiolabeled compounds, 6 stromal chemokines, 11 quality of life, sentinel lymph node
radionuclides injection, 6 superfcial inguinal lymphatics, 17 mapping on, 112–113
real-time RT-PCR assay, 59 superfcial inguinal nodes, 5, 20 radiation oncology, 120–121
regional lymphadenectomy, 6–7 superfcial intermediate group (Sf-IM), 17 technique by disease site, 46–47
regional lymph nodes, immunology, 11–12 superfcial lateral group (Sf-L), 17 vulva/vulvar
regional nodal irradiation (RNI), 93 superfcial medial group (Sf-M), 17 detection with combined technique, 66
reverse-transcriptase polymerase chain superparamagnetic iron, 51 embryology, 15
reaction (RT-PCR), 59 superparamagnetic iron oxide, 68 external female genitalia, 16
Rotter’s interpectoral group, breast and axilla, 40 surgical oncologist, lymphatic mapping, 1 femoral (deep) inguinal lymphatics,
Rotter’s nodes, 40 17–18
RxPONDER trial, 89 groin, topographic anatomy, 16
T
GROINSS-V protocol, 67, 69
S technetium-labeled colloid, 68 indication for, 67
technique by disease site inguinal lymphatics, topography of,
Sappey’s illustration, vulvar lymphatics, 1, 2 endometrial cancer, 47–48 15–17, 17
scattered keratin-positive cells, sentinel lymph vulvar cancer, 46–47 inguinofemoral lymphadenectomy, 67
node, 58 terminal lymphatics, 10 lymphatic drainage, 6, 17–18
second-echelon regional nodes, 3 triple injection technique, 51 lymphatics, 1, 2, 5, 15–19
sensitivity, 45 tumor metastases, 30–31 melanoma, 131, 131–133, 132
SENTICOL study, 136 tumor-node- metastasis (TNM) system, 85 natural history and patterns of spread,
SENTI-ENDO study, 113, 136 tumor-secreted factors, 11 18–19
sentinel lymph node biopsy (SLNB), 1, 11–12 near-infrared fuorescence imaging, 68
endometrium, 99–106 radioactive tracer, 67
U
vulva, 65–69 sentinel lymph node biopsy of, 65–69
Sentinel Lymph Nodes in Early-Stage Ovarian ultrastaging protocols, endometrial cancers, 57 sentinel node metastasis, 68–69
Cancer trial (SELLY), 36 unidirectional lymphatics fow, breast and sentinel node procedure, safety,
sentinel lymph node (SLN) mapping, 99–100 axilla, 39 65–67, 66
breast and axilla, 41, 41–42 uterine cancer superfcial inguinal lymphatics, 17
breast cancer, 83–95 carcinosarcoma, positron lymphography tissues, 16
cervical cancer, 71–79 (PLG), 106 topography, 15
cervical injection, location for, 48 cervical injection sites for mapping, 47 vulvar tissues, 16
endometrium, 99–106 uterosacral and cardinal ligaments, 24
mapping, 31, 32 uterosacral ligament, 25 W
sentinel lymph node technique, 46–47 uterus, lymphatics of
SENTInel NeoAdjuvant (SENTINA) study, 93 body lymphatic trunks, 48 whole pelvic radiation therapy (WPRT), 122

You might also like