Download as pdf or txt
Download as pdf or txt
You are on page 1of 382

Shear’s Cysts of 

the Oral and Maxillofacial Regions


Shear’s Cysts of the Oral and Maxillofacial Regions

Fifth Edition

Paul M. Speight, BDS, PhD, FDRCPS (Glasg), FDSRCS (Eng), FDSRCS (Edin), FRCPath
Professor Emeritus in Oral and Maxillofacial Pathology
School of Clinical Dentistry
University of Sheffield, UK
This fifth edition first published 2022
© 2022 John Wiley & Sons Ltd

Edition History
1e (1976); 2e (1983); 3e (1992); 4e (2007)

All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means,
electronic, mechanical, photocopying, recording or otherwise, except as permitted by law. Advice on how to obtain permission to reuse material
from this title is available at http://www.wiley.com/go/permissions.

The right of Paul M. Speight to be identified as the author of this work has been asserted in accordance with law.

Registered Offices
John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, USA
John Wiley & Sons Ltd, The Atrium, Southern Gate, Chichester, West Sussex, PO19 8SQ, UK

Editorial Office
9600 Garsington Road, Oxford, OX4 2DQ, UK

For details of our global editorial offices, customer services, and more information about Wiley products visit us at www.wiley.com.

Wiley also publishes its books in a variety of electronic formats and by print-­on-­demand. Some content that appears in standard print versions
of this book may not be available in other formats.

Limit of Liability/Disclaimer of Warranty


The contents of this work are intended to further general scientific research, understanding, and discussion only and are not intended and
should not be relied upon as recommending or promoting scientific method, diagnosis, or treatment by physicians for any particular patient.
In view of ongoing research, equipment modifications, changes in governmental regulations, and the constant flow of information relating
to the use of medicines, equipment, and devices, the reader is urged to review and evaluate the information provided in the package insert
or instructions for each medicine, equipment, or device for, among other things, any changes in the instructions or indication of usage and
for added warnings and precautions. While the publisher and authors have used their best efforts in preparing this work, they make no
representations or warranties with respect to the accuracy or completeness of the contents of this work and specifically disclaim all warranties,
including without limitation any implied warranties of merchantability or fitness for a particular purpose. No warranty may be created or
extended by sales representatives, written sales materials or promotional statements for this work. The fact that an organization, website, or
product is referred to in this work as a citation and/or potential source of further information does not mean that the publisher and authors
endorse the information or services the organization, website, or product may provide or recommendations it may make. This work is sold with
the understanding that the publisher is not engaged in rendering professional services. The advice and strategies contained herein may not be
suitable for your situation. You should consult with a specialist where appropriate. Further, readers should be aware that websites listed in this
work may have changed or disappeared between when this work was written and when it is read. Neither the publisher nor authors shall be
liable for any loss of profit or any other commercial damages, including but not limited to special, incidental, consequential, or other damages.

Library of Congress Cataloging-­in-­Publication Data


Names: Speight, P. M. (Paul M.) author. | Shear, Mervyn. Cysts of the oral
and maxillofacial regions.
Title: Shear’s cysts of the oral and maxillofacial regions / Paul M. Speight.
Other titles: Cysts of the oral and maxillofacial regions
Description: Fifth edition. | Hoboken, NJ : Wiley-Blackwell, 2022. |
Preceded by Cysts of the oral and maxillofacial regions / Mervyn Shear
and Paul Speight. 4th ed. 2007. | Includes bibliographical references
and index.
Identifiers: LCCN 2022000536 (print) | LCCN 2022000537 (ebook) | ISBN
9781119354994 (cloth) | ISBN 9781119354932 (adobe pdf) | ISBN
9781119354949 (epub)
Subjects: MESH: Jaw Cysts | Jaw Diseases–physiopathology. | Mouth
Diseases–physiopathology
Classification: LCC RC815 (print) | LCC RC815 (ebook) | NLM WU 140.5 |
DDC 617.5/22–dc23/eng/20220127
LC record available at https://lccn.loc.gov/2022000536
LC ebook record available at https://lccn.loc.gov/2022000537

Cover Design: Wiley


Cover Images: Courtesy of Paul M. Speight

Set in 9.5/12.5pt STIXTwoText by Straive, Pondicherry, India


Mervyn Shear
1931–2017
vii

Contents

Preface to the Fifth Edition  viii


Foreword  x
Acknowledgements  xi

1 Classification and Frequency of Cysts of the Oral and Maxillofacial Regions  1

2 General Considerations  6

3 Radicular Cyst  20

4 Inflammatory Collateral Cysts  47

5 Dentigerous Cyst  62

6 Eruption Cyst  83

7 Odontogenic Keratocyst  87

8 Lateral Periodontal Cyst and Botryoid Odontogenic Cyst  140

9 Gingival Cysts  155

10 Glandular Odontogenic Cyst  164

11 Calcifying Odontogenic Cyst  182

12 Orthokeratinised Odontogenic Cyst  201

13 Nasopalatine Duct Cyst  214

14 Nasolabial Cyst  230

15 Cysts of the Salivary and Minor Mucous Glands  237

16 Surgical Ciliated Cyst  262

17 Pseudocysts of the Jaws: Simple Bone Cyst and Stafne Bone Cavity  270

18 Developmental Cysts  288

Bibliography  309
Index  357
viii

Preface to the Fifth Edition

It is an honour to have prepared this fifth edition of diagnosis and management. This new edition maintains
Mervyn Shear’s classic text on cysts of the maxillofacial the same basic layout, but is restructured to present the
regions. This edition is dedicated to his memory and most common lesions first. Each chapter now includes
I am grateful to the publishers for agreeing to make the more detailed histopathology with more photomicrographs
book eponymous and have his name in the title. Mervyn and sections on radiological or histological differential
Shear wrote the first edition in 1976  in an attempt to diagnosis. There are also more detailed discussions of the
record, in a single volume, all published knowledge on historical aspects of the classification and naming of cysts
the subject. Subsequent editions were published after that I hope will be of general interest, but also provide
7 years (1983), 9 years (1992), and 15 years (2007), and some context for the global variation in terminology and
each continued to attempt to be a comprehensive record explain why there may be so much confusion in the litera-
of the literature. I was first honoured in this venture ture about some cyst types. We agreed not to include,
when I was asked to assist Professor Shear in the prepa- and to remove, detailed accounts of research findings
ration of the fourth edition that was published in 2007. that do not advance our understanding of the pathogen-
Even as we wrote it, we realised that the task was getting esis or assist in diagnosis. The applies especially to the
more difficult because of the massive proliferation of odontogenic keratocyst, where there has been a massive
new publications. The text was lengthened considerably, increase in publications in the last two decades, but little of
but the extra information was additive and we soon real- relevance to the practicalities of routine diagnosis and
ised that it was no longer possible to continue to try to treatment.
present a definitive account of the entire literature. At the outset, I had aimed to reduce the number of
Feedback from colleagues, and in particular students, references and especially to remove references to some
reported that the book had become too detailed and of the ‘old’ literature that current students and trainees
that much of the research reviewed had little relevance may not appreciate as relevant. In the final outcome
to the day-­to-­day practicalities of diagnosis and manage- about 250 references have been removed, but about
ment of cysts. 450  new references have been added. I hope that all
We agreed that a new approach was needed and in late these are relevant and helpful. Many younger students
2012 I was able to meet with Mervyn at his home near Cape may be surprised to find that I have retained many ‘old’
Town. At that time he was no longer able to participate in a papers, including some landmark studies going as far
new edition, but together we outlined a basic plan for the back as the early 1900s, but also studies carried out in
changes that we felt were necessary. This new fifth edition the two to three decades after 1950. Many of these papers
is a complete rewrite of the book, but still maintains the are freely available online and report unparalleled obser-
basic aim of providing an understanding of the pathogen- vational studies that will never be repeated, but are
esis of each cyst type as well as recording the clinical, radi- invaluable because of their original detailed observa-
ological, and histological features. The overall aim is to tions relating to the pathogenesis and histological fea-
assist pathologists and clinicians in making a correct diag- tures of many of the cyst types.
nosis and informing management, but we also wanted to As in all previous editions, I have attempted to produce
make the book more accessible to students and trainees at a book that is useful to students and trainees at all levels,
all levels, as well as to non-­specialist clinicians and general but also to practising clinicians, whether specialists or
pathologists faced with an individual lesion that requires general practitioners. Because the final diagnosis of most
Preface to the Fifth Edition ix

lesions lies in the hands of a pathologist, I have enhanced information is more accessible, allowing the reader to
the sections on pathology, histological features, and dif- rapidly ‘dip in’ to the book to find the information they
ferential diagnosis, and hope this will assist non-­specialist need. In this respect, the book may be a useful bench book
pathologists as much as oral and maxillofacial patholo- for the diagnostician, as well as a gripping read for the
gists. I have made greater use of subsections, so that keen student.
x

Foreword

­It is an honor to acknowledge and welcome the fifth edi- Cysts of the Oral and Maxillofacial Regions has become
tion of Cysts of the Oral and Maxillofacial Regions, or in the definitive source of information about this complex
today’s vernacular, Cysts of the Oral and Maxillofacial group of lesions. No other contribution covers this topic
Regions 5.0. The oral and maxillofacial region, because of more extensively and accurately than this text and there is
its anatomic complexity, is home to a variety of cysts and something here for everyone. Clearly it is applicable for
tumors unique to this site. Many of these arise from the students, both predoctoral and postgraduate, as well as to
epithelial components that form our teeth and are known radiologists, pathologists, researchers, surgeons, and other
as odontogenic cysts. Many of these cysts have proved to be clinicians.
more biologically complex than originally thought as the As scientific inquiry has exploded in recent years and the
molecular pathogenesis has been investigated and reported. publication interval of the WHO series of classification of
This has blurred the distinction between cysts and cystic tumors has been shortened significantly, so too is it likely
neoplasms and the significance of the molecular landscape that the sixth edition of this text might be just over the hori-
characterizing some of these cysts is widely and energeti- zon. But a lot has been learned since 2007, and detailed
cally debated today. description and analysis of this new knowledge are finally
The evolution of Cysts of the Oral and Maxillofacial available. One of the primary functions of a textbook is­
Regions has been remarkable: first edition (1976), second not just to convey what is new, but to analyze what has­
edition (1983), third edition (1992), fourth edition (2007), been published and then condense and summarize that
fifth ed (2022); from a concise text focusing on clinical/ information with an aim toward diagnostic and clinical­
radiographic features and standard histologic descriptions relevancy. Professor Speight has done a masterful job of
illustrated in black and white, to an exhaustive review of condensing our knowledge of these cysts into a readable
every aspect of these cystic lesions richly illustrated in and relevant format. This text is a must for anyone involved
color. Some of the historical literature has been elimi- in the diagnosis and treatment of cysts of the head and
nated, particularly where it lacked clinical relevance. The neck. Improving our understanding of this group of lesions
current literature is extensively recorded, as is the diver- improves the quality of care we can provide our patients.
sity of the histologic spectrum, including immunohisto- And ultimately, that is what it’s all about.
chemical phenotypes and molecular/genetic alterations.
Two of the world’s most experienced and respected oral John M. Wright, DDS, MS
pathologists are responsible for this text. The text was ini- Regents Professor
tiated by Professor Mervyn Shear from Johannesburg, and Diagnostic Sciences
Professor Paul Speight of the UK joined as a co-­author in Texas A&M University College of Dentistry
2007, and he currently continues the legacy of this major Dallas, Texas
contribution. USA
xi

Acknowledgements

This edition is dedicated to the memory of Mervyn Shear, their patience, for many discussions and critical com-
who must first be acknowledged for his research and schol- ments on the text, and for their assistance in retrieving
arship over many years that laid the foundations for all the and photographing cases. Daniel Brierley, Geoff Craig,
editions of this book. For this edition I specifically acknowl- Lisette Collins, Paula Farthing, Keith Hunter, and Ali
edge Mervyn’s support and permission to restructure the Khurram have commented freely on my thoughts and
book and to undertake a complete rewrite of the text, with ideas about cysts and have also provided assistance in find-
an increased emphasis on some basic principles and on ing appropriate cases for the illustrations. Chris Franklin
histopathology and differential diagnosis that may broaden and Adam Jones have allowed me access to their data on
the scope of the book, making it more accessible to a wider the incidence of cysts and oral biopsies.
range of students and clinicians. Many colleagues have selflessly provided clinical pic-
As in previous editions, we have relied heavily on the sup- tures, radiographs, and photomicrographs, and a number
port and assistance of colleagues and on the research and of publishers have allowed us to reproduce figures that
scholarship of our predecessors. In particular, I would like to have been previously published. For this we are very
draw attention to a number of outstanding giants of the sub- grateful, and each has been acknowledged in the relevant
ject on whose shoulders we stand, and who are heavily cited figure legends.
in the text or with whom, over many years, we have shared A major aim of this new edition was to broaden the
and discussed cases: Mario Altini, Jerry Bouquot, Roger readership and make the text more accessible to stu-
Browne, Roman Carlos, Geoff Craig, Ricardo Gomez, Robert dents, trainees, and non-­specialist pathologists and cli-
Gorlin, Malcolm Harris, Jos Hille, Fumio Ide, Ivor Kramer, nicians. A number of colleagues have been kind enough
TieJun Li, Hans Philipsen, Jens Pindborg, Finn Prætorius, to read early drafts of chapters and freely provided very
Peter Reichart, Paul Stoelinga, Takashi Takata, Paul Toller, helpful and constructive comments that have facilitated
Willie van Heerden, Pablo Vargas, and John Wright. Some of this aim: Daniel Brierley, Lisette Collins, Paula Farthing,
these are no longer with us, some I have never met, but most Ali Khurram, Liam Robinson, Willie van Heerden, and
have become good friends and colleagues. John Wright.
For the preparation of this fifth edition I would like to I am also especially grateful to my colleague and friend
thank my colleagues, past and present, in the Unit of Oral Professor John Wright, who very kindly agreed to write the
and Maxillofacial Pathology, University of Sheffield, for forward to this edition.
1

Classification and Frequency of Cysts of the Oral and Maxillofacial Regions

CHAPTER MENU
Classifications, 2
Cysts of the Jaws, 2
●● Odontogenic Cysts,  2
–– Odontogenic Cysts of Inflammatory Origin,  3
–– Odontogenic Cysts of Developmental Origin,  3
●● Non-­odontogenic Cysts and Pseudocysts,  3
–– Non-­odontogenic Cysts of the Jaws,  3
–– Pseudocysts of the Jaws,  3
Cysts of the Salivary and Minor Mucous Glands, 3
●● Cysts of the Major and Minor Salivary Glands,  3
●● Cysts of the Maxillary Sinus,  3
Developmental Cysts of the Head and Neck, 3
Frequency of Cysts of the Oral and Maxillofacial Regions, 4

There is no single satisfactory classification of cysts of relevant for clinicians who treat the lesions. A straightfor-
the head and neck region. In part this is because terminol- ward uniform nomenclature facilitates communication
ogy varies across the world, but also because classification between clinical specialties and enables precise reporting
systems may be developed to serve different purposes. of lesions for statistical and research purposes.
Some authors have tried to subdivide lesions into multiple International standards for classifications were first
variants based on histological or clinical features, but this developed by the World Health Organization (WHO),
has little clinical utility in terms of planning management. which set up a global project for the histological classifica-
Detailed classifications that include variants are neverthe- tion of tumours in 1952 (reviewed by Sobin 1971). The pro-
less useful for research, and occasionally a reported variant ject involved collecting samples of lesions that were
may eventually emerge as a new entity. A well-­known reviewed by groups of international experts, who agreed a
example of this is the reporting of an orthokeratinised vari- uniform nomenclature and established practical and clini-
ant of the odontogenic keratocyst (Wright  1981), which cally relevant diagnostic criteria. The first classification of
over time was shown to have distinctive clinicopathologi- cysts of the jaws was published in 1971  in Histological
cal features and is now recognised as an entity  – the Typing of Odontogenic Tumours, Jaw Cysts and Allied
orthokeratinised odontogenic cyst. Other variants, how- Lesions (Pindborg and Kramer 1971). This first edition was
ever, such as the bay or pocket variant of the radicular cyst, deliberately inclusive and provided a comprehensive clas-
have little bearing on clinical management and are of aca- sification of jaw lesions so that all neoplasms and cysts of
demic interest only. There is a tendency for classifications the odontogenic tissues could be considered in context,
to be overcomplicated by pathologists, who often describe allowing pathologists and clinicians to make an informed
subtle histological variations as new ‘entities’. The most diagnosis. The second edition, published in 1992, also
useful classifications should be simple, should use globally included cysts of the jaws (Kramer et al. 1992), but the third
recognised terminology, and should be easy to use and edition (Barnes et al. 2005) omitted cysts and restricted the

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
2 Classification and Frequency of Cysts of the Oral and Maxillofacial Regions

classification to tumours and a range of ‘tumour-­like’ epithelial lining or not, and we therefore retain the diagnos-
lesions. Subsequently, the fourth (El-­Naggar et al. 2017) and tic term mucous extravasation cyst for the non-­epithelial
fifth editions (WHO  2022a,  b) have included the odonto- lined cystic lesion caused by spillage of mucus into the
genic cysts and have restored the status of the book as a ­tissues (see Chapter 15). Similarly, we are content to use the
complete classification of lesions of the odontogenic tissues. well-­recognised diagnostic term simple bone cyst for the non-­
It is important to note that the WHO classifications still epithelial lined bone cavity that clinically and radiologically
maintain the original principles of simplicity, relevance, and presents as a cystic lesion (Chapter 17). It should be noted
a uniform and well-­recognised terminology. Thus the WHO that the definition of cyst does not include a cavity caused by
books should be regarded as a guide to terminology, defini- an accumulation of pus, which is defined as an abscess.
tions, and diagnostic criteria. They are not comprehensive In this book we do not attempt to suggest a definitive
and do not include detailed considerations of variants, unu- classification of cysts of the oral and maxillofacial regions,
sual features, or differential diagnosis. In this book we have but we divide them into cysts of the jaws, cysts of the salivary
adopted a simple working classification of the odontogenic and minor mucous glands, and developmental cysts of the
cysts using the WHO terminology. We do not include head and neck. We discuss cysts that are specific to the
detailed subdivisions or variants in the classifications, but in maxillofacial regions, and cysts that are not peculiar to
each chapter we discuss terminology and classification, and these regions are not included unless they have distinctive
include details of variants of each lesion where this might features that must be considered in the differential diagno-
have a bearing on diagnosis or management. sis. Cystic neoplasms such as unicystic ameloblastoma are
A further consideration is the use of the term ‘cyst’, not included either, although the possibility of a neoplastic
which still causes disagreement and some controversy. In origin of some of the odontogenic cysts is considered.
pathology dictionaries and general pathology textbooks, a Our classification is intended to be simple and to be par-
cyst is usually defined as a closed capsule, cavity, or sac-­like ticularly useful to pathologists and clinicians who must share
structure that may be empty or have fluid or semi-­fluid and understand definitions and terminology. Many other
contents. There is no requirement for a defined lining. classifications have been published and may well be perfectly
Most contemporary oral pathology textbooks, however, satisfactory. Although readers may use any classification they
choose to define a cyst as a pathological cavity lined by epi- find valuable as an aid to memory and understanding, they
thelium. Cyst-­like spaces not lined by epithelium have been are encouraged to facilitate communication by using the
described as pseudocysts and diagnostic terms such as ‘cav- WHO terminology and definitions (WHO 2022a, b).
ity’ have been used. Kramer (1974) defined a cyst as ‘a
pathological cavity having fluid, semifluid or gaseous con-
tents and which is not created by the accumulation of pus’. ­Classifications
He discussed the terminology and was bemused by the
requirement for an epithelial lining, stating that he was puz- In this edition of the book, the cysts are classified under
zled by ‘the definition that demands an epithelial lining’ and three broad categories:
said: ‘I am not sure how, or why, the presence of an epithelial Cysts of the jaws
lining became included in the definition: clearly it creates ●● Odontogenic cysts (Chapters 3–12)

difficulties, because so many lesions that have been accepted ●● Non-­odontogenic cysts (Chapters 13, 14, 16, and 17)

for generations as “legitimate” cysts must now be termed Cysts of the salivary and minor mucous glands (Chapter 15)
pseudocysts, or false cysts, or “cysts” in quotation marks’. Developmental cysts of the head and neck (Chapter 18)
The requirement for an epithelial lining probably became
enshrined in oral pathology because the vast majority of
cysts of the oral and maxillofacial tissues are odontogenic in
­Cysts of the Jaws
origin and are lined by epithelium. Thus, the use of alterna-
tive terms such as ‘pseudocyst’ or ‘cavity’ clearly distin-
Odontogenic Cysts
guishes cystic lesions without epithelium from odontogenic
cysts. Although we recognise the term pseudocyst for lesions The odontogenic cysts have been divided into cysts of
that are not lined by epithelium (see Chapter 17), we also inflammatory origin and cysts of developmental origin.
agree with Kramer and suggest that ‘cyst’ can be used as a These are convenient categories, since it is clearly under-
diagnostic term for lesions that are clinically or radiologi- stood that the inflammatory odontogenic cysts arise as a
cally cystic. This is in keeping with common usage and result of proliferation of odontogenic epithelium driven by
understanding among clinicians and radiologists. For exam- chronic inflammation, resulting from either pulpitis or
ple, most clinicians, especially paediatric dentists, use the pericoronitis. The pathogenesis of the developmental cysts
term cyst to describe mucoceles, whether they have an is less well understood, however, and in some cases there is
­Cysts of the Jaw  3

evidence for a neoplastic origin. The pathogenic mecha- radiolucencies and must also be considered in the differen-
nisms involved in cyst development are discussed in tial diagnosis.
Chapter 2, and details for each cyst type are presented in
each chapter. Although these categories are widely used, Non-­odontogenic Cysts of the Jaws
they are not definitive, since some cysts classified as devel- Nasopalatine duct cyst
opmental may have an inflammatory origin. In particular, Nasolabial cyst
a variant of dentigerous cyst may be inflammatory in Mid-­palatal raphe cyst of infants (Epstein pearls)
nature (see Chapter 5). In each category the cysts are listed Surgical ciliated cyst
in order of their approximate frequency (Tables 1.1–1.3).
Pseudocysts of the Jaws
Odontogenic Cysts of Inflammatory Origin Simple bone cyst
Radicular cyst Stafne bone cavity
●● Residual cyst Osteoporotic bone marrow defects
Inflammatory collateral cysts
●● Paradental cyst
Cysts of the Salivary and Minor Mucous Glands
●● Mandibular buccal bifurcation cyst

Cysts affecting the salivary and minor mucous glands of the


Odontogenic Cysts of Developmental Origin head and neck are common and may be developmental or
Dentigerous cyst reactive in nature. Retention and extravasation cysts
●● Eruption cyst (mucoceles) are the most common and may arise at any site
Odontogenic keratocyst associated with minor glands that are found throughout the
Lateral periodontal cyst submucosa of the upper aerodigestive tract and paranasal
●● Botryoid odontogenic cyst sinuses. Here we include cystic lesions of the major salivary
Gingival cyst of adults glands as well as cysts associated with minor glands of the
Gingival cyst of infants oral cavity and maxillary sinus. Ranula is included as a sepa-
Glandular odontogenic cyst rate lesion because it has distinctive and specific clinical fea-
Calcifying odontogenic cyst tures and problems of management. Cystic neoplasms are
Orthokeratinised odontogenic cyst not included. Intraoral lymphoepithelial cysts are included
in this category even though their origin is uncertain. Some
arise from intraoral tonsillar tissue, while others appear to be
Non-­odontogenic Cysts and Pseudocysts
associated with dilated ducts of minor salivary gland.
Non-­odontogenic cysts of the jaws are mostly developmen-
tal in origin and arise from vestigial epithelial remnants of Cysts of the Major and Minor Salivary Glands
ductal structures or from inclusions at the line of fusion of Mucoceles
the palatal shelves. The nasolabial and mid-­palatal raphe ●● Mucous extravasation cyst

cyst actually occur in the soft tissues, but are so closely ●● Mucous retention cyst

apposed to the maxillary bone that they are included in the ●● Ranula

classification of jaw cysts. The surgical ciliated cyst is Salivary duct cyst (of the major glands)
included here because it arises within the alveolar bone of Intraoral lymphoepithelial cyst
the maxilla. Pseudocysts are not epithelial lined, but are Lymphoepithelial cysts of the parotid gland
included because they are important in the radiological dif- Polycystic disease of the parotid gland
ferential diagnosis of cystic jaw lesions. As discussed above,
we are content to use ‘cyst’ as a diagnostic term for the sim- Cysts of the Maxillary Sinus
ple bone cyst, since this is clearly understood and widely Mucoceles
used by clinicians who recognise that they present clini- Retention cyst
cally and radiologically as a cystic lesion. Stafne bone cav- Pseudocysts
ity is neither a cyst nor a pseudocyst, but is an anatomical
anomaly causing an indentation of the mandible that
Developmental Cysts of the Head and Neck
appears as a cystic lesion on radiology or imaging. It is
often included in classifications and we include it here These cysts are mostly congenital and are usually pre-
because of its importance in the radiological differential sent at birth, although some may grow slowly and not
diagnosis of cystic lesions. Osteoporotic bone marrow become clinically apparent until later in childhood or
defects are controversial lesions, but they present as cystic adolescence. The majority arise from epithelial
4 Classification and Frequency of Cysts of the Oral and Maxillofacial Regions

remnants entrapped during fusion of the facial processes Although the actual frequencies vary, the relative frequencies
or due to incomplete obliteration of the branchial clefts and the rank order of the lesions are very similar. In these
or pouches. studies, and in all studies worldwide (Table  1.3), the most
common odontogenic cyst is the radicular cyst, followed by
Dermoid and epidermoid cysts dentigerous cyst and then odontogenic keratocyst. The naso-
Cysts of foregut origin palatine duct cyst is the most common non-­odontogenic cyst
●● Heterotopic gastrointestinal cyst and in some studies has a similar frequency among all jaw
●● Bronchogenic cyst cysts to the odontogenic keratocyst (Table 1.1). All the other
Branchial cleft cysts cyst types are relatively rare.
Thyroglossal duct cyst These data show the relative frequency of each cyst as a
Nasopharyngeal cyst proportion of all cyst types, but do not allow a clinician to
Thymic cyst determine how likely it is that they will encounter a cyst
in everyday practice. Jones and Franklin (2006a) reviewed
over 44 000 histologically diagnosed oral and maxillofa-
­ requency of Cysts of the Oral
F cial lesions in adults over a 30-­year period. Their data
and Maxillofacial Regions show that all the cyst types described in this book repre-
sent about 20% of all biopsies received (n  =  8354).
Frequency statistics differ from incidence studies in that Odontogenic cysts were by far the most commonly
they are not standardised against known population data, encountered (n  =  6052) and the overall most common
such as age, sex, and ethnicity. For data to be comparable cyst type in the maxillofacial regions was the radicular
between populations and internationally, age-­standardised cyst. They found 3793 radicular cysts (including residual
incidence rates per 100 000 are compared with a standard cyst), representing 8.6% of all biopsy specimens received.
world population. Incidence data are a requirement for all
national cancer registries, but most benign lesions, includ- Table 1.1  Distribution of 3481 jaw cysts according
ing cysts, are not registered and thus incidence data is not to diagnosis.
available for the odontogenic cysts. Epidemiological data are
therefore presented as the relative frequency of each cyst % of
type as a proportion of the total number of cysts encoun- n % of group all cysts
tered within a population, or of the total number of speci-
Odontogenic cysts
mens received. This gives clinicians an estimate of the
Radicular/residual cyst 1825 60.6 52.4
likelihood of encountering these lesions in everyday practice.
Frequency studies are rarely based on the general popula- Dentigerous cyst 599 19.9 17.2
tion, but are usually derived from archival records of diagno-   Eruption cyst 27 0.9 0.8
ses made in a hospital department, usually pathology Odontogenic keratocyst (including 355 11.8 10.2
departments. While these provide useful data on the behav- orthokeratinised odontogenic cysts)
iour and treatment of different diseases, they are of limited Inflammatory collateral cysts 109 3.6 3.1
use in international comparative studies. Table 1.3 shows the Calcifying odontogenic cyst 28 0.9 0.8
wide variation in the frequency of the three most common Lateral periodontal cyst 24 0.8 0.7
odontogenic cysts in different parts of the world. Almost Gingival cyst of adults 21 0.7 0.6
without exception, these data are derived from retrospective
Unclassified 18 0.6 0.5
analyses of pathology records and the frequency of each cyst
Glandular odontogenic cyst 6 0.2 0.2
type may depend on local protocols for patient referral and
3012 100.0
management, or even on individual pathologists’ criteria for
diagnosis. For example, a high frequency of radicular cysts Non-­odontogenic cysts
may reflect a high caries rate in the local population, or a high Nasopalatine duct cyst 404 86.1 11.6
rate of referral of periapical lesions. Conversely, a low fre- Simple bone cyst 35 7.5 1.0
quency of radicular cysts may arise if the local practice is not Nasolabial cyst 21 4.5 0.6
to submit periapical lesions for ­histological analysis. In Surgical ciliated cyst 5 1.1 0.1
Chapter 4 we discuss the very low frequency of paradental Mucosal cyst of maxillary antrum 4 0.9 0.1
cysts in some countries, where the lesion does not seem to be
469 100.0
recognised as an entity and is therefore not diagnosed.
Total 3481 100.0
Tables 1.1 and 1.2 present our experience of the frequen-
cies of jaw cysts in South Africa and the United Kingdom. Source: Data courtesy of Prof. M. Shear, University of Witwatersrand.
­Frequency of Cysts of the Oral and Maxillofacial Region  5

Dentigerous cysts represented 2.5% (n = 1081) of all biop- encountering a cyst. Common diagnoses included 6458
sies, and odontogenic keratocyst was 1.3% (n = 591). By cases of fibrous hyperplasia (14.7%), 2973 cases of lichen
comparing these data to other commonly encountered planus (6.8%), and 1901 epulides (4.3%; fibrous epulis,
lesions, a clinician can estimate the likelihood of pyogenic granuloma, or giant cell epulis). During the
same period, there were 3547 periapical granulomas,
Table 1.2  Distribution of 7121 odontogenic cysts in a United showing that the frequency of periapical granulomas
Kingdom population. (8.1%) and radicular cysts (8.6%) is similar. This was also
suggested by Koivisto et  al. (2012), who reviewed 9723
Cysts n % radiolucent lesions associated with the teeth (dentigerous
cysts and lesions in the ramus were excluded) and found
Radicular cyst 3724 52.3 that 73% were periapical granulomas or cysts. There were
  Residual cyst 573 8.0 3215 (33.1%) radicular cysts and 3931 (40.4%) periapical
Dentigerous cyst 1292 18.1 granulomas. The next most common lesion was the odon-
  Eruption cyst 15 0.2 togenic keratocyst (8.8%; n = 857).
Odontogenic keratocyst (including 828 11.6 Among other cyst types, Jones and Franklin (2006a)
orthokeratinised odontogenic cysts) found that the most common were mucoceles (3.9%;
Inflammatory collateral cysts 402 5.6 n = 1720), while all other cysts were rare (less than 1.0%).
Unclassified odontogenic cysts 210 2.9 These data show that cysts are relatively common and
Lateral periodontal cyst 28 0.4 that the most commonly encountered are the radicular
cyst, dentigerous cyst, and mucoceles. They also suggest
Calcifying odontogenic cyst 21 0.3
that when a periapical radiolucency is seen, about 50% will
Gingival cyst of adults 16 0.2
be a radicular cyst and 50% will be a periapical granuloma.
Glandular odontogenic cyst 11 0.2 Details of the frequency and incidence of each cyst type are
Gingival cyst of infants 1 0.0 illustrated and discussed in the following chapters.
Total 7121 100.00

Source: Data from Jones et al. (2006).

Table 1.3  Frequency (%) of the three most common odontogenic cysts in selected case series with a wide geographical distribution.

References Country N Radicular cysta Dentigerous cystb Odontogenic keratocyst Other Males (%)

Daley et al. (1994) Canada 6847 65.2 24.1 4.9 5.8 NR


Mosqueda-­Taylor et al. (2002) Mexico 856 42.1 33.0 21.5 3.4 53.1
Meningaud et al. (2006) France 695 58.2 22.3 19.1 0.5 65.0
Jones et al. (2006) UK 7121 60.3 18.4 11.6 8.1 55.9
Ochsenius et al. (2007) Chile 2944 61.9 18.5 14.3 5.3 52.8
Grossmann et al. (2007) Brazil 2812 63.0 26.1 7.4 3.5 50.0
Tortorici et al. (2008) Italy (Sicily) 1273 84.5 11.4 1.3 2.8 53.9
Ali (2011) Kuwait 196 52.6 26.0 15.3 6.1 57.6
Sharifian and Khalili (2011) Iran 1227 45.8 24.7 19.4 10.1 57.1
Ramachandra et al. (2011) India 252 50.3 22.4 27.4 NI 61.2
Manor et al. (2012) Israel 285 56.1 28.8 8.0 7.0 59.7
Soluk Tekkesin et al. (2012b) Turkey 5003 65.6 10.6 20.9 2.9 57.6
Tamiolakis et al. (2019) Greece 5165 73.2 14.8 8.4 3.6 61.5
Bhat et al. (2019) India 125 60.8 22.4 13.6 3.2 67.9
Kammer et al. (2020) Brazil 406 53.4 14.0 15.0 17.5 56.7
Aquilanti et al. (2021) Italy 2150 57.0 23.5 13.0 6.5 63.3

N, total number of odontogenic cysts – frequencies are proportions of odontogenic cysts only; NI, not included – proportions only given for the
three main cyst types; NR, not reported.
a
 Data for radicular cyst includes residual cysts.
b
 Data for dentigerous cyst includes eruption cysts.
6

General Considerations

CHAPTER MENU

Pathogenesis of Cysts, 6
The Cyst–Tumour Interface, 10
An Approach to the Diagnosis of Cysts of the Jaws, 12
●● Radiology of Cysts of the Jaws,  12

●● Histopathological Examination of Cysts,  15

–– Immunohistochemistry and Molecular Pathology,  15

Cysts of the oral and maxillofacial regions are common


Box 2.1  Pathogenesis: The Phases of Cyst Formation
and represent about 20% of all lesions encountered in an
oral and maxillofacial pathology department (Jones and Three elements are needed:
Franklin  2006a,b; discussed in Chapter  1). Clinicians are
●● A source of epithelium
often therefore called upon to make an informed diagnosis
●● A stimulus for epithelial proliferation
and implement correct management. Of all the cysts dis-
●● A mechanism of growth and bone resorption
cussed, those within the jaw bones are the most challenging
to diagnose. Overall the most common jaw cyst is the radicu- The cyst develops in three phases:
lar cyst, which presents as a periapical radiolucency and is
●● Phase of initiation  – a source of epithelium and
probably the most common cause of a bony swelling in the
stimulus for proliferation
tooth-­bearing areas of the jaws. The challenge is to accurately
●● Phase of cyst formation – a cyst cavity develops and
make a diagnosis and exclude other possible causes of a
becomes lined by epithelium
swelling or of a radiolucency. In most cases, a final diagnosis
●● Phase of growth and enlargement – the cyst enlarges,
usually requires histological examination of the cyst, and it is
and growth is accompanied by tissue remodelling
the histopathologist who often takes responsibility for bring-
and bone resorption
ing together the clinical, radiological, and histological fea-
tures and reporting the final diagnosis to the surgeon. Each
cyst type has characteristic features and these are discussed enlargement (Box  2.1). For the inflammatory odontogenic
and illustrated in each chapter of this book. In this chapter cysts, the processes of cyst formation and expansion are well
we consider general issues that help inform a careful and understood and are considered in detail in Chapters 3 and 4,
accurate approach to the diagnosis of cysts, and we summa- but for the developmental cysts the mechanisms are not so
rise specific radiological and histological features that have clear and many theories have been suggested, including aber-
diagnostic utility in the diagnosis of different cyst types. rant developmental processes, underlying genetic abnormali-
ties, and neoplasia. These are discussed in detail for each cyst
type in the following chapters. For most cysts, the lining is
­Pathogenesis of Cysts derived from epithelial remnants or inclusions that remain in
the tissues after developmental processes are complete.
The formation of a cyst requires three elements and can Table 2.1 shows the source of epithelium for each cyst type
be considered to develop in three phases: a phase of initia- and summarises the developmental origin. To fully under-
tion, a phase of cyst formation and a phase of growth and stand the pathogenesis of cysts, it is therefore essential to have

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
­Pathogenesis of Cyst  7

Table 2.1  Sources of the epithelial lining of cysts of the head and neck.

Source of epithelial lining Developmental origin

Odontogenic cysts
Radicular cyst Cell rests of Malassez Remnants of the epithelial root sheath of Hertwig lie in the
periodontal ligament (Figure 3.6)
Dentigerous cyst Reduced enamel Reduced enamel epithelium forms from the internal and
Eruption cyst epithelium external enamel epithelium and embraces the fully formed
Inflammatory collateral cysts crown of an unerupted tooth. This gives rise to the dentigerous
(and eruption) cyst (Chapters 5 and 6, Box 5.3, Figures 5.18 and
5.19). The reduced enamel epithelium also forms the junctional
or sulcular epithelium during tooth eruption and this gives rise
to inflammatory collateral cysts (Chapter 4)
Odontogenic keratocyst Cell rests of the dental After tooth formation is complete the dental lamina
Lateral periodontal cyst lamina (‘glands of disintegrates, but residual islands are retained in the gingival
Botryoid odontogenic cyst Serres’) mucosa and alveolar bone. Cell rests are particularly common
in the posterior mandible, where they may also be found in the
Gingival cyst of infants gubernacular cord or canal (discussed in detail in
Gingival cyst of adults Chapters 7, 8, 9, and 12; see Figures 7.12, 8.3, 9.7, and 9.9)
Glandular odontogenic cyst
Calcifying odontogenic cyst
Orthokeratinised odontogenic cyst

Non-­odontogenic cysts
Nasopalatine duct cyst Remnants of the The nasopalatine duct is a fetal structure and involutes at about
nasopalatine duct 10 weeks of intrauterine life. Residual epithelial remnants,
however, may remain in the incisive canal after birth and in
adults (Chapter 13, Figure 13.1, Box 13.1)
Nasolabial cyst Nasolacrimal duct The nasolacrimal duct forms after 6 weeks of intrauterine life
and drains tears from the lacrimal sac to the lower aspect of the
lateral nasal wall. Residual epithelial remnants may persist at
the inferior portion of the duct (Chapter 14)
Mid-­palatal raphe cyst (Epstein Epithelial inclusions The palatal shelves fuse at about 7–8 weeks of intrauterine life.
pearls) The epithelial coverings fuse and then break down into many
small islands, many of which form small ‘microcysts’ (Figure 9.7).
Most involute before birth, but up to 80% of newborns may have
cysts up to 3 months of age (Chapter 9). There is some evidence
that inclusions may arise in the bone and give rise to an
intraosseous ‘median palatal cyst’ (discussed in Chapter 13)
Surgical ciliated cyst Remnants of respiratory Fragments of sinus epithelium become implanted into a wound
epithelium following surgery involving the maxillary sinus (Chapter 16)

Cysts of the salivary and minor mucous glands


Mucous retention cysts Ducts of minor mucous The ducts of minor mucous glands become blocked and dilated,
glands most often as a result of trauma (Figures 15.5, 15.6, and 15.10)
Salivary duct cyst Intraparotid ducts The pathogenesis is uncertain, but blockage of ducts associated
Lymphoepithelial cysts with sialadenitis may cause cystic dilatation (Figure 15.12)
Intraoral lymphoepithelial cysts Tonsillar crypt The opening of intraoral tonsils may become blocked, causing
epithelium cystic dilatation of the crypt. Some intraoral lymphoepithelial
cysts may be retention cysts arising from a superficial duct of
minor salivary glands

Developmental cysts of the head and neck


Intraoral dermoid and Epithelial inclusions Epithelial remnants are sequestered into the tissues during
epidermoid cysts fusion of facial processes. Oral dermoid and epidermoid cysts
are found in the anterior oral cavity at sites of fusion of the
mandibular processes (Box 18.1)
(Continued)
8 General Considerations

Table 2.1  (Continued)

Source of epithelial lining Developmental origin

Intraoral cysts of foregut origin Epithelial inclusions Oral foregut cysts arise from epithelial remnants following
fusion of the tuberculum impar (first branchial arch) and the
posterior one-­third of the tongue (second to fourth arches)
(Box 18.2)
Branchial cleft cysts Epithelial inclusions Branchial cleft cysts arise from epithelial remnants that become
entrapped or persist due to incomplete obliteration of the
branchial clefts or pharyngeal pouches (Box 18.3)
Thyroglossal duct cyst Remnants of the The thyroid gland develops at about 4 weeks of intrauterine life
thyroglossal duct in the dorsum of the tongue. The developing gland descends
downwards into the upper neck forming the thyroglossal duct,
which then disintegrates. However, residual epithelial
remnants are found in the midline of the upper neck and
tongue in about 40% of people (Box 18.4)

an understanding of the embryology and development of the tissues adjacent to the teeth after eruption. The disintegrating
head and neck. With regard to the odontogenic cysts (and vdental lamina is illustrated in Figure 9.9. The epithelial cell
odontogenic tumours), a thorough knowledge of tooth devel- rests of the dental lamina give rise to most of the odontogenic
opment is also needed to understand the pathogenesis, since cysts (Table  2.1) as well as to most odontogenic tumours.
the complex interactions between epithelium and mesen- Dental lamina rests are particularly numerous at the poste-
chyme that underpin normal morphogenesis and tooth erup- rior aspect of the dental arches and in the tissues overlying
tion provide good models for the processes that drive cyst unerupted teeth and in the dental follicle. This accounts for
formation and growth. In addition, the histology of many the fact that the angle of the mandible is a common site for
lesions may recapitulate the features of the developing tooth many cyst types, and that many cysts (and tumours) may
and knowledge of these features facilitates the ability to reach arise in the dental follicle and embrace or surround an
an accurate diagnosis. A detailed consideration of embryol- unerupted tooth and lie in a dentigerous relationship. This
ogy and development is beyond the scope of this book, but especially affects the mandibular third molars, since these
factors relevant to each cyst type are summarised in each are the most commonly impacted teeth (Brown et al. 1982).
chapter. For up-­to-­date and expert knowledge relating to Although most types of odontogenic cyst arise from den-
development, readers should consult expert texts or reviews tal lamina, the most common cyst (radicular cyst) takes its
(Wise et  al.  2002; Nel et  al.  2015; Nanci  2017; Seppala origin from the rest cells of Malassez that lie in the perio-
et al. 2017; Diniz et al. 2017; Hovorakova et al. 2018; Bastos dontal ligament as remnants of Hertwig’s root sheath (see
et al. 2021). Figure 3.6). The second most common cyst, the dentiger-
In the majority of odontogenic cysts, the epithelial lining is ous cyst, arises from the reduced enamel epithelium that
derived from epithelial remnants of the dental lamina embraces the fully formed crown of a tooth prior to erup-
(Table 2.1). Early in the development of the jaws, the surface tion. In the case of the radicular cyst, the phases of cyst
epithelium thickens and grows downwards into the mesen- formation and growth are well understood and are driven
chyme of the future dental arches to form the dental lamina. by inflammation that is initiated by bacterial factors ema-
This extends around the arch as a band that maps the future nating from a non-­vital pulp. This process is described in
sites of tooth bud formation for both primary and secondary detail in Chapter 3. In developmental cysts, however, the
dentitions. The teeth develop as a result of complex epithe- processes are less clear, but are almost certainly driven by
lial–mesenchymal interactions that result in epithelial thick- epithelial–mesenchymal interactions that initiate the
enings or placodes, which then form the enamel organs that molecular signalling pathways that underpin normal tooth
pass through the well-­described bud, cap, and bell stages dur- development, morphogenesis, and eruption. Thus, the
ing formation of the fully developed tooth (Nanci 2017). The mechanisms of formation of developmental cysts can be
dental lamina remains as a thin band that joins the surface regarded as the aberrant expression of normal processes.
oral epithelium to the enamel organ and only disintegrates at Normal tooth eruption provides a good model of the
the late bell stage of tooth development. Disintegration of the epithelial–mesenchymal interactions that regulate tissue
dental lamina results in the formation of small epithelial remodelling, including cell proliferation and bone resorp-
islands that lie over unerupted teeth, but also remain in the tion, and involve complex cascades and networks of
­Pathogenesis of Cyst  9

cytokines, chemokines, and growth factors (Wise et al. 2002; Constitutive or aberrant activation of the HH pathway
Nel et al. 2015; Bastos et al. 2021). These biological factors can be caused by reduced expression or loss of the PTCH
are discussed in detail in the context of the radicular cyst protein at the cell surface, and this is an important mecha-
(Table 3.2), where the cascade is started by bacterial infec- nism in the pathogenesis of the odontogenic keratocyst.
tion, but the same factors are involved in normal develop- Loss of PTCH most often results from loss of heterozygo-
ment and tooth eruption and contribute to the pathogenesis sity (LOH) or point mutations in the PTCH gene, and this
of developmental cysts. For example, Il-­1α is a pivotal pro-­ is seen in up to 80% or more of keratocysts (see Table 7.6).
inflammatory cytokine that can be activated by bacterial However, although PTCH gene alterations are important in
endotoxins, but it also has an important role as a paracrine keratocysts, they are not specific, since mutations or LOH
signalling molecule in the dental follicle where, along with of PTCH or activation of the HH signalling pathway may
other factors (e.g. parathyroid hormone–related protein) it be seen in other odontogenic lesions, including orthokerati-
mediates tissue remodelling and bone resorption during nised odontogenic cyst (Vered et al. 2009; Diniz et al. 2011),
tooth eruption via its role as an activator of osteoclasts. glandular odontogenic cyst (Zhang et al. 2010), and denti-
These basic mechanisms of cytokine-­mediated tissue gerous cyst (Levanat et al. 2000; Pavelić et al. 2001; Barreto
remodelling are involved in the growth and expansion of et al. 2002; Vered et al. 2009; Zhang et al. 2010). The role of
all types of cyst. These same biological factors are also PTCH and the HH signalling pathway in these cysts is dis-
responsible for proliferation of the reduced enamel epithe- cussed in Chapters  5 (dentigerous cyst), 10 (glandular
lium that merges with the overlying oral epithelium during odontogenic cyst), and 12 (orthokeratinised odontogenic
normal tooth eruption. It is thought that proliferation of cyst). The role of the B-­catenin gene (CTNNB1) and the
the reduced enamel epithelium, in the absence of normal WNT signalling pathway in calcifying odontogenic cysts is
eruption, may be involved in the pathogenesis of the denti- discussed in Chapter 11.
gerous cyst, but proliferation of rest cells of dental lamina Overall, it appears that alterations in the PTCH gene or
within follicular tissue may also drive the formation of activation of HH signalling are common features of a num-
other lesions that are commonly associated with unerupted ber of lesions and may represent an initiating event in the
teeth. This would include some odontogenic tumours (e.g. formation of developmental odontogenic cysts, possibly in
ameloblastoma, adenomatoid odontogenic tumour) as well a progenitor epithelial cell, which then gives rise to the
as the odontogenic keratocyst and orthokeratinised odon- entire epithelial lining and drives growth and expansion. It
togenic cyst. The role of these biological factors in the has been suggested that the PTCH gene may act as a gate-
pathogenesis of dentigerous cyst, the odontogenic kerato- keeper gene and that further genetic events result in the
cyst, and the orthokeratinised odontogenic cyst is discussed formation of different cysts or tumours (Gomes and
in Chapters 5, 7, and 12, respectively. Gomez 2011). This would explain a role for PTCH in a wide
As well as activation of cytokines and other biological range of cyst types, but does not exclude a role for further
factors, there is good evidence that activation of oncogenic specific PTCH mutations in keratocysts.
signalling pathways is a common feature in the pathogen- Another unifying feature involved in the pathogenesis of
esis of odontogenic cysts and tumours (Diniz et  al.  2017; jaw cysts and probably also of soft tissue cysts is the role of
Bilodeau and Seethala 2019). These pathways are involved hydrostatic pressure. With few exceptions (odontogenic
in the normal development and morphogenesis of the keratocyst, botryoid odontogenic cyst, glandular odonto-
teeth, but aberrant activation may drive pathological pro- genic cyst), cysts in the jaws tend to be round or spherical
cesses. The most widely studied pathway is the hedgehog on radiology or imaging, suggesting that they grow slowly
(HH) signalling pathway, which is a fundamental feature in a regular and centripetal manner. It is widely accepted
of normal development with crucial roles in cell fate, dif- that hydrostatic pressure due to osmosis provides the
ferentiation, and patterning. HH activation through bind- evenly distributed internal forces that result in this growth
ing of the Sonic hedgehog (SHH) ligand is a fundamental pattern. Osmotic pressure across the cyst wall is caused by
feature of odontogenesis, regulates the development of the the accumulation of soluble proteins in the cyst lumen, so
dental lamina, and is responsible for tooth morphogenesis that the concentration of molecules inside the cyst is
and patterning (Diniz et  al.  2017; Seppala et  al.  2017; greater than in the adjacent tissues. This causes passage of
Hovorakova et al. 2018; Sasai et al. 2019). The pathway is fluid (water) into the cyst lumen and results in a high intra-
regulated by the PTCH protein, which is a receptor for luminal pressure that drives cyst expansion and growth.
SHH and under normal conditions controls and regulates The mechanisms of hydrostatic pressure and its role in the
epithelial–mesenchymal interactions, cell proliferation, radicular cyst are discussed in detail in Chapter 3, but there
and differentiation. The HH signalling pathway is dis- is good evidence that hydrostatic pressure due to osmosis is
cussed in detail in Chapter 7 and is illustrated in Figure 7.13. involved in the expansion of most, if not all, cyst types.
10 General Considerations

Although the odontogenic keratocyst grows in a multicen- is uncoordinated with that of normal tissues, and persists
tric pattern associated with cell proliferation in the wall, in the same excessive manner after cessation of the stimuli
there is also evidence of an increased intracystic pressure, that evoked the change’. No cyst described in this book
suggesting a role for osmosis in its expansion (Toller 1970b; meets these criteria and in terms of clinical behaviour none
Kubota et  al.  2004). Kubota et  al. (2004) suggested that of the cysts can therefore be described as neoplastic.
increased hydrostatic pressure was particularly important Nevertheless, in the 2005  World Health Organization
in the initiation and early growth of the keratocyst, while (WHO) classification (Barnes et al. 2005), two cysts that
cell proliferation was more important as the cyst enlarged. had hitherto been regarded as developmental in origin
This is in keeping with the observation that keratocysts were renamed ‘tumours’, with the clear intention that
tend to be unilocular when they are small, while larger they should be designated as benign neoplasms. The
cysts and lesions in older individuals are more often multi- odontogenic keratocyst was renamed keratocystic odonto-
locular or scalloped (Forssell  1980; Stoelinga  2001; genic tumour and the calcifying odontogenic cyst was
MacDonald-­Jankowski and Li 2010; Boffano et al. 2010; see renamed calcifying cystic odontogenic tumour. With regard
Chapter 7). Interestingly, Kubota’s research group (Kubota to the keratocyst, the designation as a neoplasm was based
et al. 2004; Oka et al. 2005) also showed that the increased on its ‘aggressive, infiltrative behaviour’ and the role of
pressure stimulated secretion of cytokines, including the PTCH gene in its pathogenesis. However, keratocysts
IL-­1α, suggesting another common mechanism for activa- do not meet the criteria for neoplasia suggested by Willis,
tion of biological factors that promote tissue remodelling since there is no evidence of uncoordinated independent
and bone resorption. These data are discussed in detail in growth. It is ironic that since the lesion was designated as
Chapter 7. a neoplasm in 2005, there have been almost 50 publica-
tions reporting marsupialisation (or decompression) as
an effective treatment compared to only 10 papers before
­The Cyst–Tumour Interface 2005. A number of publications have shown that marsu-
pialisation is effective and that it may result in regression
The pathogenesis of the developmental odontogenic cysts of the typical keratocyst lining to an epithelium indistin-
is still poorly understood and this has led to much debate guishable from that seen in normal oral mucosa. This
regarding the possible neoplastic nature of some of the cannot be regarded as the behaviour of a neoplasm.
cysts. This applies mostly to the odontogenic keratocyst, A further consideration is the use of the term ‘aggres-
but there is also debate regarding the nature of the calci- sive’. It is common for authors to assert that the odonto-
fying odontogenic cyst and glandular odontogenic cyst. genic keratocyst is aggressive because it has a propensity
As discussed in Chapter 7, the odontogenic keratocyst in for recurrence. While there is no doubt that the keratocyst
particular has generated an enormous literature that has (and a number of other cysts) has a tendency to recur, we
increased more than fivefold since the last edition of this do not support the well-­used assertion that ‘recurrence’
book. Many papers have explored the expression of vari- and ‘aggressive’ are synonymous. The recurrence rate is
ous markers in an attempt to show that the keratocyst is a almost entirely associated with the pattern of cyst growth
neoplasm. A common suggestion is that expression of and the type of treatment (see Chapter 7) and is similar to
proliferation markers at a higher rate than seen in other that seen in other cysts that have a multilocular growth
cyst types is evidence that the lesion is a neoplasm. Of pattern (botryoid odontogenic cyst, glandular odontogenic
more value are studies exploring aberrant gene expres- cyst). Inadequate removal may lead to residual cyst ele-
sion or activation of oncogenic signalling pathways. ments being left in the tissues and these may proliferate to
However, at the present time there is no clear marker of form new cysts. It is also true that occasionally a kerato-
neoplasia and the definition of a neoplasm has become cyst, especially a recurrent lesion, may spill into the soft
uncertain. It is not uncommon for the finding of a single tissues and cause considerable clinical problems. However,
genetic mutation to be taken as evidence of neoplasia other cysts that are known to be difficult to remove, and
regardless of the behavioural characteristics of the lesion. may have very high recurrence rates, are not described as
It must be pointed out that a single point mutation or aggressive or thought to be neoplastic. For example, inad-
genetic aberration may be the cause of developmental equately treated ranulas have a recurrence rate of up to
anomalies and is not sufficient to designate a lesion as about 65% (see Chapter  15) and thyroglossal duct cysts
neoplastic. recur in up to 50% of cases if residual ductal elements are
At the present time, most pathology textbooks still define not also removed (see Chapter  18). We believe that the
neoplasia in the terms first used by Willis in 1960, as ‘an term ‘aggressive’ is overused and may be over interpreted
abnormal mass of tissue, the growth of which exceeds and to suggest an invasive lesion or a behaviour similar to that
­The Cyst–Tumour Interfac  11

associated with truly aggressive lesions such as malignan- activation of the WNT signalling pathway (reviewed by
cies. In this context, none of these cysts behaves in a way Gomes et al. 2019). The presence of this CTNNB1 mutation
that can be described as truly aggressive. has led to suggestions that the calcifying odontogenic cyst is
A more compelling argument for the neoplastic nature a neoplasm. However, this mutation and the resulting intra-
of cysts is the finding of genetic aberrations. A key point in cellular expression of β-­catenin are seen in a range of lesions
this discussion, as already mentioned, is the finding of and a single mutation is not sufficient to justify designation
PTCH gene alterations and activation of the HH signalling as a neoplasm. Further research is needed, but at the present
pathway in a range of developmental cysts. The finding of time there is little evidence to suggest that the calcifying
a single point mutation or LOH in the PTCH gene is insuf- odontogenic cyst behaves as a neoplasm.
ficient to designate the keratocyst as a neoplasm. However, The relationship between the glandular odontogenic cyst
more recent studies (Stojanov et al. 2020) have confirmed and neoplasia has been speculative and is based largely on
earlier work that has shown that some keratocysts show its histological similarity to intraosseous mucoepidermoid
biallelic loss of both copies of the PTCH gene (Levanat carcinoma. This is discussed in detail in Chapter 10. This
et  al.  1996; Sun et  al.  2008; Pan et  al.  2010). histological similarity can make it very difficult to reach a
In their study, Stojanov et al. (2020) found that 80% of spo- definitive diagnosis and may result in misdiagnosis of
radic keratocysts showed biallelic loss of PTCH1. Biallelic lesions. A number of papers have reported lesions with his-
loss of the gene means that both copies have been lost or tological features of a glandular odontogenic cyst that have
mutated and meets the ‘two-­hit’ hypothesis of Knudson recurred as mucoepidermoid carcinomas, but some of
(Knudson  1971,  1996), which suggests that loss of both these are poorly illustrated and in others the diagnostic cri-
copies of a tumour suppressor gene causes neoplasia. teria are not clear. Nevertheless, this has led to suggestions
However, Knudson’s hypothesis related to hereditary can- that the glandular odontogenic cyst may be a precursor
cer (retinoblastoma), and it is now thought that two hits lesion, or a ‘benign variant’ of mucoepidermoid carcinoma.
may not always cause neoplasia and that further genetic The debate is further fuelled by the observation that the
aberrations may be necessary (Tomlinson et al. 2001). glandular odontogenic cyst has a high recurrence rate,
Notwithstanding this argument, the finding of biallelic leading some authors to label it ‘b­iologically aggressive’
loss of the PTCH gene in some keratocysts provide some (Greer et al. 2018). Mucoepidermoid carcinoma is charac-
evidence that at least a subset of odontogenic keratocysts terised by specific rearrangements of the MAML2 gene and
may be neoplastic. However, the line of demarcation large studies have shown that glandular odontogenic cysts
between aberrant development and neoplasia is blurred do not show this change, suggesting that the two lesions
and it seems that the keratocyst may sit on a spectrum are distinct and that the cyst is not a precursor to the carci-
between a developmental cyst and a benign cystic neo- noma (Bishop et al. 2014). More recently, two studies have
plasm. At the present time it is difficult to make a clinico- reported lesions with the features of glandular odontogenic
pathological distinction between cysts showing different cyst that have recurred as MAML2 positive mucoepider-
genetic changes and there are no defining criteria for moid carcinomas, and have suggested that this is further
which, if any, keratocysts might be neoplastic. Further evidence that mucoepidermoid carcinomas can arise from
research is needed to clearly define neoplasia and to deter- glandular odontogenic cysts (Greer et  al.  2018; Nagasaki
mine if there are any behavioural differences between cysts et al. 2018). In both cases, an alternative explanation is that
with different genotypes or clinicopathological phenotypes. the primary lesions were also mucoepidermoid carcino-
These issues and the debate about the neoplastic nature of mas, but were either misdiagnosed or were histologically
the odontogenic keratocyst are discussed in detail in Chapter 7. indistinguishable from cysts. Taken together, however,
Other lesions that sit on the cyst–tumour interface include these studies raise a number of questions and provide
the calcifying odontogenic cyst and the glandular odontogenic sufficient evidence to justify further research into the
cyst. In 2005, the WHO renamed the calcifying odontogenic relationship between glandular odontogenic cyst and
cyst the calcifying cystic odontogenic tumour, and designated mucoepidermoid carcinoma. The two possibilities to be
all odontogenic ghost cell lesions as neoplasms (Barnes explored are that glandular odontogenic cyst is a benign
et al. 2005; Prætorius and Ledesma-­Montes 2005). However, variant or precursor of mucoepidermoid carcinoma or,
more than 85% of ghost cell lesions are simple cysts and do more likely, that in some cases the two lesions are histo-
not recur, with little evidence of behaviour that would sug- logically indistinguishable. Until these issues are clarified,
gest a neoplastic origin (Ledesma-­Montes et al. 2008). It has care must be taken to use strict criteria for diagnosis and
been shown, however, that ghost cell lesions, including calci- histological assessment must be accompanied by careful
fying odontogenic cysts, are characterised by mutations in assessment of the clinical and radiological features (see
the β-­catenin gene (CTNNB1) and may be driven by Boxes 10.1 and 10.2, Tables 10.3 and 10.4).
12 General Considerations

­ n Approach to Diagnosis of Cysts


A In most cases, the clinical features of cysts are not specific,
of the Jaws since the most common presenting feature is a swelling with
few other symptoms. The surgeon therefore must rely on
An accurate diagnosis of a cyst requires knowledge, com- additional features to assist in making a provisional diagnosis.
mon sense, and a logical approach to the assimilation of all For example, a swelling associated with an absent tooth may
the information available about each lesion. With few suggest a dentigerous cyst, and a small, bluish-­coloured swell-
exceptions, it is not possible to reach a correct diagnosis ing on the lower lip of a child is almost certainly a mucocele.
without considering the clinical, radiological, and histo- Ultimately, however, a final diagnosis is usually made on his-
logical features. This is especially relevant to histopatholo- tological examination, but before this both the clinician and
gists, who may be called upon to make a diagnosis on a the pathologist should examine the radiographs or imaging.
small biopsy. To do so without considering the clinical and
radiological features often leads to diagnostic errors and
Radiology of Cysts of the Jaws
incorrect management (Barrett et al. 2017). In each chapter
of this book we present all the features of each cyst type in The characteristic radiological feature of all cysts of the jaws
context and discuss the differential diagnoses and potential is a well-­demarcated radiolucency with a well-­defined and
errors that might occur if only small biopsies are examined. often corticated margin. Further features that assist in diag-
Throughout we advise on the importance of correlating nosis include the shape and size of the lesion and the site, but
clinical, radiological, and histological features and suggest in most cases it is the relationship to the teeth that provides
that a diagnosis of an intraosseous lesion should never be the best indication of the type of cyst. A conventional plane
made without considering the radiology. We also recom- radiograph is usually sufficient to determine the extent and
mend that a diagnosis should never be made on a small relationships of jaw cysts, but computed tomography (CT)
biopsy if there is any uncertainty  – when in doubt, the and magnetic resonance imaging (MRI) are often useful and
pathologist should request a further biopsy. In this chapter, may be essential for planning surgery of larger lesions. These
we briefly discuss an approach to making a correct diagno- relationships are discussed and illustrated in each chapter,
sis and summarise the key features that can assist in mak- but here we present an overview of characteristic radiological
ing a diagnosis of cysts of the maxillofacial regions signs and the basic principles of an approach to interpreting
(Tables 2.2–2.4). With regard to the odontogenic cysts, an the radiology. Table 2.2 shows the cyst types that have charac-
accurate diagnosis is facilitated by an understanding of teristic radiological features, provides a cross reference to the
tooth development and the pathogenesis of each cyst type figures in each chapter, and summarises the diagnostic utility
(see ‘Pathogenesis of Cysts’ and Table 2.1) of each feature.

Table 2.2  Characteristic radiological features that assist in the diagnosis of cysts of the jaws.

Cyst type Radiological sign Diagnostic utility Figure references

Radicular cyst A radiolucency at the apex of All radicular cysts are located at the opening of the root Figures 2.2 and 3.4
a tooth canal, almost always at the apex. This feature can be
considered diagnostic of radicular cyst if the tooth is
also non-­vital. The radicular cyst also lies within the
lamina dura. If the tooth is vital then other lesions must
be considered, but are rare. This may include cemental
lesions (cementoblastoma, cemento-­osseus dysplasia)
or, in the anterior maxilla, nasopalatine duct cyst
Paradental cyst A radiolucency superimposed This feature is diagnostic of paradental cyst. If an intact Figures 4.2 and 4.5
over the distobuccal aspect of follicular space cannot be seen, then the radiolucency
an impacted third molar. The may be due to a hyperplastic follicle or pericoronitis
distal follicular space and
lamina dura are intact
Dentigerous Radiolucency surrounds the All dentigerous cysts show this feature. However, it is Figures 5.5–5.12, 5.14
cyst crown of an unerupted tooth not specific, since it may be seen in about 30% of (dentigerous cyst), 7.7
keratocysts, up to 50% of orthokeratinised odontogenic (odontogenic keratocyst),
cysts, and occasionally in calcifying odontogenic cysts 12.3 (orthokeratinised
odontogenic cyst)
Table 2.2  (Continued)

Cyst type Radiological sign Diagnostic utility Figure references

Odontogenic Mesiodistal extension with This appearance is almost pathognomonic for keratocysts in Figures 7.6 (odontogenic
keratocyst minimal buccolingual the mandible. Note however that glandular odontogenic cyst keratocyst), 10.4, 10.5
expansion may also show this growth pattern (see below). Other cyst (glandular odontogenic
types and ameloblastomas show ballooning expansion. The cyst, ameloblastoma)
feature is best visualised on computed tomography (CT) scans
A well-­demarcated unilocular Such a radiolucency is most likely to be an odontogenic Figure 7.11
radiolucency in the ascending keratocyst. If the cyst is associated with an unerupted
ramus not associated with a tooth a dentigerous cyst cannot be excluded, and if it is
tooth multilocular an ameloblastoma must be considered.
A keratocyst is even more likely if there is little
buccolingual expansion (see above)
Lateral Well-­defined, round corticated This feature is characteristic of lateral periodontal cyst. Figure 8.2
periodontal radiolucency lateral to the Lesions are rarely greater than 10 mm in diameter. If the
cyst tooth root. Periodontal space lamina dura surrounds the cyst, or cannot be seen, a lateral
and lamina dura are intact radicular cyst must be considered. If the radiolucency is
larger than 10 mm or is multilocular, an alternative
diagnosis must be considered: possibly botryoid
odontogenic cyst, keratocyst, or glandular odontogenic cyst
Glandular Large multilocular This is not diagnostic, but is typical of the glandular Figures 10.3 and 10.4
odontogenic radiolucency crosses the odontogenic cyst. In some reports up to 85% of cases are
cyst midline of the mandible in a located in the anterior mandible. Keratocysts and
symmetrical pattern ameloblastoma may be multilocular, but are more often
located in the posterior mandible
Calcifying A cystic radiolucency About 25% of calcifying odontogenic cysts are Figures 11.4 and 11.5
odontogenic associated with irregular associated with an odontoma and show irregular
cyst calcifications radiopacities either in or adjacent to the cyst. Note that
simple bone cyst is occasionally associated with
calcifications, but these are usually multiple and
represent florid cemento-­osseous dysplasia (Chapter 17)
A cystic radiolucency with a About 50% of calcifying odontogenic cysts contain
peripheral band of dentinoid in the wall or show dystrophic calcification in
calcifications the lining. A peripheral band of calcification is
characteristic and is best seen on CT scans
Nasopalatine A radiolucency in the midline Almost diagnostic of nasopalatine duct cyst. Very rarely Figures 13.7 and 13.8
duct cyst of the anterior maxilla a radicular cyst may be in the midline. Occasional
nasopalatine duct cysts are displaced laterally, in which
case a radicular cyst must be considered. Note that the
nasopalatine duct cyst is not associated with the
periodontal ligament and the lamina dura may be intact
A heart-­shaped radiolucency This appearance is diagnostic of nasopalatine duct cyst Figure 13.7
in the midline of the anterior and is seen in about 20% of cases
maxilla
Nasolabial cyst An upward or posterior Nasolabial cyst is a soft tissue cyst, but it may distort the Figure 14.3
convexity of the inferior margin of the nasal aperture. This ‘distorted anchor
margin of the nasal aperture appearance’ is diagnostic of nasolabial cyst. It is only
or anterior floor of the nose seen on an anterior occlusal radiograph
Simple bone A scalloped margin at the This feature is typical and almost diagnostic of simple Figure 17.1
cyst superior aspect of a bone cyst and is seen in 50% or more of cases. It has
mandibular cyst, which rises been described as the tooth roots ‘hanging’ into the cyst
up and embraces the roots of cavity (Chapter 17)
multiple teeth
A cone-­shaped margin at the This feature is specific to simple bone cyst. The margins Figure 17.1
anterior aspect of a converge at a 45° angle to form a cone. However, it is
mandibular cyst only seen in about 10% of cases
Stafne bone A corticated unilocular 85% of Stafne bone cavities are located in the posterior Figure 17.5
cavity radiolucency at the angle of mandible and are always below the ID canal. This
the mandible below the excludes a lesion of odontogenic origin. The feature can
inferior dental (ID) canal be regarded as diagnostic
A radiolucency that, in a This feature is best visualised on CT and is diagnostic of Figure 17.6
coronal view, is open on the Stafne bone cavity
lingual aspect of the mandible
14 General Considerations

Root canal

Periodontal ligament

Lamina dura

Cyst lining

Cyst lumen

Figure 2.1  In the posterior region of the mandible, the course of


the inferior dental (ID) canal (hashed lines) allows the tooth-­
bearing areas (the alveolar bone) to be clearly distinguished from
the basal bone of the mandible. Radiolucencies below the ID
canal are not odontogenic in origin (see text for details).
Figure 2.2  Diagrammatic representation of a radicular cyst.
The cyst develops from rest cells of Malassez within the
In the first instance, the site of the cyst in the jaws can periodontal ligament, and lies within the lamina dura. The
corticated margin of the cyst is continuous with the lamina
suggest an initial diagnosis. Odontogenic cysts arise in the
dura (see text for details).
tooth-­bearing areas of the jaws in the alveolar bone and in
the mandible are always situated above the inferior dental
(ID) canal (Figure  2.1). The cyst displaces the ID canal cemento-­osseous dysplasias, and cementoblastoma, must
downwards towards, and sometimes beyond, the lower also be considered in the differential diagnosis of lesions
border of the mandible. Examples of this feature can be in the tooth-­bearing area. Overall, however, odontogenic
seen in Figures  5.5, 5.11 (dentigerous cyst), 7.6, 7.7 cysts and in particular radicular cysts are by far the
(odontogenic keratocyst), 10.4 (glandular odontogenic most common.
cyst), 11.3 (calcifying odontogenic cyst), 12.2, and 12.3 The defining feature of the radicular cyst is of a radiolu-
(orthokeratinised odontogenic cyst). A cystic radiolucency cency associated with the apex of a non-­vital tooth
located below the ID canal is not an odontogenic cyst and (Table 2.2; Figure 3.4). The radicular cyst arises within the
when such a feature is seen, an alternative diagnosis must periodontal ligament from the rest cells of Malassez, and
be considered. Figure 17.5 shows a Stafne bone cavity pre- an important sign is that the cyst lies within the lamina
senting as a radiolucency below the ID canal, excluding dura that surrounds the root of the tooth. Furthermore, the
the possibility of an odontogenic origin. In the posterior corticated margin of the cyst is continuous with the lamina
region of the mandible, this judgement is easy to make dura (Figure 2.2). Although this feature is helpful in diag-
(Figure 2.1), but in the anterior mandible and in the max- nosing a radicular cyst, it is of more value in excluding a
illa, the distinction between alveolar bone and basal bone radicular cyst when another cyst type appears to be associ-
is less clear. Although radiolucencies below the ID canal ated with a tooth root. If a cystic radiolucency is associated
cannot be odontogenic, the converse is not true and a with the root of a tooth, but the lamina dura is intact, then
number of radiolucent lesions of non-­odontogenic origin a radicular cyst can be excluded and another diagnosis
may arise above the ID canal. Figure 17.1 shows an exam- must be considered. This feature is especially helpful in the
ple of a simple bone cyst that is not odontogenic, but char- diagnosis of inflammatory collateral cysts (Figures 4.2 and
acteristically lies within the alveolar bone and embraces 4.3), lateral periodontal cyst (Figure  8.2), nasopalatine
the roots of multiple teeth. Other lesions that may arise in duct cyst (Figures  13.7 and 13.8), surgical ciliated cyst
the alveolar bone and be associated with tooth roots (Figure 16.2), and simple bone cyst (Figure 17.1).
include giant cell granuloma, Langerhans cell histiocyto- It must be noted, however, that other lesions arise
sis, and ossifying fibroma. Non-­cystic odontogenic lesions, within the periodontal ligament and may lie within the
including periapical granulomas, odontogenic tumours, lamina dura. In particular, a periapical granuloma may
­An Approach to Diagnosis of Cysts of the Jaw  15

have an identical radiological appearance to a radicular but careful examination of the gross specimen will show
cyst, and it is not possible to reliably distinguish between evidence of multilocularity and reveal areas of thickening
a granuloma and a cyst (discussed in detail in Chapter 3). or luminal nodules if present. Cysts that are typically mul-
Although cysts are often larger (see Table  3.1), when a tilocular on gross examination include the botryoid odon-
radiolucency is encountered at the apex of a tooth there is togenic cyst (Figures  8.3, 8.9, and 8.10) and glandular
an equal chance that the lesion is a periapical granuloma odontogenic cyst (Figure 10.6). Thickening of the wall or
or a radicular cyst (Jones and Franklin 2006a,b; Koivisto luminal nodules are seen in lateral periodontal cyst
et al. 2012; discussed in Chapter 1). Cemental lesions also (Figure 8.6), glandular odontogenic cyst (Figures 10.6 and
arise within the periodontal ligament and, especially 10.8), and calcifying odontogenic cyst (Figures  11.8 and
when small and not fully calcified, may present as a radio- 11.9). Calcifying odontogenic cyst may also have calcified
lucency identical to radicular cyst. This feature may be material in the wall or be associated with an odontoma.
seen in lesions of cemento-­osseous dysplasia, cemento-­ Representative samples of the cyst wall, including any
ossifying fibroma, and cementoblastoma. The dentiger- areas of thickening, should be taken for histological exami-
ous cyst embraces the crown of an unerupted tooth and nation and any hard tissue should be decalcified and sam-
cannot be confused with radicular cyst, but the corticated pled for histology.
margin is continuous with the lamina dura (Figures  5.5 On dissection, most cysts contain small amounts of sero-
and 5.6). sanguinous fluid, but the odontogenic keratocyst and
orthokeratinised odontogenic cyst usually contain a
‘cheesy’ or ‘buttery’ keratinaceous material that is cream or
Histopathological Examination of Cysts
yellow coloured, and may have a characteristically unpleas-
In most cases the responsibility for a final diagnosis lies ant odour. Such contents will be familiar to many patholo-
with the histopathologist who must examine samples of gists as a characteristic feature of epidermal cysts of
tissue. As stated above, it is important that the pathologist the skin.
does not make a final diagnosis without first considering The histological features of each cyst and the histological
the clinical and radiological features of the lesion. These differential diagnosis are described in detail in each chap-
may be stated on the pathology request form, but often ter. Very few cysts have histological features that are abso-
the pathologist should read the radiology report, consult lutely diagnostic or pathognomonic, and diagnosis is
with the radiologist, or personally examine the radio- usually made by considering a combination of features in
graphs. Many cysts may reach a large size and it is good the context of the radiology. The only possible exception to
practice to establish a diagnosis before definitive surgery. this is the odontogenic keratocyst, which shows a thin reg-
This means that the histopathologist is often presented ular lining of parakeratinised epithelium with features that
with a small incisional biopsy of a large lesion. In most are unique to this cyst type (see Figures  7.15–7.17).
cases, consideration of the radiological and histological Table 2.3 provides an overview of characteristic histologi-
features together is sufficient to establish a diagnosis. The cal features and their diagnostic utility for different
key features summarised in Tables  2.2 and  2.3 should cyst types.
assist decision making in most cases. Occasionally a
definitive diagnosis is not possible on a small biopsy and Immunohistochemistry and Molecular Pathology
a final diagnosis must await examination of the whole There are very many publications reporting the expres-
specimen. Pathologists must not be afraid to withhold a sion of different proteins in odontogenic cysts. In most
final diagnosis until it has been possible to examine suf- cases the purpose has been to shed light on the pathogen-
ficient tissue. esis and mechanisms of growth of the lesions, but many
Histopathological examination of a cyst begins with papers have attempted to determine whether particular
examination and sampling of the whole specimen. If an patterns of expression can provide accurate diagnostic
associated tooth is also removed, then the relationship of markers for each cyst type. Studies of keratin expression
the cyst to the tooth can be directly observed and is of par- and proliferation markers are particularly numerous and
ticular value in the diagnosis of a radicular cyst (located at the odontogenic keratocyst has been the subject of the
the tooth apex), dentigerous cyst (attached at the cemen- majority of studies. Overall, however, immunohistochem-
toenamel junction; Figure  5.18), and paradental cyst istry has only a very small role to play in the diagnosis of
(attached to the disto-­buccal aspect of the tooth; Figure 4.6). cysts of the maxillofacial regions. Although each cyst type
In all cases it is of value to examine the cyst in its entirety may show a different pattern of cytokeratins, the expres-
and also to dissect it and examine the cut surface and the sion demonstrated by immunohistochemistry merely
lumen. Most cysts are unilocular with a thin regular lining, reflects the type of keratinisation that is easily and clearly
16 General Considerations

Table 2.3  Characteristic histological features that assist in the diagnosis of cysts of the maxillofacial regions.

Histological feature Cyst type(s) Diagnostic utility Figure references

Proliferative Radicular cyst Typical feature of radicular cyst and of inflammatory collateral Figures 3.7, 3.12
epithelium with an Inflammatory cysts (radicular cyst), 4.7
arcading pattern collateral cysts But proliferative arcading epithelium may be seen in any (paradental cyst), 5.22
odontogenic cyst that is secondarily inflamed (dentigerous cyst)
The epithelial lining is Dentigerous Virtually diagnostic of dentigerous cyst. This feature may be Figures 5.18 and 5.19
attached to an cyst seen on macroscopic examination of an intact specimen or in
unerupted tooth at the decalcified sections
cementoenamel Note: there have been reports of odontogenic keratocyst or
junction orthokeratinised odontogenic cyst attached at the
cementoenamel junction, but this is very rare and is thought
to be due to a tooth ‘erupting’ into a cyst (see discussion in
Chapter 12)
Thin regular Odontogenic Diagnostic of odontogenic keratocyst. This typical epithelium Figures 7.15–7.17
parakeratinised keratocyst is not seen in any other jaw cyst
epithelium with a
corrugated surface and
prominent basal layer
Epithelial plaques or Lateral Epithelial plaques are seen in all cases of lateral periodontal Figures 8.6, 8.7 (lateral
thickenings with a periodontal cyst cyst and are diagnostic if the cyst is unilocular and no other periodontal cyst), 8.9
whorling pattern Botryoid features are noted (botryoid odontogenic
odontogenic cyst If the cyst is multilocular, then diagnostic for botryoid cyst), 9.4 (Gingival
Gingival cyst of odontogenic cyst cyst), 10.8 (glandular
adults Gingival cyst shows similar features but is extraosseous odontogenic cyst)
Glandular Glandular odontogenic cyst may show plaques in about 65% of
odontogenic cyst cases, but must be accompanied by other features (see
Table 10.3)
Cuboidal or columnar Glandular Typical feature and seen in up to 100% of glandular Figures 10.7 and 10.8
cells at the luminal odontogenic odontogenic cysts. Diagnostic when accompanied by other
aspect of the cyst cyst features (see Table 10.3). Similar cells are occasionally seen in
lining other cyst types, including dentigerous cyst, but these lack
other features
A simple cyst with Calcifying This feature is diagnostic of calcifying odontogenic cyst. Note Figures 11.8–11.10
ghost cells in the wall odontogenic that the lining is ameloblastomatous and if ghost cells are not
cyst seen, a diagnosis of cystic ameloblastoma must be considered.
Ensure the whole lining is examined (see discussion in
Chapter 11). If the lesion is solid, then consider dentinogenic
ghost cell tumour. Ghost cells may be seen in odontomas and
rarely in ameloblastomas
Hyaline bodies Various Hyaline (Rushton) bodies are often stated as being typical Figures 3.15 (radicular
odontogenic of radicular cyst. They are seen in about 10% of radicular cyst), 7.20
cyst types cysts, but also in up to 10% of odontogenic keratocysts and (odontogenic
dentigerous cysts (see discussion in Chapter 3). However, keratocyst)
hyaline bodies are specific and diagnostic of odontogenic
cysts
Cholesterol clefts Radicular cyst Cholesterol clefts result from an accumulation of cholesterol Figure 3.16
crystals as a results of long-­standing inflammations. They are
therefore typical of radicular cyst and are seen in 30% or more
of cases (Table 3.3)
But they are not specific and may be seen in any cyst that has
become chronically inflamed, in particular in inflamed
dentigerous cysts or keratocysts
­An Approach to Diagnosis of Cysts of the Jaw  17

Table 2.3  (Continued)

Histological feature Cyst type(s) Diagnostic utility Figure references

Mucous cells Various cyst Mucous cells have been described in most types of odontogenic Figures 3.14 (radicular
types cyst and are not diagnostic. They are a result of metaplastic cyst), 5.23 (dentigerous
change and are seen in about 20% of radicular cysts, 25% of cyst), 10.10, 10.11
dentigerous cysts, and 2% of odontogenic keratocysts (glandular odontogenic
Note that mucous cells are not a diagnostic requirement for cyst), 13.10
glandular odontogenic cyst and are seen in only about 70% of (nasopalatine duct cyst),
cases (Table 10.3). Mucous cells are seen in surgical ciliated 14.5 (nasolabial cyst)
cysts and in about 50% of nasopalatine duct cysts and
nasolabial cysts
Sebaceous glands Dermoid cyst Sebaceous glands are rare in jaw cysts and when seen a Figures 18.2 and 18.3
diagnosis of dermoid cyst should be considered. If sweat (dermoid cyst)
glands and hair follicles are also present, then this is
diagnostic for dermoid cyst. Sebaceous glands have rarely
been reported in odontogenic keratocyst and orthokeratinised
odontogenic cyst
Respiratory epithelium Nasopalatine Among cysts in the jaws, respiratory epithelium is a typical Figures 3.14 (radicular
duct cyst feature of nasopalatine duct cyst and is seen in about 50% of cyst), 13.9 (nasopalatine
Various cyst cases duct cyst), 14.5
types However, this is not specific. Surgical ciliated cyst is lined by (nasolabial cyst), 16.5
respiratory epithelium and metaplastic respiratory epithelium (surgical ciliated cyst),
has been described in radicular cysts 18.5 (bronchogenic
Among soft tissue cysts, nasolabial cyst, bronchogenic cyst, cyst), 18.6 (branchial
and thyroglossal duct cyst are lined by respiratory epithelium, cyst), 18.8 (thyroglossal
and respiratory epithelium may be seen in cysts of foregut or duct cyst)
branchial cleft origin (Boxes 18.2–18.4)

visible on examination of a routine H&E (haemotoxylin patterns of keratinisation and is usually sufficient to make
and eosin)-­stained section. The best example of this an accurate diagnosis.
conundrum is the odontogenic keratocyst. Many studies Despite these limitations, there are a few instances where
have been undertaken to compare the cytokeratin profile immunohistochemistry can help establish a diagnosis and
of keratocysts with other cysts, but almost without excep- differentiate between lesions with similar histological fea-
tion, the specimens used have been selected as typical his- tures. A number of diagnostically useful applications are
tological examples of each cyst type. It is not surprising summarised in Table 2.4.
therefore that if the features are typical, there is no need Molecular studies have provided much useful and inter-
for any additional staining beyond a good H&E-­stained esting information relating to the pathogenesis of odonto-
section to make the diagnosis. One key area of diagnostic genic lesions, especially with regard to the role of the PTCH
difficulty is when the pathologist must examine a small gene in the odontogenic keratocyst and to the role of the
biopsy of an inflamed cyst. When heavily inflamed, any SHH (hedgehog), WNT, and MAPK signalling pathways in
cyst type, including a keratocyst, may become lined by a variety of cysts and tumours (Diniz et al. 2017; Bilodeau
proliferative epithelium identical to that seen in a radicu- and Seethala 2019). Molecular tests, however, have not yet
lar cyst. The lining becomes non-­keratinised and studies proven to be useful in routine diagnosis of cysts. The main
have not been able to identify immunohistochemical exception to this is the identification of MAML2 rearrange-
markers that can differentiate between an inflammatory ments that can be helpful in differentiating between the
cyst and an inflamed developmental cyst. This issue is dis- glandular odontogenic cyst and intraosseous mucoepider-
cussed in detail in Chapter 7. Our experience, supported moid carcinoma (discussed in Chapter 10). Table 2.4 sum-
by a number of studies (Rao et al. 2015), suggests that a marises a number of molecular techniques that may show
good H&E-­stained section is the most specific marker for some value as diagnostic markers.
18 General Considerations

Table 2.4  Immunohistochemical and molecular markers that might have diagnostic utility in the differential diagnosis of cysts.

Immunohistochemistry

Antibody Target Diagnostic utility

CK10 Type I keratin, found mainly in CK10 stains keratinising epithelium and is positive in the
cornified epithelia superficial layers of odontogenic keratocyst and
orthokeratinised odontogenic cyst. Of little value in
histological sections, but has some utility in cytological
smears from aspiration biopsies. Cells from keratocyst and
orthokeratinised odontogenic cyst are positive, but
dentigerous cysts and ameloblastomas are negative (August
et al. 2000). Pan-­cytokeratin antibodies (AE1/AE3) may also
stain keratin in smears from inflamed cysts (Vargas
et al. 2007) (Figure 7.22)
CK18 and CK19 Keratin intermediate filaments. Both Use of both antibodies together has been shown to be useful
are widely expressed, but CK19 is seen to distinguish a glandular odontogenic cyst from central
typically in odontogenic epithelium mucoepidermoid carcinoma (Pires et al. 2004; discussed in
Chapter 10). Glandular odontogenic cyst is CK19+/CK18-­.
Mucoepidermoid carcinoma is CK19-­/CK18+
Calretinin Calretinin, a widely expressed Calretinin has been shown to be positive in up to 100% of
calcium-­binding protein ameloblastomas, including unicystic ameloblastoma.
Odontogenic cysts including keratocysts are negative. Useful
to differentiate cystic ameloblastoma from other cysts,
especially in small biopsies from the posterior mandible
region (Altini et al. 2000; Coleman et al. 2001; De Villiers
et al. 2008; Jeyaraj 2019; Rudraraju et al. 2019; discussed in
Chapters 5 and 7)
Maspin Maspin, a member of the serine May be useful in the differentiation of glandular
protease inhibitor superfamily odontogenic cyst from central mucoepidermoid carcinoma,
especially in small biopsies where not all features may be
apparent. Maspin is widely expressed, but studies have
shown much greater expression in the mucous cells of
mucoepidermoid carcinoma than in glandular odontogenic
cyst. Note that the differences relate only to mucous
cells – other epithelial cells of the cyst lining are positive
(Vered et al. 2010; Chapter 10, Table 10.4)
β-­catenin Cell surface protein important in cell Intracellular expression of β-­catenin (cytoplasmic and
adhesion, but also in the regulation of nuclear) is characteristic of calcifying odontogenic cyst (and
the WNT signalling pathway other ghost cell lesions – see CTNNB1 gene below) and is
seen in all cases. However, it is not diagnostic, since it may
also be seen in ameloblastomas
p16 p16 is a cyclin-­dependent kinase p16 antibodies are useful in the differentiation of branchial
inhibitor, involved in regulation of the cleft cyst from a cystic metastasis in the lateral neck. p16 is
cell cycle activated by human papillomavirus (HPV) infection and
strong expression, interpreted in context, is almost
diagnostic of HPV-­associated oropharyngeal carcinoma. The
majority of cystic metastases come from HPV-­associated
squamous cell carcinomas and are p16 positive, but
branchial cysts are negative. Note that positive staining must
be carefully interpreted and the correct criteria must be used
(Pai et al. 2009; Cao et al. 2010; Müller et al. 2015;
Chapter 18, Figure 18.7)
­An Approach to Diagnosis of Cysts of the Jaws 19

Table 2.4  (Continued)

Molecular markers

Gene Alterations Diagnostic utility

PTCH Mutations or loss of heterozygosity Alteration or loss of PTCH gene is seen in up to 80% of
(LOH) of PTCH gene odontogenic keratocysts, but is not diagnostic because
altered PTCH may be seen in other cyst types. However,
biallelic loss of PTCH has been recorded in keratocysts and
not in other odontogenic cysts, and this may be diagnostic
MAML2 Rearrangements of MAML2 gene, MAML2 rearrangements are seen in mucoepidermoid
usually with CRCT1 or 3 carcinomas and their presence helps differentiate
intraosseous mucoepidermoid carcinoma from glandular
odontogenic cyst, which does not show the translocation
(Bishop et al. 2014)
CTNNB1 Mutations in the CTNNB1 (β-­catenin) CTNNB1 mutations are seen in a wide range of neoplasms,
gene but within the jaws are almost unique to ghost cell lesions
(calcifying odontogenic cyst and dentinogenic ghost cell
tumour). However, molecular analysis has not been used for
diagnostic purposes. CTNNB1 mutation results in aberrant
intracellular (cytoplasmic and nuclear) expression of
β-­catenin protein (see β-­catenin above) (Gomes et al. 2019;
Chapter 11)
20

Radicular Cyst

CHAPTER MENU
Clinical Features, 20
●● Frequency, 20
●● Age, 21
●● Sex, 22
●● Site, 22
●● Clinical Presentation,  23
–– Radicular Cyst,  23
●● Residual Cyst,  24
­Radiological Features, 25
­Pathogenesis, 26
●● Pathology of Periapical Periodontitis,  27
●● Phase of Initiation,  28
●● Phase of Cyst Formation,  31
●● Growth and Enlargement of the Radicular Cyst,  33
–– Role of Hydrostatic Pressure,  33
–– Epithelial Proliferation,  35
–– Degradation of the Connective Tissues and Bone Resorption,  35
Histopathology, 36
●● Cellular and Metaplastic Changes,  38
●● Hyaline Bodies,  40
●● Accumulation of Cholesterol,  42
●● Residual Cyst,  44
●● Pocket Cyst (Bay Cyst),  44
Malignant Change in Radicular Cysts, 45
Treatment, 46

The inflammatory odontogenic cysts arise as a result of epithe- pericoronal tissues. This lesion has been referred to as a
lial proliferation within an inflammatory focus due to a num- paradental or inflammatory collateral cyst (Main  1970;
ber of causes. Radicular cyst is the most common inflammatory Craig 1976; Speight and Soluk-­Tekkeşin 2022b). Radicular
cyst and arises due to proliferation of epithelial remnants in and residual cysts are considered in this chapter. The
the periodontal ligament as a result of periapical periodontitis inflammatory collateral cysts are considered in Chapter 4.
following death and necrosis of the pulp. Radicular cysts are
most commonly found at the apex of the involved tooth, but
may arise on the lateral aspect of the root in relation to a lateral
­Clinical Features
root canal. Quite often a radicular cyst remains behind in the
jaws after removal of the offending tooth and this is referred to
Frequency
as a residual cyst (Speight and Soluk-­Tekkeşin 2022a).
Inflammatory cysts may also occur on the lateral aspect Radicular and residual cysts are by far the most common
of a tooth as a consequence of an inflammatory process in cystic lesions of the jaw bones, probably accounting for

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
­Clinical Feature  21

more than half of all cystic lesions and about 60% of odon- In presenting these data, radicular and residual cysts
togenic cysts. In an analysis of nearly 10 000 radiolucent have been included together, but where the information is
jaw lesions, Koivisto et al. (2012) found that 73% were peri- stated, almost all the studies show that residual cysts
apical granulomas or cysts. Of these, 40.4% were granulo- account for between 5% and 15% of odontogenic cysts and
mas and 33.0% were cysts. between 10% and 20% of radicular cysts (Mosquedo-­Taylor
In Shear’s South African series, radicular and residual et al. 2002; Jones et al. 2006; Sharifian and Khalili; 2011;
cysts comprised 52.2% of cystic jaw lesions (Table 1.1) and Soluk Tekkeşin et  al.  2012b; Lo Muzio et  al.  2017;
62% of odontogenic cysts. This is similar to the frequency Tamiolakis et al. 2019; Kammer et al. 2020)
in a Sheffield study (Jones et al. 2006), where 4297 radicu-
lar and residual cysts were diagnosed over a 30-­year period,
Age
representing 60.3% of all odontogenic cysts (Table 1.2).
Data from other countries vary (Table 1.3), with a range The age distribution of 948 patients in South Africa is
from 42.1% in Mexico (Mosquedo-­Taylor et  al.  2002) to shown in Figure  3.1. Very few cases are seen in the first
84.5% in Sicily (Tortorici et  al. 2008). The reason for the decade, after which there is a fairly steep rise, with a peak
global variation in frequency is not clear, but probably frequency in the third decade and large numbers of cases
relates to variations in the prevalence of dental caries and in the fourth and fifth decades. In a Sheffield study of 1970
to methods of data collection. For example, in Mexico cases, Jones et al. (2006) found cysts in older age groups,
Mosquedo-­Taylor et  al. (2002) suggest that the low fre- with a peak in the fourth decade and a mean age of
quency is due to a large proportion of their studied popu- 37.3 years (Figure 3.2). Similar data were found in French
lation being treated in the private sector where caries is (Meningaud et al. 2006), Australian (Johnson et al. 2013),
less prevalent. Conversely, Tortorici et al. (2008) believed Italian (Lo Muzio et  al.  2017), and Greek (Tamiolakis
that the high frequency in Sicily was related to a high prev- et al. 2019) studies, with mean ages of 40.8, 50.5, 38.2, and
alence of caries in the studied population. The low 41.2 years, respectively. The older ages of these patients
­frequency in Iran was ascribed to the fact that caries and compared with the South African patients was noted in
periapical lesions are common and are not routinely sub- previous editions of this book and suggests that the South
mitted for histological diagnosis (Sharifian and Khalili African patients may have been exposed to the relevant
2011). Overall, however, these data show that radicu- aetiological factor, mainly dental caries, at a slightly
lar  (including residual) cysts are consistently the most younger age than the other groups.
common cystic lesion of the jaws, with a frequency of The wide age range of lesions, the very low frequency in
about 60%. the first decade, and a peak in the fourth decade have been

Figure 3.1  Age distribution of 948 350


South African patients with
radicular cysts.
300 289

250
225
131
No. of cases

200 Women
77
146 Men
150 131
54
100 61 83
158 148
41 41
50 92
16 70 15 14
42 3
26
0 8 8
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89
Age
22 Radicular Cyst

600 Figure 3.2  Age distribution of 1970


517 patients with radicular cysts from
Women Sheffield, England, 1990–2004 (n = 1970).
500 Men Source: Based on Jones et al. (2006).
430

400 372
257
No. of cases

308
197
300
196
129
200
144
124
260
233 56
100 56 179 59
176
6 10
68 88 33
0 5 26 5
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89
Age

confirmed in a number of studies (Tortorici et  al. 2008; of  49.4 years (n  =  218; range: 17–90) and, in their large
Sharifian and Khalili 2011; Ramachandra et al. 2011; Soluk Greek series, Tamiolakis et al. (2019) found a mean age of
Tekkeşin et  al.  2012b; Lo Muzio et  al.  2017; Tamiolakis 50.5 years (n = 749; range 11–93) for residual cysts.
et al. 2019). These studies also indicate that although dental
caries is very common in children, radicular cysts are not
Sex
often associated with deciduous teeth. Soluk Tekkeşin et al.
(2016) identified 596 odontogenic cysts in a paediatric pop- Almost all studies show that all odontogenic cysts are
ulation (age 0–17 years), of which 387 (65%) were radicular slightly more common in males than in females (Table 1.3).
cysts (including 17 residual cysts). Of these, only 3 arose in With regard to radicular cysts in the South African series,
subjects less than 6 years old, 113 were found in the mixed 555 (58.5%) were in males and 393 (41.5%) in females
dentition (age 6–12 years), and 271  were associated with (M : F 1.41 : 1), a statistically significant difference
permanent teeth (age 13–17 years). In a similar study in (P < 0.002). In the Sheffield study, 1914 (51.5%) were in
Brazil, da Silva et al. (2018) reviewed 294 odontogenic cysts males and 1801 (48.5%) in females (M : F 1.06 : 1; Jones
in subjects younger than 20 years. They identified 145 radic- et al. 2006), but this difference was not significant.
ular cysts, but only 9 arose in subjects younger than 10 years.
Residual cysts are very unusual in the first, second, or
Site
third decades and peak at a slightly older age, usually in the
fifth decade. Lo Muzio et  al. (2017) found a mean age Figure 3.3 shows that radicular cysts may arise at any site
in the jaws, but are more common in the maxilla (60%)
Box 3.1  Radicular Cyst: Epidemiology – Key Facts than the mandible, with a particularly high frequency
in  the anterior maxilla, with about 40–50% of all cases
●● Radicular cysts are the most common cystic lesion
occurring in this region. There are a number of possible
of the jaws
reasons for this. First, in addition to the hazard posed by
●● They comprise about 60% of all odontogenic cysts
dental caries, maxillary incisors have in the past, perhaps
●● Residual cysts account for 10–20% of radicular cysts
more frequently than other teeth, had silicate restorations
●● Overall radicular cysts are the most common cause
placed in them, with consequent high risk to their pulps. If
of bony swellings
this were the cause, then the prevalence of cysts at this site
●● They are slightly more common in males
might reduce in future ­generations. Second, there is a high
(M : F = 1.4 : 1)
prevalence of palatal invaginations in the maxillary lateral
●● Peak age is the third and fourth decades
incisors and a high frequency in which pulp death super-
(20–40 years)
venes in these teeth. Third, maxillary anterior teeth are
●● Rare in children
probably more prone than others to traumatic injuries,
●● Very rare on deciduous teeth
which may lead to pulp death.
­Clinical Feature  23

Figure 3.3  Site distribution of radicular cysts. S


A comparison of 1111 cases from South Africa Mandible
(Johannesburg) and 1974 cases from England J Maxilla
(Sheffield).

Percentage of cases
53
39 S J J
S

11 13
7 8

11 7 11 11
12
17

Anterior Premolar Molar

S = Sheffield series, n=1974. J = Johannesburg series, n=1111

Radicular cysts may occasionally be associated with pain,


Recent studies, however, show that this site distribu-
but this is usually pulpal in origin, or is due to secondary
tion is seen across the world and has not changed in the
infection, when there may be a sinus leading from the cyst
last two decades, suggesting that trauma and susceptibil-
cavity to the oral mucosa. However, even infected lesions
ity to caries may be the main reasons. The proportion of
may be symptomless. In a histological examination of 25
radicular cysts associated with the maxillary incisors has
radicular cysts, Vier and Figueiredo (2002) showed that 21
been reported to  be 54.4% in India (Ramachandra
contained abscess cavities that disrupted the epithelial lin-
et al. 2011), 52.8% in England (Jones et al. 2006), 47.8% in
ing. This suggests that although secondary infection may be
Turkey (Soluk ­Tekkeşin et  al.  2012b), 37.3% in Iran
quite common, it is not often a cause of pain. Conversely,
(Sharifian and Khalili 2011), 36.4% in Sicily (Tortorici
some patients complain of pain, although no evidence of
et al. 2008), and 43% in Brazil (Tavares et al. 2017). Most
infection is found clinically and no evidence of acute inflam-
studies agree that the mandibular molar area is the next
mation is seen histologically after the cyst has been removed.
most common site.
Quite often, a patient may have multiple radicular cysts.
Usually this is clearly associated with dental caries in mul-
tiple teeth, but the fact remains that radicular cysts are
Clinical Presentation
relatively rare in relation to the vast numbers of grossly
Radicular Cyst carious teeth with dead pulps. This has led a number of
An essential criterion for the diagnosis of a radicular cyst is authors to believe that there are cyst-­prone individuals
that it is always associated with a tooth with a non-­vital who show a particular susceptibility to developing radicu-
pulp, usually as a result of dental caries. lar cysts (Oehlers 1970). It is possible that in most people
Many radicular cysts are small and symptomless and are periapical infections are suppressed and cyst formation is
discovered by chance during radiological examination of inhibited, but that cyst-­prone subjects have a defective
caries or non-­vital teeth. Overall, however, radicular cysts immunological surveillance and suppression mechanism.
are probably the most common cause of swelling of the Multiple radicular cysts may also be seen in patients with
jaws and a slowly enlarging swelling is a common present- hereditary dental defects (e.g. multiple dens-­in-­dente or
ing complaint. At first the enlargement is bony hard, but as dentinogenesis imperfecta), but in these cases this is
the cyst increases in size, the cortical plate is resorbed and because of morphological defects resulting in increased
only a thin layer of subperiosteal new bone covers the caries or early exposure and death of the pulp.
lesion. The swelling then exhibits ‘springiness’ or ‘egg-­shell Radicular cysts arising from deciduous teeth appear to be
crackling’. Only when the cyst has completely eroded the very rare. Shear found that of 1300 radicular cysts recorded
bone, often due to secondary infection, will the lesion be in his department at the University of the Witwatersrand
fluctuant. In the maxilla there may be buccal or palatal over a 25-­year period, only 7 were associated with decidu-
enlargement, whereas in the mandible it is usually labial or ous teeth (0.5%). In an extensive review of the literature
buccal and only rarely lingual. from 1898 to 1985, only 28 cases were found (Lustmann
24 Radicular Cyst

and Shear  1985). Subsequent reviews have recorded 112 with increasing duration of the cyst (r  = 0.5; P < 0.005;
cases up to 2004 (Nagata et al. 2008) and a total of 122 up to High and Hirschmann 1986). There was an unexpectedly
2010 (Shetty et al. 2010). large number in the mandibular premolar region and there
The frequency is probably higher than these figures was a direct relationship between the age of the cyst and
would suggest, because many lesions may not be diagnosed the radiological and histological evidence of mineralisa-
before the deciduous teeth are extracted or exfoliate, and tion (P < 0.001). There was an overall reduction in epithe-
lesions may not be submitted for histological examination. lial thickness with cyst age and all cysts showed minimal
Nevertheless, the frequency is substantially lower than of chronic inflammatory changes. Their results support the
radicular cysts associated with permanent teeth and one may notion that the vast majority of residual cysts are slowly
speculate on the reasons for this. It may be due to the fact that resolving lesions. Nevertheless, they do persist and the
pulpal and periapical infections in deciduous  teeth tend to authors did not provide evidence of complete healing.
drain more readily than those of permanent teeth. Caries is In their second paper, High and Hirschmann (1988)
almost always the aetiological factor, although in over half of showed that symptomatic cysts were larger than asymp-
all cases reported there has been previous pulp therapy, sug- tomatic lesions, and that the negative correlation of size
gesting that medicaments used in pulpotomy may be an with cyst duration was not as strong (r  = 0.39; P < 0.05).
additional aggravating factor by perpetuating chronic Cyst ages varied from 1 month to 20 years and there was
inflammation (Grundy et al. 1984; Shetty et al. 2010) again a perplexingly high frequency in the mandibular pre-
In the study of Lustmann and Shear (1985), 23 person- molar region. Acute and chronic inflammatory cell infiltra-
ally observed cases were reported. The patients’ ages tion showed variable intensity and there was an inverse
ranged from 4 to 12 years, with one exceptional case aged relationship between the presence of acute inflammation
19 years. The M : F ratio was 1.6 : 1. The mandible was and cortication of the cyst wall radiographically (P < 0.001).
affected more frequently than the maxilla and the decidu- There were no obvious causes for the inflammation in
ous molars were the teeth most often involved. In 9 cases deeply positioned residual cysts. The authors suggested
buccal expansion was noticed and in 8 cases the permanent that in symptomatic cases chronic inflammation could per-
tooth buds were displaced. Mass et al. (1995) gave a detailed sist and gradually worsen, and that acute inflammatory
account of 36 cases, of which 22 (61%) were associated episodes may be triggered by release of pro-­inflammatory
with mandibular molars. They also noted that the lesions factors from dead or dying cells.
are not associated with the apices of the teeth, but usually Nair (1998, 2003, 2006) considered that the type of cyst
present as a periradicular radiolucency overlying the was important with regard to persistence after treatment.
crowns of the unerupted permanent premolars. This makes Although he discussed non-­healing cysts after endodontic
diagnosis uncertain, since it may be difficult to differenti- treatment, his conclusions are relevant to residual cysts. He
ate a deciduous radicular cyst from a hyperplastic follicle confirmed the work of Simon (1980), who showed that
or even a dentigerous cyst of the permanent successor, there were two types of radicular cyst. These are discussed
which in this context may be inflammatory in origin (the in more detail later in this chapter but, to summarise, there
inflammatory dentigerous cyst is discussed in Chapter 5). is the true radicular cyst, which contains a closed cavity
entirely lined by epithelium, and the periapical pocket cyst
(also called bay cyst), in which the epithelium is attached to
Residual Cyst
the margins of the apical foramen in such a way that the
Residual radicular cysts are those that remain in the jaws cyst lumen is essentially a pouch or pocket, which commu-
after removal of the cause of the lesion  –  the offending nicates directly with the affected root canal. Thus, it is
non-­vital tooth. Although the reasons for the persistence of expected that the pocket cyst would heal after treatment or
cysts are contentious, there have been relatively few publi- tooth extraction, while the true cyst, being completely
cations on the subject and the whole notion of the exist- enclosed, is ‘self-­sustaining’ and may therefore persist in
ence of such cysts has been challenged on the basis that a the absence of the cause. Nair et al. (1996) suggested that
persistent radiolucency after removal of the offending only 15% of periapical lesions were radicular cysts, but of
tooth may merely represent an ongoing healing process these 61% were true cysts and 39% were pocket cysts. If
(Walton  1996; Lee et  al.  2014). High and Hirschmann only true cysts persisted after removal of the offending
(1986, 1988) studied a series of asymptomatic and sympto- tooth, this may account for the relatively low frequency of
matic residual cysts. They were interested in the factors residual cysts.
that decide whether a radicular cyst will resolve or persist Regardless of the finer details of the biology of these
after tooth removal and the natural history and behaviour lesions, from the clinical point of view it is evident that a
of these cysts once established. With regard to the asymp- radiolucent lesion may persist and may produce symptoms,
tomatic cysts, they showed that there was a decrease in size for a substantial period of time after extraction of the
­Radiological Feature  25

offending tooth. On biopsy, many show the typical features granuloma. In one of the largest studies, which has not
of a cyst and residual cyst remains an appropriate term for been repeated, Mortensen et al. (1970) examined histologi-
these lesions. cal material of 396 periapical lesions with a diameter of
5 mm or more, that had been classified pre-­operatively as
cysts or granulomas on radiological evidence. A correct
­Radiological Features preliminary diagnosis had been made in 81% of 232 granu-
lomas, but in only 48% of 164 cysts. They also showed that
The classic description of the radiological appearance of the relative number of granulomas decreased with increas-
radicular cysts is that they are round or ovoid radiolucen- ing size of the lesion, whereas the relative number of cysts
cies surrounded by a narrow, radiopaque or corticated mar- increased. Table  3.1 summarises their data and shows
gin that extends from the lamina dura of the involved tooth that if a lesion measured between 10 and 14 mm in radio-
(Figure  3.4; see also Figure  2.2). In infected or rapidly graphic diameter, there was an equal chance of it being
enlarging cysts, the corticated margin may not be present. a granuloma or a cyst. About one-­third of lesions measur-
A residual cyst is usually round to oval with a well-­ ing 5–9 mm were cysts and one-­third of lesions measuring
demarcated and often corticated margin. They are found 15 mm or more were granulomas. However, they found
within an edentulous area of the jaws, at a site of a previous that very large lesions, over 20 mm, were almost always
tooth extraction (Figure  3.5). With a residual cyst, the cysts, a finding confirmed by others (Natkin et al. 1984).
­differential diagnosis of keratocyst must be considered. Mortensen et al. (1970) also noted that many cysts had a
A radicular cyst on the lateral margin of a root in associa- diffuse radiographic margin and therefore lacked the
tion with an accessory root canal must be differentiated
from a lateral periodontal cyst. Root resorption is not often
seen on routine radiographs, but it may occur.
Despite these well-­described features, many studies have
shown that it is not possible to reliably differentiate
­radiologically between a radicular cyst and a periapical

Figure 3.5  Radiograph of a residual cyst. The lesion is at the


site of a previously extracted tooth. The lesion must be
differentiated from an odontogenic keratocyst.

Table 3.1  The relationship between the size of a lesion on


radiological examination and the histological diagnosis of a cyst
or granuloma. The arrow illustrates the increasing chance of a
lesion being a cyst.

Size (mm) Granulomas (%) Cysts (%)

20+ <10 90+


15–19 35 65
10–14 50 50
Figure 3.4  Radiograph of a radicular cyst. The lesion is a 5–9 65 35
well-­defined radiolucency associated with the apex of a
non-­vital root-­filled tooth. Source: Based on Mortenson et al. (1970).
26 Radicular Cyst

cortication often described as typical for radicular cysts. have an indication, a priori, whether a periapical lesion is a
This suggested that a corticated margin, although com- cyst or a granuloma. There are a number of key features that
mon, was not a specific diagnostic criterion for a cyst, a together can identify that a lesion is odontogenic and can be
finding supported by others. Ricucci et  al. (2006a) exam- highly suggestive of a cyst (Box 3.2). Both granulomas and
ined 57 periapical radiolucent lesions and found that of cysts are associated with a heavily restored or carious tooth
10  lesions with a corticated outline, only 3  were cysts. and lie within the periodontal ligament. Cysts are more often
Conversely, 40 of the 47  lesions without cortication were over 15 mm in diameter, more often have a corticated margin,
granulomas. and show a greater degree of radiopacity.
Shrout et al. (1993) used radiometric methods to analyse
the grey levels on digitised images of periapical lesions.
In a pilot study of only 10 mandibular lesions, they showed ­Pathogenesis
that analysis of grey levels could correctly identify 4 of 6
granulomas and all 4 cysts. They concluded that it may be For any type of cyst to develop, three elements are needed:
feasible to differentiate between radicular cysts and peria- a source of epithelium, a stimulus for epithelial prolifera-
pical granulomas on the basis of radiographic density. With tion, and a mechanism of growth and bone resorption. In
the advent of digital radiography and powerful software to the case of a radicular cyst, the process is driven by an
routinely analyse images, it would be interesting to see if inflammatory response at the apex of a tooth (Box  3.3).
these findings could be confirmed. Early studies examining Although trauma, instrumentation, and irritation from fill-
grey levels on images from cone beam computed tomogra- ing materials may cause inflammation, in reality this is
phy (CBCT) suggest that this may provide an accurate diag- usually short lived and the chronic inflammation needed
nosis (Simon et  al.  2006). Measurement of conventional to initiate cyst formation almost always follows microbial
parameters on CBCT, however, have proved to be no more infection in a dead pulp following caries. The presence of
accurate than conventional X-­rays (Guo et al. 2013). bacteria and their products in the root canal then leads to
Although teeth may be resorbed by radicular cysts, there periapical inflammation and the formation of a periapical
is a poor correlation between radiological evidence of granuloma, which may then lead to cyst formation.
resorption and actual tooth resorption on histology. Laux It must be noted, however, that although cysts are a
et  al. (2000) compared the radiological and histological direct sequela of periapical inflammation, cyst formation is
findings in 114 periapical lesions. Ninety three (81%) not inevitable and radicular cysts are in fact quite rare rela-
showed histological evidence of tooth resorption, but only tive to the prevalence of caries and periapical lesions. In an
21 (19%) showed evidence of resorption on the radiographs. analysis of 256 periapical lesions, Nair et al. (1996) showed
It should be noted, however, that only 30 of the 93 lesions that only 15% were actually cysts, although a further 37%
with histological resorption showed dentine involvement. were granulomas with proliferating epithelium. Their cri-
In the majority (63 cases) only cementum was involved teria for diagnosis of a cyst was unusually stringent and
and it was acknowledged that this would not normally be depended on the ability to examine, in multiple serial sec-
visible on a plain radiograph. tions, the entire specimen and to be able to visualise a
The data from these studies suggest that there are no spe-
cific radiological features that can be used to distinguish
Box 3.3  Pathogenesis: Key Facts
between a radicular cyst and a periapical granuloma.
Clinicians know that it is useful in treatment planning to Three elements are needed:
●● A source of epithelium
●● A stimulus for epithelial proliferation
Box 3.2  Clinical and Radiological Features: Key Facts
●● A mechanism of growth and bone resorption
●● Always associated with a non-­vital tooth
The cyst develops in three phases:
●● Most commonly found on upper anterior teeth
●● Often symptomless and found on radiological ●● Phase of initiation – rest cells of Malassez are stim-
examination ulated to proliferate within a periapical granuloma
●● Firm or hard swelling, but large lesions may show ●● Phase of cyst formation – a cavity within the granu-
‘egg-­shell’ crackling loma becomes lined by proliferating epithelium
●● Residual cysts are found at sites of a previous tooth ●● Phase of growth and enlargement  –  growth and
extraction enlargement are driven by increased osmotic pres-
●● Radiology shows well-­demarcated, corticated lesion sure, and are associated with inflammation, cell
●● Rarely greater than 30 mm in diameter proliferation and bone resorption
­Pathogenesi  27

distinct epithelial-­lined cavity. Few studies have under- Lin et  al.  2007; Graves et  al.  2011; Graunaite et  al.  2012;
taken such meticulous examination of lesions using serial Marton and Kiss 2014; Bernardini et al. 2015).
sections, but those that have have confirmed the findings It is now well accepted that the whole process is started
of Nair et al. (1996) that cysts form the minority of periapi- by bacterial infection in the necrotic pulp, and that the
cal lesions. The actual frequencies reported have been key  initiating event is the action of bacterial endotoxins
17.1% (Simon 1980), 15.2% (Nair et al. 1996), 32.0% (Ricucci (lipopolysaccharides, LPS) on periodontal ligament fibro-
et al. 2006b), and 24.2% (Ricucci et al. 2020). blasts, which then secrete a range of pro-­inflammatory
It is convenient first to consider the pathology of periapical cytokines and chemokines. Meghji et  al. (1996) studied
periodontitis and then to discuss the pathogenesis of radicu- cyst fluids and cultured cyst explants from radicular cysts,
lar cysts in three phases: the phase of initiation, the phase of keratocysts, and follicular cysts. They showed high levels
cyst formation, and the phase of growth and enlargement. of endotoxins in radicular cysts compared with negligi-
ble  levels in the other cyst types. These endotoxins were
LPS derived from Actinobacillus actinomycetemcomitans,
Pathology of Periapical Periodontitis
Porphyromonas gingivalis, and Escherichia coli. Wayman
Radicular cysts develop within a pre-­existing periapical et al. (1992) were able to cultivate bacteria from 51 periapi-
granuloma where proliferation of the epithelial cell rests cal lesions, 23 of which had had previous endodontic ther-
of  Malassez is initiated by inflammation caused by apy. They found a total of 50 different species, of which
the  necrotic debris and bacterial factors derived from over 90% were anaerobes. Bacteria were cultured from both
the  dead pulp. One can think of this process as part of granulomas and cysts with equal frequency, but were seen
­normal healing, where the host response acts to prevent histologically in only 8 (13%) cases. Tek et al. (2013) were
dissemination of bacteria and stimulates epithelial regen- only able to culture bacteria from about one-­third of peri-
eration. Histologically a periapical granuloma is composed apical lesions. Nair (1987), however, in a detailed ultras-
of granulation tissue at various stages of development tructural analysis, found bacteria in 100% of the root canals
and maturation. Early lesions may show clear evidence of of teeth affected by periapical lesions, but in only 4 of
acute inflammation with polymorphonuclear leukocytes 31 cases were bacteria found in the extraradicular tissues.
(PMNs), while later lesions become dominated by lympho- In these cases, 1  was infected with Actinomyces and in 3
cytes and plasma cells and may develop a fibrous outer cap- the bacteria were isolated on the dentinal wall close to the
sule. The process involves many cell types that give rise to foramen and were associated with abscess formation with
a massive array of pro-­inflammatory mediators, cytokines, accumulations of PMNs. In a later review of the literature,
chemokines, and growth factors, which work together in Nair (2004) concluded that periapical lesions rarely har-
complex interactions to pursue the final goal of elimina- bour bacteria unless secondarily infected. This is supported
tion of the cause and healing. However, as well as being by Ricucci and Siqueira (2010), who studied 106 biopsy
protective to the host, these immunomodulatory pathways specimens of tooth roots with apical lesions. They found
are also destructive, especially if the bacterial insult per- evidence of bacteria in only 6% of extraradicular lesions.
sists, as is often the case in periapical periodontitis (Marton However, bacterial biofilms were found in 77% of the root
and Kiss  2014). Many studies have examined periapical canals and were more common in untreated (80%) than
lesions to determine which cells and pro-­inflammatory treated canals (74%). These authors also showed that
mediators may be important in the development of cysts, ­biofilms were significantly associated with epithelialised
and it is difficult and unnecessary to review them all. For lesions, with a frequency of 95%, 83%, and 69.5% in cysts,
example, Bernardi et al. (2015) undertook a review of the abscesses, and granulomas, respectively.
literature and identified 187 references to epithelium in Taken together, these data indicate that periapical perio-
periapical lesions between 1975 and 2014. After removing dontitis is a result of endodontic infection, following death
duplicates and simple case reports, they found 42 publica- of the pulp due to caries. Although bacteria proliferate
tions that reported the expression of various biological within the root canal, they are rarely encountered in the
­factors that might be involved in the formation of radicu- periapical lesions, confirming the view that cyst formation
lar  cysts. Because the overall biological process is one of is initiated by leakage of endotoxins through the apical
inflammation and the immune response, the factors are foramen.
not specific to radicular cysts, and the whole field is With regard to the host response, all the expected
­constantly evolving. Readers interested in the finer points immune cells have been reported in periapical granulo-
of the associated immunopathological processes should mas, with most studies indicating that T lymphocytes
search for relevant up-­to-­date papers. There are a number predominate and that among these, T-­helper cells (CD4)
of detailed reviews that have helped to inform the follow- predominate over cytotoxic/suppressor cells (CD8) (Skaug
ing discussion of the key points (Nair 2004; Silva et al. 2007; et al. 1984; Nilsen et al. 1984; Liapatas et al. 2003). However,
28 Radicular Cyst

the proportions of different cell types may vary between abscess formation may be a key factor in cyst formation
sites in the same lesion, or may depend on the stage (or (see later in this chapter).
‘age’) of the lesion (Matthews and Browne 1987). Of par- Although the main initiating factor of a periapical lesion
ticular importance, especially in the balance between pro- is bacterial LPS, the lesions are then sustained by a com-
tective and destructive tissue responses, is the relative plex network of inflammatory mediators, including
number of the T-­helper cell subsets Th1, Th2, Th17, and cytokines, chemokines, and the eicosanoids (mainly pros-
regulatory T cells (Tregs) (Marton and Kiss  2014). Th1 taglandins). These have multiple roles in cell activation
cells, which secrete a range of pro-­inflammatory cytokines, and migration, in epithelial proliferation, and in bone
always predominate in periapical granulomas. Th17 cells resorption. In early lesions there is good evidence that the
are the primary source of interleukin (IL)-­17, which is chemokine CXCL8/IL-­8 is important since it is absent in
also pro-­inflammatory (Graunaite et al. 2012; Marton and normal pulp, but found in about 95% of periapical lesions.
Kiss  2014), but their action is moderated by Treg cells, LPS stimulates periodontal ligament fibroblasts to secrete
which are a major source of anti-­inflammatory transform- CXCL8/IL-­8, which has a pivotal role in PMN migration
ing growth factor (TGF)-­β. The Th17/Treg ratio seems to be and therefore in lesion initiation (reviewed in Silva
an important regulator of the intensity of the inflammatory et al. 2007; Graunaite et al. 2012; Marton and Kiss 2014).
response and may thus determine the extent of tissue This chemokine is also  important in bone resorption.
destruction (Marton and Kiss  2014). Kontiainen et  al. Other pivotal cytokines are  the interleukins, especially
(1986), Babál et al. (1987), and Gao et al. (1988a) found that IL-­1 and IL-­6, which have roles in activating PMNs and
suppressor/cytotoxic cells (CD8) dominated. Gao et  al. lymphocytes, and in bone resorption (reviewed in
(1988a), however, also noted that although suppressor/ Graunaite et al. 2012). Table 3.2 lists a number of selected
cytotoxic cells outnumbered T-­helper cells in periapical biological factors that may be involved in the pathogene-
granulomas, T-­helper cells predominated in the cysts that sis of radicular cysts. Mostly this list is derived from the
they examined. A more recent study (Liapatas et al. 2003) reviews mentioned previously (Bernardini et  al.  2015;
has confirmed most of these findings. It showed that T cells Marton and Kiss  2014; Graunaite et  al.  2012; Graves
predominated and that helper cells predominated over et  al.  2011; Silva et  al.  2007; Lin et  al. 2007; Nair  2004).
cytotoxic/suppressor cells in most periapical granulomas The actions of those factors thought to have important or
and cysts. In cysts, however, they found increased numbers specific roles in the formation of radicular cysts are dis-
of plasma cells, suggesting that humoral immune reactions cussed in the sections that follow.
may take on a more important role in cysts. These studies
illustrate the complex nature of the immune interactions
Phase of Initiation
in periapical lesions and may explain the discrepancy in
the results reported by different workers. It is generally agreed that the source of epithelium for a
The proportion of B lymphocytes has been reported to radicular cyst is the epithelial cell rests of Malassez. During
be about 20% (Kontiainen et al. 1986), with plasma cells tooth development the epithelial root sheath of Hertwig
varying from 2% (Kontiainen et  al.  1986) to 13% (Stern maps out the shape of the roots and initiates dentine for-
et al. 1982). As would be expected, the vast majority (over mation. When tooth formation is complete, the sheath
75%) of plasma cells express immunoglobulin (Ig)G, with ­disintegrates and epithelial remnants remain in the perio-
15–20% IgA and very few IgE or IgM (Stern et  al.  1981; dontal ligament as the rest cells of Malassez. There are two
Smith et  al.  1987). Other cell types that play a role in common misconceptions about these cells: first, that they
­periapical lesions include macrophages, mast cells, and are small islands; and second, that they have no normal
Langerhans cells (Pulver et al. 1978; Kontiainen et al. 1986; function. It is now clear that the epithelial remnants form
Drazic et al. 2010). Langerhans cells may be of particular a network or mesh that lies in the periodontal ligament and
interest because they have been found in the epithelial lin- surrounds or embraces the tooth root. Only in histological
ings of radicular cysts (Contos et al. 1987; Matthews and sections do they appear to be isolated islands (Figure 3.6).
Browne 1987; Gao et al. 1988a; Liapatas et al. 2003) and It is now also apparent that the cell rests of Malassez
are most prominent in areas of heavy inflammation. have a number of important functions in normal periodon-
Although they are known to be important in antigen pres- tal homeostasis, including cementogenesis, healing and
entation, it has been suggested that they may also be asso- regeneration (Keinan and Cohen 2013; Xiong et al. 2013).
ciated with epithelial proliferation (Carillo et  al.  2010). They are thus important in maintaining periodontal
Non-­immune cells, including fibroblasts, endothelial health during all the normal challenges of tooth move-
cells, and epithelial cells, are also present and may be ment and function as well as in responses to trauma, ortho-
involved in producing relevant cytokines and growth fac- dontic tooth movement, and periodontitis. These properties
tors. Lesions also contain PMNs and primary or secondary are being exploited in the development of therapeutic
­Pathogenesi  29

Table 3.2  Biological factors that have a role in the pathogenesis of radicular cyst.

Factors Cell(s) of origin Target cell (ligand(s)) Key function

Bacterial factors
LPS Bacteria Fibroblasts and many Induces a wide range of mediators, including
(endotoxin) cell types (CD14/ CXCL8/IL-­8, TNF-­α, IL-­1, RANKL, OPG. Indirectly
TLR) stimulates bone resorption
Cytokines
IL-­1α Macrophages, PMN, Attracts and activates PMNs; stimulates production
osteoclasts, epithelial cells, of prostaglandins, proteolytic enzymes, cytokines
dendritic cells IL-­6, IL-­8; stimulates bone resorption and inhibits
bone formation (originally called osteoclast-­
activating factor, OAF)
IL-­1β Macrophages Monocytes Inhibits osteoclast formation and bone resorption
IL-­6 Macrophages, epithelial Activates and stimulates PMNs and T cells;
cells, PMN, Th2 cells, B stimulates differentiation of B lymphocytes into
lymphocytes, endothelial plasma cells; stimulates osteoclasts and bone
cells, fibroblasts resorption; down-­regulates production of IL-­1
TNF-­α Macrophages, Th1 cells, Activates lymphocytes and macrophages; stimulates
PMN, fibroblasts bone resorption
IL-­17 Th17 Up-­regulates secretion of IL-­1, IL-­6, TNF-­α, and IL-­8
secretion; attracts PMNs; stimulates osteoclasts and
bone resorption
GM-­CSF Macrophages, T Functionally activates macrophages and PMNs
lymphocytes, endothelial
cells, PMN
TGF-­β Lymphocytes (Treg), PMNs, macrophages Anti-­inflammatory; suppresses T and B lymphocytes;
macrophages, fibroblasts, down-­regulates production of IL-­1, IL-­6, TNF-­α, and
osteoblasts, osteoclasts, IFN-­γ; blocks production of nitric oxide by
epithelial cells macrophages; inhibits bone resorption; inhibits Th17
and promotes Treg formation
IFN-­γ Th1 cells, dendritic cells Th lymphocytes Activates macrophages; induces IL-­1 production;
inhibits RANKL and bone resorption
IL-­12 Th1 cells, macrophages, Up-­regulates IL-­1 and IFN-­γ; stimulates Th1
dendritic cells differentiation; suppresses Th2 differentiation
IL-­10 Macrophages, dendritic cells, Anti-­inflammatory; down-­regulates IL-­1 and IFN-­γ;
lymphocytes (Treg) inhibits action of RANKL and bone resorption
IL-­4 Th2 cells Inhibits bone resorption; inhibits Th17 formation;
down-­regulates IL-­1
RANKL Normally present on Osteoclasts and Activates osteoclasts; positively regulates bone
osteoblasts; also Th1cells, precursors (RANK) resorption
endothelial cells, fibroblasts,
PMNs, epithelial cells; may
be soluble
OPG Osteoblasts, some epithelial Osteoblasts (RANKL) Decoy receptor for RANKL and blocks RANK/
cells, endothelial cells, B RANKL pathway; inhibits osteoclastogenesis and
cells negatively regulates bone resorption
Chemokines
CXCL8 (IL-­8) Macrophages, PMN, Th1 CXCR1-­2 Attracts PMNs and macrophages; chemotaxis and
cells, Th17 cells differentiation of osteoclasts
CXCL12 Endothelial cells Osteoclast precursors Chemotaxis and differentiation of osteoclasts;
(SDF-­1α) (CXCR4); PMNs attracts PMNs; up-­regulates MMPs
CCL7 (MCP-­3) Endothelial cells, Osteoclasts and Chemotaxis and differentiation of osteoclasts
lymphocytes, fibroblasts, precursors
plasma cells

(Continued )
30 Radicular Cyst

Table 3.2  (Continued)

Factors Cell(s) of origin Target cell (ligand(s)) Key function

CCL5 T cells, fibroblasts, Osteoclasts and Chemotaxis and differentiation of osteoclasts


(RANTES) osteoclasts, osteoblasts precursors (CCR1,
CCR5)
CCL-­2 Osteoblasts Osteoclasts and Chemotaxis and differentiation of osteoclasts
(MCP-­1) precursors (CCR2)
CCL3 Fibroblasts, osteoclasts, Macrophages Chemotaxis and differentiation of osteoclasts;
(MIP-­1α) osteoblasts (CCR1), attracts macrophages
lymphocytes/Th1
(CCR5)
CCL4 Th1 cells Chemotaxis and differentiation of osteoclasts;
(MIP-­1β) activates macrophages
Prostaglandins
Prostaglandins Macrophages, fibroblasts, Osteoclasts Stimulates osteoclasts and bone resorption
(PGE2) inflammatory cells, epithelial (receptors EP1–EP4)
cells, endothelial cells

GM-­CSF, granulocyte-­macrophage colony-­stimulating factor; IFN, interferon; IL, interleukin; LPS, lipopolysaccharides; MCP, monocyte
chemoattractant protein; MIP, macrophage inflammatory protein; MMP, matrix metalloproteinase; OPG, osteoprotegerin; PMN,
polymorphonuclear leukocyte; RANK, receptor activator of nuclear factor kappa B; RANKL, receptor activator of nuclear factor kappa B ligand;
RANTES, regulated upon activation, normal T cell expressed and presumably secreted; SDF, stromal cell-­derived factor; TGF, transforming
growth factor; TLR, Toll-­like receptor; TNF, tumour necrosis factor; Treg, regulatory T cell.

approaches to promote periodontal regeneration in the


management of periodontal diseases (Xiong et al. 2013).
There is no doubt that the start of the process of cyst for-
mation involves the proliferation of the epithelial cell rests
within the inflamed tissues of a periapical granuloma. As
discussed previously, LPS is the key factor that initiates the
inflammatory and immune response, but it may also
directly cause epithelial proliferation. In their study of flu-
ids and explants from radicular cysts, keratocysts, and fol-
licular cysts, Meghji et al. (1996) showed high levels of LPS
in radicular cysts and demonstrated that it could directly
stimulate epithelial proliferation in a dose-­dependent man-
ner. They proposed that bacterial LPS, derived from the
necrotic pulp, is the key initiating factor in the pathogene-
sis of radicular cysts. In this same study they also provided
evidence that cytokines can directly stimulate epithelial
proliferation. All cysts contained IL-­1α and IL-­6, but radic-
ular cyst explants produced significantly more IL-­6 than
either keratocysts or follicular cysts. Further experiments
showed that IL-­1 and IL-­6, and culture supernatants from
cyst fibroblasts, were able to stimulate epithelial prolifera-
tion in a dose-­dependent manner. Significantly, this activ-
ity was further enhanced by the addition of LPS.
In earlier experiments, the same research group (Meghji
Figure 3.6  Rest cells of Malassez appear as multiple small et al. 1989; Bando et al. 1993) used immunocytochemistry
islands of epithelium (arrows) within the periodontal ligament. to localise cytokines and adhesion molecules in the walls
­Pathogenesi  31

of radicular cysts. All cysts showed positive staining for 1) Central necrosis or nutritional deficiency theory. This
IL-­1α, IL-­1β, and IL-­6 in the epithelial lining and in vascu- proposes that a cyst cavity forms within a proliferating
lar endothelial cells. Tumour necrosis factor (TNF) and mass of epithelial cells due to loss of nutrition followed
CXCL8/IL-­8 were occasionally seen in macrophages. The by degeneration and death of cells in the centre of
cell adhesion molecules ICAM-­1 and ELAM-­1 (E-­selectin) the mass.
were also identified in all lesions and were localised to 2) Abscess theory. This postulates that the proliferating epi-
endothelial cells, epithelium, and inflammatory cells. thelium surrounds an abscess cavity, in effect walling
Kusumi et  al. (2004) produced further evidence that off the central focus of inflammation. Essentially this
IL-­6 has an important role. They used reverse transcriptase represents the process of normal wound healing, where
polymerase chain reaction (RT-­PCR) to study a number of epithelium proliferates to cover denuded connective
cytokines in tissues from 19 radicular cysts and compared tissues.
expression with normal gingivae and periodontal ligament. 3) Merging epithelial strands theory. This proposes that the
They found variable expression of cytokines in all tissues, proliferating epithelium forms a three-­dimensional ‘ball
but most cysts expressed IL-­1β, IL-­6, CXCL8/IL-­8, TNF-­α, mass’ (Lin et al. 2007), which entraps inflamed connec-
interferon (IFN)-­γ, and TGF-­β1, and most of these showed tive tissue. This connective tissue then breaks down due
increased expression compared with normal tissues. All to loss of a blood supply and a cyst cavity forms.
the cytokines were expressed at low levels except for IL-­6,
Proponents of each process often promote their favoured
which showed high levels of secretion from fibroblasts
theory, while suggesting that others are not tenable (Lin
extracted from the radicular cysts.
et al. 2007; Nair et al. 2008; Huang 2010), but in fact they
These studies confirmed that the walls and epithelial lin-
are not mutually exclusive and all three have a similar
ing of radicular cysts synthesise a range of cytokines and
premise  –  that epithelium proliferates to surround and
growth factors that are known to be involved in epithelial
encase a focus of inflamed and degenerating or necrotic tis-
proliferation (Table 3.2).
sue. There is little difference, for example, between an
The role of abscess formation in the formation of a cyst
abscess (theory 2) and necrotic connective tissue (theory
will be discussed below, but there is good evidence that an
3), apart perhaps from the degree or stage of necrosis and
acute inflammatory cell infiltration may be directly associ-
the types of cells present. Similarly, the necrotic centre
ated with epithelial proliferation, since many early studies
of  an epithelial mass (theory 1) may contain PMNs and
were able to demonstrate large numbers of PMNs in the
essentially become an abscess, a feature familiar to tumour
proliferating epithelium (Shear 1963a, 1964; Cohen 1979;
pathologists in the form of comedo necrosis. Thus on histo-
Johannessen  1986). As mentioned above, LPS from the
logical examination, the presence of an abscess cavity lined
root canal stimulates CXCL8/IL-­8 secretion from perio-
by epithelium may support theory 1 or 2. Also, in the
dontal fibroblasts via the co-­receptors CD14 and Toll-­like
receptor (mainly TLR4). In addition, epithelial cells in peri-
apical lesions have been shown to express CD14/TLR4
(Leonardi et al. 2015), and there is evidence that CXCL8/
IL-­8  may directly cause epithelial proliferation as well as
being a chemoattractant for PMNs (Silva et al. 2007; Marton
and Kiss 2014). This would explain the association of pro-
liferating epithelium and PMNs in early lesions.

Phase of Cyst Formation


The next phase in the pathogenesis of a radicular cyst is
the  process by which a cavity comes to be lined by the
­proliferating odontogenic epithelium. Many will think it
astonishing that the actual process of cyst formation is so
poorly understood, and although a number of theories
have been debated for decades, there are very few observa-
tional or experimental data to support or refute any of
Figure 3.7  Arcades and rings of proliferating epithelium in a
them. There are three proposed mechanisms of cyst forma-
periapical granuloma. The epithelium surrounds islands of
tion (Shear  1963a; Summers  1974; Valderhaug  1974; Lin inflamed connective tissue that may break down to initiate early
et al. 2007; Nair et al. 2008; Huang 2010): cyst formation.
32 Radicular Cyst

previous edition of this book we showed a photomicro- in earlier lesions. In lesions examined after 200 days, 11 of
graph (Figure  3.7) of arcading epithelium surrounding 16 (69%) developed cysts. Even after 300 days, 3 of 8 lesions
inflamed connective tissue in support of theory 1 – but in showed no inflammation, but 4 had developed into cysts.
fact this pattern of proliferating epithelium, with entrap- Of relevance to the proposed mechanisms of cyst forma-
ment of connective tissue, is very similar to the merging tion is that he did not see evidence of intraepithelial
epithelial strands theory of Lin et al. (2007) (theory 3). degeneration with formation of microcysts, but observed
In support of theory 1, there is some histological evi- long strands of proliferating epithelium lining surfaces of
dence for central necrosis or nutritional deficiency. In granulation tissue, or arcades surrounding cores of vascu-
some periapical lesions, distinct clefts may be seen in larised or degenerating granulation tissue. These observa-
sheets of epithelial cells (Shear  1963a) (Figure  3.8), and tions support the merging epithelial strands theory (theory
the proliferating epithelial masses often show considerable 3). However, in most cases Valderhaug also observed that
intercellular oedema. These intercellular accumulations the proliferating epithelium was associated with PMNs,
of  fluid coalesce to form microcysts containing epithelial but he did not describe frank abscess formation. He also
and inflammatory cells, including PMNs (Figure  3.9). showed that in most cysts the epithelial lining was closely
These central areas have been shown to contain high lev- connected to the roots around the apical foramen, sup-
els of  acid  phosphatase activity and proteolytic enzymes porting the notion that the epithelium is reforming an
­consistent with autolysis or necrosis (Lutz et  al.  1965; intact integument, and that the cysts are pocket or bay
Summers 1972, 1974). Microcysts may increase in size by cysts. In a very similar study, Valderhaug (1974) examined
coalescence with adjacent microcysts and, once established, 52 primary teeth, and although periapical inflammation
the cyst increases in size by mechanisms that are dis- was common, he found small cysts only in ‘a few cases
cussed later. with long observation periods’. Of relevance to the theo-
Most information regarding cyst formation is taken ries of cyst formation is that he found that abscess forma-
from histological observation of biopsy specimens taken tion, often with oral fistulas, was common and more
from humans and there are very few longitudinal studies. frequently seen than in his previous study on permanent
In  experiments that have not been repeated, Valderhaug teeth (Valderhaug  1972). His findings, however, did not
(1972) induced radicular cysts in monkeys. Because of the support the abscess theory, since ‘proliferating epithelium
rarity of such experiments, it is worth considering some of was not observed in the periapical area in connection with
the details of his findings. He induced pulpal necrosis abscess formation’.
sequentially in 39 teeth in 4 animals and was able to histo- Despite this, as described previously, there is good
logically examine periapical inflammation and cyst forma- ­evidence that PMNs are a prominent feature of periapi-
tion for up to 360 days after the pulps were removed. Of cal  granulomas and are associated with epithelial
interest is that he did not observe any cysts until after
200 days, although proliferating epithelium was observed

Figure 3.9  Degeneration of cells in the centre of a mass of


proliferating epithelium in a periapical granuloma. There is an
intense infiltration of lymphocytes and polymorphonuclear
Figure 3.8  Sheet of epithelial cells in a periapical lesion. A leukocytes. Accumulations of intercellular fluid coalesce to form
distinct cleft has formed and this may initiate a radicular cyst. a microcyst.
­Pathogenesi  33

proliferation  (Valderhaug  1972; Shear  1963a,  1964; exclusive. There is good observational or experimental evi-
Cohen 1979; Johannessen 1986; Marton and Kiss 2014). In dence for each but, conversely, there is little evidence to
their examination of 256 periapical lesions, Nair et  al. refute any of them.
(1996) showed that 90 (35%) were an ‘abscess’ and that
two-­thirds of these contained epithelium. However, the Growth and Enlargement of the Radicular Cyst
definition of an abscess was of a collection of PMNs in a
Role of Hydrostatic Pressure
pre-­existing periapical granuloma – making distinction of
The third phase in the pathogenesis of the radicular cyst is
a primary abscess from a collection of PMNs in a necrotic
its growth and enlargement, which must involve a mecha-
lesion difficult. In  support of the abscess theory, Nair
nism for expansion and for resorption of alveolar bone.
et  al. (2008) were able to demonstrate that epithelium
Almost without exception, radicular cysts, especially when
implanted into experimentally induced abscesses could
small, are seen as round or spherical radiolucencies on
form cysts. However, the relevance of these experiments
radiographs and on 3D imaging (CT or CBCT). This implies
is uncertain, since the abscesses were induced in the skin
that growth of the cyst is regular and centripetal, and it is
of experimental rats, and cysts only developed in 2 of 16
widely accepted that hydrostatic pressure, due to osmosis,
animals (6%). In addition, it is known that implanted epi-
provides the slow and evenly distributed forces necessary
thelium may cause cysts even in the absence of infection
to achieve this growth pattern. This was first noted by
or abscess formation. Nevertheless, we have seen cases of
Warwick James in 1926 in an address to the Royal Society
abscess formation in periapical granulomas where the
of Medicine. He also reported that he had measured the
abscess cavity has become encased in epithelium to form
increased pressure in cysts and was probably the first to
a cyst filled with PMNs (Figure  3.10). This supports the
suggest that ‘The increase in tension may be partly due to
abscess theory, but not to the exclusion of the other pos-
osmosis’ (Warwick James 1926). (Many of these very early
sible mechanisms.
papers are freely available online and are recommend for
It seems, therefore, that there is little difference between
their clarity, insight, and the quality of the scientific
the three proposed mechanisms  –  cyst formation occurs
observations.) The evidence for this was provided by early
due to a ‘walling-­off’ of inflamed connective tissue by a
experiments carried out by Paul Toller, more than half a
process of epithelial proliferation similar to healing at an
century ago, which have never been repeated or bettered
epithelial surface. This is due to the innate property of epi-
(Toller 1948, 1966b, 1967, 1970a, 1970b). In his first paper,
thelium to form an external protective integument. The
Toller noted that early surgeons had observed that when
cyst lumen therefore represents the external environment,
opened, jaw cysts appeared to be under pressure, and that
and in the case of a pocket or bay cyst (see later in this
marsupialisation checked further growth and led to a
chapter) is continuous with the outside through the root
reduction in size of the lesions (Toller  1948). He quoted
canal. All three theories are thus tenable and not mutually
Potts, who in 1927 had noted that cysts displaced the roots
of teeth ‘as if by pressure’. This led to his experiments to
measure the hydrostatic pressure in jaw cysts. Using a can-
nula and a manometer, he showed that the intracystic pres-
sure in radicular and dentigerous cysts averaged 65–70 cm
of water, which was considerably higher than capillary
blood pressure, which was estimated to be lower than
10 cm of water (Toller  1948). In the same paper, he sug-
gested that this increased pressure was a result of ‘osmotic
tension’ and then went on to test whether the cyst lining
acted as a semi-­permeable membrane. He used freshly
­dissected walls from radicular cysts and clamped them
between two cylinders of Ringer’s solution with 5% albu-
min added to one side. In all cases fluid passed through the
cyst wall towards the albumin, showing that the wall acted
as a semi-­permeable membrane.
In later studies, Toller (1970b) showed that the mean
Figure 3.10  A periapical granuloma at the apex of a molar
osmolality of the fluid from 21 apical and residual cysts
tooth root. There is a central focal accumulation of
polymorphonuclear leukocytes that has become surrounded by was 290 ± 14.93 mOsm and was greater than the mean
epithelium (inset). serum osmolality of 279 ± 4.68 mOsm (P < 0.01). Lytic
34 Radicular Cyst

products of the epithelial and inflammatory cells in the Ward et al. (2004) used mathematical modelling to simu-
cyst cavity provided the great numbers of smaller mole- late odontogenic cyst growth. They assumed a spherical
cules that raised the osmotic pressure of the cyst fluid. cyst lined by a semi-­permeable membrane and with a
Toller believed that the upper limit of permeability in most ­central osmotic pressure as a result of accumulation of
cysts was close to the molecular size of albumin (molecular degraded cellular material. The model supported the con-
weight 69 kDa) and that particles of larger size would find clusions of the early experimental work, that osmotic pres-
difficulty in diffusing across a cyst lining. These findings sure played an important part in cyst growth. Interestingly,
were confirmed by Skaug (1976a), who conducted similar the model also confirmed the findings of Kubota et  al.
experiments and showed that the intracystic pressure (2004), and suggested that as the cyst became larger,
ranged from 25 to 66 mmHg and that these pressures were osmotic pressure played a lesser part and cell proliferation
restored 3–7 days after aspiration, suggesting that there was became more important.
constant movement of fluid into the cyst lumen. Osmotic pressure must be maintained by a high concen-
This pioneering work on the role of hydrostatic pressure tration of soluble proteins in the cyst fluid. Electrophoretic
in the growth of odontogenic cysts was mostly undertaken studies (Toller and Holborow 1969; Toller 1970a) demon-
during the 1970s, but it has not been superseded or refuted strated that radicular cyst fluids contained small molecular-­
by more contemporary experiments. Nair (1998, 2004) has sized albumin and β1-­globulin in quantities comparable
suggested that osmotic pressure as a factor in the develop- with the patient’s serum, but had fewer, if any, of the larger
ment of radicular cysts has been ‘eliminated’. As evidence protein molecules. α-­and β2-­globulins were greatly dimin-
for this he cites the fact that the lumen of pocket or bay ished or absent, and γ-­globulins (mainly immunoglobu-
cysts is open to the root canal, and cannot therefore sus- lins) varied greatly in quantity, but were most often found
tain an increased internal pressure. He suggests that cyst in inflamed cysts. They showed that more than half display
growth is sustained by molecular mechanisms. This is levels of immunoglobulins much higher than the patient’s
undoubtedly correct – a cascade of biological factors under- own serum. In 19 cyst fluids in which levels of IgG, IgA,
pins the processes of epithelial proliferation, extracellular and IgM were measured independently, all three were sig-
matrix destruction, and bone resorption, but there is still nificantly raised in most of the non-­keratinising cysts.
good evidence that there is increased pressure within the Immunofluorescent staining showed that lymphoid cell
cyst lumen, and that osmosis is the driving force. This does aggregates in the walls of radicular cysts often included
not exclude other factors, although it should be noted that numerous plasma cells.
even in pocket cysts it is unlikely that the root canal Skaug (1973,  1974,  1976b,  1977) confirmed that fluid
remains empty and that the lumen is truly open to the oral from non-­keratinising jaw cysts contained high concen-
cavity. It is also well recognised that release of this pres- trations of proteins, including immunoglobulins, but
sure, through decompression, has long been and still is a supported the view that accumulation of cyst fluid
common and effective form of treatment (Castro-­Núñez resulted essentially from inadequate lymphatic drainage
2016). More recently this issue has been re-­evaluated, but of the cyst cavity. He suggested that plasma protein exu-
the outcome remains that hydrostatic pressure is still con- date and hyaluronic acid, as well as the products of cell
sidered to be of primary importance in the growth of all breakdown, contributed to the high osmotic pressure of
cyst types. Kubota et  al. (2004) measured the intracystic the cyst fluid.
fluid pressure of odontogenic keratocysts, dentigerous These data suggest that the accumulation of proteins in
cysts, and radicular cysts. They confirmed the earlier the cyst lumen is a combination of a serum exudate, an
results of Toller (1970b) and Skaug (1976a), that the pres- inflammatory exudate, and breakdown products of cells,
sure was greater than the local blood pressure and that including luminal epithelial cells and inflammatory cells.
there were no differences between the three cyst types. It is likely therefore that the luminal pressure, necessary
They also measured cyst volume and showed that volume for cyst expansion, is greatest when the cyst is most
correlated to the area of the cysts measured on panoramic inflamed, at earlier stages of development. As a cyst gets
radiographs. They correlated the pressure to the areas of the larger and matures, inflammation may subside, the rate of
cysts and found pressures of 337.6 ± 126.0, 258.2 ± 160.9, growth will slow, and a state of equilibrium may be reached.
and 254 ± 157.3 mmHgcm−2 for keratocysts, dentigerous This is supported by the study of Kubota et al. (2004) and
cysts, and radicular cysts, respectively. Furthermore, these the model proposed by Ward et al. (2004), who both show
authors showed that the intracystic pressure in all cyst that pressure and the rate of growth decrease with size of
types was inversely correlated to the cyst size. They there- the lesion.
fore concluded that increased pressure played a pivotal Although osmotic pressure may provide the stimulus for
part in early cyst growth. expansion, it does not operate alone and, as mentioned
­Pathogenesi  35

above, a complex cascade of molecular events is responsi- Degradation of the Connective Tissues
ble for growth. Growth must be accompanied by further and Bone Resorption
epithelial proliferation, and by degradation of adjacent The next crucial element of cyst growth is degradation of
connective tissues and bone resorption. the connective tissues. Among the most important and
widely studied factors are the matrix metalloproteinases
Epithelial Proliferation (MMPs). These are a large family of calcium-­dependent
The stimuli for epithelial proliferation have been discussed and zinc-­containing proteases, capable of degrading a wide
previously, but in an established cyst LPS plays a less range of extracellular matrix proteins. A number of studies
important role, and epithelial growth is sustained by have shown expression of MMPs in odontogenic cysts,
cytokines and growth factors resulting from the inflamma- including the gelatinases (MMP-­2 and MMP-­9; Teronen
tory response (Table  3.2) (Bernardini et  al.  2015; Silva et  al.  1995a; Kubota et  al.  2000; D’addazio et  al.  2014;
et al. 2007; Lin et al. 2007; Nair 2004). However, as the cyst Alvares et al. 2017; Andrade et al. 2017) and collagenases
matures and inflammation subsides, the rate of cell prolif- (MMP-­1, MMP-­8, and MMP-­13; Teronen et al. 1995b; Lin
eration may be relatively low. Soluk ­Tekkeşın et al. (2012a) et  al.  1997; Wahlgren et  al.  2001; D’addazio et  al.  2014;
showed that only about 1.0% of cells were positive for Andrade et al. 2017), suggesting a role for these enzymes in
Ki-­67 in the epithelial lining of radicular cysts, compared cyst growth and development. Furthermore, MMP activity
to 2.0% or more in odontogenic keratocysts. They also increases with the intensity of inflammation and is often
found that radicular cysts showed higher levels of the pro-­ more prominent in periapical granulomas than in estab-
apoptotic protein Bax and low levels of bcl-­2, suggesting lished cysts (Lin et al. 1997; Andrade et al. 2017). In keep-
that low levels of proliferation are accompanied by high ing with this, inflammatory cytokines, especially IL-­1,
levels of apoptosis. Others have identified apoptotic mark- up-­regulate active MMP-­9  in odontogenic cysts (Kubota
ers in radicular cysts and have suggested that this contrib- et al. 2000) and increased numbers of mast cells are associ-
utes to their slow growth (Loreto et al. 2013). As the cyst ated with MMP activation in odontogenic cysts (Teronen
continues to expand, this slow proliferation and high levels et  al.  1996; Rodini et  al.  2008; Andrade et  al.  2017).
of apoptosis probably lead to the thinning of the epithelial Inflammation in radicular cysts is also associated with
lining that is seen in long-­standing and residual cysts secretion of neutrophil collagenase (MMP-­8) and with
(Figure 3.11). increased expression of plasminogen activator (Tsai et al.
2004), which indirectly forms plasmin that may also acti-
vate MMPs.
All odontogenic cysts are encased in the bone of the jaws,
and therefore any expansion must be associated with bone
resorption. Normal bone undergoes continuous remodel-
ling controlled by a number of factors, the most important
of which are receptor activator of nuclear factor kappa B
(RANK), its ligand (RANKL), and osteoprotegerin (OPG,
also known as osteoclastogenesis inhibitory factor). These
form the RANKL/RANK/OPG system, which is primarily
responsible for bone homeostasis (Graves et al. 2011; Kular
et  al.  2012). RANK is expressed on the surface of osteo-
clasts and their precursor cells and when bound by
RANKL promotes osteoclast formation and maturation.
OPG secreted by osteoblasts acts as a decoy receptor and
binds RANKL to prevent activation by RANK. RANKL is
primarily cell bound on the surface of osteoblasts and in
normal bone osteoblast–osteoclast interactions is the main
method of bone turnover regulation. In pathological condi-
tions, however, RANKL may be secreted by osteoblasts and
a number of other cell types, including Th1 lymphocytes,
endothelial cells, and epithelial cells, and may function as
Figure 3.11  A long-­standing radicular cyst. The cyst wall has
a soluble receptor. It should also be noted that RANKL is
become fibrous with little inflammation and the epithelial
lining is relatively thin and regular. Note the considerable the only cytokine that can stimulate osteoclastogenesis,
cellular and proteinaceous debris in the lumen (top left). although a number of other factors support recruitment of
36 Radicular Cyst

osteoclast precursors, or promote osteoclast maturation originally called osteoclast-­activating factor, OAF), IL-­6,
and activation (Table 3.2). and TNF-­α. A range of chemokines are also responsible for
Perturbation of the RANKL/RANK/OPG system is chemotaxis of osteoclast precursors and differentiation of
involved in all pathological conditions associated with osteoclasts, including CXCL8/IL-­8, which has been men-
bone resorption, and many studies have now provided evi- tioned previously as an important initiator of the process,
dence for its role in increased osteoclast activity in many since it is up-­regulated by LPS and also chemotactic to
odontogenic lesions, including radicular cysts (Tay et  al. PMNs. Prostaglandins have also been shown to be impor-
2004; Menezes et al. 2006; da Silva et al. 2008; de Moraes tant mediators of bone remodelling and may act to stimu-
et al. 2011; Graves et al. 2011; Soluk ­Tekkaşin et al. 2011; late both bone deposition and resorption. There are
Belibasakis et al. 2013). a  number of prostaglandins, but PGE2 is particularly
Tay et  al. (2004) were the first to demonstrate that important in bone resorption, since it stimulates expres-
RANKL was expressed in osteolytic lesions of the jaws, sion of RANKL and inhibits OPG on osteoblasts (Blackwell
including radicular cysts. RANKL was expressed in the et al. 2010). Early studies by Harris and his research group
fibrous wall of radicular cysts, and its association with oste- (Harris and Goldhaber 1973; Harris et al. 1973; Harris 1978;
oclast recruitment was confirmed by the demonstration of Meghji et al. 1989) were the first to demonstrate that cul-
tartrate-­resistant acid phosphatase (TRAP) and calcitonin-­ tures of cyst walls had potent bone-­resorbing activity, and
receptor positive osteoclasts adjacent to the RANKL-­ identified prostaglandins as a key factor. Matejka et  al.
positive cells. In a more detailed analysis, Menezes et  al. (1985a,b, 1986) confirmed these findings and also demon-
(2006) confirmed these findings, but were also able to dem- strated that the granulation tissue and the inflammatory
onstrate that RANKL and OPG were expressed by a range cells in the wall of radicular cysts were the main source
of cell types, including PMNs, lymphocytes, macrophages, of  PGE2. Further studies from Harris’s group (Bando
endothelial cells, and the epithelial lining of radicular et  al.  1993; Meghji et  al.  1996) reinforced these findings
cysts. They found that RANKL was more highly expressed and also showed that IL-­1 and IL-­6 were the predominant
than OPG and that expression was greater in cysts than cytokines with bone-­resorbing activities. In immunocyto-
granulomas. Other studies have shown similar levels of chemical studies, Bando et  al. (1993) showed that these
expression, but have also shown, as would be expected, cytokines appeared to be synthesised primarily by the epi-
that RANKL expression is not specific to radicular cysts thelial cells of the cyst lining. Kusumi et  al. (2004) also
and is also found at similar or even greater levels in denti- showed that IL-­6 was the predominant cytokine in radicu-
gerous cysts, odontogenic keratocysts, and ameloblastomas lar cysts and found evidence for synthesis by fibroblasts in
(Tay et al. 2004; Menezes et al. 2006; de Moraes et al. 2011; the cyst wall.
Soluk ­Tekkaşin et  al.  2011). It is likely that the level of
expression at any moment in time will be related to the
degree of ‘maturation’ of the cyst or to the extent of inflam- ­Histopathology
mation. Kawashima et  al. (2007) studied the kinetics of
RANKL, RANK, and OPG expression in experimentally From the foregoing account of pathogenesis, it is clear that
induced periapical lesions in rats. They showed that all on histological examination a periapical lesion may show a
three factors peaked at between two and three weeks, but wide variety of features. Over 100 years ago Thoma (1917)
that the greatest increase was in RANKL, with the highest described five types of lesion that may be encountered
RANKL/OPG ratios at this time. They also found that in  association with a non-­vital tooth. To paraphrase his
RANKL was expressed in a range of cell types, but these descriptions into contemporary terminology, the five lesions
were close to the alveolar bone and were associated with were periapical granuloma, periapical abscess, periapical
activated osteoclasts. The up-­regulation of the RANKL/ granuloma with proliferating epithelium, periapical gran-
RANK/OPG system also correlated to increased expression uloma with early cyst formation, and radicular cyst.
of pro-­inflammatory cytokines, known to be able to acti- However, we should not regard these as individual lesions,
vate RANKL, including IL-­1α and IL-­1β. Similar data have but as a continuum of changes that reflect the pathogenic
been presented in a number of subsequent studies (reviewed pathway, with a fully developed cyst as the end result.
in da Silva et  al.  2008; Graves et  al.  2011; Belibasakis Some have suggested that a diagnosis of a cyst can
et al. 2013), confirming the role of inflammatory cytokines only be made after examination of multiple serial sections
and the close association between the inflammatory to confirm the presence of a true cavity with an epithe-
response and bone resorption. lial  lining (Simon  1980; Nair et  al.  1996; Ricucci and
The most important cytokines involved in the process are Bergenholtz 2004), but this is not practical or sustainable
indicated in Table  3.2, and include IL-­1α (which was in routine clinical practice. The diagnostic histopathologist
­Histopatholog  37

rarely receives an intact specimen, or a specimen still in depend on the age or stage of development of the cyst, or
continuity with the offending root apex. This is because a on the intensity of the inflammation. In early cysts, the epi-
periapical lesion is often surgically removed during an api- thelial lining may be proliferative and show a characteristic
cectomy operation, where the apex of the tooth and associ- arcading with an intense associated inflammatory process
ated soft tissue are removed and a filling material is placed (Figure  3.12), but as the cyst enlarges the inflammation
in the root canal, thus enabling preservation and restora- may subside and the lining becomes thin and regular, with
tion of the affected tooth. Therefore, the pathologist often a certain degree of differentiation to resemble a simple
receives the lesion as a fragmented or irregular curettage non-­keratinised stratified squamous epithelium. In well-­
specimen from which the diagnosis of a cyst must be developed cysts, the lining may vary. At sites of heavy
deduced, in the context of the orientation of the tissues and inflammation, often adjacent to the tooth root, the epithe-
the clinical and radiological findings. An intact specimen lium may be proliferative and thickened, but away from the
may be a spherical or ovoid cystic mass, but often they are tooth the wall is often less inflamed and the lining becomes
irregular and collapsed. Lesions are usually 10–15 mm in thin and regular (Figure  3.11). Long-­standing cysts and
diameter and rarely exceed 30 mm. The walls vary from residual cysts in particular may be lined by a thin, regular
extremely thin to a thickness of about 5 mm. As with all lining of stratified squamous epithelium (Figure 3.13).
cystic lesions, it is important to examine the inner surface The inflammatory cell infiltrate in the cyst wall and the
of the cyst. This may be smooth or corrugated, but mural epithelial lining may vary considerably, since it reflects
nodules or thickenings may be seen, which often represent a  single moment of the pathogenic pathway caught in a
accumulations of cholesterol or hyaline (Rushton) bodies histological section. Thus any of the inflammatory cells
projecting into the lumen. The fluid contents are usually involved in the development of the lesion may be seen.
watery and brown from the breakdown of blood, but when In  early lesions the proliferating epithelial lining usually
cholesterol crystals are present they impart a shimmering contains many PMNs, whereas the adjacent fibrous cap-
gold or straw colour. Pathologists must also recall that a sule is infiltrated mainly by chronic inflammatory cells
radicular cyst is always associated with a non-­vital tooth. If (Shear  1963a,  1964; Cohen  1979; Matthews and Browne
this is not the case, then an alternative diagnosis must be 1987). The proliferating epithelial lining shows a consider-
sought, usually needing careful correlation with clinical able degree of inter-­ and intraepithelial oedema or
and radiological findings. spongiosis.
On histological examination, the key diagnostic criterion The cyst wall is essentially composed of granulation
is to identify a variably inflamed fibrous cyst wall lined ­tissue at various stages of maturation, depending on the
wholly or in part by stratified squamous epithelium age of the lesion and on the proximity to the source of the
(Figure 3.12). The epithelial lining may be discontinuous inflammation. The wall is therefore often zoned, with
and ranges in thickness from 1 to 50 cell layers. The major- heavily inflamed granulation tissue adjacent to the epithe-
ity are 6–20 cell layers thick. The nature of the lining may lial lining, less inflamed maturing granulation tissue cen-
trally, and collagenous fibrous tissue at the peripheral
margin adjacent to the bone (Figure 3.12). Inflammation is
also more prominent adjacent to the root apex  –  where

Figure 3.12  Radicular cyst. The cyst has a thick fibrous wall. Figure 3.13  Quiescent epithelium lining a mature, long-­
The lumen is lined by proliferating and arcading epithelium. standing residual cyst.
38 Radicular Cyst

accumulations of polymorphs may be seen, even in long-­ Table 3.3  Characteristic histopathological features found
standing lesions. The predominant cell population, how- in radicular cysts, with their approximate frequency (see text
for explanation).
ever, is of chronic inflammatory cells, mostly lymphocytes;
plasma cells are also seen, and on occasion may predomi-
Feature Frequency (%)
nate or may form dense focal accumulations.
Remnants of odontogenic epithelium and occasional sat- Keratinisation 2
ellite microcysts may be found in the fibrous capsule and
Ciliated cells 10
there have been reports of examples where epithelial pro-
Hyaline bodies 10
liferation is so extensive that it resembles squamous odon-
Foamy histiocytes 10
togenic tumour (Wright  1979; Simon and Jensen  1985;
Unal et al. 1987; Chrcanovic and Gomez 2018a). These pro- Mucous cells 20
liferations are reactive in nature and should not be inter- Cholesterol 30
preted as a co-­existent neoplasm. The behaviour is that of
the cyst of origin and no further treatment is required if
this observation is made during histological examination of radicular cysts may show some keratinisation and that
of the cyst wall (Chrcanovic and Gomez 2018a). orthokeratin with evidence of a granular cell layer is most
Some cyst walls are markedly vascular. Haemorrhage is common. More recently, Maheswaran et al. (2014) analysed
invariably present and haemosiderin deposits are seen in 38 radicular cysts and 9 residual cysts using Papanicolaou
many specimens (Shear  1963c). Calcifications of various stain and found evidence of keratinisation in 12 (31.6%)
kinds may also be seen. Dystrophic calcifications associated radicular cysts and 6 (66.7%) residual cysts. Orthokeratin
with necrotic and degenerative material in the cyst lumen was only found in 1 residual cyst and only 2 cysts showed
are a particular feature of residual cysts that  have been typical parakeratin. In all other cases the keratin was
present for a long time (High and Hirschmann  1986). described as focal. However, little detail was given and the
Hyaline bodies may also calcify either within the epithelial findings were not illustrated. We interpret this to mean that
lining or among deposits that have extruded into the lumen the Papanicolaou technique revealed occasional superficial
or into the wall. In curettage specimens, trabeculae of reac- orange-­stained cells. Although this may suggest early
tive woven bone and occasionally lamellar bone are often keratinisation, it should be noted that this technique is pri-
found at the periphery of the lesion. Occasionally a well-­ marily a cytological stain and may not be as reliable as a
formed rim of woven bone may be seen. routine haematoxylin and eosin (H&E) stain for identifica-
Although well-­formed colonies of actinomycosis are well tion of keratin in histological sections (Rao et  al.  2015).
described in case reports, this is a rare finding. Hirschberg A more cautious interpretation of Maheswaran et al.’s data
et al. (2003) found colonies of Actinomyces in only 17 of 936 may suggest that only three of their cysts showed clearly
(1.8%) periapical lesions examined, 4 of which were radicu- identifiable keratinisation (6.4%). Our experience would
lar cysts. Nair (2006) found a similar frequency in a review support this, since we rarely see true keratinisation in radic-
of the literature, and postulated that established colonies ular cysts, and when present it affects only a small section
of Actinomyces may persist and be an important cause of the lining. This, and attention to the clinical and radio-
of  endodontic failure and persistent or recurrent lesions. logical findings (association with a non-­vital tooth), should
Ricucci and Siqueira (2008), however, found no evidence prevent the lesion being misinterpreted as odontogenic
for this and suggested that provided the root canal was keratocyst. Also, when present the parakeratin seen in a
properly cleaned, the presence of actinomycosis was not radicular cyst is different morphologically from that seen
associated with treatment failure. in keratocysts, since it lacks the typical corrugated surface
and affects only a small portion of the lesion.
Metaplastic changes, in the form of mucous cells or
Cellular and Metaplastic Changes
­ciliated cells, are frequently found in the epithelial linings
Radicular cysts may show variable histological features of radicular cysts (Table  3.3; Shear  1960b; Browne  1972;
due to reactive changes and to metaplastic changes of Browne and Smith  1991; Slabbert et  al.  1995; Takeda
the lining epithelium. These additional features are charac- et al. 2005; Tsesis et al. 2016). Mucous cells are seen in the
teristic and occasionally assist in diagnosis (Table  3.3, surface layer of the stratified squamous epithelial lining,
Figure 3.14). either as a continuous row (Figure  3.14d) or as scattered
Keratin formation may occasionally be seen in radicu- cells (Figure  3.14c). Ciliated cells may also be seen, but
lar  cysts, but when present it affects only part of the cyst are  always found in association with mucous cells and
wall (Figure 3.14a). Browne and Smith (1991) stated that 2% together they sometimes form quite well-­developed
­Histopatholog  39

(a) (b)

(c) (d)

Figure 3.14  Cellular changes in the lining of radicular cysts. (a) A portion of the lining showing a focal area of orthokeratinisation.
(b) Respiratory-­type epithelium with cilia and occasional mucous (goblet) cells. (c, d) Mucous cells in the surface layer may be
scattered (c) or may form a continuous row (d).

respiratory-­type (pseudostratified columnar ciliated) epi- (5.8%) cysts, and were also more frequent in asymptomatic
thelium (Figure 3.14b). cysts and in cysts with well-­demarcated radiographic mar-
Browne (1972) examined 402 radicular cysts and found gins. This suggests that metaplasia takes time and is more
mucous cells in 159 (39.6%), but cilia were only found in likely to be encountered in well-­established or older cysts.
3 cases (0.7%). Takeda et al. (2005) found mucous cells in This view is supported by the observation of Browne (1972)
18% of radicular cysts, and in most cases they were arranged that there was an increasing frequency of mucous cells
along the surface of the epithelium, but occasional intraep- with age, at the rate of 7% per decade.
ithelial gland-­like structures were also noted, most often Slabbert et  al. (1995) studied 154  mandibular radicular
in  areas where the epithelium was hyperplastic. Browne and residual cysts and found unequivocal mucous meta-
(1972) found no difference in frequency of mucous cells plasia in 15 (10%). In many cases they found that the
between mandibular and maxillary lesions, but Takeda mucous cells were associated with vacuolated cells, many
et al. (2005) found that they were more common in maxil- of which were empty, but some contained fine granules or
lary lesions (21%) than mandibular lesions (14%). In an networks of periodic acid–Schiff (PAS)–positive material.
analysis of 711 radicular cysts, Tsesis et  al. (2016) found The authors observed that the vacuolated cells resembled
mucous cells in 5.3% and 7.4% of mandibular and maxil- those described by Fell (1957) in the process of metaplasia
lary lesions, respectively, but this difference was not signifi- from stratified squamous to ciliated epithelium in explants
cant. They also found that mucous cells were significantly of chick embryo skin grown under the influence of excess
more likely to be found in residual (23.5%) than radicular vitamin A. By analogy to Fell’s findings, Slabbert et  al.
40 Radicular Cyst

(1995) suggested that the vacuolated cells represented an CK18+/CK8+/CK13– phenotype was only found in nasal
intermediate stage in the process of mucous metaplasia. epithelium. Only three cysts showed this phenotype and
Takeda et  al. (2005) found similar clear cells and agreed all  three were large maxillary lesions protruding into the
with Slabbert et al. (1995) that mucous cell differentiation maxillary sinus. They concluded that occasional maxillary
is a process of metaplasia. This view is also supported by radicular cysts may not be odontogenic in origin, but that
the fact that mucous cells are found in mandibular lesions. their epithelium may derive from nasal or antral respira-
Some have suggested that an origin from antral mucosa tory mucosa.
(see below) may explain mucous cells in radicular cysts. From these studies, it seems certain that some maxillary
This may happen on occasion, but the mucous cells in cysts may derive at least part of their epithelial lining from
mandibular lesions are almost certainly explained by true the antral mucosa, and this may explain the occurrence of
metaplasia, and provide good evidence for the metaplastic mucous and ciliated cells or respiratory-­type epithelium
potential of odontogenic epithelium. Browne (1972) also found in maxillary cysts. Such an occurrence could also be
found mucous cells in dentigerous cysts and lateral perio- deduced from the radiological appearance of large maxil-
dontal cysts, with a similar frequency between mandibular lary cysts, which often clearly protrude into the maxillary
and maxillary lesions. sinus. However, the presence of secretory and ciliated epi-
Cilia are found in radicular cysts with reported frequen- thelium in mandibular radicular cysts also confirms that
cies of 0.7% (Browne 1972), 11.4% (Takeda et al. 2005), 4.8% mucous and ciliated cells may arise as a result of metaplasia.
(Tsesis et  al.  2016), and 8.2% (Ricucci et  al.  2014). In his
careful ultrastructural studies, Nair examined 39 cysts
Hyaline Bodies
and  found 3 (7.6%) that were lined by ciliated columnar
epithelium (Nair et al. 2002). All were found in the maxilla In approximately 10% of radicular cysts, hyaline bodies are
and he suggested that the cyst linings were derived in part found in the epithelial linings (Figure 3.15). Although ini-
from cell rests of Malassez, but also from antral mucosa. tially noted by Dewey in 1918, they were first described in
However, although cilia do appear to be more common in detail by Rushton (1955) and are often referred to as
the maxilla, ciliated epithelium has also been found in Rushton bodies. They measure up to about 0.1 mm and are
cysts in the anterior and posterior regions of the mandible. described as having two morphological patterns. The first
In the study of Takeda et  al. (2005), ciliated cells were pattern appears like linear, straight, or curved rods, some-
found overall in 11% of radicular cysts, but in 12% and 9% times with a ‘hairpin’ shape (Figure  3.15b). These may
of maxillary and mandibular lesions, respectively. Tsesis have a pale centre and a darker eosinophilic or basophilic
et  al. (2016) found cilia in 4.8% of 711 cysts, but only in periphery. Serial sections, however, will show that these
2 (0.2%) mandibular lesions compared to 32 (8.9%) maxil- are not actually rods, but are folded sheets. The second pat-
lary lesions. Furthermore, 16 were found in the maxillary tern is of circular or polycyclic bodies (Figure 3.15), which
molar regions, 12 in the anterior region, and 4 associated often have a clear or pale centre, surrounded by concentric
with premolars. Browne (1972) also found that cilia were laminations that are variably eosinophilic or basophilic.
more frequently encountered in the maxilla, with 2 of 3 Both patterns may exist together, as can be seen in
being of maxillary origin. Figure 3.15. Hyaline bodies are brittle and frequently frac-
Gao et  al. (1988b) and Lu et  al. (2002) investigated ture during histological processing, so that only small frag-
cytokeratin (CK) expression in radicular cysts. Gao et  al. ments may be seen. Occasionally they may form masses
showed strong CK19 expression in rest cells of Malassez that protrude into the cyst lumen (Figure 3.15a), but only
and in the epithelium of periapical granulomas and radic- very rarely are they seen in the fibrous capsule. The pres-
ular cysts, supporting an odontogenic origin for the cyst ence of hyaline bodies may be suspected if, on examination
lining. As an early change, proliferating epithelium in peri- of the gross specimen, the pathologist sees small, smooth,
apical granulomas also uniformly and strongly expressed white, dome-­shaped swellings of the epithelial surface pro-
CK14 and subsequently CK13 and CK4. Further epithelial truding into the cyst cavity.
changes to form a cyst lining were associated with a more Although they are typically seen in radicular cysts, they
clearly differentiated phenotype of non-­keratinised strati- may also be encountered in dentigerous cysts or odonto-
fied squamous epithelium expressing CK8 and CK18. Lu genic keratocysts, but are always associated with areas of
et  al. (2002) confirmed some of these findings and also inflammation. Lam and Chan (2000) reported hyaline
showed that most cysts expressed CK8 and CK18, as well as ­bodies in 7% of a series of 69 odontogenic keratocysts and
CK13. Of relevance to the above discussion, they compared Lin et al. (2013) found them in 11% of dentigerous cysts.
keratin protein and mRNA expression in radicular cysts to Ide et  al. (1996) reported an unusual glandular odonto-
normal nasal and oral epithelium. They showed that a genic cyst with hyaline bodies, and Takeda et  al. (1985)
­Histopatholog  41

(a) (b)

Figure 3.15  Hyaline bodies in the epithelial lining of a radicular cyst. (a) The bodies accumulate in the epithelial lining and form
nodules that protrude into the lumen. (b) The bodies are eosinophilic and form circular, folded, and curved ‘rod’ shapes.

described their presence in a plexiform ameloblastoma. microscopic studies (Rühl et al. 1989; Philippou et al. 1990)
They are, however, specific to odontogenic epithelium and also found that the bodies were associated with cell debris,
have not been reported in non-­odontogenic cysts. suggesting that foreign material may irritate the epithe-
Occasionally similar structures may be found associated lium to form a cuticle-­like substance. These findings are
with epithelial rests in dental follicles. consistent with the observation that hyaline bodies are
Hyaline bodies are intriguing and enigmatic, but they always associated with areas of chronic inflammation. In a
have little diagnostic or prognostic significance. More than rarely cited paper dating back to 1943, Bauer described the
100 years after they were first noted, the origin and patho- histology of  the enamel cuticle associated with an
genesis of these bodies is still debated and no consensus unerupted third molar overlying the carious roots of the
has been reached. There have been two competing theories second molar (Bauer 1943). There was a periapical granu-
of their origin: first, that they derive from epithelium; and loma containing proliferating epithelium that was in conti-
second, that they are of haematogenous origin. Rushton nuity with the reduced enamel epithelium of the unerupted
(1955) believed that the hyaline bodies resembled, in tooth. Within the epithelium Bauer described ‘bands, rods,
appearance and the liability to fracture, the keratinised sec- rings, and spherically shaped bodies’, which were continu-
ondary enamel cuticle of Gottlieb. Wertheimer (Wertheimer ous with the primary enamel cuticle of the unerupted tooth
et  al.  1962; Wertheimer  1966) directly compared enamel and with the ‘dental cuticle’ surrounding the cementum of
cuticle and hyaline bodies and showed similar staining the decayed roots. He referred to these structures as ‘horny
for a number of histochemical stains, also concluding that hyaline-­like bodies’ and his illustrations show structures
the bodies represented a keratin-­like epithelial product identical to what we now regard as hyaline bodies. Bauer
equivalent to dental cuticle. also noted that these bodies were associated with prolifer-
A number of other studies, which have included electron ating epithelial strands that were a ‘product’ of chronic
microscopy and X-­ray microanalysis, have also supported inflammation. These simple, but careful, observations are
an epithelial origin (Allison  1974,  1977a,  1977b; Jensen consistent with the more detailed later studies described
and Erickson  1974; Morgan and Johnson  1974; Morgan above and provide good evidence that hyaline bodies can
and Heyden 1975; Rühl et al. 1989; Philippou et al. 1990). derive from odontogenic epithelium and are similar to
Morgan and Johnson (1974) also found cell debris and par- enamel cuticle.
ticulate matter and concluded that the bodies are a secre- Others, however, have believed that hyaline bodies are
tory product of odontogenic epithelium that is deposited of  haematogenous origin (Bouyssou and Guilhem  1965;
on a hard surface in a manner analogous to the formation Sedano and Gorlin  1968; El-­Labban  1979). Although the
of dental cuticle on the unerupted portions of enamel mechanism is not clear, these studies suggest that the
­surfaces. X-­ray microanalysis and scanning electron ­bodies derive from thrombi or degenerate red blood cells
42 Radicular Cyst

within vessels that have become entrapped in the prolifer- allow this hypothesis to be properly tested and provide
ating epithelial lining of the cyst. Browne and Matthews ­further clues to the origin of hyaline bodies.
(1985) tested this hypothesis using immunohistochemistry
to stain cysts for keratin, factor VIII-­related antigen, hae-
Accumulation of Cholesterol
moglobin, and fibrinogen. The hyaline bodies were nega-
tive for all these antigens, but fibrinogen was detected in Deposition of cholesterol is a characteristic feature of long-­
the cores of some circular and polycyclic forms. Browne standing chronic inflammation at any site and is due to
and Matthews concluded that hyaline bodies were not deposition and crystallisation of lipids, probably derived
keratinous in nature, nor did they arise from erythrocytes from degenerating cell walls. It is, for example, a ­prominent
or capillary endothelium. They tentatively proposed that feature in atherosclerotic plaques, arthritis, appendicitis,
the presence of fibrinogen in the cores of some hyaline and cholecystitis.
bodies could support the notion of a haematogenous origin. Deposits of cholesterol crystals are found in many radic-
Although we agree that the circular or polycyclic forms ular cysts, but by no means in all. The reported frequency
are sometimes of a morphology that suggests a transversely varies from study to study and appears to be between about
sectioned blood vessel, there are some puzzling features 10% and 45%. In one of the largest studies, Browne (1971b)
about their distribution if they were of vascular or haemo- examined 402 radicular cysts and found evidence of
togenous origin. For one thing, they are often seen in epi- ­cholesterol deposition in 43.5%. In other studies, the fre-
thelium overlying connective tissue devoid of any blood quencies have been 9.4% (Lin et  al.  2010), 28.5%
vessels. For another, they are very rarely found in the (Shear 1963b), and 30% (Trott and Esty 1972). However, it
fibrous capsules, and we have never seen them in this situ- is likely that if entire cyst linings were examined instead of
ation. Third, if their pathogenesis is as described, it is most random sections, the frequency would be higher. Browne
surprising that they are only found in lesions of odonto- (1971b) showed that there was a statistically significant
genic origin. correlation (P < 0.01) between the presence of cholesterol
More recently a novel solution to the debate has been and haemosiderin, and suggested that the main source of
suggested. Sakamoto et  al. (2012) proposed that hyaline cholesterol was from disintegrating red blood cells in a
bodies are of both haematogenous and epithelial origin. form that readily crystallises in the tissues. This was con-
They carried out immunohistochemistry for anti-­hair kera- firmed by Arwill and Heyden (1973), who showed that the
tin (CK40; AE13), CK17, CK19, and anti-­haemoglobin crystals may form in congested capillaries in the inflamed
alpha chain on 10 cysts containing hyaline bodies. They areas as they appear to be enveloped by endothelial cells.
found that hair keratin and haemoglobin alpha chain were Trott et  al. (1973) also found a close correlation between
specifically expressed in all the hyaline bodies, but not the occurrence of cholesterol and haemosiderin-­
in adjacent epithelium. Hyaline bodies were also positive containing macrophages as well as free haemosiderin in
for orcein and Congo red and occasionally for Prussian the tissues. However, their regression analysis showed that
blue. Sakamoto et al. suggested that dysregulated epithelial only 35% of the cholesterol may be formed from this asso-
differentiation results in the formation of hair keratin, ciation, suggesting that accumulation of cholesterol may
and that keratins and haemoglobin released from dead epi- also derive from degenerating lymphocytes, plasma cells,
thelial cells and red blood cells may aggregate to form hya- and macrophages.
line bodies. They further proposed that these aggregates Cholesterol crystals are small, rectangular or rhomboid
undergo β-­sheet conversion to form Congo red–positive plate-­like crystals, which characteristically have one cor-
amyloid, and that hyaline bodies are therefore a new and ner  notched or cut off. They can be visualised in fresh
novel amyloidogenic protein. This unifying hypothesis was smears of fluids or cyst contents, where they are strongly
further developed by Sarode et  al. (2016), who proposed birefringent, producing an array of colours from red/
that inflammation and osmotic pressure drive an inflam- orange to pale blue. Once the cholesterol crystals have been
matory exudate and red blood cells into the epithelium. deposited in the tissues, they behave as foreign bodies and
Degenerated red cells and accumulations of fibrinous exu- excite a foreign body giant cell reaction. They cannot, how-
date may then calcify and initiate production of enamel ever, be seen in routine histological sections, because they
cuticle. This is an attractive hypothesis and is founded on are dissolved out of the tissues by the solvents used during
the previous observations described, in particular that hya- processing. This leaves characteristic cholesterol clefts, sur-
line body production is analogous to secretion of cuticle rounded by fibrous tissue and dense aggregations of multi-
onto a surface. It is also known that hair keratin may be a nucleate giant cells (Figure  3.16b). In radicular cysts the
component of enamel (Duverger et al. 2016). Further stud- cholesterol masses form in the fibrous wall and are associ-
ies of keratins and enamel proteins in these structures may ated with heavily inflamed granulation tissue, and are
­Histopatholog  43

(a) (b)

Figure 3.16  Cholesterol clefts in a radicular cyst. (a) An accumulation of cholesterol has formed a nodule that ruptures the cyst
lining and protrudes into the lumen. (b) Multinucleate foreign body giant cells on the surface of cholesterol clefts in the wall of a
radicular cyst.

sometimes referred to as ‘cholesterol granulomas’. These


masses are often extruded from the fibrous wall through
the epithelial lining into the cyst lumen and appear mac-
roscopically and microscopically as a ‘mural nodule’
(Figure  3.16a). Once the entire mass has passed into the
cavity, the epithelial breach may heal and cholesterol crys-
tals may lie free in the cyst lumen (Shear 1963b).
Occasionally isolated or scattered clefts may be seen
within the cyst wall. High-­power examination shows that
each cleft is surrounded by one or more large multinucle-
ated giant cell (Figure 3.16b), an indication that the crys-
tals are recognised as foreign bodies. The persistence of
cholesterol crystals in the tissues and the accumulation of
macrophages may be an important cause of persistent
chronic inflammation, and is thought to be a major factor
in the persistence of non-­healing periapical lesions after
Figure 3.17  A focal accumulation of plump, foamy histiocytes
endodontic treatment of the offending tooth (Nair  2006).
in the wall of a radicular cyst.
Slutzky-­Goldberg et  al. (2013) compared the incidence
of  cholesterol in periapical lesions in young and elderly
patients, and found that the elderly (over 60 years) had a radicular cysts and noted a correlation with cholesterol and
significantly higher incidence of cholesterol than young also with accumulation of foamy histiocytes. Haemosiderin
(13–21 years) patients. They suggested that this may be is often found within histiocytes and Lin et al. (2010) found
associated with increasing serum cholesterol with age, and haemosiderin-­laden and foamy histiocytic in 6.0% and
that it may also be a factor in non-­healing of periapical 6.8% of radicular cysts, respectively (Figure 3.17). Buchner
lesions in older age groups. It is equally likely, however, and David (1978) demonstrated the presence of pigmented
that the increased frequency in the elderly is associated cells in 42% of radicular cysts. However, in this study the
with the length of time the lesion, and associated chronic pigment was not haemosiderin, but was identified as lipo-
inflammation, has been present. fuscin. It was found in association with haemosiderin
A number of other features are typically seen in radicu- deposits and cholesterol, and Buchner and David suggested
lar cysts, but these are not specific and are also associated it was also associated with the inflammatory process in the
with long-­standing chronic inflammation. As noted above cyst wall. Lipofuscin is pale yellow-­brown in colour, is posi-
(Browne  1971b), deposition of haemosiderin pigment is tive with PAS stain, negative for Perls Prussian blue, and
common. Browne (1971b) found haemosiderin in 61.4% of shows a yellow autofluorescence under ultraviolet (UV)
44 Radicular Cyst

light. It can thus be differentiated from haemosiderin,


which is dark brown in colour, PAS negative, and positive
for Perls Prussian blue.
As mentioned above, foamy histiocytes are also com-
monly seen in the walls of radicular cysts (Figure  3.17).
Most often they are scattered or form small focal accumula-
tions in areas of inflammation or are associated with cho-
lesterol clefts. Occasionally, however, sheets of large, pale
foamy histiocytes may be encountered, which appear dis-
concertingly like a xanthogranulomatous reaction.

Residual Cyst
The histopathological features of the residual cyst are simi-
lar to those described above for conventional radicular
cysts. However, because the cause of the cyst has been
removed, residual cysts may progressively become less
inflamed so that eventually the cyst wall is composed of
uninflamed collagenous fibrous tissue (Figures  3.11
and  3.13). The epithelial lining may be thin and regular
and indistinguishable from a developmental cyst, such as a
dentigerous cyst or lateral periodontal cyst. In these cases,
it is important to establish the relationship of the lesion to
the teeth and recall that a residual cyst must arise at a site
of an extracted tooth.

Figure 3.18  A low-­power view of a pocket cyst. The cyst lining


Pocket Cyst (Bay Cyst) is attached to the apex of the tooth (inset). Although in this
plane of section the apical foramen is not visible, it can be seen
With regard to the relationship between the tooth and the that the tooth apex protrudes into the cyst lumen.
cyst lining, there is some evidence that there are two distinct
histological types of radicular cyst. Brief mention has already
been made of work by Simon (1980), who found that in shown that pocket cysts comprise about 50% of all periapi-
some lesions a cavity may be present that is open to the root cal cysts. The actual frequencies reported have been 50.0%
canal and is lined by epithelium that is attached to the root (Simon  1980), 38.5% (Nair et  al.  1996), 56.3% (Ricucci
apex. Simon examined 35 extracted teeth with apical lesions et al. 2006b), and 52.2% (Ricucci et al. 2020).
still attached. Each specimen was decalcified and serial sec- In the largest study, Nair et al. (1996) undertook meticu-
tioned through the whole block. He found that the majority lous serial sectioning of 256 periapical lesions and found
of lesions (27 cases: 75.2%) were periapical granulomas and that only 39 (15%) were radicular cysts. They also confirmed
that 8 (22.9%) of these contained strands of proliferating epi- Simon’s observations and showed that of the 39 cysts,
thelium without evidence of cyst formation (e.g. see 24 (61.5%) were true cysts and 15 (38.5%) were pocket cysts.
Figure  3.7). Only 6 cases (17.2%) were diagnosed as cysts. In further detailed examination of these lesions, Nair
Three cysts showed an epithelial lining that was attached to (1998,  2003) showed that in pocket cysts the epithelium
the root apex, so that the root protruded into the cyst lumen, was attached at the root apex, so that the lumen formed an
and the lumen was in direct continuity with the root canal, extension of the root canal, and the intact epithelium acted
forming an open ‘pocket’ (Figure 3.18). Simon (1980) called as a barrier to seal off the contents of the infected root
these lesions bay cysts. The remaining cysts showed cavities canal from the adjacent tissues (Figure  3.18). The cyst
entirely lined by epithelium with an inflamed fibrous wall, grows through accumulation of debris and necrotic tissue
with no epithelial connection to the root apex. These were in the thus-­formed ‘bay’ or ‘pocket’. Simon (1980) and Nair
regarded as true cysts. Simon’s data also suggest that cysts (1998, 2003) attached considerable clinical significance to
only comprise about 17% of periapical lesions and that bay the distinction of the two cyst types and suggested that
cysts and true cysts have an equal frequency. This has been pocket cysts were likely to heal after tooth extraction or
confirmed in a number of more recent studies that have endodontic treatment, whereas a true cyst may be
­Malignant Change in Radicular Cyst  45

must be excluded. Intraosseous carcinomas are derived


Box 3.4  Histopathology: Key Features
from odontogenic epithelium and may arise de novo, with
●● The cyst is lined by proliferating non-­keratinised no identifiable precursor lesion, or may be preceded by an
stratified squamous epithelium odontogenic cyst or a benign odontogenic tumour. In a
●● In long-­standing and residual cysts, the epithelium review of the world literature, Thomas et  al. (2001) were
may become thin and regular only able to identify 35 cases of de novo lesions, suggesting
●● The cyst wall is composed of inflamed fibrous and that the majority of intraosseous squamous carcinomas
granulation tissue arise in a pre-­existing lesion and most probably arise in
●● Cholesterol clefts and deposits of haemosiderin odontogenic cysts. Eversole et al. (1975) reviewed series of
are often seen cases of central squamous cell carcinoma and found that
●● Hyaline bodies are characteristic, but only seen in 75% were associated with a cyst lining. In a detailed review
about 10% of lesions of the literature, Gardner (1969) examined the evidence
●● Other features include mucous cells, cilia, focal presented with each of 63 cases reported during the period
keratinisation, and accumulation of foamy histiocytes 1889–1967 and concluded that 25 (39.7%) fulfilled the crite-
ria for an origin from odontogenic cyst.
Bodner et al. (2011) reviewed 116 cases of primary intra-
self-­sustaining and can persist after treatment. From his osseous squamous cell carcinomas that had arisen in odon-
data, Nair (1998) suggested that apical true cysts only togenic cysts. Of these, 70 cases (60.3%) were associated
account for about 10% of all periapical lesions, and this with radicular or residual cysts, 19 (16.4%) with dentiger-
may explain the frequency of about 10% of persistent peri- ous cysts, and 16 (13.8%) with an odontogenic keratocyst.
apical lesions after conventional root canal treatment, and Only 1 lesion was found to be associated with a lateral peri-
the low prevalence of residual cysts. Ricucci and odontal cyst and 10 could not be classified. The majority of
Bergonholtz (2004) consider that the pocket cyst may be cases (92, 79.3%) were found in the mandible and there was
problematic for the endodontist, because the cyst fluid may a male : female ratio of 2 : 1.
be under pressure and may continuously wet the root Before the diagnosis of carcinoma arising from a cyst lin-
canal, making instrumentation and filling difficult. ing can be established, a number of alternative possibilities
More recently, however, it has been suggested that true must be excluded (Bodner et al. 2011; Woolgar et al. 2013;
cysts and pocket cysts do not differ in any clinically signifi- Koutlas and Sloan  2021). It is possible that a benign cyst
cant way, and that the management can be the same. and the carcinoma may have developed independently
Ricucci et al. (2020) undertook a detailed clinicopathologi- adjacent to one another and ultimately fused in some parts.
cal analysis of 11 true cysts and 12 pocket cysts and found Careful questioning of the patient and clinical examination
no significant differences in any of the clinical or histologi- are necessary to exclude the possibility that the neoplasm
cal features studied, including signs and symptoms, sex, arose primarily from the oral mucosa, or that it is a meta-
location, size, or degree of acute inflammation. They also static deposit in the jaw. A further possibility to be consid-
examined the presence of bacteria in the root canal or the ered is that the lesion was initially an epithelial neoplasm
cyst lumen and found no differences. The authors con- that underwent secondary cystic change. To be certain of
cluded that there are no clinically important differences the diagnosis, histological evidence of frank squamous cell
between the two cyst types and that there is no real need to carcinoma directly in transition from a typical odontogenic
draw a diagnostic distinction between true cysts and pocket cyst lining is regarded as the gold standard diagnostic crite-
(bay) cysts. Furthermore, they also found that both pocket rion, but in some cases the malignancy may have over-
cysts and true cysts were infected, and suggested that there grown and effaced the precursor lesion. It is also important
is little evidence for the idea that true cysts may be self-­ not to overinterpret an inflamed or proliferating cyst lining
sustainable independent of the infected root canal. as malignant. On occasions, the arcading epithelium of a
radicular cyst may give a pseudoepitheliomatous appear-
ance and islands of odontogenic epithelium in the wall are
­Malignant Change in Radicular Cysts not uncommon. Attention to the cytology of the islands is
helpful. Often the epithelium is bland and resembles squa-
Primary intraosseous squamous cell carcinoma is defined mous odontogenic tumour rather than the atypical epithe-
as a central lesion in the jaw bones, which cannot be lium of malignancy (Parmar et  al.  2011; Chrcanovic and
­categorised as any other type of carcinoma (Koutlas and Gomez 2018a).
Sloan 2022). To establish the diagnosis, an oral lesion that Despite the undoubted examples that occur from time to
has invaded the jaws, or metastatic lesions to the jaw bones, time, the frequency of neoplastic change is exceptionally
46 Radicular Cyst

rare in relation to the large numbers of cysts that are seen. root canal. Occasionally the offending tooth may be removed,
There is no evidence, however, that cyst epithelium is at but if the cyst is not curetted, then a residual cyst may arise.
particular risk and there is therefore no justification for Many periapical radiolucencies are treated non-­surgically
regarding cysts as pre-­cancerous lesions. The pathogenesis by endodontic procedures that remove the necrotic pulp
of malignant change is not known, but some have specu- and disinfect and seal the root canal. In this way, about
lated that it is induced by the long-­standing chronic inflam- 95% or more of apical lesions heal or do not progress. In
mation (Bodner et  al.  2011; Jain et  al.  2013). This would this case, however, the lesion is not removed for histologi-
explain the higher frequency of malignant change in radic- cal examination and, as discussed above, the majority will
ular cysts, and is in keeping with the now well-­established be periapical granulomas. In this respect, the lesion heals
association between carcinogenesis and inflammation because the causative root canal infection has been
(Crusz and Balkwill 2015). removed and it is not necessary to make any distinction
between periapical granulomas and cysts, nor between
true cysts and pocket cysts (Ricucci et al. 2020).
­Treatment Useful analyses or reviews on the non-­surgical treatment
of periapical lesions have been presented by Natkin et al.
Radicular cysts may be removed by conservative surgery (1984), Nair (2003, 2004, 2006), Santos-­Junior et al. (2019),
involving simple curettage of the lesion. Often this is carried and Ricucci et  al. (2020), and in systematic reviews by
out by an apicectomy procedure involving removal of the Sathorn et al. (2005), Torabinejad et al. (2005), and Glynis
cyst and the root apex and sealing of the apical portion of the et al. (2021).
47

Inflammatory Collateral Cysts

CHAPTER MENU

Classification and Terminology of Inflammatory Collateral Cysts, 47


Clinical Features, 49
●● Frequency, 49
●● Age,  52
●● Sex, 52
●● Site, 52
●● Clinical Presentation,  53
Radiological Features, 54
●● Radiological Differential Diagnosis,  56
Pathogenesis, 58
●● Relationship of the Paradental Cyst to the Dentigerous Cyst,  60
Histopathology, 61
Treatment, 61

Inflammatory collateral cysts are cysts of inflammatory ori- discrepancies may be the very nature of the cyst itself. The
gin that are found towards the buccal aspect of the roots of lesion presents as a ballooning of the pericoronal tissues to
partially or recently erupted teeth. They arise as a result of form a pocket (Ackermann et  al.  1987; Slater  2003) that
inflammation in the pericoronal tissues, but the pathogen- some may not regard as a true cyst, while others regard it as
esis and the epithelium of origin remain uncertain. a simple variant of a dentigerous cyst. Nevertheless, the
Although it is possible for a collateral cyst to arise on any clinical and radiological features are those of a cystic lesion
erupting tooth, the vast majority are associated with man- and the inflammatory collateral cysts are widely accepted as
dibular molars, with about 60% on third molars and most of an entity and are included in the World Health Organization
the remainder seen on the buccal aspect of first or second (WHO) classification of jaw cysts (WHO 2022a; Speight and
molars. Thus, two main types of inflammatory collateral Soluk ­Tekkeşin 2022b).
cyst are recognised: the paradental cyst, found predomi-
nantly on mandibular third molars, and the mandibular
buccal bifurcation cyst, found on first or second molars ­ lassification and Terminology
C
(Speight and Soluk Tekkeşin 2022b). In some respects the of Inflammatory Collateral Cysts
collateral cysts remain controversial, since it is clear that in
some quarters their very existence is questioned. There are In 1970 Main proposed a classification of jaw cysts based
a number of papers reporting lesions associated with on a review and analysis of 274 odontogenic cysts. He
impacted third molars that do not include a single case of found eight cysts (2.9%) that arose ‘alongside a vital tooth
paradental cyst (Curran et al. 2002; Stathopoulos et al. 2011), involved in pericoronitis’, which he called an inflammatory
while other series of pericoronal lesions on mandibular collateral cyst, in recognition of their association with
third molars have suggested that more than 50% may be chronic inflammation and a location on the lateral aspect
paradental cysts (Costa et  al.  2014). One reason for these of a tooth. This term also distinguishes it from a radicular

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
48   Inflammatory Collateral Cysts

cyst associated with a lateral accessory root canal of a non-­ molars. Stoneman and Worth (1983) did not refer to the
vital tooth, and from a developmental lateral periodontal reports of Main or Craig and although they emphasised
cyst. Seven of his eight cases arose on mandibular third their lesion as being on first and second molars, three of
molars, and one on an impacted upper canine (Main 1970). their cases involved the third molar region. It seems there-
Main’s 1970 paper is often cited as the first report of this fore that the mandibular infected buccal cyst and paraden-
cyst, but Philipsen et  al. (2004) found that Hofrath in tal cyst share similar clinical and histological features and
1930  had reported several jaw cysts on mandibular third should be regarded as variants of the same lesion. This is
molars affected by pericoronitis. Hofrath called these cysts suggested by more recent papers that have reappraised
‘marginal wisdom tooth cyst’, but all the features were con- these cysts and consider them to be the same entity (Packota
sistent with the inflammatory collateral cyst described by et al. 1990; Wolf and Hietanen 1990; Thurnwald et al. 1994;
Main (1970) and the paradental cyst as currently defined. Pompura et  al.  1997; Thompson et  al.  1997; Chrcanovic
The first detailed account of inflammatory collateral et al. 2011; Ramos et al. 2012). Pompura et al. (1997) sug-
cysts was by Craig (1976), who described a cyst of inflam- gested the term mandibular buccal bifurcation cyst for these
matory origin that occurred on the lateral aspect of the lesions, since it described the location of the cyst and
roots of partially erupted mandibular third molars where reflected the fact that not all lesions are overtly infected.
there was an associated history of pericoronitis. He recog- None of these cysts appeared in the first (1972) edition of
nised that these cysts were the same clinicopathological the WHO classification of odontogenic tumours and cysts,
entity as the inflammatory collateral cyst described by but they were included in the second edition (Kramer
Main (1970), but he was the first to suggest the term et  al.  1992). The 1992  WHO classification used the term
paradental cyst. He felt this was more appropriate for this paradental cyst, but included inflammatory collateral cyst
lesion because it emphasised the odontogenic associations and mandibular infected buccal cyst as synonyms. The clas-
of the cyst and its location adjacent to a tooth. sification did however make a distinction between cysts aris-
Craig’s series consisted of 49 cysts in 48 patients, which ing in association with third molars and a distinctive variant
represented about 5% of odontogenic cysts seen in his arising on the buccal aspect of first molars in children.
department over a 21-­year period. In all cases the involved In a review of the world literature, Philipsen et al. (2004)
tooth was partially erupted and was associated with a his- concurred with this view, but found that the literature
tory of pericoronitis. In terms of location, 26 cysts were on included at least 16 names for these cysts, including marginal
the buccal aspect of the roots, 19 were distal, and 4 were wisdom tooth cyst (Hofrath  1930), inflammatory collateral
mesial, but Craig was of the opinion that there was some cyst (Main 1970), paradental cyst (Craig 1976), mandibular
buccal involvement even in those cysts designated clini- infected buccal cyst (Stoneman and Worth 1983), and man-
cally as of mesial or distal location. Macroscopically, the dibular buccal bifurcation cyst (Pompura et al. 1997). Slater
cysts were firmly attached to the bifurcation area on the (2003) had suggested eruption pocket cyst, which nicely
buccal aspects of the roots, and extended up to the cemen- describes the association with an erupting tooth as well the
toenamel junction. Craig’s description of the paradental morphological feature of a pocket cyst. Nevertheless, this
cyst associated with third molar teeth has become the name was never used again in the literature. Philipsen et al.
defining feature of this lesion. (2004) preferred to use the term inflammatory paradental
Craig’s paper on the paradental cyst was, for a number of cyst to encompass all the collateral cysts of inflammatory ori-
years, the only detailed account of the entity. Subsequently, gin. They pointed out, however, that differences in clinical
however, Ackermann et al. (1987) described 50 cases, all of presentation and appearance justify separating the cyst into
which arose on impacted third molars and had similar fea- clinicopathological variants. In the literature up to 2004 they
tures to those described by Craig. This was soon followed identified reports of 342 patients with 377 cysts. The most
by a report of 6 cases by Fowler and Brannon (1989) and 15 common lesions, representing 61.4% of cysts, arose in adults
cases by Vedtofte and Praetorius (1989), and then by and were associated with a mandibular third molar. The sec-
numerous case reports and a number of case series or ond group, comprising 35.9% of cysts, were related to the
reviews (de Sousa et al. 2001; Colgan et al. 2002; Philipsen first and second molars and arose in younger individuals
et al. 2004; Mohammed et al. 2019). with a characteristic clinical presentation. A further 10 cysts
In 1983, Stoneman and Worth described 17 cases of a (2.7%) were described as occurring in the gobulomaxillary
lesion that was similar to the inflammatory collateral cyst region between the second incisor and canine. Eight of these
(Main 1970) and the paradental cyst (Craig 1976), but that ten cases were reported by Vedtofte and Holmstrup (1989).
arose primarily on the buccal aspect of mandibular first and There are also reports of four cysts arising in association
second molars in children. They named this entity the man- with mandibular premolars (Morimoto et al. 2004) and one
dibular infected buccal cyst to emphasise its origin in case arising on a maxillary second molar (Vedtofte and
inflamed periodontal tissues of partially or fully erupted Praetorius 1989).
­Clinical Feature  49

It is apparent from this discussion that all these terms ­Clinical Features
refer to a similar lesion and have been used synonymously
to describe an inflammatory collateral cyst that arises Most descriptions of inflammatory collateral cysts are to be
towards the buccal aspect of a partially, or recently, erupted found in single case reports or in small case series. In a
tooth. It is also clear that the vast majority of cysts (about review of the world literature, Philipsen et al. (2004) found
97%) fall into two main groups  – those associated with 23 case reports and 18 case series reporting 377 cysts in 342
third molars and those associated with first or second patients. More recently, Ramos et  al. (2012) reviewed 16
molars. In the previous edition of this book we suggested reports (57 cases) of buccal bifurcation cyst. Selected case
that these two variants should be called paradental cyst, series, and the review of Philipsen et al. (2004), reporting
using the criteria of Craig (1976), for lesions associated paradental cysts and mandibular buccal bifurcation cysts
with third molars and juvenile paradental cyst for lesions are summarised in Tables 4.2 and 4.3, respectively.
in younger individuals associated with mandibular first or
second molars. Subsequently, however, the 2017  WHO
Frequency
classification (Speight and Soluk Tekkeşin  2017) adopted
the term inflammatory collateral cysts for all cysts found Table 4.1 summarises the reported frequencies of inflam-
towards the buccal aspect of the roots of partially or matory collateral cysts in selected series from a wide geo-
recently erupted teeth, and described the two main types as graphical distribution. Data are taken from the same
paradental cyst and mandibular buccal bifurcation cyst. references shown in Table 1.3 (Chapter 1) and from the
This terminology has been retained in the latest edition of frequencies encountered in the larger series shown in
the WHO classification (WHO  2022a; Speight and Soluk Table 4.2. Almost without exception, the authors of the
­Tekkeşin 2022b). papers cited in Table  1.3 used ‘paradental cyst’ as a
Although the paradental cyst is usually defined as aris- generic term for all inflammatory collateral cysts, so
ing on mandibular third molars, identical lesions have these frequencies include both paradental cysts and
occasionally been described on second permanent molars mandibular buccal bifurcation cysts.
when the third molar is absent and the second molar is The frequency ranges from 0 to 13.6% of odontogenic
the last standing tooth (Vedtofte and Praetorius  1989; cysts (Table  4.1). Those authors who presented series of
Maruyama et  al.  2015). Thus the definition of the para- paradental cysts found that they comprised between 3.0 and
dental cyst might more appropriately refer to the ‘last 4.7% of odontogenic cysts encountered in their departments
standing mandibular molar tooth’. The vast majority, (Craig 1976; Ackermann et al. 1987; de Sousa et al. 2001).
however, are located on third molars. The key features of In Shear’s large series of 3496 jaw cysts from South Africa
the two main variants are summarised in Box  4.1 and (see Table  1.1), there were 109  inflammatory collateral
later Tables 4.2 and 4.3. cysts representing 3.1% of all jaw cysts and 3.6% of

Box 4.1  Key Features of the Two Main Clinical Variants of Inflammatory Collateral Cysts

Paradental cyst Mandibular buccal bifurcation cyst


Site Mandibular third molar Mandibular first or second molar
a
% of ICC 61% 36%
b
% of OC 4% 0.2%
Location Distal or disto-­buccal Buccal
Proportion in males 70% 55%
Proportion bilateral 4% 25%
Clinical features History of pericoronitis on a partially Swelling, painful. May be signs of infection
erupted tooth. Cyst is continuous with and suppuration. Tooth tipped buccally,
the pericoronal pocket deep pockets in continuity with cyst lumen
Radiology Well-­demarcated disto-­buccal radiolu- Well-­demarcated radiolucency over buccal
cency, superimposed over roots. Distal aspect of roots, buccal expansion with cor-
follicular space preserved ticated outline
a
 Frequency as a proportion of inflammatory collateral cysts.
b
 Frequency as a proportion of all odontogenic cysts.
50   Inflammatory Collateral Cysts

Table 4.1  Selected papers, with a wide geographical odontogenic cysts. These data suggest that paradental cysts
distribution, showing the frequency of inflammatory collateral are about 10–20 times more common than mandibular buc-
cysts as a proportion of total odontogenic cysts. Based on
Philipsen et al. (2004). cal bifurcation cysts.
Overall, the data from these studies (Table 4.1) suggest
Country n ICC (%)
that inflammatory collateral cysts represent about 4% of
odontogenic cysts. It is probable, however, that they are
Data from references in Table 1.3 under-­reported, because many cases received within
Meningaud et al. (2006) France 695 0 pathology departments may have insufficient clinical or
Tortorici et al. (2008) Italy (Sicily) 1273 0 radiological information to establish the diagnosis and
many may have been diagnosed as inflamed dentigerous
Ali (2011) Kuwait 196 0
cysts, pericoronitis, or inflamed follicles. In departments
Ramachandra et al. (2011) India 252 0
where the diagnosis is made on a regular basis, the para-
Manor et al. (2012) Israel 285 0 dental cyst appears to be a common lesion. In the series of
Bhat et al. (2019) India 125 0 Colgan et  al. (2002), paradental cysts comprised 15 of 60
Soluk Tekkeşin et al. (2012b) Turkey 5003 0.2 (25%) cystic lesions associated with lower third molars.
Daley et al. (1994) Canada 6847 0.5 This was the second most common diagnosis after denti-
Grossmann et al. (2007) Brazil 2812 0.7 gerous cyst (30%).
Tamiolakis et al. (2019) Greece 5165 1.1 In an analysis of pericoronal tissues from extracted third
molars, Costa et  al. (2014) found that 73.5% (83 of 113)
Mosqueda-­Taylor et al. (2002) Mexico 856 1.4
showed pathological changes and of these, 55 (66.2%) were
Aquilanti et al. (2021) Italy 2150 1.5
diagnosed as paradental cysts and 21 (25.3%) as dentiger-
Sharifian and Khalili (2011) Iran 1227 1.8
ous cysts. It should be noted, however, that the majority of
Ochsenius et al. (2007) Chile 2944 3.8 lesions were associated with erupted or partially erupted
Jones et al. (2006) UK 7121 5.6 teeth (85.5%), and only seven teeth were unerupted, sug-
Kammer et al. (2020) Brazil 406 13.6 gesting that a number of dentigerous cysts were diagnosed
Data from references in Table 4.2 in association with partially erupted impacted third molars.
Ackermann et al. (1989) South Africa 1852 3.0 Mohammed et  al. (2019) undertook a similar study and
Craig (1976) UK 1051 4.7
examined 407 tissue specimens associated with impacted
teeth from 390 patients. The most common diagnosis was
De Sousa et al. (2001) Brazil 1256 4.3
dentigerous cyst (56.5% of lesions; n  =  230), followed by
ICC, inflammatory collateral cysts; n, total number of odontogenic odontogenic keratocyst (6.1%) and then paradental cyst
cysts in each study.
(5.7%; n = 23).
It is interesting that these figures contrast starkly with
odontogenic cysts. In Craig’s original series (Craig  1976), data from other studies that have shown that paradental
paradental cysts comprised 4.7% of 1051 odontogenic cysts, cysts are exceedingly rare or are not diagnosed at all. Six of
and in a recent study from the same department (Jones the studies shown in Table 4.1 did not record any inflam-
et al. 2006), a diagnosis of paradental cyst was made on 402 matory collateral cysts. Costa et al. (2014) also reviewed 11
occasions over a 30-­year period (1975–2004), representing studies that reported histological findings associated with
5.6% of 7121 odontogenic cysts. third molars in 8464 patients. The most common finding
Few studies have determined separately the frequencies of was of normal dental follicle (76%), but the most common
paradental cysts and mandibular buccal bifurcation cysts. lesions were dentigerous cysts, found in 410 cases (11%).
Tamiolakis et  al. (2019) reviewed 5165 odontogenic cysts Only one paper (Al-­Khateeb and Bataineb 2006) reported
and found only 57 (1.1%) inflammatory collateral cysts. Of finding any paradental cysts and there were only 2, sug-
these, 53  were paradental cysts on third molars and only gesting an overall prevalence of 0.05%. In a systematic
4 were mandibular buccal bifurcation cysts in children, sug- review of the prevalence of odontogenic cysts and tumours
gesting frequencies of 1.0% and 0.1%, respectively. Jones associated with impacted third molars, Mello et al. (2019)
et  al. (2006) used the term paradental cyst to encompass reviewed 16 studies reporting histological diagnoses asso-
both types of inflammatory collateral cyst, but found only 15 ciated with more than 50 000 teeth. There were 1371 cysts,
cases in their paediatric population (patients 16 years), rep- of which 783 (57.1%) were dentigerous cysts, 400 (29.2%)
resenting 2.7% of odontogenic cysts in children (n  =  553) radicular cysts, and 150 (10.9%) odontogenic keratocysts.
and only 0.2% of all odontogenic cysts (n = 7121). Paradental These authors also found the same single study (Al-­
cysts in adults comprised 376 cases, representing 5.2% of Khateeb and Bataineb  2006) that had reported only 2
­Clinical Feature  51

Table 4.2  Paradental cysts. Age, sex, and site distribution from selected reports, and from the review of 222 cases by Philipsen
et al. (2004).

References n Mean agea Age range Male (%) % Bilateral

Craig (1976) 48 3rd decade NR 83.0 2.1


Ackermann et al. (1987) 50 3rd decade 17–62 70.0 6.0
Vedtofte and Praetorius (1989) 15 24.4 18–34 60.0 0.0
b
de Sousa et al. (2001) 54 3rd decade 13–47 39.0 NR
Colgan et al. (2002) 15 27.4 18–43 46.6 6.6
Jones et al. (2006) 376 29.6 17–74 57.3 NR
Philipsen et al. (2004) 11 29.9 18–46 63.6 18.0
Tamiolakis et al. (2019) 53 29.3 18–56 52.8 NR
Mohammed et al. (2019) 23 30.2 11–51 69.5 NR
Philipsen et al. (2004) 222c 27.6 18–47 70.6 4.1

n, number of patients; NR, not reported.


a
 In some reports, the mean age is not given, so the peak decade is included.
b
 Includes three cases on first or second molars.
c
 Total cases reviewed, n for each parameter varies.

Table 4.3  Mandibular buccal bifurcation cysts. Age, sex, and site distribution from selected reports, and from the review of 110 cases
by Philipsen et al. (2004) (see text for discussion).

References First molars Second molars

n % Male % Bilateral n Mean age Age range n Mean age Age range

Vedtofte and Praetorius (1989) 12 33.3 16.6 5 8.0 7–9 7 13.3 11–15
Wolf and Hietanen (1990) 6 16.6 NR 3 7.3 6–9 3 13.0 12–14
Thurnwald et al. (1994) 10 70.0 40.0 10 7.7 5–9 – – –
Pompura et al. (1997) 32 43.7 37.5 32 7.5 5–11 – – –
Philipsen et al. (2004) 110a 55.2 23.9 36 8.7 5–47 13 17.4 10–40

n = number of patients; NR, not reported.


a
 Total cases reviewed, n for each parameter varies.

paradental cysts. These data are similar to other studies textbooks on oral and maxillofacial pathology (Neville
that have examined tissues associated with impacted third et al. 2016) recognises the buccal bifurcation cyst in chil-
molars and have not reported a single paradental cyst (e.g. dren, but is uncertain about the paradental cyst. Its authors
Curran et al. 2002 [USA]; Stathopoulos et al. 2011 [Greece]; agree that the features may be due to chronic pericoronitis,
Patil et al. 2014 [India]). but suggest that most are probably diagnosed as examples
These data cannot be taken to represent the true preva- of inflamed dentigerous cysts. Two other excellent and
lence of paradental cysts because many studies were based widely used textbooks take a similar stance and suggest
in surgical units, the criteria for diagnosis are not given, that the paradental cyst is an inflamed dentigerous cyst
and although most report ‘impacted’ teeth, it is rarely that has become buccally or distally displaced, presumably
stated whether the tooth is fully unerupted or partially as a result of eruption of the tooth (Woo  2016; Regezi
erupted. These studies do, however, suggest that many cli- et al. 2017).
nicians may not recognise or diagnose paradental cysts. In a study from the United States of 2646 pericoronal
There is some evidence that many clinicians and pathol- lesions associated with impacted teeth, Curran et al. (2002)
ogists in the United States may not recognise the paraden- did not identify a single case of paradental cyst. They
tal cyst as an entity, but rather regard it as a variant of reported that 67% (1776 lesions) were diagnosed as normal
dentigerous cyst. One of the best and most widely used follicular tissue and of the remaining pathologically
52   Inflammatory Collateral Cysts

significant lesions (n  =  872), 86.6% (n  =  752) were diag- Paradental cysts on third molars present in an older age
nosed as dentigerous cysts. This suggests that in this cen- group (Table 4.2). The mean age of presentation is about
tre, paradental cysts were not recognised as an entity and 28 years and all studies show a peak in the third decade.
were probably diagnosed as dentigerous cysts. The paper Two-­thirds (66.7%) of the cases in Craig’s (1976) series
was challenged on this issue by Slater (2003), who sug- arose in the third decade. In the study by Ackermann et al.
gested in correspondence that some of the dentigerous (1987), 48 of the 50 cysts occurred between the ages of
cysts diagnosed by Curran et al. (2002) may in fact be para- 10 years and 39 years, with 34 cases (68%) in the third dec-
dental cysts. It is not stated, however, how many of their ade. There was only one case in the fifth decade and one
impacted teeth were unerupted or partially erupted. patient aged 62 years. Five of the six cases in the study of
These discrepancies in frequency or prevalence are almost Fowler and Brannon (1989) affected patients in the third
certainly due to uncertainty about the criteria for diagnosis decade. In their review of the world literature, Philipsen
and the fact that pericoronal radiolucencies associated with et al. (2004) reported similar age distributions with a peak
partially erupted third molars are often given a clinical or in the third decade (38% of cases) and a mean age of
radiological diagnosis of ‘dentigerous cyst’, ‘pericoronitis’, or 27.6 years. The age range of lesions associated with third
‘hyperplastic follicle’. The pathologist may receive only frag- molars is wide, overall ranging from 11 to 74 years
ments of inflamed tissue that may be consistent with any of (Table  4.2), again suggesting that occasional lesions may
these diagnoses, and paradental cyst may not be considered. remain undetected and symptomless for long periods.
The differential diagnosis of the paradental cyst and criteria Inflammatory collateral cysts arising at other sites also
for diagnosis will be discussed later in this chapter. reflect the age of eruption of the associated teeth: the four
premolar cases reported by Morimoto et  al. (2004) pre-
sented at ages 9 or 10 years, and 7 of the 8 cases reported
Age
in the globulomaxillary region were found between the
The mean age of presentation of inflammatory collateral ages of 10 and 19 years (Vedtofte and Holmstrup 1989).
cysts correlates well with the chronological stage of erup-
tion, with lesions most often presenting a few years after
Sex
the eruption of the associated tooth. The mandibular buc-
cal bifurcation cyst presents in children, but the mean age Inflammatory collateral cysts show a slight preponderance
of presentation depends on the tooth affected (Table 4.3). for males, but this is most apparent for paradental cysts
Lesions on first molars are found at a mean age of about (Table  4.2), where about 70% of cases overall affected
8 years, with most studies showing a narrow range between males. Mandibular buccal bifurcation cysts affecting chil-
5 and 11 years. Cysts on second molars are found at a mean dren show a more even distribution between males and
age of about 13 years. In their review of more than 40 females (Table 4.3), with only one series reporting a male
papers, Philipsen et al. (2004) found limited demographic preponderance (70%: Thurnwald et  al.  1994). Philipsen
data, but showed a wider age range than suggested in the et al. (2004) found 87 cases where sex had been recorded,
literature. The age range for lesions on first molars and sec- with a slight male preponderance of 55%. In their analysis
ond molars was reported as 5–47 years and 10–40 years, of odontogenic cysts, Jones et al. (2006) found 14 cases in
respectively. However, careful reading of their paper shows children where sex was recorded and showed a male :
that there was only one first molar cyst in a patient over female ratio of 1.33 (57% males).
11 years (a male age 47) and this was from their own files.
They also found only three cases over the age of 16 on sec-
Site
ond molars, and two of these were from their own files.
Philipsen et al. (2004) also suggested that the ages differ Inflammatory collateral cysts are almost exclusively found
for males and females, with males affected slightly later. in the mandible and over 60% are paradental cysts involv-
They showed that lesions on the first molar affect individu- ing a mandibular third molar. Only a handful of cases
als with a mean age at presentation of 9.0 years for males have been reported in the maxilla. In the original descrip-
(range 5–47; n = 24) and 8.1 years for females (range 6–11; tion of the lesion, Main (1970) described 1 case associated
n = 12). Lesions on the second molar presented with mean with an upper canine and Vedtofte and Holmstrup (1989)
ages of 19.8 years for males (range 10–40; n  =  8) and subsequently reported a series of 8 cases arising between
13.6 years for females (range 12–16; n = 5). These differences the upper canine and second incisor (globulomaxillary
were not shown to be significant and the higher mean age in region). In their large review, Philipsen et al. (2004) found
males is almost certainly due to outlying single cases at an that 97.3% of cases arose in the mandible, with only 10
older age. Nevertheless, the data show that occasional cysts (2.7%: including the 8 cases of Vedtofte and Holmstrup) in
may persist undetected into later adulthood. the maxilla. Jones et  al. (2006) found 402  inflammatory
­Clinical Feature  53

collateral cysts in their files, but only 1 case was recorded on a second molar when it is the last standing tooth in the
in the maxilla, associated with a third molar. Ochsenius mandible. This is rare, but when it occurs the clinical and
et al. (2007) found 113 paradental cysts in their series of radiological features are identical to a paradental cyst aris-
odontogenic cysts, 6 (5.3%) of which were associated with ing on a third molar (Vedtofte and Praetorius 1989; de Sousa
maxillary third molars. The 4 premolar cases reported by et al. 2001; Maruyama et al. 2015).
Morimoto et al. (2004) were all in the mandible. In contrast to the paradental cyst, the mandibular buccal
Paradental cysts are located almost exclusively on man- bifurcation cyst usually presents with symptoms  – most
dibular third molars, but 2 cases have been recorded on sec- often a complaint of swelling, with pain or tenderness.
ond molars when the third molar is absent and the second Stoneman and Worth (1983) first reported this lesion as
molar is the last standing tooth (see later Figure 4.2; Vedtofte ‘mandibular infected buccal cyst’, and although some cases
and Praetorius 1989; de Sousa et al. 2001 [2 cases]; Maruyama produced few or no clinical symptoms, they felt that the
et al. 2015). These unusual examples arose at an average age lesion was characterised by signs of infection, including
of 14.5 years (range 13–17 years), but were otherwise identi- discomfort, pain, tenderness, and painful occlusion. Rarely,
cal to paradental cysts encountered on third molars. there may be suppuration with formation of a facial abscess
Mandibular buccal bifurcation cysts are located on the that may point (Figure 4.1). Swelling and pain are the clini-
first or second molar teeth, but the relative frequency of cal features most likely to induce the patient to seek advice.
occurrence on each tooth is rarely reported. In the review by Pompura et al. (1997) reported 32 patients with cysts on
Philipsen et al. (2004), cysts on the first and second molars mandibular first molars and found that all cases presented
were usually reported together, but for 49 cases the site dis- with pain or tenderness in the affected area. Only
tribution was known. Of these cases, 36 (73.5%) were 14 patients (43.7%) were aware of swelling of the cheek,
located on first molars and 13 (26.5%) on second molars. but in the remaining 18 cases intraoral swelling of the alve-
This suggests that mandibular buccal bifurcation cysts are olus was evident on clinical examination. A foul-­tasting
about twice as common on first molars as on second molars. discharge consistent with infection was reported by 20
Inflammatory collateral cysts may be bilateral, but this is patients (62.5%). Vedtofte and Praetorius (1989) reported
more common for the mandibular buccal bifurcation cyst 12 cases associated with first or second molars and found
than for paradental cysts. Case series have shown that that the most common symptoms were pain and swelling
between about 16 and 40% of mandibular buccal bifurca- associated with discharge of pus. In all cases the cyst lumen
tion cysts present as bilateral lesions (Table  4.3), while communicated with the periodontal pocket on the buccal
bilateral lesions are only seen in about 4% of patients with aspect of the tooth. Philipsen et  al. (2004) reported that
paradental cysts (Table 4.2) buccal swelling was rarely associated with cysts on the sec-
ond molar, but is a characteristic feature of lesions on the
first molar.
Clinical Presentation The mandibular buccal bifurcation cyst is always situated
on the buccal aspect of the root of the affected tooth and the
Over 60% of all inflammatory collateral cysts are paradental
tooth is usually tilted buccally so that the apices are
cysts involving mandibular third molar teeth with a history
adjacent to the lingual cortical plate, a feature that is best
of recurrent or persistent pericoronitis, although lesions
may be symptomless at presentation. The third molar is
always partially or recently erupted and most are impacted.
In Craig’s (1976) series of 48 patients, all cases were associ-
ated with a history of pericoronitis, but bony expansion or
swelling was not a feature and most lesions were chance
findings on extraction of the impacted tooth. Ackermann
et al. (1987), Fowler and Brannon (1989), and Colgan et al.
(2002) found that all their cases were associated with a par-
tially erupted third molar with a history or presence of peri-
coronitis. Symptoms of pain or swelling were rarely
encountered, although there may be signs of persistent
infection. On examination, all lesions are found on the buc-
cal side of the tooth and most are orientated towards the
distal aspect. On probing, the cyst lumen is usually found to
Figure 4.1  Young boy with mandibular buccal bifurcation cyst
communicate with the associated periodontal pocket or
involving a recently erupted mandibular first permanent molar.
with the pericoronal space beneath the inflamed opercu- Infection has extended through the bone, resulting in a facial
lum. As mentioned previously, a paradental cyst may arise abscess. Source: Courtesy of Dr D.W. Stoneman.
54   Inflammatory Collateral Cysts

seen in occlusal radiographs (Figure  4.4) or cone-­beam The paradental cyst shows a number of quite specific but
computed tomography (CBCT). The tooth is always vital, subtle features, which were first described by Craig (1976).
which allows a lateral radicular cyst to be excluded. On conventional radiographs the cyst presents as a well-­
With regard to inflammatory collateral cysts presenting demarcated radiolucency, usually with a corticated mar-
at other sites, the clinical features are very similar to the gin. This is well illustrated in Figure  4.2, which shows
mandibular buccal bifurcation cyst. Morimoto et al. (2004) radiographs from the paper by Vedtofte and Praetorius
found that all four of their cases on lower premolars pre- (1989). Of note is that the case illustrated in Figure 4.2a is
sented with swelling and three were also painful. of bilateral paradental cysts associated with second molars,
Inflammatory collateral cysts in the globulomaxillary in a 13-­year-­old girl where the third molars are absent. The
region probably arise in association with the erupting cyst is usually displaced distally, but in all cases the radio-
canine and present as an inverted pear-­shaped radiolu- lucency is superimposed over the buccal aspect of the tooth
cency between the incisor and canine teeth, which are vital and overlies the roots and bifurcation area. Most paraden-
and show divergent roots. Of the eight cysts in the globulo- tal cysts are 10–15 mm in diameter (Philipsen et al. 2004)
maxillary region reported by Vedtofte and Holmstrup and rarely exceed 20 mm. Larger lesions may appear to be
(1989), five were asymptomatic chance findings and three periapical, but it is important to note that the periodontal
presented with signs of acute infection. ligament space is not widened and the lamina dura is intact
around the roots (Figures 4.2 and 4.3). Most cysts extend
distally, but the distal element is well defined and is dis-
­Radiological Features tinct from the distal follicular space (Figure 4.2). This fea-
ture was first noted by Craig (1976) and was confirmed by
The clinical features of inflammatory collateral cysts are Colgan et al. (2002), who identified it in 9 of their 15 cases
not specific and an accurate diagnosis can only be made and considered it to be an important and helpful diagnostic
after consideration of the radiological features, which are criterion, since it indicates that the dental follicle is not
characteristic (Box 4.2). involved in the development of the cyst.

Box 4.2  Inflammatory Collateral Cysts: Key Features and Diagnostic Criteria


Paradental cyst
●● Arises on the last standing mandibular molar – almost always a third molar
●● There is a history or presence of pericoronitis
●● May be swelling and discomfort, but often symptomless
●● The associated tooth is vital
●● Well-­demarcated and corticated radiolucency
●● Usually 10–15 mm in diameter
●● Lies on the buccal aspect of the tooth root and bifurcation, but often orientated distally
●● An important diagnostic feature is that the distal follicular space is intact and distinct from the cyst
●● The periodontal space and lamina dura are intact
Mandibular buccal bifurcation cyst
●● Arises on mandibular first or second molars
●● Often has symptoms – swelling, pain, and there may be suppuration
●● The associated tooth is vital
●● Well-­demarcated and corticated radiolucency
●● 10–20 mm in diameter
●● Lies on the buccal aspect of the tooth root and bifurcation
●● The periodontal space and lamina dura are intact
●● Buccal expansion is common
●● Subperiosteal new bone (visible especially on occlusal radiographs) may be deposited in a laminated pattern
●● The tooth is tilted buccally and the root apices may abut onto the lingual cortical plate
­Radiological Feature  55

(a)

(b)

Figure 4.2  Paradental cysts. (a) Bilateral cysts in a 13-­year-­old associated with recently erupted second molars. The third molars
were absent. (b) Two cysts in different patients associated with partially erupted (left) and recently erupted (right) third molars. In all
cases, the cysts are distal and buccal to the involved teeth. The distal part of the cyst is separate from the distinct distal follicular
space (arrows in a) and the periodontal ligament space and lamina dura are intact (arrowheads). Source: Vedtofte P 1989, p. 182–188 /
with permission of Elsevier. Courtesy of CV Mosby Co.

Although most lesions are located in a buccal or disto-­ mesioangular, these data suggest that a distally impacted
buccal location (Figure 4.2), Colgan et al. (2002) suggested tooth is more likely to give rise to a paradental cyst than a
that the precise site of the cyst may depend on the angle of mesially angled tooth.
impaction of the associated tooth. A distoangular impac- Radiological examination of the mandibular buccal
tion was shown in 10 of their 15 teeth and all had cysts bifurcation cyst should include occlusal and panoramic
located towards the distal aspect. Two of their cysts were radiographs or CBCT. The cyst presents as a well-­
located towards the mesial aspect of the teeth, but both demarcated and usually corticated radiolucency overlying
showed mesioangular impactions. Two teeth that were ver- the buccal aspects of the roots of an erupting or recently
tically impacted had cysts located purely on the buccal erupted first or second molar tooth (Figure 4.3). The infe-
aspect. The vast majority of reports of paradental cysts rior margin of the cyst is concave and rarely the cyst may
record that cysts are located towards the distal aspect of the extend to the inferior border of the mandible, but does not
teeth and that a mesial orientation is rare (Craig  1976; lead to distortion of the lower border or to an external
Ackermann et  al.  1987; Vedtofte and Praetorius  1989; deformity (Figure 4.3a). There may be involvement of the
Philipsen et al. 2004). Since most impacted third molars are bone in the furcation and inter-­radicular bone may be lost.
56   Inflammatory Collateral Cysts

(a) (b)

Figure 4.3  Mandibular buccal bifurcation cysts involving (a) an erupting first permanent molar and (b) a second permanent molar.
The cysts are well demarcated and corticated and overly the buccal and periapical aspects of the tooth roots. The periodontal
ligament and lamina dura are intact (arrows). Sources: (a) Courtesy of Dr Douglas W Stoneman. (b) Courtesy of Prof Paul Speight
(Previously published: El-Naggar AK 2017, Courtesy of IARC).

In the inter-­radicular region, therefore, the lamina dura Inflammatory collateral cysts at other sites show similar
may be lost, but the periodontal ligament space and the radiological features. The four cases associated with pre-
lamina dura are intact on the mesial and distal aspects of molars, reported by Morimoto et al. (2004), showed well-­
the roots (Figure  4.3), a feature that enables distinction demarcated radiolucencies overlying the buccal aspect of
from a radicular cyst. erupting and incompletely formed premolars. There was
Buccal expansion is often apparent and with involve- buccal expansion and in all cases the teeth were displaced
ment of the periosteum new bone may be laid down, either lingually. Cysts in the globulomaxillary region present as a
as a single linear band or in a laminated pattern (Figure 4.4). well-­demarcated radiolucency between the lateral incisor
This pattern of laminated new bone is seen as a result of and the canine tooth. The cyst has an ‘inverted pear’ shape
periostitis and is also seen in chronic osteomyelitis. Wolf and the tooth roots are displaced and diverge. All eight
and Hietanen (1990) noted this feature and suggested that cases reported by Vedtofte and Holmstrup (1989) showed
clinically and radiographically, the mandibular buccal these features and in all cases both the canine and incisor
bifurcation cyst may be similar to osteomyelitis, and that in tooth were fully erupted.
some cases the cyst may be a cause of chronic osteomyeli-
tis, (sometimes referred to as Garré’s osteomyelitis or peri-
Radiological Differential Diagnosis
ostitis ossificans).
CBCT is especially useful in the diagnosis of mandibular The histological features of inflammatory collateral cysts
buccal bifurcation cysts because it allows the buccal expan- are not specific, but taken together the clinical and radio-
sion and extent of the lesion to be fully visualised (Ramos logical features should allow an accurate diagnosis
et al. 2012; Bautista et al. 2019; Dave et al. 2020). Magnetic (Box 4.2). In virtually all cases the cysts arise on the buccal
resonance imaging (MRI) can also be useful, but although or disto-­buccal aspect of a partially erupted or recently
it has the advantage of lower radiation dosage, it has very erupted tooth with characteristic radiological features.
limited use in routine examination of jaw lesions in den- The associated tooth is vital. Nevertheless, as discussed
tistry. CBCT shows a hypodense, well-­demarcated, and previously, there is great variation in the relative frequency
often spherical cystic lesion with a corticated outline, of these cysts in different studies and it is apparent that
which is always located on the buccal aspect of the tooth. some workers do not recognise it as an entity. For the most
CBCT and occlusal radiographs show that the affected part this is probably due to lack of clear diagnostic criteria
tooth is tilted buccally and transverse views show the api- and uncertainty about the pathogenesis. The key features
ces abutting and occasionally eroding the lingual cortical of these lesions and diagnostic criteria are suggested
plate (Figure 4.4; Bautista et al. 2019). in Box 4.2.
­Radiological Feature  57

radiolucency may result from an inflamed or hyperplastic


dental follicle or pericoronitis (Figure 4.5). In such a case,
the pathologist may be presented with fragments of
inflamed tissue and a clinical diagnosis of ‘pericoronitis’,
and therefore a diagnosis of paradental cyst may not be
considered. In addition, some workers consider that a per-
icoronal radiolucency distal to a partially erupted third
molar may arise as a result of lateral displacement of a
dentigerous cyst during tooth eruption, or that a cyst that
arises in a pericoronal relationship is a mere variant of
dentigerous cyst. In this case, the final diagnosis may be of
an ‘inflamed dentigerous cyst’.
However, careful examination of radiographs will show
that the paradental cyst is distinct from the pericoronal fol-
licular tissues and the criteria for diagnosis must include
the finding of a well-­demarcated cyst that is distinct from
the distal aspect of the dental follicle (Figures 4.2 and 4.5a).
If the follicular space is merely expanded, then the lesion
may be a hyperplastic dental follicle, or bone loss associ-
ated with chronic pericoronitis. Figure  4.5b shows an
Figure 4.4  Occlusal view of a mandibular buccal bifurcation expanded pericoronal space associated with a partially
cyst on a mandibular first molar tooth. There is resorption of
erupted but vertical third molar. There is also evidence of
bone on the buccal aspect of the tooth and buccal expansion.
Subperiosteal new bone has been deposited buccally, giving a pocketing with alveolar bone loss. This appearance is con-
laminated appearance. Note also the displacement and tilting of sistent with chronic long-­standing pericoronitis, but with-
the tooth, so that its apices abut onto, and appear to erode, the out cyst formation. The relationship to the dentigerous cyst
lingual cortical plate. Source: Courtesy of Dr Douglas W
is discussed further below.
Stoneman (Previously published: Dental Radiography and
Photography (1983) 56, 1–14. Courtesy of Eastman Kodak Co.) Both the paradental cyst and mandibular buccal bifurca-
tion cyst overly the tooth roots and may resemble a periapi-
With regard to the paradental cyst, the presence of a cal radicular cyst, or a radicular cyst lying lateral to the root
pericoronal radiolucency associated with a partially associated with a lateral root canal. However, in the case of
erupted third molar is not specific, since a similar an inflammatory collateral cyst, the periodontal space and

(a) (b)

Figure 4.5  Differential diagnosis of the paradental cyst. (a) The cyst is well demarcated and corticated (arrows) and is distinct from
the distal follicular space (arrowhead). (b) Chronic pericoronitis on a vertically impacted and partially erupted third molar. The distal
follicular space is dilated (arrowhead) and there is evidence of alveolar bone loss, but there is no cyst.
58   Inflammatory Collateral Cysts

the lamina dura remain intact (Figures 4.2 and 4.4), a fea- tooth erupts, the reduced enamel epithelium fuses with the
ture that excludes an inflammatory cyst of endodon- oral epithelium and forms the dentogingival junction or
tic origin. the early epithelial attachment, which is composed of the
A number of other cysts may on occasion arise on the junctional epithelium that binds to the tooth crown and
lateral aspect of a tooth, including odontogenic keratocyst the sulcular epithelium towards the opening of the gingival
and glandular odontogenic cyst. However, these do not sulcus. Since inflammatory collateral cysts are associated
embrace the crown of partially erupted teeth, are not asso- with partially or recently erupted teeth, Craig (1976) actu-
ciated with pericoronitis, and have characteristic features ally proposed that the epithelium of origin was the early
that clearly distinguish them from inflammatory collateral epithelial attachment that is derived from the reduced
cysts (see Chapters 7 and 10). Developmental lateral peri- enamel epithelium. Thus the cyst is derived from the epi-
odontal cysts may also be considered, but the clinicopatho- thelium that lines the inflamed periodontal or pericoronal
logical features are quite different (see Chapter 8). Although tissues associated with pericoronitis. Although classified as
lateral periodontal cysts are most common in the mandi- an odontogenic cyst, it probably does not arise directly
ble, they are found in the canine/premolar region and they from odontogenic remnants, but rather from pocket or per-
lie lateral to the roots of fully erupted teeth in middle-­aged icoronal epithelium that is itself of odontogenic origin.
adults. They are not associated with inflammation. Craig (1976) and others (Main 1970; Ackermann et al. 1987;
A number of workers have drawn attention to the fact Fowler and Brannon 1989; Vedtote and Praetorius 1989; de
that the clinical and radiological features of mandibular Sousa et al. 2001; Philipsen et al. 2004) also considered the
buccal bifurcation cyst may be similar to the chronic form rest cells of Malassez as a potential source of epithelium, but
of osteomyelitis often referred to as Garré’s osteomyelitis or this has largely been dismissed on the basis that, when seen
periostitis ossificans (Stoneman and Worth 1983; Wolf and in histological sections, the rests of Malassez always appeared
Hietanen 1990). This is typified by buccal expansion with inactive, and such an origin would not account for the con-
laminated depositions of new subperiosteal bone – a fea- sistent buccal distribution of the lesions (Craig 1976; Fowler
ture that is also seen in cases of mandibular buccal bifurca- and Brannon 1989).
tion cyst (Figure 4.4). Periostitis ossificans of the mandible Ackermann et  al. (1987) agreed with Craig (1976) that
is most often seen in association with first molars in young the paradental cyst arises as a consequence of inflamma-
people and is usually secondary to periapical infection. The tory destruction of bone that is followed by proliferation of
mandibular buccal bifurcation cyst is of inflammatory ori- epithelium and cyst expansion. They also discussed an ori-
gin and it is probable that in some cases, persistent chronic gin from reduced enamel epithelium or from sulcular (or
inflammation may give rise to a low-­grade osteomyelitis crevicular) epithelium, and suggested that cyst formation
with periostitis. However, a diagnosis of collateral cyst can occurs as a result of unilateral expansion of the dental fol-
be made on the basis of a vital tooth, a well-­defined buccal licle secondary to inflammatory destruction of periodon-
radiolucency, and evidence of continuity between the buc- tium and alveolar bone. This, they proposed, was different
cal periodontal pocket and the cyst lumen. from the histogenesis of a dentigerous cyst, which is not
inflammatory and where expansion of the follicle is the
primary event with consequent bone destruction.
­Pathogenesis Vedtofte and Praetorius (1989) agreed that inflammatory
collateral cysts are of inflammatory origin, and are associ-
There is no unanimity with regard to the pathogenesis of ated with pericoronitis on third molars or with a deep and
inflammatory collateral cysts. Craig (1976), however, pro- inflamed periodontal pocket when found on first or second
posed a pathogenic process that has become widely molars. While accepting that the paradental cyst was an
accepted. He suggested that the cyst arises within a focus of entity, Fowler and Brannon (1989) suggested that it may be
inflammation associated with pericoronitis around a par- a variant of the dentigerous cyst, in that it is derived from a
tially erupted tooth. In this respect the pathogenesis is sim- cystic expansion of follicular reduced enamel epithelium
ilar to the radicular cyst, in that chronic inflammation secondary to pericoronitis.
initiates the cyst and is the stimulus for proliferation of the Fowler and Brannon (1989) also proposed that the para-
epithelial lining. Craig (1976) is widely cited as having pro- dental cyst may arise as a result of expansion of an occluded
posed that the epithelium of origin is the reduced enamel periodontal or pericoronal pocket. This is consistent with
epithelium, but in fact his proposal was more nuanced the proposal of Craig (1976) and others, if one recalls that
than this. The reduced enamel epithelium embraces the the epithelium lining a pocket or pericoronal tissues
crown of an unerupted tooth and is derived from the rem- around a partially erupted tooth are derived from the same
nants of the inner and outer enamel epithelium. As the source – the reduced enamel epithelium.
­Pathogenesi  59

More recently, Maruyama et al. (2015) found that inflam- opening of the associated periodontal pocket or from
matory collateral cysts consistently expressed cytokeratins beneath the operculum (Figure 4.6b). Some workers require
K13, K14, and K19, and perlecan and UEA-­1. Dentigerous an opening to the surface as a diagnostic criterion (de Sousa
cysts showed similar expression, but also expressed K10, et al. 2001). A number of workers (Craig 1976; Ackermann
which was negative in the collateral cysts. Other cyst types et al. 1987; de Sousa et al. 2001) have obtained sections of
(odontogenic keratocyst and radicular cyst) showed minor the cyst in continuity with the associated teeth and showed
differences in keratin expression, but the expression of the that the cyst linings were attached at the cementoenamel
markers in lateral periodontal cyst and in normal junc- junction and were continuous with the oral epithelium
tional or sulcular epithelium was similar to the collateral (Figure 4.6).
cysts. Overall, the expression of cytokeratins was variable These studies suggest that inflammatory collateral
and the results of this study cannot be regarded as conclu- cysts are equivalent to a dilated follicle or pocket lined by
sive. Nevertheless, the similarity of expression patterns hyperplastic and proliferative epithelium derived from
between the inflammatory collateral cysts and junctional/ reduced enamel (follicular) epithelium. Thus, a descrip-
sulcular epithelium supports the view that the cysts arise tive designation of ‘inflammatory pocket cyst’ may be
from the epithelium lining a periodontal or pericoronal appropriate, and Slater (2003) has suggested that the
pocket. The authors suggest that their results contradict third molar lesions should be called ‘eruption pocket
the findings of Craig (1976), who they say proposed an ori- cysts’. It is possible that swelling associated with inflam-
gin from ‘reduced enamel epithelium’, but the findings mation leads to occlusion of the opening of the pocket,
actually support Craig’s proposal that the epithelium of thus allowing accumulation of debris and cyst growth by
origin is from the early epithelial attachment (i.e. the junc- osmotic pressure in a similar process to that described for
tional or sulcular epithelium) that derives from the reduced radicular cysts.
enamel epithelium. From this discussion it can be seen that there is agree-
An origin from pocket or pericoronal epithelium is fur- ment that collateral cysts are of inflammatory origin and
ther supported by the observation that the cyst lining is that the initiating factor is inflammation within the peric-
often continuous with junctional or sulcular epithelium, or oronal tissues of an erupting or partially erupted tooth.
with the periodontal or pericoronal pocket around the asso- However, pericoronitis is common and this pathogenic
ciated tooth (Figures  4.6 and  4.7; Craig  1976; Ackermann process does not easily explain why paradental and man-
et al. 1987; de Sousa et al. 2001; Colgan et al. 2002; Philipsen dibular buccal bifurcation cysts are so rare.
et al. 2004). The cyst therefore presents as a dilated pocket, Craig (1976) was able to examine the teeth in 28 of his
whereby a probe can be placed into the lumen through the 49 cysts. In 20 of the 28 cases (71.4%) he found a

(a) (b)

Figure 4.6  Gross specimen of a paradental cyst on the buccal aspect of a partially erupted third molar, which has been received
intact. (a) The cyst is attached to the cementoenamel junction and involves the distal and buccal surfaces of the roots. (b) Careful
probing shows that the lumen is open towards the coronal aspect and is in continuity with the lining of the pericoronal pocket.
Source: Courtesy of Prof G.T. Craig.
60   Inflammatory Collateral Cysts

developmental enamel projection or spur extending from cusps and the buccal inclination may predispose to food
the cementoenamel junction towards the bifurcation of impaction on the buccal aspect.
the roots on the buccal aspect of the tooth. He suggested
that the downwards extension of the reduced enamel epi-
Relationship of the Paradental Cyst to the
thelium at the site of an enamel spur may provide a site of
Dentigerous Cyst
stagnation or focus of inflammation, and predispose to
the development of a paradental cyst at this site. Fowler As discussed previously (see ‘Frequency’), there is evidence
and Brannon (1989) found enamel projections on two of that some clinicians and pathologists do not recognise the
the three teeth they were able to examine and agreed with paradental cyst as an entity, but rather regard it as a variant
Craig that such projections may localise inflammation of dentigerous cyst. Ackermann et al. (1987) suggested that
and cyst formation on the buccal aspect. In their study, although the paradental cyst may arise from follicular
Ackermann et al. (1987) were only able to examine eight (reduced enamel) epithelium, the histogenesis is quite dif-
of the associated teeth. In all cases the cyst was attached ferent and it should not be regarded as a variant of denti-
at the cementoenamel junction, and two showed enamel gerous cyst. They believed that the dentigerous cyst should
spurs. Others, however, have been unable to show an be defined as a cyst enclosing the crown of a completely
association between these cysts and enamel spurs, but unerupted tooth.
often this is because the associated teeth were not availa- Despite this, many clinicians and pathologists diagnose
ble for examination (Vedtofte and Praetorius  1989; de collateral lesions associated with partially erupted third
Sousa et al. 2001). Studies have shown that enamel pro- molars as ‘inflamed dentigerous cysts’. Fowler and Brannon
jections may be seen on up to 70% of teeth and are more (1989) agreed with Ackermann et al. (1987) that a dentiger-
common on mandibular molars (Chan et al. 2010). Risnes ous cyst is, by definition, associated with an unerupted
(1974) found that 11.7% of third molars had enamel pro- tooth, but also believed that the paradental cyst is a variant
jections and that 99% occurred on the buccal aspect. of dentigerous cyst. In this respect, a relationship to the
These data may explain in part why lesions are found pre- dentigerous cyst may be suggested on the basis of a com-
dominantly on the buccal aspect of mandibular molars. mon origin from epithelium lining the dental follicle, with
Colgan et al. (2002) suggested that food impaction may intraosseous (dentigerous cyst) and extraosseous (paraden-
have an important part to play. In 13 of their 15 cases the tal cyst) variants associated with totally unerupted and par-
associated tooth was opposed by a maxillary molar and tially erupted teeth, respectively.
they proposed that the angulation of the affected tooth One possible pathogenic mechanism for the paradental
(usually distal) could promote food impaction into the per- cyst is that it represents a developmental dentigerous cyst
icoronal tissues around the crown. As further evidence for that has become laterally (and buccally) displaced by the
this they showed that four cases contained giant cells con- eruption of the associated tooth, with subsequent inflam-
sistent with a foreign body reaction. mation. This seems unlikely, however, since the typical
With regard to the mandibular buccal bifurcation cyst, radiology of the paradental cyst (see Figures 4.2 and 4.5a)
studies have shown that the affected tooth is almost always suggests that the dental follicle remains intact and is not
tilted buccally (Figure 4.4), giving prominence to the lin- dilated, and such a mechanism would not explain the con-
gual cusps and often associated with increased pocket sistent buccal location. An alternative proposal is that the
depth on the buccal aspect (Pompura et al. 1997; Philipsen paradental cyst is simply an inflammatory type of dentiger-
et al. 2004). Stoneman and Worth (1983) suggested that the ous cyst that arises as a result of inflammation of the peri-
mesio-­buccal cusp of the first molar is the first to penetrate coronal tissues surrounding the partially embedded crown
the oral mucosa during eruption, and that this would of an impacted or erupting tooth. This would be a perfectly
explain the buccal location of the cyst, but this explanation acceptable proposal, but for the fact that an inflamed and
is at odds with the fact that the lingual cusps may erupt expanded pericoronal follicle would be indistinguishable
first in the buccally displaced tooth. However, it is not from pericoronitis associated with inflammatory osteoly-
known whether this buccal inclination occurs as a result of sis. Yet paradental cysts show a very distinctive radiology
displacement of the tooth by the cyst, or is present at erup- with a buccally orientated radiolucency that is quite dis-
tion and may thus predispose to cyst formation. Since the tinct from an expanded dental follicle (see ‘Radiological
cysts are rare, it is most likely that the associated tooth is Differential Diagnosis’ and Figures 4.2 and 4.5). For these
buccally displaced at eruption and that this predisposes to reasons, we believe that inflammatory collateral cysts and
inflammation in a buccal pocket and subsequent cyst for- dentigerous cysts show distinctive diagnostic features and
mation. It is possible that the prominence of the lingual should be designated as separate entities (Box 4.2).
­Treatmen  61

­Histopathology

Paradental cysts associated with third molars are removed


by enucleation of the cyst along with extraction of the asso-
ciated tooth. Occasionally, the whole specimen is received
intact (Figure 4.6) and will show that the cyst is a sac-­like
mass attached at the cementoenamel junction and located
on the buccal aspect of the tooth roots. This is in contrast to
a dentigerous cyst, which is also attached to the cementoe-
namel junction, but surrounds the crown of the tooth (see
Figures  5.18 and 5.19). Careful examination with a probe
will show that the lumen of the cyst is continuous with the
periodontal or pericoronal pocket (Figure 4.6b). This appear-
ance is virtually diagnostic of an inflammatory collat-
eral cyst.
Mandibular buccal bifurcation cysts are usually removed
by enucleation or curettage and the tooth is left in situ.
In this case the pathologist will often receive fragments of
soft tissue that in places may resemble a cyst wall.
Histological examination shows non-­specific features
that are indistinguishable from radicular cyst and it will
be impossible to make a diagnosis without consideration
of the clinical findings and review of the radiology. The
cysts are lined by a hyperplastic, non-­keratinised, strati-
fied squamous epithelium, which may be spongiotic and
of varying thickness. There is an intense chronic or mixed
inflammatory cell infiltrate associated with the hyper-
plastic epithelium and in the adjacent fibrous capsule
(Figure 4.7). As in radicular cysts, haemosiderin deposits,
hyaline bodies, or accumulations of cholesterol crystals Figure 4.7  Paradental cyst adjacent to the root of an impacted
may be seen. An opening into the cyst lumen may also be mandibular third molar. The cyst is lined by non-­keratinised
stratified squamous epithelium of variable thickness and
seen and the epithelial lining may be continuous with
showing areas of proliferation and inflammation in the wall.
sulcular or gingival epithelium at the periphery. Source: Courtesy of Prof G.T. Craig.
Occasional cases may show focal accumulations of for-
eign body–type giant cells, consistent with impaction of
food particles (Fowler and Brannon  1989; Colgan
et al. 2002).
normally. Nevertheless, removal of the cyst may not be
necessary, since there is good evidence that it may resolve
­Treatment after more conservative treatment. Marsupialisation of the
cyst has been found to be effective and has resulted in com-
Inflammatory collateral cysts are simple cysts and there is pete regression, with normal eruption of the associated
no evidence to suggest that recurrence is a problem. tooth and restoration of a normal gingival profile (Lizio
Paradental cysts associated with third molars are usually et  al.  2011). Cysts have also been shown to resolve after
enucleated along with the offending tooth. However, if the simple probing and irrigation, or even to heal spontane-
tooth is not impacted or angulated and appears to be erupt- ously as the tooth erupts (David et  al.  1998; Gomez
ing, it may be preserved and should continue to erupt into et  al.  2001; Zadik et  al.  2011). That these cysts are self-­
a normal occlusion. limiting and may resolve spontaneously is supported by the
Mandibular buccal bifurcation cysts are often enucleated fact that they rarely reach a large size and they do not occur
without removal of the associated tooth, which then erupts in adults.
62

Dentigerous Cyst

CHAPTER MENU
Clinical Features, 63
●● Frequency, 63
●● Age, 63
●● Sex, 65
●● Site, 66
●● Clinical Presentation,  67
Radiological Features, 67
●● Radiological Differential Diagnosis,  70
●● Distinguishing Dilated Follicles from Dentigerous Cysts,  71
Pathogenesis, 72
●● Source of Epithelium and Phase of Initiation,  72
●● Phase of Cyst Formation,  73
●● Growth and Enlargement of the Dentigerous Cyst,  73
–– Degradation of the Connective Tissues and Bone Resorption,  74
●● Dentigerous Cysts and the PTCH Gene,  74
●● Inflammatory Dentigerous Cyst,  75
●● ‘Extrafollicular Dentigerous Cyst’,  76
●● Dentigerous Cyst as a Potential Ameloblastoma,  76
Histopathology, 78
●● Immunohistochemistry and Biomarker Studies,  80
●● Malignant Change in Dentigerous Cysts,  81
Treatment, 82

A dentigerous cyst is an odontogenic cyst that encloses the for any radiolucent lesion that envelops the crown of an
crown of an unerupted tooth and is attached to the cervical unerupted tooth. The lesion is referred to as being in a ‘den-
region at the cementoenamel junction (Nelson and tigerous relationship’ and the radiological differential diag-
White 2021; see later Figures 5.18 and 5.19). The cyst devel- nosis may be quite wide, but would include an odontogenic
ops as a result of expansion of the dental follicle and keratocyst, an orthokeratinised odontogenic cyst, or an
follicular cyst has been used as a synonym. There has been ameloblastoma, all of which are commonly encountered at
debate with regard to terminology, but the term dentigerous the angle of the mandible and may envelop the crown of an
is most widely used and is preferred since it is unique to unerupted third molar (Müller 2021). It is important there-
this lesion, asserts an association with the teeth, and avoids fore that the diagnosis of dentigerous cyst is not made
any confusion with follicular cysts at other sites such as the uncritically on radiographic evidence alone. The criteria for
ovary, or hair follicles (Browne and Smith  1991; Nelson diagnosis will be discussed later in this chapter.
and White 2022). An eruption cyst is a variant of dentigerous cyst that is
Unfortunately, the term dentigerous is also used in a found in the soft tissues overlying an erupting tooth.
broader sense, especially in radiology, as a descriptive name Eruption cysts will be considered in Chapter 6.

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
­Clinical Feature  63

­Clinical Features Soluk Tekkeşin et al. (2012b) found that 21.3% of cysts were
dentigerous in their paediatric population, compared to only
Frequency 9.1% in adults. Other studies have shown a frequency of den-
tigerous cysts of 53.8% and 19.3% (Manor et al. 2012), 66.4%
Dentigerous cyst is the second most common cyst of the and 12.5% (Ochsenius et  al.  2007), and 30.3% and 17.5%
jaws and comprises about 20% of odontogenic cysts. In a (Jones et al. 2006) in children and adults, respectively. These
large series of cysts recorded over a 46-­year period at the data appear to suggest that children may be more susceptible
University of the Witwatersrand, Shear found that 17.1% to dentigerous cysts, but a more probable explanation is that
(599) of 3498 jaw cysts were dentigerous cysts (Table 1.1). children are less susceptible to radicular cysts, resulting in a
If only odontogenic cysts are included, dentigerous cysts relatively higher frequency of dentigerous cysts.
represented 19.7% and were the second most common
odontogenic cyst. This is similar to the frequency of 18.1%
Age
found in a large series of 7121 odontogenic cysts in the
United Kingdom (Jones et al. 2006; Table 1.2). Dentigerous cysts show a wide age distribution, but most
The frequency varies between series and countries reports suggest a mean age of between 30 and 40 years.
(Table  1.3), with a range from 10.6% in Turkey (Soluk However, there is some variation between studies and coun-
Tekkeşin et  al.  2012b) to as high as 33.0% in Mexico tries. This is illustrated in Figures 5.1 and 5.2, which show
(Mosqueda-­Taylor et al. 2002). Three studies found that the contrasting data from two different countries. In Shear’s
frequency of dentigerous cysts was lower than that of odonto- series of 343 patients from South Africa (Figure 5.1), most
genic keratocysts (Soluk Tekkeşin et al. 2012b; Ramachandra cases arose in the second to fourth decades, but with a slight
et al. 2011; Kammer et al. 2020), but this is unusual and no peak in the 20–29-­year age group, followed by a gradual
clear explanation has been given. In a pooled analysis of 17 decline. A large Sheffield series of 1274 cases shows a similar
studies, Kammer et  al. (2020) found that dentigerous cysts wide age range (Figure  5.2), but with most cases in the
represented 22.9% of odontogenic cysts compared to 12.0% fourth to sixth decades and a slight peak in the 40–49-­year
for odontogenic keratocysts (and 54.3% for radicular cysts). age group. The mean age was 40.8 years (Jones et al. 2006).
A number of studies have shown that the relative fre- Both series showed that the frequency of dentigerous cysts
quency of dentigerous cysts is greater in paediatric popula- in the first decade was considerably lower than in the sub-
tions (16 years and less). In an Israeli study of 57 paediatric sequent three decades. This is because the most frequently
patients with odontogenic cysts, Bodner (2002) found that affected teeth are the mandibular third molars and maxillary
31 (54.3%) were dentigerous cysts and a further 15 (26.3%) permanent canines, which are more likely to be impacted in
were eruption cysts. Only 9 cysts (15.8%) were radicular. adolescents and young adults. Figure  5.3 illustrates the

Figure 5.1  Age and sex distribution of 343 100


patients with dentigerous cysts (South
African series). 90 86

80 76

70 34

59 Females
60
No. of cases

41 Males
50 18

40 38

32 8
30 28
10 52
11 20
20 41
4
35
30
10 22
17 16
3
1
0
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89
Age
64 Dentigerous Cyst

300

Females
252
250 Males
230
218

200 90
78 66
No. of cases

166
158

150 141
61
60
56
100
162

152 152
50 49
50 105 98
14 85 20

36 10
29
4
0 6
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89
Age

Figure 5.2  Age and sex distribution of 1274 patients with dentigerous cysts (Sheffield series).

0-9 10-19 20-29 30-39


Tooth Tooth Tooth Tooth
8 7 6 5 4 3 2 1 8 7 6 5 4 3 2 1 8 7 6 5 4 3 2 1 8 7 6 5 4 3 2 1

12
10
5 5
3 2
1 1 1 1 1
1

2 1 1 6 1 1 1 1
4 4
6 8 8

23 22

40-49 50-59 60-69 70-79


Tooth Tooth Tooth Tooth
8 7 6 5 4 3 2 1 8 7 6 5 4 3 2 1 8 7 6 5 4 3 2 1 8 7 6 5 4 3 2 1

5
2 1 2 2
1

1 1 1 1
2
4

10
12

Figure 5.3  Distribution of the location of dentigerous cysts in different decades (n = 174). The distribution closely follows the
chronology of tooth development and eruption.
­Clinical Feature  65

Table 5.1  Age and sex distribution from selected large series


Box 5.1  Dentigerous Cyst: Epidemiology and Key Facts of dentigerous cysts.
●● Dentigerous cysts are the second most common
cystic lesion of the jaws Mean Peak
References n age decade(s) Male (%)
●● They comprise about 20% of all odontogenic cysts
●● They are more common in males. Overall about 60% Daley et al. (1994) 1662 NR 3rd 59.9
are found in males (M : F 1.5 : 1)
Ledesma-­Montes et al. 108 NR 2nd 64.8
●● Peak age is in the third and fourth decades (2000)
(20–40 years)
Meningaud et al. (2006) 154 44.9 NR 70.0
●● Upto about 70% are associated with impacted lower
Jones et al. (2006) 1292 40.8 5th 65.1
third molars
Ochsenius et al. (2007) 546 23.5 2nd 63.0
●● Very rare on deciduous teeth
Tortorici et al. (2008) 149 31.0 2nd 61.1
Zhang et al. (2010) 2082 35.0 2nd 61.0
distribution of 174 cases from the South African series for Sharifian and Khalili 303 21.5 2nd and 3rd 57.3
which both age and the involved tooth had been recorded. (2011)
No cases were recorded on deciduous teeth. Only 17 cases Soluk Tekkeşin et al. 529 32.8 NR 60.4
(2012a,b)
occurred in the first decade and these were found most
commonly on mandibular premolars. Maxillary canines Lin et al. (2013)a 338 33.0 2nd 63.8
are the second most affected tooth and these were found to Tamiolakis et al. (2019) 766 38.7 4th–6th 65.3
be involved mostly in the second (12 cases) and third (10 Lo Muzio et al. (2017) 806 34.8 3rd and 4th 60.5
cases) decades. Mandibular third molars were found to be Karabas et al. (2020) 114 NR 5th 66.0
affected predominantly in the third and fourth decades
NR, not reported.
(23 and 22 cases, respectively). Dentigerous cysts were a
 Lin et al. reported a second peak for males in the fourth to sixth
rarely encountered in the seventh or eighth decades, but decades.
even then impacted third molars or canines were found to
be involved.
Most studies that have reported large a number of denti- with 213 (62%) in males and 130 (38%) in females, a ratio of
gerous cysts show a similar wide age range and a mean age 1.6 : 1. In the Sheffield sample (Figure 5.3; Jones et al. 2006),
of between 30 and 40 years, with a peak incidence in the 819 (64%) cases were in males and 455 (36%) in females
second or third decades (Table  5.1). Three studies have (M : F 1.8 : 1).
reported dentigerous cysts in slightly older age groups, with Figures 5.1 and 5.2 show that the proportions of males
mean ages of 44.9 (Meningaud et al. 2006) and 38.7 years in each decade are approximately the same and most
(Tamiolakis et al. 2019), or with peaks in the fourth to sixth studies that have undertaken this analysis have a found
decades (Tamiolakis et al. 2019). In the Sheffield data there a similar distribution (Ochsenius et  al.  2007; Zhang
is a small peak in the fifth decade (Figure 5.2), but overall et al. 2010; Tamiolakis et al. 2019). Tamiolakis et al. (2019)
there was a wide age range, with the majority of cases found also found no significant differences in the mean ages of
between 30 and 59 years. male and female patients (39.8 and 36.6 years, respec-
These data also show that dentigerous cysts may arise tively). Lin et al. (2013), however, found a significant dif-
in children. At this age, many may be inflammatory in ference between males and females, with mean ages of
origin and are associated with maxillary canines or 35.2 and 29.1 years, respectively. They also showed that
lower premolars (see ‘Inflammatory Dentigerous Cyst’). although the frequency in both sexes peaked in the sec-
The youngest reported case is in a 1-­year-­old child ond decade, there were further peaks among male patients
(Suresh et al. 2011). in the fourth to sixth decades. Their overall proportion of
males was 63.8% (M : F 1.8 : 1), but in the fourth, fifth, and
sixth decades the proportions of males were 68.9%, 75.0%,
Sex
and 72.5%, respectively.
Without exception, the literature shows that there is a Although it might be suggested that the more frequent
male predilection for dentigerous cysts, with males affected occurrence in males is because they have a higher preva-
in about 60% of cases (M : F 1.5 : 1; Table  5.1). The fre- lence of unerupted or impacted teeth, there is mixed evi-
quency of dentigerous cysts in the South African sample dence for this in the literature. In a recent study in Canada,
(Figure 5.1) was significantly greater in males than females, MacDonald and Yu (2020) examined 6252 consecutive
66 Dentigerous Cyst

dental panoramic radiographs and found that the most com- confidence intervals were wide and overall there were no
mon incidental finding was the presence of impacted teeth. significant differences between males and females.
These were found in 1470 patients (23.5%), with a slight These data show that the higher frequency of dentiger-
male predilection of 797 cases (12.7%) compared to 673 ous cysts in males cannot be explained simply by a higher
(10.8%) in females. The proportion of males with impacted rate of impacted teeth, and suggest that other, unknown
teeth was 54.2% and the proportion with impacted third factors may influence the development of dentigerous cysts.
molars was 54.4%. These authors also identified 18 inciden-
tal dentigerous cysts, of which 61.0% (11 cases) arose
Site
in males.
A Greek study, however, showed the opposite findings The anatomical distribution of 245 dentigerous cysts, in
(Gisakis et al. 2011). These authors identified 425 patients relation to the tooth involved, is shown in Figure  5.4. A
with impacted teeth from consecutive panoramic radio- substantial majority (119 cases: 48.6%) involved the man-
graphs. A total of 940 impacted teeth were identified, of dibular third molar. The maxillary permanent canine was
which 91.6% (841) were third molars, and of these 508 were next in order of frequency with 42 cases (17.1%), followed
in the mandible, representing 54.0% of all impactions. Of by the mandibular premolars and the maxillary third molar.
interest, however, is that of all the impacted teeth only 441 Similar distributions have been reported in other large
(46.9%) were found in males and 499 (53.1%) in females. series. The proportions of dentigerous cysts affecting the
Of the 425 patients with impacted teeth, 202 (47.5%) were mandibular third molar and maxillary canine regions, respec-
male and 223 (52.5%) were female (M : F 0.9 : 1). Considering tively, have been reported as 77.0% and 6.6% (Daley and
only lower third molars, 46.3% were found in men. Wysocki 1995), 73.2% and 10.6% (Jones et al. 2006), 77.0% and
These data support previous large series of radiographs 5% (Zhang et al. 2010), and 45.3% and 11.2% (Lin et al. 2013).
that have found no evidence of sex differences in the fre- Dentigerous cysts may be associated with supernumerary
quency of impacted teeth (Mourshed  1964a,b; Brown teeth, but the overall frequency appears to be between 1 and
et al. 1982). With regard only to third molars, Carter and 5% (Lustmann and Bodner  1988). In Shear’s series that
Worthington (2016) undertook a systematic review and contributed to Figure  5.4, there were 2 additional cysts
identified 49 studies that had investigated the prevalence of associated with supernumerary teeth (<1%). In their series
third molar impaction. Worldwide, the prevalence was of 2082 cysts, Zhang et al. (2010) found only 11 on supernu-
24.4%, but ranged from only 3.1% in a Nigerian population merary teeth (0.5%). Lin et  al. (2013) found 46 of 338
to 68.6% in Singapore. The odds ratio of impaction in males dentigerous cysts (13.6%) associated with supernumerary
versus females was examined in 14 studies. Interestingly, 9 teeth, but this appears to be an unusually high frequency.
of the 14 studies showed that the odds of having an Lustmann and Bodner (1988), in a review of 42 such cases
impacted tooth were greater in females, but the 95% from their own material and those reported in the

Tooth Figure 5.4  Anatomical distribution


8 7 6 5 4 3 2 1 of 245 dentigerous cysts.

42

11
Numbers of cases

8 7 7
2 1 2 Maxilla

3 5 0 2 Mandible
10 10
16

119
­Radiological Feature  67

literature, found that about 90% were associated with a max- Denti­gerous cysts may occasionally be painful, particularly
illary mesiodens. In a more recent review, Anthonappa et al. if infected. Patients may give a history of a slowly enlarging
(2018) identified more than 40 case reports of dentigerous swelling, and this is the common form of presentation
cysts associated with supernumerary teeth. In this study, 41 with edentulous patients when unerupted teeth have
out of 64 (64.1%) reported cases were found in the anterior inadvertently been retained. Occasionally a cyst may grow
maxilla, and only 5 were found at any site in the mandible. to a large size before it is diagnosed. This is particularly the
Dentigerous cystsvmay also be associated with odontomas. case in maxillary lesions, which may expand and displace
In their large Canadian series, Zhang et al. (2010) found 5 the maxillary antrum. Lesions in the mandibular third
cases (0.25%) associated with odontomas. Kaugars et  al. molar region, as they grow, may quickly become trauma-
(1989b) reported a series of 351 odontomas and found that tised during mastication or may become continuous with
27.6% were associated with dentigerous cysts. the oral cavity through a periodontal pocket distal to the
Occasional patients present with dentigerous cysts at second molar. They may therefore be inflamed or infected
multiple sites (see later Figure  5.11). Most occurrences at presentation and give rise to pain or discomfort, and
have been recorded as single case reports and have most occasionally may be associated with a purulent discharge.
often been found in patients with syndromes associated
with impacted teeth, including cleidocranial dysplasia and
the mucopolysaccharidoses. Non-­syndromic cases can be ­Radiological Features
seen, however. In the Canadian, Taiwan, and Turkish
series (Zhang et al. 2010; Lin et al. 2013; Karabas et al. 2020), A dentigerous cyst, by definition, is found in a dentigerous
the proportion of patients with multiple cysts was 2.5% relationship with an unerupted tooth. Radiographs there-
(51 cases), 1.8% (6 cases), and 1.7% (3 cases), respectively. fore show a radiolucent area enveloping the crown of an
Devi et al. (2015) reviewed the literature and recorded 19 unerupted tooth (Figure 5.5). The unerupted tooth may be
case reports of bilateral dentigerous cysts between 1943 impacted as a result of inadequate space in the dental arch
and their own case in 2015. Of these, 8 involved bilateral or as a result of malpositioning, for example a horizontally
mandibular third molars, 2  involved maxillary third impacted mandibular third molar or an inverted tooth.
molars, and 1 reported a case involving bilateral upper The typical radiological appearance of a dentigerous cyst
canines. Other teeth involved included mandibular premo- is illustrated in Figure 5.5. The lesion envelops the crown of
lars and second molars, and 9 cases were in children, an impacted tooth and is attached at the neck or cervical
including 6 under 10 years. A number of single case reports region (where the tooth crown meets the root), so that
continue to be published, including 2 involving 10-­year-­olds only the crown of the tooth protrudes into the cyst lumen.
(Khandeparker et al. 2018; Pant et al. 2019). Dentigerous cysts are unilocular and have a corticated
More recently, Boussouni et al. (2020) reviewed the litera- margin. The corticated outline of the cyst is continuous
ture and found 36 papers reporting 43 cases of multiple or with the lamina dura surrounding the tooth roots (Figure 5.5),
bilateral dentigerous cysts between 2009 and 2019. Of these, indicating that the cyst arises and is contained within the
23 cases (53.5%) involved the mandible, 12 (27.9%) the dental crypt of the developing tooth. Cysts may grow to a
maxilla, and in 8 patients (18.6%) both jaws were affected. large size and cause considerable buccal or lingual expan-
Bilateral mandibular third molars were affected in only 12 sion of the cortical plates (Figures 5.6 and 5.8).
patients (27.9%), with mandibular premolars affected in 14
cases (32.6%) and maxillary canines in 13 (30.2%).
There have been five patients reported with bilateral den- Box 5.2  Clinical and radiological Features: Key Facts
tigerous cysts involving all four quadrants (Devi et al. 2015;
●● Always associated with an unerupted or impacted tooth
Jeon et  al.  2016; Boussouni et  al.  2020) and one patient
●● Most commonly found on lower third molars and
reported with five cysts affecting a maxillary incisor and
then upper canines
premolar, and mandibular canine and second and third
●● Often symptomless and found on radiological
molars (Moturi and Kaila 2018).
examination
●● Slowly growing swelling, may cause bucco-­lingual
Clinical Presentation expansion
●● Radiology shows well-­demarcated, corticated lesion
The vast majority of dentigerous cysts are symptomless
enveloping the crown of the affected tooth
and are discovered on radiographs that have been taken
●● A pericoronal space greater then 10 mm is highly
because a tooth has failed to erupt, or is missing, or because
suggestive of a dentigerous cyst
teeth are tilted or are otherwise out of alignment.
68 Dentigerous Cyst

Figure 5.5  Radiograph of a typical dentigerous cyst, associated


with a lower third molar. The cyst is well defined and has a
corticated margin attached at the cementoenamel junction (arrow).

Figure 5.6  Computed tomographic scan of a central type of


dentigerous cyst, showing the extent of bucco-­lingual expansion
and clear evidence of a corticated margin.

(a) (b) (c) Figure 5.7  Diagram illustrating the manner in which


the cyst may expand to produce the radiographic
appearances of (a) central, (b) lateral, and
(c) circumferential types of dentigerous cyst.

This typical radiological appearance is referred to as the


central type of dentigerous cyst (Figure  5.7). In this type
the crown is enveloped symmetrically and the pressure of
cyst growth is applied to the crown of the tooth and may
push it away from its direction of eruption. In this way,
mandibular third molars are often displaced towards the
angle or lower border of the mandible or into the ascend-
ing ramus (Figure  5.8), and a maxillary canine may be
forced into the maxillary sinus. A maxillary cyst may dis-
place the antrum and impinge on the floor of the nose
(Figure 5.9).
More often, and especially when third molars are
involved, a dentigerous cyst becomes displaced as the tooth
tries to erupt and may be found lateral to the tooth, or the
whole cyst may envelop the tooth in a circumferential pat-
tern (Figure  5.7). The lateral type of dentigerous cyst
(Figures 5.10 and 5.11) is a radiographic appearance that Figure 5.8  A central type of dentigerous cyst has displaced the
results from dilatation of the cyst on one aspect of the third molar into the ascending ramus. This large cyst has
expanded bucco-­lingually, but on this plain radiograph only a
crown, or if the cyst is displaced by the tooth as it tries to shadow can be seen (arrows). Bucco-­lingual expansion is best
erupt (Figure 5.10). seen on computed tomography (Figure 5.6).
­Radiological Feature  69

The so-­called circumferential type of dentigerous cyst, (1976) observed root resorption in 11 of 20 dentigerous cysts
in which the entire tooth appears to be enveloped by the (55%) (Figures  5.12 and  5.13), compared to only 6 of 33
cyst (Figure  5.12), results when the cyst expands in the radicular cysts (18%), and in none of their sample of 26
manner illustrated in Figure 5.7c. It is important that this odontogenic keratocysts. They suggested that the dentiger-
variety be differentiated from other lesions that may ous cyst potential for root resorption may be due to its origin
envelop a tooth. from dental follicle and the ability of the follicle to resorb
Dentigerous cysts appear to have a greater tendency than the roots of the deciduous predecessors of the affected teeth.
other simple jaw cysts to produce some resorption of the Most dentigerous cysts can be diagnosed on plain radio-
roots of adjacent teeth. In a radiographic study of root graphs, but computed tomography (CT) is useful and is now
resorption produced by jaw cysts, Struthers and Shear widely used since the accessibility of cone beam computed
tomography (CBCT). CT is especially useful to appreciate
the three-­dimensional extent of a lesion in a bucco-­lingual
direction (Figure  5.6), or to determine the relationship of
the cyst to the complex anatomy of the maxilla (Figure 5.9)
(MacDonald 2016; Allison and Garlington 2017).
As discussed above, the majority of dentigerous cysts are
associated with impacted lower third molars followed by
maxillary canines, so it might be expected that cysts are
commonly encountered when radiographs are taken for
routine diagnostic purposes or for orthodontic surveillance.
However, this is not the case and overall the prevalence of

Figure 5.9  Computed tomographic scan of a maxillary Figure 5.10  Radiograph of a lateral type of dentigerous cyst.
dentigerous cyst extending to, and impinging on, the floor of the The cyst is displaced to the medial aspect of the tooth, which is
nose. Source: Courtesy of Dr Mark Cohen. now partially erupted.

Figure 5.11  A rare example of bilateral


dentigerous cysts in a child, affecting
both developing second molars. Both
cysts are large, but have been displaced
in a medial direction to the lateral aspect
of the teeth.
70 Dentigerous Cyst

Figure 5.13  A dentigerous cyst wall with resorption of the


contiguous root of an adjacent tooth.

alone, without histological confirmation. The most impor-


tant lesions that must be considered, and that are frequently
Figure 5.12  Radiograph of a circumferential type of associated with unerupted third molars, are odontogenic
dentigerous cyst associated with a mandibular third molar. The
cyst has enveloped the tooth so that both the crown and roots keratocyst, orthokeratinised odontogenic cyst, and amelo-
appear to be in the lumen. Note also that the cyst wall has blastoma. These arise from remnants of the dental lamina
resorbed part of the root of the adjacent molar. that may be found in the connective tissues overlying the
dental follicle, within the gubernacular canal or the dental
cysts on routine radiographs appears to be low. MacDonald crypt. Thus, as the cyst or tumour expands, it may envelop
and Yu (2020) undertook a retrospective review of 6252 con- the tooth and appear radiologically to be in a dentigerous
secutive panoramic radiographs and found that 32% showed relationship with the crown (Figure 5.14).
at least one incidental finding. In 1470 (23.5%) cases this In their systematic review of lesions associated with
was an impacted tooth, but evidence of a widened pericoro- third molars, Mello et  al. (2019) found that 5.3% were
nal space suggestive of a dentigerous cyst was only found in affected by odontogenic cysts or tumours. Dentigerous
18 cases (1.2%). The prevalence of possible dentigerous cysts were found in 2.1% of cases, odontogenic keratocysts
cysts in the total cohort was only 0.3%. in 0.5%, and ameloblastomas in 0.4%. Other lesions that
In a similar study, Karabas et  al. (2020) reviewed 6758 were occasionally encountered included calcifying odonto-
CBCTs and found that only 5.9% (400 images) showed evi- genic cyst and odontogenic myxoma.
dence of a radiolucent lesion (a pericoronal space of >2.5 mm) In a series of 5486 impacted third molars (in 4133 patients),
around impacted teeth. Of these, 190 were examined histo- Patil et al. (2014) found 132 dentigerous cysts (2.4%) as well
logically and 114 dentigerous cysts were confirmed. Thus, as 16 odontogenic keratocysts (0.3%) and 31 ameloblastomas
only 1.6% of patients from the whole cohort were confirmed (0.6%). Overall, dentigerous cysts, odontogenic keratocysts,
to have a dentigerous cyst. and ameloblastomas comprised 67%, 8%, and 16%, respec-
In a systematic review of the prevalence of odontogenic tively, of the 197  lesions detected. Other lesions included
cysts and tumours associated with impacted third molars, calcifying odontogenic cyst (2), hyperplastic follicles (5),
Mello et al. (2019) identified 16 studies where lesions had odontogenic fibroma (3), and odontomas (6).
been noted on radiographs and the diagnosis confirmed by Curran et al. (2002) reported a sample of 2646 pericoro-
histological examination. A total of 50 969 impacted third nal lesions over a six-­year period, and found that 1776
molars were identified and meta-­analysis showed a pooled (67%) were follicular tissue with no evidence of pathology.
prevalence of cysts and tumours of 5.3% and a prevalence Of the remaining cases, 752 (28.4%) were dentigerous cysts,
of dentigerous cysts of only 2.1%. 71 (2.7%) were odontogenic keratocysts, and 13 (0.5%) were
ameloblastomas. The remaining lesions included odon-
toma, squamous carcinoma, calcifying odontogenic cyst,
Radiological Differential Diagnosis
calcifying epithelial odontogenic tumour, and odonto-
Although the radiographic features of a dentigerous cyst are genic myxoma.
characteristic, other lesions may also envelop a tooth and it At other sites, an important lesion to consider is adenoma-
is not possible to give a definitive diagnosis on radiology toid odontogenic tumour, up to 75% of which may be found,
­Radiological Feature  71

Figure 5.14  A cyst envelops the crown of a lower third molar, and on radiology is in a dentigerous relationship with the tooth.
Histology, however, showed an odontogenic keratocyst.

on radiographs, to be in a dentigerous relationship with an predilection. They concluded that a pericoronal space of
impacted tooth – most often an upper canine (Reichart and 4 mm or greater was suggestive of a dentigerous cyst, but
Philipsen 2004; Chrcanovic and Gomez 2019b). that distinguishing reliably between a small dentigerous
Diagnostic errors can be avoided if the criteria for diag- cyst and a large dental follicle may be resolved only by
nosis are followed (see later Box 5.4) and the clinical, radio- identifying a cyst cavity at the surgical operation.
logical, and histopathological features are reviewed and The approach taken by Damante and Fleury (2001) was
correlated (Barrett et al. 2017; Müller 2021). Barrett et al. to correlate the relationship between the width of the
(2017) showed that histological features of dentigerous pericoronal space and the microscopic features of the
cyst, keratocyst, and cystic ameloblastoma may be similar, follicular tissue. Their sample comprised 130 unerupted
especially in small incisional biopsies. In a review of and 35 partially erupted teeth that were radiographed and
101  lesions diagnosed histologically as dentigerous cysts, then extracted. The pericoronal spaces were measured on
they found that five keratocysts and four ameloblastomas the radiographs and the widths were compared with the
had been missed because of a lack of knowledge of the cor- results of the histological examination of the removed tis-
rect histology, or failure to properly consider the radiologi- sue. The width of the pericoronal space ranged from 0.1 to
cal features. 5.6 mm. The most frequently observed lining of the folli-
cles was a reduced enamel epithelium in 68.4% of
unerupted teeth, and a hyperplastic stratified squamous
Distinguishing Dilated Follicles
epithelium in 68.5% of the partially erupted teeth.
from Dentigerous Cysts
Inflammation was present in 36.1% of the unerupted
All unerupted or impacted teeth show a zone of radiolu- teeth and 82.8% of the partially erupted group. There was
cency around the crown that represents the soft tissues of a statistically significant association between the pres-
the dental follicle. In the differential diagnosis, therefore, it ence of stratified squamous epithelium and pericoronal
is important to be able to distinguish between a normal or space enlargement for unerupted teeth. They also found a
hyperplastic dental follicle and a dentigerous cyst. There is trend in the association between inflammation and
much debate about the size or width of a normal pericoro- enlargement of the pericoronal space.
nal space, but most authors suggest that a space of greater Surgically, the authors detected no bone cavitation or
than 3–5 mm is abnormal. An additional consideration is luminal cystic contents, and were unable to diagnose any
that some erupting teeth have a dilated or hyperplastic fol- dentigerous cysts in pericoronal spaces smaller than
licle in the pre-­eruptive phase. 5.6 mm. They proposed that a pericoronal space up to 3 mm
Daley and Wysocki (1995) have pointed out that it can be in width corresponds to a normal dental follicle and a
difficult to distinguish between a small dentigerous cyst width between 3 and 5.6 mm suggests an inflamed and
and a large dental follicle, despite the availability of both hyperplastic dental follicle. They agreed with Daley and
radiographic and histological information. Their compara- Wysocki (1995) that the criteria for diagnosis of a dentiger-
tive study of 1662 dentigerous cysts and 824 dental follicles ous cyst must include the presence of a cystic cavity and
showed considerable overlap in age distribution and site luminal contents. Some regard the presence of stratified
72 Dentigerous Cyst

squamous epithelium as diagnostic, but both these studies Source of Epithelium and Phase of Initiation
(Daley and Wysocki  1995; Damante and Fleury  2001)
The epithelial lining of the dentigerous cyst arises from the
found non-­keratinised stratified squamous epithelium lin-
reduced enamel epithelium that overlies the crown of a
ing normal or hyperplastic dental follicles.
tooth after enamel formation is complete, but before erup-
Stathopoulos et al. (2011) examined 417 ‘lesions’ (defined
tion. Apart from the histological appearance of the lining
as a pericoronal space >3 mm) in a consecutive series of
of some cysts (see ‘Histopathology’), the evidence for this
7782 impacted third molars. Of these, 202 (48.4%) proved
comes from animal studies, mostly carried out in the 1970s,
to be normal follicles and 138 (33%) were dentigerous cysts.
that are unlikely to be repeated (reviewed by Harris and
Other detected lesions included 29 odontogenic kerato-
Toller  1975; Browne  1991a). These early studies largely
cysts (7.0%) and 21 (5.0%) ameloblastomas.
aimed to investigate the possibility of tooth transplants, but
More recently, Namgyel et al. (2020) compared the his-
it soon became apparent that implanted teeth often devel-
tological features of pericoronal tissues from follicles
oped cysts around the crowns – a serendipitous model of
measured on radiographs as being less than 2.5  mm or
dentigerous cyst formation.
2.5 mm and above. Of the 206 specimens less than 2.5 mm,
The work of two research groups was especially informa-
105 (51%) showed normal dental follicle and 101 (49%)
tive. Atkinson (1972, 1976, 1977) and Al-­Talabani and
showed pathological changes. There were 92 specimens
Smith (1980) transplanted tooth germs subcutaneously
from spaces greater than 2.5 mm, of which 41% were normal
into mouse skin or hamster cheek pouches, and found
follicles and 59% pathological. There were no significant
cystic change as early as 7 days after transplantation. Cyst
correlations between the width of the pericoronal space
formation only occurred after enamel formation was com-
and pathological changes.
plete and was preceded by hyperplasia of the reduced
Edamatsu et al. (2005) compared 80 dental follicles with
enamel epithelium and the formation of intraepithelial
27 dentigerous cysts and found that 76% (61) of the follicles
clefts overlying the occlusal surfaces of the teeth. Cystic
were less than 3 mm in width and the remainder were
degeneration within the hyperplastic epithelium produced
between 3 and 10 mm. There were no follicles greater than
cavities lined with a thick, parakeratotic, stratified squa-
10 mm. In contrast, 20 of the cysts (74%) were greater than
mous epithelium, but as the cysts enlarged, the lining
10 mm and 7 (26%) were between 3 and 10 mm. No cysts
changed to a thin, non-­keratinised, stratified squamous
were less than 3 mm.
epithelium. After about 50 days the cysts may envelop the
Taken together, these studies suggest that the width of
tooth crown and extend to the cementoenamel junction.
the follicle is not a reliable indicator of pathological
Al-­Talabani and Smith (1980) found that many of the
change. Although a pericoronal space of greater than
affected teeth showed enamel hypoplasia. They went on to
2.5–5 mm is suggestive of pathological change, it may also
examine teeth associated with human dentigerous cysts
be an enlarged follicle. Only pericoronal spaces of 10 mm
and found that in 50% of cases there was evidence of
or more are most likely to be dentigerous cysts.
enamel hypoplasia on the occlusal surfaces or incisal edges
of the affected teeth. They proposed that there was a direct
­Pathogenesis relationship between the development of cysts and the
occurrence of enamel hypoplasia, and suggested that there
In Chapter  3, the pathogenesis of the radicular cyst was may be two types of dentigerous cyst, with different causes
discussed in detail and was presented in three phases: the and arising at different stages of tooth development. One
phase of initiation, the phase of cyst formation, and the type would arise by cystic degeneration of the stellate retic-
phase of growth and enlargement (including bone resorp- ulum at an early stage of development and is likely to dis-
tion). These three phases are the basis for the pathogenesis rupt amelogenesis and cause enamel hypoplasia. In the
of all cysts, but whereas the initiating phase of inflamma- second type, cyst formation commences after completion
tory cysts is quite well understood, the initiating factors of the crown and is initiated by accumulation of fluid,
behind the formation of dentigerous cysts are poorly either between the layers of the reduced enamel epithe-
understood or even unknown. The pathogenesis of odonto- lium or between the epithelium and the tooth crown.
genic cysts has not been of great interest to researchers and Enamel hypoplasia would not be a significant feature of
little new knowledge has been added to the literature since this variety. This association of dentigerous cyst with
the seminal studies of Harris and Toller in the 1960s and enamel hypoplasia of the contained tooth is an interesting
1970s (reviewed in Harris and Toller 1975) and of Browne one, since it would explain the frequent finding of enamel
and colleagues (reviewed in Browne  1991a; Browne and hypoplasia on affected teeth. However, another explana-
Smith 1991). tion that was not considered by Al-­Talabani and Smith
­Pathogenesi  73

(1980) may be that the presence of enamel hypoplasia Growth and Enlargement of the
reduces the adhesion of reduced enamel epithelium to the Dentigerous Cyst
crown and provides the starting point for cyst development.
Once a cystic cavity has formed, the mechanisms of further
Great care must be taken in extrapolating animal experi-
growth and enlargement are similar to those described for
ments to the human situation, but these studies do indicate
radicular cysts. In particular, expansion is almost certainly
that the reduced enamel epithelium may give rise to a den-
mediated by hydrostatic pressure, and the early studies,
tigerous cyst and suggest that cleft formation may be an
especially those of Toller (1948, 1966b, 1967, 1970a,b), have
initiating event. An account of normal tooth eruption is
been described in detail in Chapter 3.
beyond the scope of this chapter, but an understanding of
A number of studies have shown that dentigerous cyst
the basic mechanisms is helpful (Wise et  al.  2002; Nel
fluids have soluble protein and immunoglobulin (Ig) levels
et al. 2015; Bastos et al. 2021). The processes that cause a
similar to those in serum, suggesting a simple transudate
tooth to move towards the surface of the alveolus are still
(Skaug 1973; Skaug and Hofstad 1973; Browne 1975). There
poorly understood, but do involve a complex network of
is also evidence that immunoglobulins are produced in the
inductive signals between the tissues involved. The overall
capsule of dentigerous cysts, since plasma cells are found
outcome is movement of the tooth that is associated with
in the cyst wall and there is intense extracellular staining of
pressure on the adjacent structures. As a tooth moves
IgG, suggesting that the immunoglobulins found in cyst
towards the surface, the reduced enamel epithelium prolif-
fluids may be derived from local synthesis in the cyst cap-
erates upwards, merges with the overlying mucosa, and
sule as well as from a transudate (Smith et al. 1987).
forms the gingival cleft through which the tooth emerges
Many dentigerous cysts show evidence of acute and
into the mouth. If tooth eruption is impeded the epithe-
chronic inflammation in their walls, and in these instances
lium may still proliferate, but without upward movement it
an inflammatory exudate may contribute to the accumu-
may then degenerate and form clefts that represent the ear-
lation of luminal proteins and play some part in the
liest stage of cyst formation. An alternative scenario is that
expansion of the cyst. Moreover, desquamated epithelial
if eruption is impeded, the reduced enamel epithelium
cells and the passage of inflammatory cells into the cyst
does not proliferate at all, but becomes separated from the
cavity must contribute to an increase in luminal osmotic
tooth crown to form a space that then develops into a cyst.
tension and thereby promote further expansion of the
cyst. Glycosaminoglycans, predominantly hyaluronic
acid, heparin, and chondroitin-­4-­sulphate, have been
Phase of Cyst Formation
found in the fluids and walls of dentigerous cysts (Skaug
Whether the cyst starts as a cleft within the reduced enamel and Hofstad  1972; Smith et  al.  1984, 1988), and are also
epithelium or between the reduced enamel epithelium and thought to have an important role in expansile cyst growth
the tooth, it is thought that further cyst development results raising the internal hydrostatic pressure.
from accumulation of fluid as a result of pressure in the Toller (1970b) showed that the mean osmolality of denti-
tooth follicle producing a fluid transudate. Main (1970) gerous cyst fluid (291 ± 14.42 mOsm) was similar to that
suggested that the pressure exerted by an erupting tooth on found in radicular cysts (290 ± 14.93 mOsm) and odonto-
an impacted follicle obstructs the venous outflow and genic keratocysts (296 ± 15.16 mOsm). He believed that
thereby induces rapid transudation of serum across the raised osmolality of the cyst fluid made an important con-
capillary walls. The increased hydrostatic pressure of this tribution to expansion of odontogenic cysts. These findings
pooling fluid separates the reduced enamel epithelium were supported by Kubota et  al. (2004), who also found
from the crown. With time, capillary permeability increases similar fluid pressures in keratocysts, dentigerous cysts,
and permits the passage of greater quantities of protein and radicular cysts. Furthermore, the latter authors showed
above the low concentration of the pure transudate. Such a that the intracystic pressure in all cyst types reduced as the
mechanism is supported by the observation that during cyst increased in size. They concluded that increased pres-
normal tooth development and eruption, there is apoptosis sure played a pivotal part in early cyst growth, but was less
of cells in the enamel organ and this stimulates the release important as the cyst got bigger.
of paracrine factors, including interleukin (IL)-­1α, which As the cyst expands, there must be some compensatory
are pro-­inflammatory and may mediate capillary permea- epithelial proliferation to cover the greater surface area of
bility as well as osteoclastogenesis (see later; Nel et al. 2015). the cyst lumen. Similar mechanisms occur in all cyst types
Thus in the impacted tooth, rather than facilitating normal and studies have shown that the proliferation rate of the
eruption, these paracrine signalling events may promote lining epithelium is similar in dentigerous and radicular
cyst formation. cysts (Martins et al. 2011). There is also evidence that the
74 Dentigerous Cyst

PTCH gene and the Sonic hedgehog (HH) signalling path-


Box 5.3  Pathogenesis: Key Facts
way are activated in a number of cyst types, including den-
tigerous cyst (see below), and this may contribute to cell Three elements are needed for cyst formation:
proliferation and growth.
●● A source of epithelium
●● A stimulus for epithelial proliferation
Degradation of the Connective Tissues
●● A mechanism of growth and bone resorption
and Bone Resorption
Growth and enlargement of all odontogenic cysts must be Dentigerous cysts are developmental in origin and
accompanied by degradation of the adjacent connective the initiating factors are poorly understood. However,
tissues and resorption of surrounding bone. Mechanisms cyst development can still be considered in three phases:
of bone resorption have been considered in detail in
●● Phase of initiation – the reduced enamel epithelium
Chapter  3, and these apply equally to bone resorption in
overlying an unerupted tooth proliferates and
dentigerous cysts. Of particular interest is the role of IL-­1
forms clefts, either within the epithelium or
and prostaglandins, both of which are produced in denti-
between the epithelium and the tooth crown
gerous cysts and appear to have a pivotal role in mediating
●● Phase of cyst formation –  accumulation of debris
osteoclastogenesis and bone resorption (Harris and
and proteins in the clefts increases luminal osmotic
Goldhaber  1973; Harris et  al.  1973; Harris  1978; Meghji
pressure, resulting in cyst formation and expansion
et  al.  1989; Bando et  al.  1993; Meghji et  al.  1996). In the
●● Phase of growth and enlargement – expansion of the
case of radicular cysts, it is inflammation that produces the
cyst is associated with epithelial cell proliferation
network of biological factors that underpin growth of the
and bone resorption. These events may be facilitated
cyst (Table  3.2), but in dentigerous cysts inflammation
by perturbed bone remodelling and by associated
plays a lesser role and the initiating factors are less clear.
inflammation
During normal tooth eruption there is turnover of adja-
cent connective tissues and bone around the erupting
tooth, with bone resorption above the erupting tooth and they may be traumatised by the occlusion. It is almost cer-
redeposition below it. This is thought to be mediated by a tainly the case, therefore, that inflammation plays a role in
cascade of paracrine signalling events in the follicular tis- cyst growth and destruction of bone and adjacent tissues.
sues surrounding the developing tooth. These events may
be initiated by tissue remodelling and apoptosis of the fol-
Dentigerous Cysts and the PTCH Gene
licular tissues after enamel formation is complete, result-
ing in production of cytokines, including IL-­1 and The role of the PTCH gene and the hedgehog (HH) signal-
parathyroid hormone related protein (PTHrP), both of ling pathway in the pathogenesis of the odontogenic
which influence osteoclastogenesis and bone resorption keratocyst is discussed in Chapter 7. The point is made that
(Wise et al. 2002; Nel et al. 2015; Bastos et al. 2021). If tooth although PTCH alterations are important in keratocysts,
eruption is impeded, these events will still occur, but rather they do not seem to be specific, and there is some evidence
than facilitating tooth eruption, they may result in bone that PTCH may be involved in other odontogenic lesions
resorption that allows a developing cyst to grow. This sug- (Gomes and Gomez  2011), including orthokeratinised
gests that bone resorption in dentigerous cysts is similar to odontogenic cyst (Diniz et  al.  2011), calcifying epithelial
the normal bone remodelling that might occur during odontogenic tumour (Peacock et al. 2010), glandular odon-
eruption, but is perturbed by the impaction of the tooth. togenic cyst, and dentigerous cysts (Levanat et  al.  2000;
This idea is supported by the observation that there is no Pavelić et al. 2001; Barreto et al. 2002; Zhang et al. 2010).
difference in the expression of bone resorption–related Although much more research is needed, the studies show-
matrix metalloproteinases (MMPs) between dentigerous ing loss of heterozygosity (LOH) of the PTCH gene in den-
cysts and normal dental follicles (Suojanen et al. 2014). tigerous cysts (Levanat et al. 2000; Pavelić et al. 2001) have
Although dentigerous cysts are developmental in origin, not yet been repeated and sequencing studies to investigate
most become inflamed as they get larger. This may be a mutations have not been performed.
direct result of the signalling events described above, Levanat et al. (2000) examined seven each of dentiger-
which will facilitate recruitment of monocytes and other ous cysts, radicular cysts, and odontogenic keratocysts for
inflammatory cells, but may also be a result of trauma or LOH in two regions of the PTCH gene. Three of seven
infection. As cysts grow, especially in the third molar dentigerous cysts and four of seven keratocysts showed
region, they may become continuous with the oral cavity LOH in the 9q22.3 region of the gene and one from each
through a periodontal pocket distal to the second molar, or group showed loss at 9q31. LOH was not detected in any
­Pathogenesi  75

radicular cysts. In a further study, the same group (Pavelić


et al. 2001) confirmed these findings in a larger series and
found LOH in the same region in five of ten dentigerous
cysts. They also used RT-­PCR in two cysts to show that the
PTCH gene was still expressed despite evidence of
LOH. They interpreted this to mean that the remaining
allele must be mutated, otherwise normal PTCH protein
would have blocked the normal signalling pathway.
Inactivation of the PTCH gene and reduced expression of
the PTCH protein may lead to activation of the hedgehog
signalling pathway. This is discussed in detail in Chapter 7
in the context of the odontogenic keratocyst, but there is
good evidence that hedgehog signalling contributes to the
pathogenesis of the dentigerous cyst and may promote
growth and cell proliferation. Vered et  al. (2009) exam-
ined the expression of the hedgehog pathway–associated
proteins PTCH, SMO, and Gli1 in keratocysts, orthokerati-
nised odontogenic cysts, dentigerous cysts, and radicular
Figure 5.15  An inflammatory dentigerous cyst. The crown of
cysts. PTCH and Gli1 were expressed strongly in all cyst the developing first premolar is surrounded by a well-­
types; SMO was found in all keratocysts and in 33% of demarcated radiolucency associated with the roots of the
orthokeratinised odontogenic cysts and 40% of dentiger- overlying deciduous teeth.
ous cysts. In a similar study, Zhang et al. (2010) examined
20 dentigerous cysts and found homogeneous expression developing permanent tooth by periapical inflammation
of SHH, PTCH, SMO, and Gli1 in the epithelial lining of from a carious, non-­vital deciduous precursor. Thus, inflam-
all lesions. They also found similar expression in all cases matory dentigerous cysts arise in children, in the mixed
(n = 12) of glandular odontogenic cyst examined. dentition, and most frequently are associated with develop-
These studies suggest that aberrations in the PTCH recep- ing premolars (Figure  5.15). Most cases are single case
tor and activation of the HH signalling pathway are not spe- reports, but some series of inflammatory dentigerous cysts
cific to keratocysts. PTCH gene alterations may represent a have been published (Benn and Altini  1996; Shibata
decisive initiating event in the formation of developmental et al. 2004; Marques et al. 2017; Huang et al. 2019).
odontogenic cysts, possibly in a progenitor epithelial cell, Benn and Altini (1996) reported 15 patients ranging in age
which then gives rise to the entire epithelial lining. The from 5 to 12 years. In 13 cases the cyst was associated with
resultant activation of the hedgehog signalling pathway may periapical inflammation from a non-­vital deciduous prede-
then drive growth and expansion of the cyst. Such a scenario cessor, while in 2 patients there was evidence of osteomyeli-
could explain a role for HH signalling in many odontogenic tis. The mandible was involved in 10 cases and premolars
lesions, including dentigerous cysts, but does not exclude a were the most commonly affected teeth (9 cases), followed
further role for specific PTCH mutations in keratocysts. by canines (4 cases). The mandibular second permanent
Gomes and Gomez (2011) supported this view and sug- molars were involved in the 2 cases associated with osteomy-
gested that PTCH1 may act as a gatekeeper gene for many elitis. Of note is that two-­thirds (10) of the cases presented
types of odontogenic lesions, and that further genetic with pain and swelling. On pathological examination, all the
events drive the formation of different cysts or tumours. cysts were attached to the cervical region of the teeth and
were composed of an inflamed wall lined by non-­keratinised
stratified squamous epithelium, with varying degrees of pro-
Inflammatory Dentigerous Cyst
liferation and hyperplasia. In most cases part of the cyst lin-
As described above, the vast majority of dentigerous cysts ing, adjacent to the tooth, showed a thin layer of cuboidal
are developmental in origin, and although inflammation epithelium, which they interpreted as evidence of an origin
may play a role in growth and enlargement, initiation of the from the reduced enamel epithelium.
epithelial proliferation remains speculative. However, in Shibata et al. (2004) reviewed 70 patients under the age of
some cases there is good evidence that cyst development 16 years who had histologically confirmed dentigerous cysts
may be initiated by inflammation, resulting in a true inflam- that had developed between the central incisors and the sec-
matory dentigerous cyst. The pathogenesis of these cysts ond premolars. Of these cases, 54 (77%) affected the premo-
involves stimulation of the reduced enamel epithelium of a lar teeth. In 7 cases the deciduous teeth had been extracted,
76 Dentigerous Cyst

but of the remainder, 44 (94%) showed radiological evidence unerupted teeth (similar to Figure 5.14). In all cases the cysts
of a periapical lesion and 98% (53 of the 54) of cases were showed typical histological features of an odontogenic
inflamed on histological examination. Marques et al. (2017) keratocyst. In five cases they were able to process the cyst and
reported similar findings and found that 6 of their 12 cases tooth in continuity, and in all five they found a keratocyst lin-
affected the maxillary canines and 6 affected the mandibular ing, but at the point where the cyst joined the cervical region
second premolars. Radiology showed well-­demarcated radi- of the tooth, the keratocyst lining merged with a typical
olucencies ranging from 10 to 27 mm in diameter. All cysts reduced enamel epithelium. They postulated that the cyst
were inflamed and lined by variably hyperplastic non-­ develops from epithelial rests outside of the dental follicle
keratinised stratified squamous epithelium. and that the tooth then erupts into the cyst and the lining
These studies support the concept of an inflammatory merges with the reduced enamel epithelium. In their 1987
aetiology in the pathogenesis of some dentigerous cysts. paper (Altini and Cohen 1987), they demonstrated in animal
Nevertheless, individual cases need to be assessed criti- studies that this mechanism can occur. An earlier study by
cally. Attachment of the cyst wall to the neck of the associ- Browne (1971a) had investigated 139 odontogenic kerato-
ated tooth is an essential feature, and microscopically the cysts and found 56 cases associated with an unerupted tooth.
cyst lining should demonstrate a readily identifiable com- Of these, 8 (14%) were found to have a keratocyst lining that
ponent of reduced enamel epithelium before a diagnosis of was attached at the cementoenamel junction of the tooth.
dentigerous cyst is made. Altini and Cohen (1982) and Browne (1991b) have called
these cysts ‘envelopmental primordial cyst (keratocyst)’
and clearly state that, although they have a dentigerous
‘Extrafollicular Dentigerous Cyst’
relationship with the teeth, they are not dentigerous cysts.
In 1958, Gillette and Weinmann (1958) reported two cysts They are odontogenic keratocysts and should be managed
that enveloped the crowns of unerupted teeth and on radi- as such. A similar mechanism may give rise to other lesions
ology were seen to be in a ‘dentigerous relationship’. They being in a dentigerous relationship, including cystic amelo-
proposed that an alternative mechanism for the pathogen- blastoma, orthokeratinised odontogenic cyst, adenomatoid
esis of dentigerous cysts is that they may arise from epithe- odontogenic tumour, and calcifying odontogenic cyst.
lium outside of the dental follicle (presumably dental With regard to the inflammatory dentigerous cyst dis-
lamina rests) and then envelop the tooth. They introduced cussed above, consideration must also be given to the pos-
the term ‘extrafollicular dentigerous cyst’. Since their paper, sibility that these arise due to the crown of a permanent
it has often been reported that a dentigerous cyst may have tooth erupting into a radicular cyst formed at the apex of its
an extrafollicular origin. However, examination of their deciduous predecessor. This phenomenon possibly does
paper reveals that both of their cysts show the histological occur, but only exceptionally rarely, because radicular cysts
features of what we now recognise as odontogenic kerato- involving the deciduous dentition are so uncommon
cyst, and both were separated from the tooth follicle (and (Lustmann and Shear 1985). In such a case, the erupting
reduced enamel epithelium) by a fibrous cyst wall. As dis- tooth may indent rather than penetrate the wall of the
cussed previously (see ‘Differential Diagnosis’; Figure 5.14), radicular cyst and this should be apparent histologically, if
a number of lesions, including odontogenic keratocysts, not macroscopically (Gebhardt and Lenz  1985; Wood
may arise from within the dental crypt and embrace an et al. 1988). Figure 5.16 illustrates how this might occur.
unerupted tooth in a dentigerous relationship. Therefore,
there is no evidence for an extrafollicular origin for denti-
Dentigerous Cyst as a
gerous cysts.
Potential Ameloblastoma
In this context it is worth reviewing Browne’s definition
of a dentigerous cyst (Browne 1991b) as a cyst that ‘by defi- A simple search of the literature will show many case
nition is one which encloses the crown of an unerupted reports suggesting that dentigerous cysts are a precursor
tooth lying within bone and cannot be assigned to any of various neoplasms, usually ameloblastoma. Ameloblas­
other classification’. Thus the criteria for diagnosis must tomas are of odontogenic epithelial origin and therefore it
include a consideration of the histological features (see is possible that they may arise from reduced enamel epi-
‘Histopathology’ and Box 5.4) and if these show a defined thelium or a dentigerous cyst lining, as well as any other
lesion, such as a keratocyst or ameloblastoma, then the odontogenic epithelium. However, the belief that it com-
lesion is not a dentigerous cyst. monly arises in this situation and that the dentigerous cyst
A nice historical discussion of this is given by Altini and should be regarded as pre-­ameloblastomatous should be
Cohen (1982,  1987), who reported 17 cases of odontogenic viewed with caution. Much of the confusion has probably
keratocyst that presented in a dentigerous relationship with arisen for three reasons. First, an ameloblastoma (like an
­Pathogenesi  77

(a) (b)

Figure 5.16  (a) A radicular cyst associated with a deciduous mandibular second molar appears to be in a dentigerous relationship
with the erupting premolar. Source: Courtesy of Dr J. Lustmann. (b) A diagrammatic representation of the probable relationship, where
the erupting tooth has indented the radicular cyst wall.

Figure 5.17  Radiograph of a unilocular


ameloblastoma that appears to be in a
dentigerous relationship with unerupted teeth.

odontogenic keratocyst) may envelop an unerupted tooth, expanded locule lined by a thin layer of epithelium. If the
particularly a third molar at the angle of the mandible, surgeon’s provisional diagnosis is dentigerous cyst because
and this may be incorrectly interpreted as a dentigerous of the radiological picture, the pathologist may well regard
cyst on radiographs (Figure 5.17). When subsequently the such histological features as consistent with this diagnosis.
lesion is removed and diagnosed histologically as an When the tumour is removed entirely and a diagnosis of
ameloblastoma, the erroneous conclusion may be reached ameloblastoma is made, once again this may be misinter-
that the ameloblastoma developed from a dentigerous preted as having developed from a dentigerous cyst.
cyst. In this situation, it is considered that the ameloblas- A third reason may be misinterpretation of epithelial
toma arises from dental lamina rests within the crypt of islands within a cyst wall. Isolated islets or follicles of
the developing tooth and envelops the tooth crown in a odontogenic epithelium are sometimes found in the wall of
dentigerous relationship. a dentigerous cyst some distance from the epithelial lining.
The second possible reason for believing that many If prominent or enlarged, these may be interpreted as
ameloblastomas develop from a dentigerous cyst is that ameloblastoma, although they bear only a superficial
biopsies of a radiolucent lesion may be taken of a superficial resemblance to the tumour.
78 Dentigerous Cyst

­Histopathology tooth at the cementoenamel junction. Thus, the lumen of


the cyst is partially lined by the enamel of the tooth.
Pathologists very rarely receive a specimen where the cyst is If the associated tooth is decalcified with the cyst
intact and often may not receive the associated tooth. attached, histology will show that the cyst is attached at the
Usually the thin cyst wall is torn or fragmented during the cementoenamel junction of the tooth (Figure 5.19). Typical
surgical procedure. Nevertheless, pathologists should per- histology shows a thin, fibrous cyst wall that, being derived
form a careful dissection of the gross specimen and care- from dental follicle, consists of fine stellate fibroblasts
fully examine the associated tooth to determine that the widely separated by stroma and ground substance to give a
cyst surrounds the crown of the tooth, and is attached at the slightly myxoid appearance (Figures  5.20 and  5.21). The
cementoenamel junction. Figure 5.18 shows an example of epithelial lining is usually thin, non-­keratinised stratified
a dentigerous cyst attached to the cervical region of the squamous epithelium about two to five cell layers thick
(Figure 5.20). This is derived from the reduced enamel epi-
thelium and occasionally, especially in small cysts, a sim-
ple bilayer of cuboidal cells resembling reduced enamel
epithelium may be seen (Figure 5.21).
Although a typical developmental cyst is uninflamed, as
discussed previously inflammation may play a role in cyst
growth and occasional inflammatory cells are seen in the
wall. Often, however, by the time they present clinically,
dentigerous cysts have become traumatised and areas of
heavy inflammation are seen. This is often associated with
epithelial proliferation and the cyst may then resemble, or
be indistinguishable from, a radicular cyst (Figure  5.22).
Similar to radicular cysts, inflammation may be accompa-
nied by accumulation of granulation tissue and cholesterol
clefts are not uncommon. In most analyses about 70–80%
of dentigerous cysts show evidence of inflammation in the
wall (Lin et al. 2013; Huang et al. 2019) and this is more
common and intense in paediatric cases, where an inflam-
matory origin may be apparent (Benn and Altini  1996;
Shibata et al. 2004; Huang et al. 2019). In their analysis of
338 dentigerous cysts, Lin et al. (2013) found that 71% of
Figure 5.18  A dentigerous cyst and the associated tooth, in dentigerous cysts were inflamed, 26.6% contained choles-
this case a canine. The cyst is attached to the cervical region at terol clefts, and 30% had foamy or hemosiderin-­laden
the cementoenamel junction. macrophages.

Figure 5.19  A low-­power view of a decalcified section of a dentigerous cyst and its associated tooth. The cyst envelops the crown
and is attached to the tooth at the cementoenamel junction. Source: Courtesy of Prof. Paul Speight (Previously published: El-Naggar AK
2017 / Courtesy of IARC).
­Histopatholog  79

Figure 5.20  The wall of a dentigerous cyst lined by a thin epithelium of two to four layers of undifferentiated cells derived from the
reduced enamel epithelium. The fibrous cyst wall is relatively uninflamed and sparsely cellular.

Figure 5.21  The wall of a dentigerous cyst, composed of loose myxoid fibrous connective tissue lined by two-­layered cuboidal
epithelium, resembling the reduced enamel epithelium from which it is derived.

Figure 5.22  An inflamed region of a dentigerous cyst wall, lined by hyperplastic epithelium. The features are indistinguishable from
those seen in inflammatory odontogenic cysts.
80 Dentigerous Cyst

Figure 5.23  A portion of the lining of a dentigerous cyst shows mucous metaplasia.

In some cysts, part of the epithelial lining may contain


Immunohistochemistry and Biomarker Studies
mucous-­producing cells (Figure 5.23). Browne (1972) found
them in 36% of mandibular and 53% of maxillary dentiger- Numerous immunohistochemical studies have been
ous cysts, and made the interesting observation that the fre- undertaken to compare the expression of cytokeratins (CK)
quency of such mucous cells increased in proportion to the and other markers in dentigerous cyst epithelium with that
age of the patient. In a sample of 130 dentigerous cysts, of the odontogenic keratocyst, radicular cyst, and amelo-
Takeda et al. (2005) found mucous cells in the linings of 31 blastoma. The purpose of these studies is not always well
(23.8%) cysts and ciliated cells in 14 (10.8%), indicating that articulated, but overall they represent attempts to shed
both can be present together. It has been thought that light on the pathogenesis or to assist in diagnosis. In par-
mucous and cilia are more likely to be found in maxillary ticular, many recent studies have used markers in an
cysts, and may be associated with proximity to the epithelial attempt to highlight differences between dentigerous cysts
lining of the maxillary antrum. Takeda et al. (2005), how- and keratocysts, with a view to establishing the neoplastic
ever, found that ciliated cells were more frequently encoun- nature of the keratocyst. This is discussed in Chapter 7.
tered in the mandible, but overall there were no significant With regard to diagnosis, the clinical, radiological, and
differences between the mandible and maxilla. histological features of the individual cyst types are char-
Nests, islands, and strands of odontogenic epithelium acteristic, and if strict diagnostic criteria are followed
are often seen in the fibrous cyst wall. These are almost diagnosis is rarely a problem (Box 5.4). A particular diag-
certainly epithelial remnants of the dental lamina and nostic challenge may arise when a pathologist is faced
have been reported in about 20–25% of cases (Lin with a small biopsy from a large radiolucent lesion, and in
et al. 2013; Huang et al. 2019). Wright (1979) reported the this situation a diagnostic marker may be useful.
presence of larger epithelial proliferations that resem- Furthermore, if the cyst lining is inflamed, then all cyst
bled squamous odontogenic tumour. In a review of the types (as well as a cystic ameloblastoma) may show identi-
literature, Chrcanovic and Gomez (2018b) found reports cal features of a proliferative or hyperplastic epithelial lin-
of 60 cases of squamous odontogenic tumour–like prolif- ing and therefore be indistinguishable from each other.
erations in odontogenic cysts. Most were seen in radicu- Overall, however, immunocytochemistry appears to have
lar cysts (51 cases; 85.0%), but 6 dentigerous cysts with a limited role in the diagnosis of odontogenic lesions
squamous odontogenic tumour–like proliferations had (Hunter and Speight 2014).
been reported. Bhakhar et al. (2016) investigated the expression of CK18
Another rare example of metaplasia in dentigerous cysts and CK19  in dentigerous cysts, odontogenic keratocysts,
is the occasional presence of sebaceous glands in their and radicular cysts, and found that while more dentigerous
walls (Chi et al. 2007). Hyaline bodies (Rushton 1955) are cysts than keratocysts expressed both cytokeratins, expres-
also sometimes seen (Lin et  al.  2013). Takeda and sion in radicular cysts was the same as in dentigerous cysts.
Yamamoto (2001) have reported a case of a dentigerous These authors also reviewed 5 studies that had investigated
cyst containing granules of melanin pigment and dendritic CK18, and 17 studies that had investigated CK19. These
melanocytes in the basal cells of the lining epithelium. showed either no differences between the different cyst
­Histopatholog  81

Box 5.4  Dentigerous Cyst: Criteria for Diagnosis


●● A correct diagnosis is facilitated by careful correlation of clinical, radiological, and histological features. The patholo-
gist should review the radiographs or the radiologist’s report to confirm the relationship of the cyst to the crown of
an unerupted tooth
●● Dentigerous cysts always present as a radiolucency around the crown of an unerupted or impacted tooth
●● The radiolucency, or pericoronal space, is greater than 5 mm, although a space of 10 mm or greater is most likely to
be a dentigerous cyst
●● A cystic cavity or lumen can be demonstrated at surgery or on macroscopic examination
●● The cyst lining is attach to the cervical region of the tooth at the cementoenamel junction. This can be seen on
macroscopic examination or microscopically if the tooth and cyst are processed together
●● On histological examination the cyst is lined by thin, regular non-­keratinised stratified epithelium. Areas of bilayered
cuboidal epithelium resembling reduced enamel epithelium may be seen
●● Inflammation is often present
●● A lining showing features of an odontogenic keratocyst or ameloblastoma excludes a diagnosis of dentigerous cyst,
even if the lining is attached to the tooth. These would be ‘envelopmental’ lesions

types or wide variability within or between the studies. cysts, and 20 dentigerous cysts, all of which were nega-
This degree of variability suggests that the use of cytokerat- tive for calretinin, compared to 81.5% of unicystic amelo-
ins as a diagnostic test on a single isolated case would be blastomas that had been shown to be positive in their
inappropriate and grossly inaccurate. previous study (Altini et al. 2000). Subsequently, a num-
It is also notable that in most studies the specimens are ber of studies have confirmed that odontogenic cysts,
chosen to show the typical features of each cyst type, in including dentigerous cysts, are negative for calretinin,
seeming disregard of the fact that if the features are typical, but that ameloblastomas (including unicystic ameloblas-
then there is no requirement for staining beyond a good toma) are positive in up to 100% of cases (De Villiers
haematoxylin and eosin (H&E) stain to differentiate et al. 2008; Jeyaraj 2019; Rudraraju et al. 2019).
between the lesions most commonly encountered around
unerupted teeth (Müller 2021). Few studies have been able
Malignant Change in Dentigerous Cysts
to identify markers that may differentiate between the his-
tologically identical features of an inflammatory cyst and Intraosseous squamous cell carcinoma is a central jaw car-
an inflamed developmental cyst  –  this is because when cinoma that arises from odontogenic epithelium (Koutlas
inflamed, the histology and CK expression are the same. and Sloan  2022). Although these may arise from odonto-
Many studies have also investigated proliferation mark- genic epithelial rests, the majority probably arise from the
ers, but rarely for diagnostic purposes. Rather, these stud- epithelium of odontogenic cysts. Overall they are exceed-
ies have sought to compare the proliferative potential of ingly rare, but they most commonly arise in radicular cysts
the lining of the odontogenic keratocyst to other cyst types followed by dentigerous cysts and then odontogenic kerato-
(reviewed by Shear 2002b; Kichi et al. 2005). Almost with- cysts. However, this does not reflect a particular propensity
out exception, studies have shown that keratocyst linings to malignant change within the epithelium of the different
have a greater proliferation rate than dentigerous or radic- cyst types, but rather their relative frequency.
ular cysts, although they may be similar to cystic amelo- Bodner et  al. (2011) reviewed the literature and found
blastomas. These studies support the role of mural growth reports of 116  intraosseous carcinomas that had arisen in
in the expansion of keratocysts, but add little to our under- odontogenic cysts between 1938 and 2010. Of these, 19
standing of the diagnosis or pathogenesis of dentiger- cases (16%) arose in a dentigerous and 70 (60%) in a radicu-
ous cysts. lar cyst, and 16 (14%) in a keratocyst. Well or moderately
An exception to this discussion may be the expression differentiated squamous cell carcinomas represented 85%
of calretinin, which in the context of odontogenic lesions of lesions, and there were no differences in lesions from dif-
appears to be specific for ameloblastomas and is there- ferent cyst types. In their systematic review, Borrás-­Ferreres
fore useful in the differential diagnosis of cystic lesions, et al. (2016) identified 48 reports of carcinomas arising in
especially in small biopsies (Coleman et  al.  2001; De odontogenic cysts, but they excluded keratocysts (which
Villiers et al. 2008; Hunter and Speight 2014). Coleman they defined as benign tumours, keratocystic odontogenic
et al. (2001) studied a series of 22 keratocysts, 26 residual tumour or KCOT). They identified 53 cases of which 27
82 Dentigerous Cyst

(51%) arose in dentigerous cysts, 4  in radicular cysts, and occlusion (Bodner 2002; Motamedi and Talesh 2005; Hauer
10 in residual cysts. They found 6 cases in orthokeratinised et  al.  2020). Marsupialisation or decompression may be
odontogenic cysts. In both series, males accounted for used when attempting to preserve the teeth (Koca et al. 2009;
60–70% of the cases, and the average age was about 60 years, Hyomoto et al. 2003). In their series, Hyomoto et al. (2003)
a decade or two older than the average age for benign cysts. performed a retrospective investigation into the eruption
of teeth associated with dentigerous cysts involving
47 mandibular premolars and 11 maxillary canines in pre-­
­Treatment adolescent children. In one group, 81% of the mandibular
premolars and 36% of the maxillary canines erupted suc-
The mainstay of treatment of the dentigerous cyst is enu- cessfully about 100 days after marsupialisation without
cleation of the cyst with or without removal of the involved traction. Koca et al. (2009) successfully treated 35 children
tooth. In the case of impacted third molars, the tooth is usu- with marsupialisation followed by eruption of the associ-
ally extracted, but when canines or premolars are involved ated tooth. The eruption potential of the teeth was closely
in children, the emphasis is on conservative enucleation, related to root formation, so that teeth with incomplete root
often combined with orthodontics, in order to retain the formation had good potential to erupt, whereas those with
involved tooth and to ensure eruption into normal fully formed roots may not.
83

Eruption Cyst

CHAPTER MENU
­Clinical Features, 83
●● Frequency, 83
●● Age, 83
●● Sex, 84
●● Site, 84
●● Clinical Presentation,  84
­Radiological Features, 84
­Pathogenesis, 85
­Histopathology, 85
­Treatment, 86

An eruption cyst is a variant of dentigerous cyst occurring in It is likely that eruption cysts are seen much more fre-
the soft tissues overlying an erupting tooth. Whereas the den- quently clinically and because many rupture spontane-
tigerous cyst develops around the crown of an unerupted or ously, they are not excised and are therefore not submitted
impacted tooth within the bone, the eruption cyst is associ- for histological examination. Bodner (2002) reported a
ated with a tooth that might otherwise erupt normally. series of 69 paediatric patients with cystic lesions of the
jaws, diagnosed clinically as well as on histology, and found
that 15 (22%) were eruption cysts. In their large series of 66
­Clinical Features cases, Şen-­Tunç et  al. (2017) showed that only 10 (15%)
required surgical intervention. This suggests that the true
Frequency prevalence of eruption cysts might be 10 times, or more,
higher than that reported from pathology series.
Eruption cysts are usually treated conservatively and tissue
is rarely sent for histological examination. Since most
series of cysts are derived from pathology departments, the
Age
reported frequency and prevalence appear, falsely, to be
low. Tables 1.1 and 1.2 show that in our series, eruption Eruption cysts are found in children and rarely in adults.
cysts comprised only 0.8% and 0.2% of cysts, respectively. In the three largest series reported, the mean ages of the
Of those studies shown in Table 5.1 that reported eruption patients affected were 6.7 years (n  =  27; range 13 months
cysts, the frequency ranged from 0.1% to 1.0% of odonto- to  11 years; Aguilo et  al.  1998), 4.4 years (n  =  24; range
genic cysts, but all studies only reported cases that had 1 month to 12 years; Bodner et  al.  2005), and 5.4 years
been ­submitted for histological examination (Daley and (n = 53; range 5 months to 11 years; Şen-­Tunç et al. 2017).
Wysocki 1995; Ledesma-­Montes et  al.  2000; Jones Occasional cases have been reported in adults, usually
et al. 2006; Ochsenius et al. 2007; Sharifian and Khalili 2011; associated with teeth that had been impacted but erupted
Soluk Tekkeşın et  al.  2012b; Lo Muzio et  al.  2017; late. Seward (1973) recorded a case in a 21-­year-­old and
Tamiolakis et al. 2019). Woldenberg et  al. (2004) documented an unusual case

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
84 Eruption Cyst

that occurred in a 40-­year-­old woman associated with an 10 of which were in the primary dentition and were sym-
impacted canine erupting ectopically in the buccal mucosa. metrically bilateral. Kimura et al. (2014) described a child
Eruption cysts have also been reported at birth, either who had had six eruption cysts. The first two presented
associated with natal teeth (Bodner 2002) or presenting as around mandibular central incisors at 7 months old, and
a swelling in the anterior mandible associated with devel- regressed spontaneously. At 18 months the child devel-
oping deciduous incisors (de Oliveira et al. 2018). oped four cysts simultaneously on all four deciduous
first molars.
Sex
Clinical Presentation
In almost all series, boys have been affected more often
than girls. In the three large series the proportions of males The eruption cyst produces a smooth swelling over the
were 55.6% (Aguilo et al. 1998), 66.6% (Bodner et al. 2005), erupting tooth, which in most cases is blue or translucent
and 58.4% ­(Şen-­Tunç et al. 2017). and blueish (Figure 6.1). Occasional cases are the colour
of normal gingiva or clear. It is usually painless unless
infected and is soft and fluctuant. In Şen-­Tunç et  al.’s
Site
(2017) series of 53 patients, only 7 (13.2%) reported symp-
Deciduous and permanent teeth may be involved, most fre- toms and these comprised dull aching pain, usually appar-
quently anterior to the first permanent molar. Aguilo et al. ent during eating.
(1998) found that over 90% of their cases arose in the per- There is often a brief history of about three to four
manent dentition and 80.6% were in the maxilla. This is weeks’ duration during which they enlarge to approxi-
unusual, however, and other large series and the majority mately 1–1.5 cm. They are usually exposed to masticatory
of case reports have shown an almost equal distribution trauma, which causes haemorrhage and the blue colour,
between the primary and permanent dentition. Bodner and are sometimes referred to clinically as an ‘eruption
et  al. (2005) and Şen-­Tunç et  al. (2017) found 50% and haematoma’ (Figure  6.1). The majority of cases resolve
58.5%, respectively, in the permanent dentition. The teeth spontaneously or are ruptured during mastication and
most frequently involved in the permanent dentition have heal uneventfully.
been maxillary central incisors and first molars, and in the
primary dentition, maxillary first molars and maxillary or
mandibular incisors (Aguilo et al. 1998; Bodner et al. 2005; ­Radiological Features
Şen-­Tunç et al. 2017).
Multiple eruption cysts are not unusual (Figure  6.1). The cyst may throw a soft-­tissue shadow, but there is usu-
Aguilo et al. (1998) found two or more eruption cysts in 6 ally no bone involvement, except that the dilated and open
of their 27 patients, and in 3 of these the lesions were crypt may be seen on the radiograph.
bilateral, symmetrical, and concurrent. Şen-­Tunç et  al.
(2017) found multiple cysts in 13 of their patients (24.5%),
Box 6.1  Eruption Cyst: Key Facts
●● Eruption cysts may constitute up to 10% of odonto-
genic cysts
●● The clinical features are typical, and radiology and
histopathology play only a minor role in diagnosis
●● They arise in children, with most seen between
5 and 7 years
●● About 60% are found in males, with a M : F ratio of
about 2 : 1
●● Deciduous teeth and permanent teeth are almost
equally affected
●● In the primary dentition incisors and maxillary first
molars are most often affected
●● In the permanent dentition first molars and maxillary
central incisors are most often affected
Figure 6.1  Eruption cysts involving both maxillary permanent ●● Most lesions present as soft, fluctuant bluish swellings
incisors.
­Histopatholog 85

Figure 6.2  Histological features of an


eruption cyst. The surface epithelium is
at the top right and the cyst lumen at the
bottom of the photomicrograph. The cyst
lining is thin and has become fragmented
(arrows). Focal infiltrates of chronic
inflammatory cells are seen in the wall.

­Pathogenesis general aetiological role in eruption cyst formation, but are


important to note as a possible side effect when cyclosporine
The pathogenesis of the eruption cyst is similar to that of medication is being considered. Cyclosporine is known to
the dentigerous cyst. The difference is that tooth eruption cause gingival fibrosis and it may be that this impedes erup-
in the case of the eruption cyst is impeded in the soft tis- tion and has a role in cyst formation.
sues of the gingiva rather than in the bone. As Browne and
Smith (1991) have argued, ‘as the crown of the tooth associ-
ated with an eruption cyst usually projects into the cyst ­Histopathology
lumen, it is widely believed that the epithelium is derived
from the reduced enamel epithelium, as in the dentigerous There are no specific histopathological features of the
cyst’. The factors that actually impede eruption in the soft eruption cyst and histological examination plays only a
tissues are not known. It is possible that cyst formation minor role in diagnosis. Few cysts are actually biopsied and
may be associated with trauma to the overlying alveolus as when tissue is made available it is usually as a result of
a result of mastication. Trauma may cause gingival fibrosis, surgical treatment, which involves marsupialisation. The
and any associated inflammation might initiate epithelial pathologist receives only the superficial part of the cyst
proliferation and cyst formation, as discussed for dentiger- wall (Figure 6.2). This is covered by the keratinised, strati-
ous cyst in Chapter 5. fied squamous epithelium of the overlying gingiva, sepa-
Interestingly, there is some evidence that fibrosis may rated from the cyst by a strip of dense connective tissue of
have a role, by showing that the administration of cyclo- varying thickness, which usually shows a chronic inflam-
sporine may lead to the development of eruption cysts. The matory cell infiltrate. Sometimes it is possible to distin-
first article (O’Hara et al. 2002) reported the development of guish a line of demarcation between gingival and follicular
multiple eruption cysts in neonatal dogs given oral cyclo- connective tissues. The gingival connective tissue is rela-
sporine as an immunosuppressant, as part of an investiga- tively acellular and densely collagenous, while the follicu-
tion of the efficacy of adenoviral-­mediated gene therapy in lar connective tissue is more cellular, less collagenous, and
a canine model of Duchenne muscular dystrophy. The cysts may be myxoid with a basophilic hue.
resolved within one month when cyclosporine administra- The epithelial lining of the cyst is of reduced enamel epi-
tion was discontinued. In the second report (Kuczek thelial origin and characteristically consists of two to three
et al. 2003), eruption cysts developed in a boy treated with cell layers of squamous epithelium, with a few foci where it
cyclosporine A following a heart transplant. The child was may be a little thicker. Invariably, however, the epithelial
given periodontal treatment and the cyclosporine A was lining is fragmented (Figure  6.2). If the cyst is heavily
switched to tacrolimus, following which no further erup- inflamed, the epithelial lining may be proliferative and
tion cysts developed. These findings cannot impute any hyperplastic.
86 Eruption Cyst

­Treatment (2005), 12  were treated by marsupialisation, and 10


resolved without treatment. In two cases, involved neona-
Eruption cysts should be treated conservatively and in tal teeth were extracted. Şen-­Tunç et al. (2017) reported 66
many cases only reassurance is needed. The cysts often cysts, of which 56 (85%) were treated with no intervention
rupture or may resolve spontaneously and the involved except for advice to ‘massage’ the area. In all cases the
tooth then erupts normally. When surgical intervention is teeth erupted normally between 15 days and 5 months
required, then simple marsupialisation with removal of later. The other 10 cysts were associated with symptoms
the ‘top’ of the cyst almost always has a satisfactory out- and were treated surgically, and in all cases the teeth
come. In the series of 24 cases reported by Bodner et  al. erupted within a few weeks.
87

Odontogenic Keratocyst

CHAPTER MENU
Classification and Terminology: A Brief Historical Review, 88
Clinical Features, 89
•• Frequency, 89
•• Age, 92
•• Sex, 93
•• Site, 93
•• Clinical Presentation,  95
•• Peripheral (Extraosseous) and Soft-­Tissue Keratocysts,  96
–– Peripheral Odontogenic Keratocyst,  96
–– Soft-­Tissue Keratocyst,  96
Naevoid Basal Cell Carcinoma Syndrome (NBCCS), 98
Radiological Features, 100
•• Computerised Tomography and Magnetic Resonance Imaging,  103
•• Radiological Differential Diagnosis,  104
Pathogenesis, 106
•• Source of Epithelium,  106
–– Rest Cells of Dental Lamina,  106
–– Epithelial Islands Derived from Oral Epithelium,  107
–– Summary and Conclusions,  108
•• Hedgehog Signalling Pathway, PTCH Gene Alterations, and Odontogenic Keratocyst,  108
•• Is the Odontogenic Keratocyst a Neoplasm?,  111
–– Summary and Conclusions,  113
•• Growth and Enlargement of the Odontogenic Keratocyst,  113
Histopathology, 117
•• Odontogenic Keratocysts Associated with Naevoid Basal Cell Carcinoma Syndrome,  124
•• Histological Differential Diagnosis,  125
•• Solid Odontogenic Keratocyst,  125
•• Cytology and Aspiration Biopsy,  129
•• Immunohistochemistry and Biomarker Studies,  131
•• Malignant Change in Odontogenic Keratocysts,  132
Recurrence of Odontogenic Keratocyst, 133
•• Factors Associated with Recurrence,  134
–– Clinical and Radiological Factors,  134
–– Histological Factors,  135
Treatment and Recurrence of Odontogenic Keratocyst, 136
•• Treatment Methods,  136
–– Enucleation and Curettage,  136
–– Marsupialisation and Decompression,  137
–– Resection, 137
–– Adjunctive Methods,  137
•• Evaluation of Treatment Methods,  138
–– Summary and Conclusions,  138

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
88 Odontogenic Keratocyst

The odontogenic keratocyst has aroused more interest and (Browne  1970,  1971a). A number of studies also demon-
debate than any other cyst of the jaws. This is partly because strated that the site distribution of keratinising cysts dif-
it has a particular tendency to recur following surgical fered from that of non-­keratinised cysts (Browne  1970;
treatment, and because it may grow to a large size before it Hjørting-­Hansen et al. 1969; Rud and Pindborg 1969), sup-
manifests clinically. More recently, however, interest has porting the view that the odontogenic keratocyst was a
been fuelled by the debate regarding terminology and the separate and distinct entity.
possible neoplastic nature of the lesion. An internet search Before Philipsen introduced ‘odontogenic keratocyst’ in
of publication numbers showed that between 1963 and 1956, a variety of terms had been used for keratinising cysts
2006 there was an average of 10 publications per year with (Pogrel 2003a; Ide et al. 2020), including the term primor-
odontogenic keratocyst in the title. Since the last edition of dial cyst, which was suggested by Robinson in 1945 when
this book (2007) there have been more than 750 papers he proposed a comprehensive classification of cysts of the
with either odontogenic keratocyst or ‘keratocystic odonto- jaws (Robinson 1945). He suggested that these cysts were
genic tumour (KCOT)’ in the title, and the average number ‘formed through degeneration of the stellate reticulum in
of publications per year rose almost fivefold to 47. This enamel organs before any calcified structures had been laid
chapter will start with a brief review of terminology and down’. In other words, they developed from the tooth ‘pri-
will later include a discussion on the evidence that the mordium’, replaced a developing tooth, and clinically
odontogenic keratocyst may be a benign cystic neoplasm. would present at a site where a tooth was absent, or at the
site of a putative supernumerary tooth. Robinson did not
however describe any distinctive histological features and
­ lassification and Terminology:
C regarded the primordial cyst and dentigerous cyst as both
A Brief Historical Review arising from the dental lamina, but before and after hard
tissue formation, respectively. Over time it became appar-
It is well established that any epithelial-­lined cyst of the ent that if primordial cysts (cysts found to have replaced a
jaws may, on occasion and often as a result of metaplasia, developing tooth) were examined as a group, the vast
show some evidence of keratinisation of the lining. In the majority were found to be parakeratinised, with cuboidal
early literature there was no real distinction between what or columnar palisaded basal cells (Shear  1960a; Forssell
we now know as odontogenic keratocyst and any jaw cyst and Sainio 1979). These authors suggested that these were
with a keratinised epithelial lining. ‘genuine keratocysts’ and that cysts lined by parakerati-
Pogrel (2003a) and Ide et al. (2020) have presented his- nised epithelium should not be regarded as ‘primordial
torical reviews of the odontogenic keratocyst and described cysts’. The studies of Browne (1970,  1971a) showed that
how early researchers recognised keratinising cysts in the dentigerous cysts and ‘primordial cysts’ were clinically and
jaws, but often used terms borrowed from other anatomical pathologically distinguishable as separate entities, and that
sites such as cholesteatoma or dermoid cyst. Ide et  al. the latter were characterised by a wholly parakeratinised
(2020) traced the first description of keratocysts to 1855, lining and a higher recurrence rate. He proposed that ‘pri-
when they were referred to as ‘buttery cysts’ by mordial cyst’ should be abandoned in favour of the more
Maissonneuve, the term ‘buttery’ referring to the thick appropriately descriptive term ‘odontogenic keratocyst’. It
keratinaceous contents that resemble butter. The term thus became accepted that ‘primordial cyst’ and ‘odonto-
‘odontogenic keratocyst’ was introduced by Philipsen genic keratocyst’ were in fact the same cystic entity charac-
(1956), but in this and a subsequent paper (Pindborg terised by a parakeratinised epithelial lining. In 1971, the
et al. 1962) the term was still used to describe any jaw cyst first edition of the World Health Organization (WHO) clas-
in which keratin was formed to a large extent. Some denti- sification of odontogenic cysts described odontogenic
gerous, radicular, and residual cysts were therefore keratocyst and primordial cyst as synonymous and defined
included in the category of odontogenic keratocyst. Browne it as being characterised by ‘a thin fibrous capsule and a
(1971a) pointed out that although a few radicular and lining of keratinized stratified squamous epithelium rarely
residual cyst linings may become keratinised by metapla- exceeding about five cells in thickness and having no rete
sia, the pattern of keratinisation and the histology of the processes’ (Pindborg and Kramer 1971). The term primor-
linings was distinctly different from the characteristic par- dial cyst (as a synonym for odontogenic keratocyst) gradu-
akeratinised lining epithelium of the odontogenic kerato- ally fell out of favour and last appeared in the literature in
cyst. In early studies he showed that the parakeratinised 1987 (Partridge and Towers 1987). It may, however, still be
cysts occurred at a significantly younger age than other found as a synonym for odontogenic keratocyst in some
cysts, making it unlikely that they may have arisen by textbooks and also appeared in the second and third edi-
metaplasia in long-­standing dentigerous or radicular cysts tions of the WHO classification (Kramer et  al.  1992;
­Clinical Feature  89

Philipsen  2005). Nevertheless, it does not appear in the Li  2022; WHO  2022a). This is not without controversy
fourth or fifth editions of the WHO classification (Speight (Stoelinga 2019), but the name does not deny the possible
et al. 2017a; Wright and Li 2022), and the term primordial neoplastic nature of the odontogenic keratocyst, and man-
cyst should no longer be used as a synonym for odonto- agement decisions should be based on an understanding of
genic keratocyst. the pathobiology of the lesion, not on the name alone.
Yet it should be noted that ‘primordial cyst’ is still occa- Finally, previous definitions of the odontogenic kerato-
sionally used for other categories of odontogenic cyst. In cyst (Wright  1981; Kramer et  al.  1992) have included an
the 2012 Armed Forces Institute of Pathology (AFIP) fasci- orthokeratinised variant. However, it has long been recog-
cle (Robinson and Vincent  2012), a primordial cyst was nised that this ‘variant’ showed different and characteristic
defined as an ‘odontogenic cyst of undetermined origin’ clinical and pathological features. In the 2005  WHO
and is described as being a non-­keratinised cyst that forms classification, it was clearly stated that cysts lined by
in place of a developing tooth. They assert that a missing orthokeratinised epithelium were not part of the spectrum
tooth is not needed for the diagnosis, since the definition of the ‘KCOT’ (Philipsen 2005), and in 2017 the orthokerati-
includes supernumerary teeth. They suggest that it is a nised odontogenic cyst was included as a separate entity in
diagnosis of exclusion and should be used when a ‘develop- the classification of odontogenic cysts (Speight et al. 2017b;
mental cyst’ cannot be identified as any other defined Speight and Takata  2018). The orthokeratinised odonto-
cystic entity. More recently, Argyris et al. (2016) suggested genic cyst is discussed in Chapter 12.
the term primordial cyst for an unusual cystic lesion that
had developed at the site of a congenitally missing second
molar in an 18-­year-­old. This cyst showed evidence of ­Clinical Features
inductive change and hard tissue formation, suggesting
that it may in fact be an unusual hamartoma or odontoma. Most descriptions of the odontogenic keratocyst are to be
A further change in terminology was initiated by the found in case reports and small case series. In a systematic
debate regarding the possible neoplastic nature of the review, MacDonald-­Jankowski (2011) found a total of 151
odontogenic keratocyst. This is discussed later in this chap- case series of odontogenic keratocyst between 1976 and
ter. However, some regarded the evidence that the odonto- 2010. In his review, he excluded incomplete descriptions,
genic keratocyst is a neoplasm to be so compelling that a referred cases and repetitions, and finally reviewed only 49
change of name was warranted. Thus, in the third edition papers that reported 4795 cysts. The case series ranged in
of the WHO classification (Philipsen 2005), the term odon- size from 3 to 588 lesions. In his analysis of these papers, he
togenic keratocyst was replaced by ‘keratocystic odonto- also excluded orthokeratinised cysts and cysts associated
genic tumour (KCOT)’. This change was made on the basis with the naevoid basal cell carcinoma syndrome (NBCCS).
that the odontogenic keratocyst ‘behaves as a neoplasm’ Subsequently there have been a number of large case
and may show genetic changes (primarily in the PTCH series, including a multicentre study of 2497 cases reported
gene) that are associated with the naevoid basal cell carci- from Brazil (Schuch et al. 2020), Selected case series with a
noma syndrome and with neoplasia at other sites. Although broad geographical spread and MacDonald-­Jankowski’s
the possibility that the odontogenic keratocyst is a neo- review are summarised in Tables 7.1 and 7.2.
plasm had been long debated, this change in terminology
was controversial and resulted in widespread debate,
Frequency
which is reflected in the huge increase in publications
mentioned previously. However, KCOT was not adopted The odontogenic keratocyst is the third most common cyst
worldwide and the literature continued to be dominated by of the jaws, representing 10.2% of all jaw cysts in our South
the term odontogenic keratocyst (Bhargava et  al.  2012). African series (Table 1.1) and 11.6% of odontogenic cysts in
Many felt that the evidence of neoplasia was insufficient to the Sheffield study (Table  1.2; Jones et  al.  2006). Studies
warrant a change in designation. Also, the term odonto- from other countries, however, show a wide variation
genic keratocyst is well established and is so widely used by (Table 1.3), with a range from only 1.3% in Sicily (Tortorici
clinicians and pathologists that a good case can be made et  al.  2008) to 27.4% in India (Ramachandra et  al.  2011)
for retaining it even if it is indeed a neoplasm. For the (Table 1.3). A systematic review of frequencies showed an
fourth edition of the WHO classification, the consensus overall combined frequency of 11.7% (Johnson et al. 2014).
group felt that odontogenic keratocyst was the most appro- The reasons for the variation are not always clear, but it
priate name for this lesion and the term was reinstated must be noted that the studies are not population studies,
(Speight et  al.  2017a; Speight and Takata  2018) and has but are mostly retrospective analyses of pathology records.
been retained in the latest classification (Wright and The prevalence and frequency of different lesions may
90 Odontogenic Keratocyst

Table 7.1  Age, sex, and site distribution from selected case series of odontogenic keratocyst, and from the systematic review of Data
from MacDonald-­Jankowski (2011). Wherever possible, data are presented for solitary, non-­syndromic cysts only.

References Country n Mean age Peak decade Male (%) Mandible (%)

Brannon (1976) USA 283 37.8 3rd 56.9 65.4


Ahlfors et al. (1984) Sweden 255 41.0 3rd and 4th 65.5 75.0
Woolgar et al. (1987b) UK 379 40.4 3rd and 4th 61.7 74.0
Crowley et al. (1992) USA 387 39.6 NR 61.9 70.3
Myoung et al. (2001) S Korea 256 30.8 3rd 58.6 76.5
Jones et al. (2006) UK 828 41.3 3rd 56.0 71.1
Jing et al. (2007) China 588 34.9 3rd 63.0 81.5
Habibi et al. (2007) Iran 74 27.1 2nd 59.5 67.5
Gonzalez-­Alva et al. (2008) Japan 183 32.8 3rd 51.3 70.5
Luo and Li (2009) China 507 36.0 3rd 63.7 74.7
Boffano et al. (2010) Italy 241 43.3 4th 67.6 73.2
Jattan et al. (2011) New Zealand 223 37.0 3rd 61.0 65.9
Azevedo et al. (2012) Brazil 158 32.2 3rd 44.7 69.3
Siriwardena et al. (2012) Sri Lanka 431 31.3 3rd 55.5 74.4
Bello (2016) Saudi Arabia 65 31.1 3rd 64.6 84.6
Tamiolakis et al. (2019) Greece 436 42.5 NR 56.7 71.7
Kahraman et al. (2018) Turkey 204 46.6 NR 55.5 81.0
Schuch et al. (2020) Brazil 2497 34.2 3rd 52.2 77.3
MacDonald-­Jankowski (2011) SR 4795a 37.8 3rd 60.0 72.1

n, number of patients affected; NR, not reported; SR, systematic review.


a
 Total cases in review, but n for each parameter varies depending on number of reports.

therefore be affected by local clinical practice. Radicular cyst, and showed frequencies of 7.4% (Grossmann
cysts, for example, are often a straightforward diagnosis et  al.  2007; Brazil), 15.3% (Ali  2011; Kuwait), 8.4%
based on clinical and radiological findings and since the (Tamiolakis et  al.  2019; Greece), 13.6% (Bhat et  al.  2019;
treatment may often be tooth extraction, the soft tissue may India), 15.0% (Kammer et  al.  2020; Brazil), and 12.9%
be discarded and not submitted for histological examina- (Aquilanti et  al.  2021; Italy). Early papers that discussed
tion. This may explain the lower frequency of radicular orthokeratinised cysts as a variant of odontogenic kerato-
cysts and higher frequency of keratocysts in some coun- cyst suggested that they only represented about 10% of the
tries, including Mexico (Mosqueda-­Taylor et al. 2002), Iran total (Brannon 1977; Wright 1981; Crowley et al. 1992; Li
(Sharifian and Khalili  2011), and India (Ramachandra et al. 1998; MacDonald-­Jankowski 2011).
et al. 2011) (Table 1.3). Conversely, some countries, includ- Since 2005, a number of workers have redefined odonto-
ing Sicily (Tortorici et  al.  2008) and Greece (Tamiolakis genic keratocyst as a neoplasm (KCOT) and have reana-
et al. 2019), have shown a low frequency of keratocysts, but lysed the frequency of odontogenic cysts without
a very high level of radicular cysts, which they ascribed to keratocysts included, or have presented the frequency of
high levels of dental caries in the region. The French study keratocysts (KCOT) among odontogenic tumours (Jaeger
(Meningaud et al. 2006) analysed only cases that had been et al. 2017; Gaitán-­Cepeda et al. 2010). This has had little
operated on under general anaesthesia, which may bias the overall effect on the frequency of cysts, since keratocysts
results towards greater numbers of larger cysts, including only represent about 10% of the total and there is great vari-
the odontogenic keratocyst. ation. However, others have suggested that the frequency
Most studies recording the relative frequency of odonto- of odontogenic tumours doubled after 2005 from about 2.5
genic cysts (Table  1.3) have included orthokeratinised to 5% (Gaitán-­Cepeda et al. 2010), or even as high as 11.5%
odontogenic cyst in with odontogenic keratocysts, but this (Jaeger et  al.  2017), and that the KCOT represented the
does not appear to affect the overall frequencies. Six of the most common tumour type with frequencies of 38.9%
reports in Table 1.3 excluded orthokeratinised odontogenic (Gaitán-­Cepeda et al. 2010) and 41% (Jaeger et al. 2017). In
Table 7.2  Frequency (%) of clinical features reported in selected case series. Totals may exceed 100% because patients show multiple symptoms. Based on Brannon, 1976; Chow,
1998; Myoung et al., 2001; Chirapathomsakul et al., 2006; González-Alva et al., 2008; Bello, 2016 and Sundaragiri, 2018.

Associated with
Country n Swelling Pain Infection or discharge Incidental finding Paraesthesia NBCCS Multiple cystsa impacted tooth

Brannon (1976) USA 283 41.3 16.7 NR 49.3 1.4 3.9 3.5 26.6
Crowley et al. (1992) USA 387 27.9 15.0 10.3 14.5 2.1 4.7 NR 47.8
Chow (1998) Singapore 70 51.4 9.9 18.5 15.7 1.4 1.4 2.8 52.8
Myoung et al. (2001) S Korea 256 62.5 36 6.6 5.5 0.8 4.3 10.9 27.1
Chirapathomsakul et al. (2006) Thailand 51 72.5 45.0 15.7 13.7 2.0 0 11.8 31.3
Habibi et al. (2007) Iran 74 60.2 13.2 19.3 24.1 NR 6.1 0 33.7
Simiyu et al. (2013) Kenya 22 81.8 13.6 NR 18.2 NR 4.5 0 9.1
Bello (2016) Saudi Arabia 75 75.7 13.7 8.4 10.0 1.1 4.0 9.3 30.8
MacDonald-­Jankowski (2011) SR 4795b 58.3 32.4 12.2 21.0 1.9 6.4 NR 35.2

n, number of patients affected; naevoid basal cell carcinoma syndrome (NBCCS); NR, not reported; SR, systematic review.
a
 Relates to multiple cysts in patients without NBCCS.
b
 Total cases in review, but n for each parameter varies depending on number of reports.
92 Odontogenic Keratocyst

their systematic review, Johnson et  al. (2014) showed an This bimodal distribution may not be apparent in all
overall combined frequency of odontogenic keratocyst of populations. In a series of 223 cases from New Zealand,
11.7% of odontogenic cysts. However, papers that defined it most cases occurred in the second (40 cases: 18%) and third
as a neoplasm showed a combined frequency of 36.6% of decades (59 cases: 26.5%), followed by a gradual decline
odontogenic tumours (range 19.5–38.7%), which was simi- with no second peak (Jattan et al. 2011). In a sample of 256
lar to ameloblastoma (37.9%). When interpreting the litera- cases in South Korea, Myoung et al. (2001) found a similar
ture, it is important to ensure that the classification and distribution, with 53.9% of cases in the second (25%) and
the denominator used for calculating frequencies are third (28.9%) decades, followed by a gradual reduction with
clearly stated. no second peak. Very similar data were reported from
Japan, with 53.5% of 183 cases in the second and third dec-
ades, and no second peak (González-­Alva et al. 2008).
Age
To address the question of whether two different types of
Odontogenic keratocysts are seen in young adults with a keratocyst might exist, one in younger and one in older age
mean age of about 35 and a peak incidence in the third groups, Rachanis et al. (1979) reviewed the clinical and his-
decade (Figure 7.1 and Table 7.1). However, there is a wide topathological features of a series of odontogenic kerato-
age range and cases have been recorded as early as the first cysts from patients in the age groups 10–29 and 50–64 years,
decade and as late as the ninth (Meara et  al.  1996; Jones but no significant differences were observed between the
et al. 2006; Jattan et al. 2011). Jones et al. (2006) recorded two groups. The authors concluded that it was unlikely
one case in the tenth decade (female, age 92). In most series that these were different varieties of odontogenic kerato-
there has been a pronounced peak incidence in the second cyst and agreed with Browne (1975) that the cysts in older
and third decades, with 40–60% of patients being in this age groups have probably been present but undiagnosed
age group, but many workers have also demonstrated a for many years. This is in keeping with the observation dis-
bimodal age distribution with a second, although smaller, cussed later in this chapter that keratocysts may involve the
peak in the fifth decade or later (Toller 1967; Vedtofte and body and ascending ramus of the mandible extensively,
Prætorius  1979; Ahlfors et  al.  1984; Partridge and with little or no bony expansion.
Towers  1987; Woolgar et  al.  1987b,c; Jones et  al.  2006; Although the odontogenic keratocyst is more common in
Azevedo et  al.  2012). This is illustrated by an analysis of young people, it is very rare in the first decade. In our series
1007 consecutively diagnosed odontogenic keratocysts in (Figure  7.1) we found only 9 of 1007 cases in children
our Sheffield department (Figure 7.1). In this series, 37.3% under 10 years. In a Turkish study, Soluk Tekkeşin et  al.
of cases arose in the second and third decades, with smaller (2016) reviewed 745 odontogenic lesions (including 596
peaks in the sixth (13.5%) and seventh (12.7%) decades. cysts) in patients aged 17 years and under and found

200 Figure 7.1  Age distribution of 1007


odontogenic keratocysts diagnosed in
Males
180 Sheffield.
Females
160
102
140
101

120
No. of cases

100
70 82

59
80 93

53
60
93
40 80
60
55
20 54
35 40 6
5 14 1
0 4

0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89 90+
Age
­Clinical Feature  93

64  keratocysts (10.7% of cysts). All cases were associated cases. In the Sheffield series (Figure 7.1), 571 cases (56.7%)
with the mixed or permanent dentition, 44 were found in arose in males, a male: female ratio of 1.3 : 1. Table 7.1 sum-
the 13–17-­year age group, and 20  in the 6–12-­year age marises a number of selected studies that show that males
group. No lesions were found in patients under the age of predominate, although there are small variations between
6. Similar frequencies have been found in other studies. In studies and in different parts of the world. Occasional stud-
South Korea, Myoung et  al. (2001) found only 11 (4.3%) ies, including from Brazil (Azevedo et  al.  2012) and
cases under the age of 10 years, and only three (1.7%) were Thailand (Chirapathomsakul et  al.  2006), have shown
found in the Japanese study, one of which (age 8 years) was female predilections of 55.3% and 54.9%, respectively, but
associated with the NBCCS (González-­Alva et al. 2008). this is unusual and the reasons for the discrepancy are
In an analysis of biopsy specimens received over a 30-­ not clear.
year period, Jones and Franklin (2006b) found 73 odonto- There is some evidence that keratocysts in association
genic keratocysts in a paediatric population (0–16 years) with NBCCS may present more frequently in females.
compared to 591 cases in the adult population (Jones and Rehefeldt-­Erne et  al. (2016) found that 56.6% of their
Franklin 2006a). Only two cases were found to be associ- NBCCS patients were male, but in a review of four other
ated with NBCCS. However, the data also suggested that studies they found that females predominated in three,
keratocysts were relatively more common in children. As a with a male : female ratio of 1 : 1.3. Woolgar et al. (1987a)
proportion of the total specimens received, keratocysts found that 61.7% of patients with non-­syndromic solitary
constituted 1.6% of specimens received in the paediatric cysts were male (Table 7.1; n = 379; M : F 1.63 : 1), but this
group compared to 1.3% in the adult group. As a proportion was significantly different (P < 0.025) from patients affected
of total odontogenic cysts, keratocysts represented 13.8% of by the syndrome (n  =  60), in which 55.0% were females
odontogenic cysts in children compared to only 9.8% in the (M : F 1 : 1.22). This is in keeping with the evidence that
adult group. This apparent higher frequency in children females may be more frequently affected by
arises because radicular cysts are relatively uncommon in NBCCS. Macdonald’s (2015) systematic review of 14 case
children, but represent a larger proportion of cysts in adults. series of NBCCS found that in 3 of 7 East Asian studies,
Woolgar et al. (1987b) compared the age of patients with and in 5 of 7 European studies, there was a predilection for
sporadic keratocysts with those arising in association with females.
NBCCS and suggested that cysts arise at a younger age in
syndromic patients. In patients without the syndrome
Site
(n = 379), the mean age at removal of the first diagnosed
keratocyst was 40.4 years, compared to 26.2 years in patients The mandible is involved far more frequently than the
with NBCCS (n = 60; P < 0.001). In a systematic review of maxilla (Table  7.1 and Figure  7.2). In his systematic
14 case series, MacDonald (2015) found that although review, MacDonald-­Jankowski (2011) found that the man-
NBCCS was diagnosed at a mean age of 29.2 years in East dible was affected more than twice as often as the maxilla,
Asian patients (7 reports; n  =  251) and 36.2 years in although he was able to demonstrate geographical varia-
European patients (7 reports; n  =  406), the mean age of tions. In studies from East Asia, the mandible was affected
presentation of the first odontogenic keratocyst was 31.4 in 77% of cases, compared to 73% in Latin America and
and 16.9 years, respectively. Rehefeldt-­Erne et  al. (2016) 63% in Western studies. In our own material from South
also found that NBCCS was diagnosed in the fifth decade, Africa, 149 of 193 cysts (77%) for which data were availa-
but in a review of four studies (including their own) they ble occurred in the mandible. The high frequency of man-
found that the first jaw cyst was always found in the second dibular involvement is borne out in many series from
decade, with mean ages between 15.5 and 19.8 years. throughout the world (Table 7.1) and is reported from 65%
From these studies it can be concluded that odontogenic (USA; Brannon  1976) to as high as 85% (Saudi Arabia;
keratocysts in patients with NBCCS arise in a younger age Bello 2016).
group, that the age difference is an intrinsic feature of the More recently, Slusarenko da Silva et al. (2019a) under-
syndrome, and that it may be in keeping with the underly- took a systematic review and meta-­analysis of 34 studies
ing inherited genetic changes known to be associated with that had accurately recorded the site of lesions. Of
NBCCS (see also later Table 7.5). 3126 keratocysts included, 2376 (76%) were located in the
mandible, and 1401 (44.8% of all keratocysts) of these were
found in the posterior mandible – including the third molar
Sex
region, angle, and ramus. Conversely, lesions in the maxilla
Similar to most odontogenic cysts, keratocysts show a were more likely to be found in dentate areas of the jaw. Of
slight predilection for males, who account for about 60% of the 750 (24%) maxillary cysts, 75.5% were found anterior to
94 Odontogenic Keratocyst

Figure 7.2  The approximate site distribution


of odontogenic keratocysts (see text for
details).

5%
15% 25%
5%
50
%

20% 75%
5%

the second molar and only 24.5% (184 lesions) were associ- without the syndrome, while only 44% arose at this site in
ated with the third molar or tuberosity. Ali and Baughman patients with NBCCS. Of their patients with the syndrome,
(2003) in an analysis of 398 keratocysts found only 7.5% in 18 (30%) initially had only one cyst, but in all except 3 patients
the posterior maxilla, but of the remainder, the majority further cysts developed subsequently; 24 syndrome patients
(54 lesions: 13.6%) were located in the canine region and had cysts in two quadrants, 13 of which were bilateral man-
half of these (27 cysts) were in a periapical position, resem- dibular cysts related to third molar teeth; 5 patients had cysts
bling a radicular cyst. in three quadrants and 13  had cysts in all four quadrants.
Figure  7.2 illustrates the approximate distribution of These authors emphasised that the term ‘multiple’, when
odontogenic keratocysts in the jaws. These data are esti- applied to cysts occurring in patients with NBCCS, referred
mated from the publications in Table  7.1 and from to the lifetime history of the patient and not necessarily that
Slusarenko da Silva et al. (2019a), and figures are rounded more than one cyst was present at any one time. More
to the nearest 5% for simplicity and to aid recall. The right recently, Karhade et al. (2019) supported these findings, and
side of the diagram shows that about 75% of all keratocysts found that 13 of 18 (72.2%) syndromic cysts arose in the max-
arise in the mandible, while the left side illustrates the dis- illa, compared to only 11 of 32 (34.3%) sporadic cysts.
tribution in different regions of the jaws. A total of about Woolgar and colleagues (Woolgar et  al.  1987b; Rippin
50% are located at the angle of the mandible, some of and Woolgar 1991) have also suggested that the finding of
which may extend into the ramus, and only 20% are found multiple keratocysts is sufficient for a diagnosis of NBCCS
anterior to the second molars. About 5% of lesions extend even if other features are not evident, since on full exami-
across multiple mandibular sites. In the maxilla, the major- nation other features of the syndrome, albeit only minor
ity (15%) are located in the dentate regions of the jaw, and anomalies, may be revealed. Others, however, do not sup-
only about 5% of all keratocysts are associated with a max- port this and have reported multiple (not recurrent) kerato-
illary third molar or the tuberosity. About 5% may extend cysts in up to about 10% of non-­syndromic patients
across multiple sites. (Table 7.2; Brannon 1976; Chow 1998: Myoung et al. 2001;
A number of studies have suggested that the site distribu- Chirapathomsakul et  al.  2006; González-­Alva et  al.  2008;
tion of NBCCS-­associated keratocysts is different to sporadic Bello  2016; Sundaragiri  2018). Nevertheless, clinicians
cysts, with a higher frequency in the maxilla. Woolgar et al. need to be aware of the probability that if a patient has
(1987b) reviewed 164  keratocysts from 60 patients with more than one odontogenic keratocyst, other features of
NBCCS. Of these, 34 (21%) arose in the posterior maxilla in the syndrome should be investigated. Furthermore, if a
patients with NBCCS, compared to only 11% at this site in patient, especially a young patient, has a single obvious
patients without the syndrome (n = 379). In addition, 60% of keratocyst, careful scrutiny of appropriate radiographs
cysts were found in the posterior mandible in patients should be performed to exclude the possibility of other cysts.
­Clinical Feature  95

with numbness or tingling (paraesthesia) has been reported


Box 7.1  Odontogenic Keratocyst: Clinical Features
to occur in less than 2% of cases. Between 10 and 56% of
and Key Facts
cysts may be associated with a missing or impacted tooth
●● Third most common of the odontogenic cysts with a and this will be discussed below in the section on radiology.
frequency of 10–15% In most cases, swelling is accompanied by expansion of
●● Mean age is about 35 years, with a peak in the the bone. Browne (1970) found that expansion of bone
third decade occurred in about 60% of cases, with about half of man-
●● Slightly more common in males (60%) dibular lesions causing buccal expansion and one-­third lin-
●● 75% arise in the mandible gual expansion. One-­third of maxillary cysts caused buccal
●● 50% arise at the angle and ramus region of the expansion, but palatal expansion was very rarely seen. In
mandible his systematic review, MacDonald-­Jankowski (2011) found
●● Lesions in the posterior maxilla are rare (~5%) only two papers that detailed the pattern of expansion, and
●● Up to 10% of patients without naevoid basal cell car- these recorded that 62% of cases produced bucco-­lingual
cinoma syndrome (NBCCS) may have multiple cysts expansion and 71% displaced the lower border of the man-
●● About 5% of keratocysts are associated with NBCCS dible. The review also found that 100% of maxillary lesions
●● Keratocysts in patients with NBCCS show a different expanded into the antrum. Forssell (1980) observed expan-
profile: sion of bone in 53% of his cases, and this occurred signifi-
–– Almost always multiple and in multiple quadrants cantly more frequently at the angle or ascending ramus of
–– More often female (55%) the mandible than in the maxilla or body of the mandible.
–– Often younger (<20 years) Perforation of bone, as observed in orthopantomograms,
–– More frequently encountered in the posterior max- occurred in 39% of his cases. As with other intraosseous
illa (up to ~20%) jaw lesions, the enlarging cyst may cause resorption or dis-
placement of teeth (Borghesi et  al.  2018). MacDonald-­
Jankowski and Li (2010) reported tooth displacement and
Clinical Presentation
resorption in 69% and 41% of cases, respectively.
The clinical features are summarised in Table 7.2. The sin- With regard to the incidental presentation of odonto-
gle most common presentation is of a painless swelling, genic keratocysts, patients may be remarkably free of
although a significant number of cases arise as an inciden- symptoms until the cyst has reached a large size, often with
tal finding during radiological examination for other pur- extensive involvement of the entire ascending ramus,
poses. In one of the largest series (256 patients), Myoung including the condylar and coronoid processes or with
et  al. (2001) found that 62.5% of odontogenic keratocysts extension into the maxillary sinus. This occurs because the
presented with a swelling and 36% with pain. However, odontogenic keratocyst tends to extend antero-­posteriorly
only 42 patients (16.4%) had both swelling and pain through the cancellous bone and clinically observable
together. This means that almost half of patients (46%) pre- expansion of the jaws occurs late.
sented with swelling only and 20% with pain only. They Odontogenic keratocysts are often described as showing
also found 17 cases (6.6%) that presented with a discharge ‘aggressive’ behaviour, but this must not be overinterpreted
and 5.5% presented as an incidental finding. Only 2 cases to suggest invasion, or a growth pattern similar to that seen
(0.8%) showed signs of paraesthesia. in truly aggressive lesions such as malignancies. It is cer-
In a large Italian study of 241 patients (Boffano tainly true that keratocysts may grow to a large size and
et  al.  2010), only 37.2% presented with symptoms, while have a tendency to recur, but even when large, the lesions
the remainder (62.8%) were incidental findings during rou- are usually well defined on radiology, suggesting slow
tine radiographic examination. In a systematic review of 49 growth. We do not support the concept that clinical recur-
case series, MacDonald-­Jankowski (2011) found that fewer rence alone is synonymous with, or a defining feature of,
than 10 papers reported clinical signs and symptoms. The aggressiveness. The size and extent of the lesion appear to
most common clinical sign, affecting up to about 60% of be associated with late presentation of a cyst that has
patients, was the presence of swelling, with or without grown slowly over time. This may be a feature of the fact
pain. Overall, about 20% of keratocysts were an incidental that they tend to grow within the bone of the mandible
finding, either secondary to a pathological fracture or, with late cortical expansion.
more commonly, discovered fortuitously during dental Extensive growth may also occur within the maxilla,
examination when radiographs are taken. Other features where the lesion can expand unimpeded within the
included infected lesions, which may present with a puru- antrum. Lund (1985) described a large keratocyst involving
lent discharge, in up to 18% of cases. Nerve involvement the maxillary sinus that led to displacement and
96 Odontogenic Keratocyst

perforation of the floor of the orbit and proptosis of the eye. They identified 23 cases, but 3 were not fully documented
Vencio et al. (2006) also reported a case that had extended and 3 appeared to be in soft tissues. Data were available for
into the maxillary antrum, destroying the floor, and a more 17 peripheral gingival lesions (Chehade et al. 1994; Fardal
recent case has been shown to extend through the posterior and Johannesssen 1994; Dayan et al. 1988; Ide et al. 2002;
wall of the antrum and involve the pterygoid process of the Chi et  al.  2005; Preston and Narayana  2005; Faustino
sphenoid bone (Goto et  al.  2020). Other authors have et  al.  2008; Vij et  al.  2011; Gröbe et  al.  2012; Sakamoto
reported on aggressive behaviour of keratocysts to the et al. 2014). Vázquez-­Romero et al. found that peripheral
extent that they perforated the cortical bone and involved lesions showed an average age of 51.3 years (range 24–83),
surrounding soft tissues (Emerson et  al.  1972; Partridge with a slight predilection for females (11 lesions: 65%). The
and Towers 1987). Jackson et al. (1993) reported two cases most common site was the canine/premolar region, with a
that showed extensive growth. One lesion in a 47-­year-­old slight predilection for the maxilla (9/17  lesions: 52.9%).
male extended from the maxilla and eventually involved Clinically the peripheral odontogenic keratocyst is indis-
the base of the skull. However, it should be noted that the tinguishable from the gingival cyst of adults (see Chapter 9)
lesion recurred over a period of 10 years after first present- and presents as a gingival nodule that may be normal or
ing as a suppurating infection that was curetted, but not blue/grey in colour and ranges from 3 to 15 mm in diame-
diagnosed or removed. Subsequently there were multiple ter (Chi et al. 2005). All reported lesions have arisen on the
recurrences associated with multiple cysts, infection, and buccal or labial aspect of the gingivae. Only two cases have
persistent draining sinuses. There are many similar reports been shown to recur. One case has been reported in asso-
of extensive lesions, including involvement of the infratem- ciation with the NBCCS, in a 24-­year-­old female (Sakamoto
poral fossa (Chuong et al. 1982) and the temporalis muscle et al. 2014)
(Worrall  1992). These cases are often cited to illustrate The peripheral odontogenic keratocyst has a clinical
aggressive behaviour, but most are unusual, involving presentation identical to gingival cyst of adults and proba-
inadequate initial treatment, multiple recurrences, and bly has a similar pathogenesis, arising from rest cells of
infection. They cannot be regarded as representing the typ- dental lamina. Some have suggested that they are part of a
ical behaviour of a keratocyst, but do illustrate how a recur- spectrum of the same lesion. However, the peripheral
rent lesion can cause significant management problems. keratocysts are histologically identical to their intraosseous
counterpart and occasionally may show daughter or satel-
lite cysts (Chi et al. 2005). Given its characteristic features,
Peripheral (Extraosseous)
we would regard the peripheral odontogenic keratocyst as
and Soft-­Tissue Keratocysts
a separate and distinct entity.
It is well recognised that a typical intraosseous odonto-
genic keratocyst may recur within the soft tissues adjacent Soft-­Tissue Keratocyst
to the site of initial surgery. In addition, however, there In addition to peripheral lesions presenting on the gingi-
have been reports of primary keratocysts arising within the vae, there are a number of reports of lesions with the typi-
gingivae or in the soft tissues. Lesions in the gingivae are cal histological features of odontogenic keratocyst, but
regarded as peripheral odontogenic keratocysts that arise arising within the soft tissues of the maxillofacial region.
from odontogenic epithelium within the alveolar mucosa These lesions are of uncertain origin and are often referred
or gingivae overlying bone. Lesions with features of a to as soft-­tissue keratocyst.
keratocyst arising within soft tissues away from bone have The total number of cases of soft-­tissue keratocyst reported
also been described, but are of uncertain origin. is uncertain, but appears to be fewer than 20. A number are
reported under different names or are included in larger
Peripheral Odontogenic Keratocyst series of other cyst types. Table  7.3 summarises 12  well-­
Gingival or peripheral odontogenic keratocysts appear to be documented cases. The majority occurred in males (10 cases;
rare, with fewer than 20 fully documented cases. The first 83.3%) and the average age at presentation was 52.5 years
well-­characterised case was reported by Dayan et al. (1988), (range 16–74). Although this is older than for intraosseous
who described a lesion entirely within the gingiva, which odontogenic keratocysts, it is possible that the soft-­tissue
had the clinical features of a gingival cyst of adults but the lesions present late because they grow slowly and are embed-
histological characteristics of an odontogenic keratocyst ded deep in the soft tissues. The one case reported in a
with the typical parakeratinised lining. They suggested the 16-­year-­old (Ide et  al.  2010) was a chance finding during
term ‘peripheral odontogenic keratocyst’ for this rare pres- microscopic examination of a biopsy of a ‘traumatic fibroma’.
entation of the lesion. Vázquez-­Romero et  al. (2017) It was only 3 mm in diameter and if left, might well have
reported a case and reviewed the literature up to 2016. grown for a long period before becoming clinically apparent.
­Clinical Feature  97

Table 7.3  Soft-­tissue keratocyst: summary of the clinical features of 12 well-­documented cases.

Age Sex Location Max dimension Notes

Precheur and Krolls (2009) 59 M Buccal space 31 mm


Ide et al. (2010) 60 M Buccal mucosa 30 mm Multicystic
16 M Buccal mucosa 3 mm Microcyst associated with ‘traumatic
fibroma’
Gröbe et al. (2012) 52 M Buccal mucosa 20 mm
Yamamoto et al. (2013) 74 M Buccal mucosa 35 mm
Kaminagakura et al. (2013) 37 M Buccal mucosa 20 mm
Abé et al. (2014) 46 M Temporalis muscle 21 mm Extended into buccal mucosa; multicystic
Zhu et al. (2014) 44 F Soft palate, pharynx 40 mm
69 M Buccal mucosa 20 mm Also involved the pterygopalatine fossa
Makarla et al. (2015) 62 M Buccal space 48 mm
Witteveen et al. (2019) 63 M Buccal space 18 mm
48 F Buccal space Not reported

The most common presentation was of an intraoral have proposed an origin from sebaceous glands (Ide
swelling of normal colour, varying from about 20 to 40 mm et  al.  2010; Makhija  2015). Cutaneous keratocysts are an
in diameter. One case presented in the soft palate and occasional finding in NBCCS and show similar histological
extended into the pharynx (Zhu et al. 2014) and one case features. Makhija (2015) noted this similarity and proposed
appeared to arise in the temporalis muscle (Abé et al. 2014), that steatocystoma (simplex) and cutaneous keratocysts
but all the others arose in the ‘cheek’. Those described in should be unified as sebaceous duct cysts. The soft-­tissue
Table 7.3 as presenting in the ‘buccal mucosa’ appeared to keratocyst of the maxillofacial region may therefore be
be primarily intraoral and were submucosal, lying deep to derived from sebaceous glands. In this respect it must be
the buccinator muscle (the oral side), and all were located noted that sebaceous glands are commonly found in the
posterior to the opening of the parotid duct. Those reported buccal mucosa (‘Fordyce spots’). Against this concept is the
as presenting in the ‘buccal space’, however, were primarily fact that sebaceous glands have never been reported in the
extraoral and lay superficial to the buccinator muscle, walls of these cysts. On the rare occasions that steatocysto-
towards the skin or facial aspect (Precheur and Krolls 2009; mas have been reported in the oral tissues, they have been
Makarla et al. 2015; Witteveen et al. 2019). associated with sebaceous glands and have shown seba-
Histologically, all the lesions showed the typical features ceous differentiation in the lining (Olsen et  al.  1988;
of an odontogenic keratocyst, with a thin lining of par- Daley 1993).
akeratinised epithelium. Two lesions (Ide et al. 2010; Abé These studies suggest that soft-­tissue keratocysts in the
et al. 2014) were described as multicystic, but others were oral tissues are most likely derived from ectopic odonto-
simple cysts with no evidence of satellite cysts or epithelial genic epithelium, but some may occur in the subcutaneous
islands. Occasional lesions were associated with salivary tissues, be analogous to cutaneous keratocysts, and arise
tissue, but sebaceous glands or dermal appendages were from dermal appendages. Table  7.3 indicates that lesions
not seen. may be superficial (facial) or deep (oral) to buccinator mus-
Most authors have noted the similarity of the cysts to cle, and it may be that these represent two similar cyst
conventional odontogenic keratocyst and assumed there- types with different pathogenic mechanisms.
fore that they are of odontogenic origin. The presumptive A rare multicystic lesion, called keratocystoma, with fea-
source of epithelium would be cell rests of dental lamina tures resembling odontogenic keratocyst has been
that have become displaced during development. This is described in the parotid gland (Seifert et  al.  1999).
perfectly possible and heterotopic odontogenic tumours, Histologically the keratocystoma is composed of multiple
including ameloblastoma and odontomas, have occasion- cystic spaces lined by stratified squamous epithelium
ally been reported in the soft tissues. showing para-­ and orthokeratinisation, but without a
Others have noted that the parakeratinised lining is simi- prominent basal or granular cell layer. Solid islands of
lar to the lining of steatocystoma or sebaceous ducts and squamous epithelium are also seen. The cysts are often
98 Odontogenic Keratocyst

irregular and multilocular. Keratocystoma resembles the the feet, and calcification of the falx cerebri. The features of
so-­called solid odontogenic keratocyst (see the syndrome and the criteria for diagnosis are shown in
‘Histopathology’), but the lesion is only found in the Table 7.4 (Bree et al. 2011) and Table 7.5 summarises the
parotid gland. The true nature of the lesion is unknown prevalence of the key features taken from four representa-
and the literature has variously described it as a benign tive large series (Evans et  al.  1993; Shanley et  al.  1994;
cyst, a choristoma, or more frequently as a benign neo- Kimonis et al. 1997; Endo et al. 2012) and from a system-
plasm. Only 11 cases have been reported (reviewed by atic review of seven reports from East Asia (n = 251) and
Aresta et al. 2019; Komatsu et al. 2020) and most authors seven reports of patients of European origin (n  =  406)
regard the lesion as a benign neoplasm that arises from (MacDonald 2015).
intercalated ducts that have undergone squamous meta- Overall, about 75% of patients present with odontogenic
plasia. Keratocystoma and soft-­tissue keratocyst are proba- keratocysts, but this may be higher in East Asian patients.
bly completely unrelated lesions, but their histological In the vast majority of cases these are multiple and present
similarity may cause diagnostic difficulties in small biop- in a synchronous and metachronous manner. The number
sies. Lesions located in the parotid gland should be diag- of cysts that each patient may have over a lifetime ranges
nosed as keratocystoma, not as soft-­tissue keratocyst. The from 1 to 28, with an average of 4–6. Odontogenic kerato-
keratocystoma is discussed further in Chapter 15. cysts are often the first manifestation of the syndrome,
with 75% or more of patients presenting with their first cyst
before the age of 20 years. In the totality of odontogenic
­ aevoid Basal Cell Carcinoma
N keratocysts, however, only about 6% or less are associated
Syndrome (NBCCS) with NBCCS (Table 7.2). Basal cell carcinomas tend to pre-
sent later, with an average age of 21.4 years, but the full
In 1960, Gorlin and Goltz described a syndrome comprised syndrome is often not diagnosed until the middle of the
of cysts of the jaws, multiple basal cell ‘epitheliomas’, and fourth decade (Table 7.5; Kimonis et al. 1997).
bifid ribs, a combination that is frequently referred to as The important fact to note is that dental healthcare work-
the ‘Gorlin–Goltz syndrome’, ‘Gorlin syndrome’, basal cell ers are in a pivotal position to identify the first and early
nevus syndrome, or NBCCS. Gorlin et  al. (1963) and signs of the syndrome and refer accordingly. As well as
Meerkotter and Shear (1964) later identified the jaw cysts odontogenic keratocysts, patients may present with frontal
as odontogenic keratocysts. Subsequently, numerous clini- bossing, hypertelorism, or cleft lip or palate. Furthermore,
cal and molecular studies have shown that there is a wide the basal cell carcinomas are often encountered on the face,
range of possible clinical features with variability in clini- often as clusters around the eyes and nose. There is some
cal presentation (reviewed by MacDonald 2015). However, evidence that the prevalence of basal cell carcinomas may
the most commonly encountered clinical manifestations of be affected by ethnicity or geographical factors. While the
the syndrome include odontogenic keratocysts, basal cell overall prevalence is about 75%, the prevalence in Japanese
carcinomas, pitting of the palms of the hands or soles of patients was only 38% (Endo et al. 2012) (Table  7.5) and

Table 7.4  Diagnostic criteria for naevoid basal cell carcinoma syndrome. Two major criteria, or one major criterion and two minor
criteria, or one major criterion and genetic confirmation are required for diagnosis.

Major criteria Minor criteria

1)  Multiple basal cell carcinomas (BCCs) or 1)  Rib anomalies (bifid, fused, or splayed)
one BCC in a person younger than 20 years
2)  Odontogenic keratocyst before age 20 2)  Other specific skeletal and radiological abnormalities (vertebral anomalies,
kyphoscoliosis, short fourth metacarpals, postaxial polydactyly)
3)  Palmar or plantar pitting 3)  Macrocephaly
4)  Calcification of the falx cerebri 4)  Cleft lip or palate
5)  Medulloblastoma 5)  Ovarian or cardiac fibroma
6)  First-­degree relative with basal cell nevus 6)  Lymphomesenteric cysts
syndrome (BCNS)
7)  Ocular anomalies (congenital cataract, coloboma, glaucoma, hypertelorism)

Source: Based on Bree et al. (2011).


­Naevoid Basal Cell Carcinoma Syndrome (NBCCS  99

Table 7.5  Clinical features of naevoid basal cell carcinoma syndrome (NBCCS) from four representative case series and a large
systematic review. Data from MacDonald 2015.

Evans Shanley Kimonis Endo MacDonald MacDonald


et al. (1993) et al. (1994) et al. (1997) et al. (2012) (2015)a (2015)a

Country UK Australia USA Japan East Asian European


No. of patients 84 118 105 157 251 406
OKC (%) 66 75 74 86 90 70
No of OKC – mean (range) NR 6 (1–28) 5.1 (1–28) 3.7 (1–10) NR NR
Age at first cyst (mean) NR 15.5 17.3 19.8 31.4 16.9
Age at diagnosis of NBCCS NR 35 34.5 33.1 29.2 36.2
(mean)
BCC (%) 47 76 73 38 31 71
Palmar/plantar pitting (%) 71 80 87 69 68 79
Falx cerebri calcification (%) NR 92 65 79 61 78
Rib anomalies (%) NR NR 26 36 37 39
Cleft lip/palate (%) 5.7 NR 2.8 9.0 9 4

OKC, odontogenic keratocyst; BCC, basal cell carcinoma; NR, not reported.
a
 Systematic review.

Kimonis et al. (1997) found only 38% in black patients com- an overall prevalence of about 1 in 60 000 live births. The
pared to 80% in whites. In a systematic review, MacDonald most common mutation is in the PTCH1 gene, but muta-
(2015) found a prevalence of 31% in East Asian patients tions in PTCH2 and SUFU have also been implicated. So
compared to 71% in patients of European origin (Table 7.5). far, almost 450 mutations have been identified in PTCH1
The current criteria for diagnosis of NBCCS are shown in (Reinders et al. 2018) and this genetic heterogeneity may
Table 7.4. A diagnosis should be suspected on the basis of contribute to the lack of genotype–phenotype correlations
(i) one major criterion confirmed by molecular analysis, or and to the variability of clinical presentation. The PTCH
(ii) two major criteria, or (iii) one major and two minor cri- gene encodes a transmembrane protein that acts as a
teria. If these criteria are met, a patient should be referred receptor for the hedgehog family member SHH (Sonic
to a medical geneticist (Bree et al. 2011). Furthermore, the hedgehog) (Figure 7.13). The function of the PTCH pro-
association with odontogenic keratocyst is so strong that tein is to inhibit activation of SMO (smoothened). When
any patient under age 20 years with even a single keratocyst SHH binds to the PTCH receptor, SMO is activated and in
should ‘trigger’ further investigations following an estab- turn activates the Gli transcription factors that up-­regulate
lished diagnostic protocol that includes a detailed history cell proliferation and also have roles in development and
and skeletal and dermatological examinations (Bree et normal odontogenesis (Diniz et  al.  2017; Seppala
al.  2011). Karhade et  al. (2019) reviewed odontogenic et  al.  2017; Reinders et  al.  2018). Mutation of PTCH1
keratocysts in 50 children and young adults (18 years and results in constitutive activation of the hedgehog (HH) sig-
under), 18 of whom (36%) had NBCCS, 8 with a previously nalling pathway with a plethora of downstream conse-
known family history, and 10 based on further investiga- quences, including increased cell proliferation or failure
tions following diagnosis of the odontogenic keratocyst. Of of apoptosis. PTCH1 is thus regarded as a tumour-­
the 18 patients who were diagnosed with NBCCS, 8 (44%) suppressor gene.
had presented with a single keratocyst. Odontogenic It has been suggested that the developmental anoma-
keratocysts associated with NBCCS may also show charac- lies associated with NBCCS are caused by a germline
teristic histological features that might alert an examining mutation in one allele of PTCH1, while tumours are a
pathologist to the possibility that a patient has the syn- result of mutations in both PTCH alleles (the ‘two-­hit
drome. These are discussed later in this chapter (see hypothesis’). The hedgehog signalling pathway and
‘Histopathology’ and Table 7.8) molecular studies on solitary (sporadic) and syndrome-­
NBCCS is an autosomal-­dominant inherited syndrome, related odontogenic keratocyst are discussed in detail
with strong penetrance, but with variable expressivity and later in this chapter (see ‘Pathogenesis’ and Figure 7.13).
100 Odontogenic Keratocyst

­Radiological Features when larger, have a scalloped or lobulated margin and may
show incomplete bone septae within the lesion (Borghesi
A typical odontogenic keratocyst appears as a well-­ et  al.  2018; Figure  7.4). These are sometimes misinter-
demarcated unilocular radiolucency with a corticated mar- preted as multilocular. A truly multilocular lesion is com-
gin (Figure 7.3). However, a wide variation in appearances posed of multiple cysts, often described as having a
may be seen, resulting in considerable diagnostic difficul- ‘soap-­bubble’ appearance (Figure 7.5; Borghesi et al. 2018;
ties. Cysts range from small, round or ovoid and well-­ MacDonald-­Jankowski and Li 2010).
demarcated to large, extensive lesions with poorly defined In a series of 75 keratocysts, Stoelinga (2001) was careful
borders. to make this distinction, and carefully defined cysts as uni-
Case series show that between about 70 and 85% of locular, scalloped, multilobular, and multilocular. In par-
keratocysts are unilocular (Chirapathomsakul et al. 2006; ticular, multilobular comprised two or more lobes, but with
Boffano et  al.  2010; MacDonald-­Jankowski  2011; Schuch
et al. 2020; Kitisubkanchana et al. 2021) and that 95% or
more are described as well demarcated with a corticated or
sclerotic margin (MacDonald-­Jankowski and Li  2010;
Schuch et  al.  2020). Many unilocular lesions, especially

Figure 7.3  A small odontogenic keratocyst. Radiology shows a Figure 7.5  A large keratocyst at the angle and ramus of the
well-­demarcated radiolucency with a corticated margin. mandible. This cyst is truly multilocular.

(a) (b)

Figure 7.4  (a) Radiograph of an odontogenic keratocyst that is unilocular but lobulated with scalloped margins. (b) The gross
specimen shows the irregular growth pattern of the lesion that results in the scalloped radiological appearance.
­Radiological Feature  101

no bony septae dividing the lobules (e.g. Figure  7.4), significant association between multilocular lesions and
whereas multilocular showed locules or lobules separated size greater than 31 mm. Forssell (1980) found that uniloc-
by intact bony septae (e.g. Figure 7.5). He found that only 7 ular cysts that were scalloped or multilobular were also sig-
(9.3%) cases were truly multilocular, 35 (46.7%) were uni- nificantly larger than unilocular cysts with a smooth margin.
locular, but 15 (20.0%) were scalloped and 18 (24.0%) were These data suggest that unilocular lesions as they grow
multilobular. All the multilocular cysts were located in the may become scalloped and then multilocular (compare
posterior mandible. A review of many case reports shows Figures  7.3–7.5). This is feasible, but difficult to prove.
that most authors describe both multilobular and multi- Multilocular lesions are more common in the mandible
locular lesions as multilocular, and these combined in and it is possible that the restricted growth imposed by the
Stoelinga’s series gives a proportion of 33%, in keeping with cortical plates results in a multicystic lesion. In the maxilla,
other reports. however, growth is relatively unimpeded and the cyst is
A number of authors agree with Stoelinga, and have enabled to grow centripetally in a ballooning pattern, often
shown that larger lesions and lesions in the mandible are filling the antrum (see later Figures 7.8 and 7.9).
more often multilocular. Chirapathomsakul et  al. (2006) The pattern of growth in the mandible produces a char-
found that 34.8% of mandibular lesions were multilocular acteristic feature of the odontogenic keratocyst. Lesions
compared to only 14.3% in the maxilla. Browne (1970) may show marked mesio-­distal extension, but with rela-
found 19 of 83 cysts (22.9%) to be multilocular, all in the tively little evidence of bucco-­lingual expansion
mandible, and Forssell (1980) observed a frequency of (Figure  7.6). This feature is best visualised on computed
25.0% in a series of 135, also all in the mandible. Forssell tomography (CT) scans, but is so characteristic that
showed that about 50% of his cysts were 40 mm or more in MacDonald (2016) regarded it as pathognomonic of the
diameter and that this was particularly the case with cysts odontogenic keratocyst. This is discussed further later in
of the ascending ramus and angle of the mandible, com- this section.
pared with cysts of the maxilla or body of the mandible. The most common site of presentation of odontogenic
MacDonald-­Jankowski and Li (2010) found that 75.0% of keratocysts is the posterior aspect of the mandible
mandibular cysts were multilocular compared to only (Figure 7.2), and both unilocular and multilocular lesions
15.4% of maxillary lesions. These authors also found that may involve the body and ascending ramus of the mandi-
multilocular lesions (especially in the mandible) were ble extensively (Figures 7.4–7.6). Up to 50% of lesions are
larger than unilocular lesions, and that unilocular lesions associated with an impacted tooth (Table  7.3) and it is
presented at a younger age (24.5 years) than multilocular common for the odontogenic keratocyst to present in a
lesions (36.1 years). Boffano et  al. (2010) also found a radiological dentigerous relationship, particularly with

(a) (b)

Figure 7.6  A large odontogenic keratocyst. (a) A conventional radiograph shows the lesion arising at the angle of the mandible, with
extensive mesio-­distal extension from the first molar and almost filling the ramus. (b) The coronal computed tomography (CT) scan,
however, shows relatively little bucco-­lingual expansion.
102 Odontogenic Keratocyst

impacted third molars. (Figure 7.7; see also Figure 5.14). and Weinmann 1958). The relationship between the den-
Ali and Baughman (2003) found that 137 of their 398 tigerous cyst and odontogenic keratocyst is discussed in
cysts were located in the posterior mandible, of which 52 detail in Chapter 5.
(37.9%) were in a pericoronal relationship with an Downward displacement of the inferior alveolar canal is
impacted third molar. Such lesions are frequently misdi- almost always seen and about 70% of mandibular lesions
agnosed as dentigerous cysts and this has given rise to also cause downward displacement of the lower border of
two misconceptions. One is that dentigerous cysts may the mandible (MacDonald-­Jankowski  2011; Figure  7.5).
have a keratinised epithelial lining similar to that found Maxillary lesions displace the floor of the maxillary sinus
in odontogenic keratocysts; and the second is that denti- upwards, but may still show a corticated margin (Figures 7.8
gerous cysts may have an extrafollicular origin (Gillette and 7.9). Displacement of teeth (70% of cases) and perfora-
tion of the cortical plate (50% of cases) are also common
(Chirapathomsakul et al. 2006; MacDonald-­Jankowski 2011).
Resorption of the roots appears to be infrequent and is rarely
mentioned in reports (Struthers and Shear  1976).
Chirapathomsakul et al. (2006) found that only 1 of 67 (1.5%)
cysts showed radiological evidence of root resorption, and
Kitisubkanchana et al. (2021) found resorption in 7 of 100
cases (7%). Others, however, have found a higher incidence.
Forssell (1980) noted varying degrees of root resorption in
24% of his series of 90  keratocysts, but pointed out that a
large proportion of these showed only a slight degree of
resorption. Partridge and Towers (1987) observed resorption
in 9 of their sample of 82 cases (11%), and MacDonald-­
Jankowski and Li (2010) found root resorption in 13 of
32 lesions (40%).
Very rarely (less than 4% of cases) an odontogenic kerato-
cyst may show a mixed radiological pattern with small
areas of radiopacity (Azevedo et  al.  2012; Borghesi
et al. 2018; Schuh et al. 2020). Almost certainly, this is due
to dystrophic calcification within the keratinaceous or
Figure 7.7  An odontogenic keratocyst that has enveloped an
necrotic luminal contents, or within the cyst wall.
unerupted tooth and lies in a ‘dentigerous’ relationship.

(a) (b)

Figure 7.8  Sagittal computed tomography (CT) scans of odontogenic keratocysts in two different patients. (a) The keratocyst has
expanded into the maxillary antrum, but still has a corticated margin (arrows). (b) In this case, the CT scan shows the multilocular
nature of the lesion and buccal expansion.
­Radiological Feature  103

Computerised Tomography and Magnetic showed a ballooning pattern of expansion with a similar


Resonance Imaging degree of extension mesio-­distally and bucco-­lingually. In
contrast, four of seven odontogenic keratocysts that had
CT and cone beam CT (CBCT) have advantages in the diag-
expanded bone showed a fusiform pattern of expansion,
nosis of odontogenic keratocysts since they allow high-­
with mesio-­distal extension much greater than bucco-­
resolution three-­dimensional assessment of the lesion. CT
lingual. This pattern is characteristic of odontogenic
is particularly valuable for visualising the bucco-­lingual
keratocyst and has been described as pathognomonic
expansion of mandibular cysts and for determining the
(MacDonald 2016). It should be noted, however, that a sim-
extent of maxillary lesions, which may be particularly dif-
ilar pattern of growth can be seen in glandular odontogenic
ficult to see on conventional radiographs (Figures  7.8
cyst; this is discussed in Chapter 10. Figure 10.5 illustrates
and  7.9; MacDonald  2016; Borghesi et  al.  2018; Alves
the typical ballooning expansion of an ameloblastoma,
et al. 2018). Three-­dimensional imaging is now regarded as
which is not seen in odontogenic keratocysts. Alves et al.
essential for proper surgical planning of large lesions,
(2018) also found that root resorption was more common
although it should be noted that CT is not ideal for evaluat-
in ameloblastomas and was seen in six cases compared to
ing extension into soft tissues.
only one case of keratocyst. However, both lesions showed
MacDonald (2016) showed how CBCT can be used to
a similar association with unerupted teeth (four of nine
visualise the three-­dimensional extent of mandibular
lesions) and with cortical bone resorption (six of nine
odontogenic keratocysts and to show how marked mesio-­
lesions)
distal extension may be associated with minimal bucco-­
Magnetic resonance imaging (MRI) is less often used or
lingual expansion (Figure  7.6). This was also shown by
necessary, but may complement CT and is especially use-
Alves et  al. (2018), who compared nine cases each of
ful in selected cases to evaluate soft-­tissue extension. By
ameloblastoma and odontogenic keratocyst by conven-
using selective T1 and T2 weighting, it is also possible to
tional radiographs and CT. Radiographs enabled accurate
clearly distinguish between odontogenic keratocysts and
evaluation of location, margins, and the relationship to the
more solid lesions such as ameloblastoma (Borghesi
teeth, but CT showed a higher resolution and allowed addi-
et al. 2018). The use of MRI to differentiate odontogenic
tional assessment of bucco-­lingual expansion and perfora-
keratocysts from ameloblastomas and other cysts was the
tion of the cortical bone (Figure 7.9). All the ameloblastomas
basis of a study by Minami et  al. (1996). Their sample
included 19 keratocysts, 11 ameloblastomas, and 13 other
jaw cysts. They found that MRI of the ameloblastomas
differed from the keratocysts in displaying a mixed solid
and cystic pattern and irregularly thickened walls in all 11
cases, papillary projections in 7, and strong enhancement
of solid components in 9. Odontogenic keratocysts
showed a purely cystic pattern in 13 of the 19 cases, while
4  were mixed cystic and solid and 2 appeared wholly
solid. The walls of the keratocysts were uniformly thin
with weak enhancement compared to thicker walls and
stronger enhancement in ameloblastomas. These authors
suggested that MRI was able to accurately differentiate
odontogenic keratocysts from ameloblastomas in all their
cases, although some other cysts showed similar findings
to the keratocysts. T1 and T2 weighting of images shows
that keratocysts tend to be heterogeneous, but ameloblas-
tomas are homogeneously hypointense (grey) on
T1 weighting and hyperintense (white) on T2 weighting
(Minami et al. 1996; Van Rensburg et al. 2003; Borghesi
Figure 7.9  A coronal computed tomography (CT) scan enables et al. 2018). Figure 7.10 shows a maxillary lesion that has
accurate visualisation of the extent of this large keratocyst. The been visualised using a T2-­weighted MRI image with het-
lesion has filled the antrum with displacement of the lateral erogeneous intensity. The lesion is well demarcated and
wall of the nose and has raised the floor of the orbit. There is
its extent and lobulated outline can be clearly seen. Such
also evidence of perforation of the cortical bone (arrows). This
lesion was diagnosed as a solid odontogenic keratocyst (see a clearly demarcated image would not be possible using
Figure 7.21). conventional radiographs.
104 Odontogenic Keratocyst

Radiological Differential Diagnosis The most common single site for odontogenic keratocyst
is the posterior mandible and at this site the three most com-
The clinical features of the odontogenic keratocyst
mon lesions that must always be considered are dentigerous
(Table  7.2; Boxes  7.1 and  7.2) are not diagnostic and the
cyst, keratocyst, and ameloblastoma. About 35% of kerato-
first opportunity to make a preoperative diagnosis is usu-
cysts are associated with an unerupted tooth (Table 7.2) and
ally after examination of radiographs or imaging. The radi-
dentigerous cyst is probably the most common provisional
ological features of a well-­demarcated radiolucency are
radiological diagnosis. Myoung et  al. (2001) and Brannon
shared by many lesions, and for simple unilocular lesions
(1976) found that 27.1% and 25.1%, respectively, of kerato-
the differential diagnosis may include a wide range of
cysts were provisionally diagnosed as dentigerous cyst.
benign cysts and tumours of the jaws as well as other
Other diagnoses included ameloblastoma (11.6% and 9.5%,
lesions, including giant cell granulomas. For multilocular
respectively), residual cyst (9.7% and 19.2%), and radicular
lesions the differential diagnosis may be more limited.
cyst (3.1% and 5.3%). Myoung et al. (2001) also found that a
majority of lesions (39.9%) were given a correct preoperative
diagnosis of odontogenic keratocyst (although the term pri-
mordial cyst was also used). Stoelinga (2001) found that
51.2% of his cases presented with radiographic features sug-
gestive of a keratocyst, while the remainder could not be dis-
tinguished from other odontogenic cysts, including 13.4% of
cases that were in a dentigerous relationship.
The differential diagnosis of lesions at the angle of the
mandible and in a pericoronal relationship with impacted
third molars has been discussed in detail in Chapter 5. At
this site dentigerous cysts are by far the most common,
comprising more than 70% of lesions (Curran et al. 2002;
Patil et  al.  2014; Mello et  al.  2019), but the frequency of
odontogenic keratocysts and ameloblastomas may be simi-
lar, reported as about 9.5% and 7.5%, respectively (Mello
Figure 7.10  Coronal magnetic resonance imaging (MRI) et  al.  2019). Patil et  al. (2014), however, reported that
showing the lobularity (arrow) of a large maxillary odontogenic ameloblastomas were twice as common (16%) at this site as
keratocyst. The image is T2 weighted and shows a
heterogeneous signal intensity. Source: Prof van Rensburg 2003, keratocysts (8%), a finding also noted by MacDonald-­
p.311-466 / With permission of Elsevier. Jankowski and Li (2010). Dentigerous cysts are always

Box 7.2  Odontogenic Keratocyst: Clinical Presentation and Radiology


Clinical presentation
●● Over 50% present with swelling, with or without pain
●● Up to about 20% present as an unexpected incidental finding on radiographs
●● 10–15% may be infected or have a discharge
●● Nerve involvement (numbness or paraesthesia) is rare (<2%)
●● Peripheral lesions are rare, but presentation is indistinguishable from gingival cyst
Radiology
●● A characteristic feature is marked mesio-­distal extension with minimal bucco-­lingual expansion
●● About 75% are well demarcated and unilocular
●● 40–50% are unilocular, but with a scalloped or lobulated outline
●● Most lesions (over 90%) have a corticated margin
●● Only about 10% are truly multilocular
●● Multilocular or multilobular lesions are most frequently seen at the angle of the mandible
●● Larger lesions are more frequently multilobular or multilocular
●● Up to 50% are associated with a missing or impacted tooth
●● About 30% may be in a dentigerous relationship with an unerupted tooth.
­Radiological Feature  105

unilocular with a corticated margin, and are attached to often rely on an incisional biopsy. For larger lesions, how-
the tooth in the cervical region. Larger cysts may become ever, it may be possible to predict more accurately a diagno-
lobulated, but they rarely show significant extension into sis of keratocyst. The fusiform expansile pattern of the
the ramus. They tend to grow in a ballooning fashion and keratocyst has been mentioned above, and this contrasts
to show a similar degree of bucco-­lingual and mesio-­distal markedly with the ball-­like or ballooning expansion of the
expansion. Odontogenic keratocysts may be very similar, ameloblastoma (MacDonald  2016; Alves et  al.  2018;
especially while small, and an accurate diagnosis may not Kitisubkanchana et  al.  2021). This pattern of growth in
be possible (Figure  7.7; see also Figure  5.14). However, ameloblastoma is illustrated in Chapter  10 (Figure  10.5)
keratocysts are more often scalloped or lobulated and about and is so characteristic as to be almost diagnostic. The only
10% are multilocular. Keratocysts may also grow to larger other lesion that may show this pattern is odontogenic
sizes and mesio-­distal extension often exceeds bucco-­ myxoma, but the myxoma tends to be more radiolucent
lingual expansion. As mentioned previously, this fusiform and may often show incomplete bony septae. To fully
swelling is so characteristic of the odontogenic keratocyst appreciate these features, three-­dimensional imaging
that MacDonald (2016) regarded it as pathognomonic. would now be regarded as essential and the role of CT and
Odontogenic keratocysts may also grow in the ramus MRI has been discussed.
posterior to the third molars. At this site an ameloblastoma Kitisubkanchana et al. (2021) compared the radiological
is the most likely alternative diagnosis. Ameloblastomas, features of odontogenic keratocysts (n = 100) and amelo-
however, are more often multilocular, and so a unilocular blastomas (n = 101) and showed that keratocysts were sig-
radiolucency in the ramus of the mandible that is not asso- nificantly more often associated with an impacted tooth
ciated with an unerupted tooth is most likely to be an odon- (47% compared with 19% of ameloblastomas), but less
togenic keratocyst (Figure 7.11). often caused tooth displacement (29% compared with 53%)
The differential diagnosis of multilocular lesions is also or root resorption (6% compared with 62%). Keratocysts
problematic, but the number of possible diagnoses is more were also more frequently unilocular (82% compared with
limited. Ameloblastoma is the most important, but for 32%) and with a smooth or non-­scalloped outline (60%
small lesions on plane radiographs it is difficult to accu- compared with 23%).
rately differentiate between an ameloblastoma and an Other lesions to consider in the differential diagnosis of
odontogenic keratocyst. The lesions show a similar age, a multilocular radiolucency are botryoid odontogenic cyst
site, and sex distribution and so a definitive diagnosis will and glandular odontogenic cyst. These are discussed in
Chapters 8 and 10, respectively. Botryoid odontogenic cysts
are always lateral to a tooth, present in the canine–premo-
lar region, and rarely reach 40 mm in diameter. The glan-
dular odontogenic cyst may grow to a large size and may
show a multilocular pattern identical to an ameloblastoma
or keratocyst. Its growth pattern is also very similar to the
keratocyst, with extensive mesio-­distal extension and rela-
tively little bucco-­lingual expansion (e.g. see
Figures  10.3–10.5), and 75% arise in the mandible.
However, glandular odontogenic cyst has a predilection for
the midline and canine–premolar region, and the average
age of presentation is about 50 years with a peak in the
sixth decade. They are very rare below 20 years of age.
Finally, a number of authors have drawn attention to the
potential for misdiagnosis of keratocysts as radicular cyst.
Small unilocular lesions in the dentate areas of the jaws
often present in association with the roots of the teeth. The
key diagnostic feature of the radicular cyst is the associa-
tion with a non-­vital tooth, but this is not specific and vital-
ity testing is not always reliable. There are a number of case
reports of the chance finding of keratocysts in the periapi-
cal region, and Ide et al. (2009) estimated that 0.03–0.1% of
Figure 7.11  A well-­demarcated unilocular radiolucency in the
ascending ramus, not associated with an unerupted tooth. Such periapical biopsies may show an odontogenic keratocyst.
a lesion is most likely to be an odontogenic keratocyst. Garlock et al. (1998) reviewed 239 odontogenic keratocysts
106 Odontogenic Keratocyst

and found that 8.8% (21 cysts) were found at the periapex, there have been suggestions that some keratocysts may
and in 12 of these (57.1%) the tooth was either non-­vital or arise from epithelial islands originating from the basal
had undergone prior root canal treatment. Two-­thirds of aspect of the overlying oral epithelium. The evidence for
patients presented symptoms of pain and/or swelling con- each will be briefly reviewed.
sistent with pulpal and periapical pathology. Most lesions
were small (less than 20 mm), but presented at an average Rest Cells of Dental Lamina
age of 56 years. From these data it can clearly be appreci- The dental lamina begins to form at about 37 days of devel-
ated why keratocyst would not be the most favoured diag- opment as a thickening of the epithelium in the early oral
nosis for a small radiolucency at this site. Ali and Baughman cavity (the stomatodeum). It develops as a series of out-
(2003) found a high frequency of periapical keratocysts in growths into the underlying mesenchyme, each of which
the maxilla, especially in the canine region. At this site develops through the bud, cap, and bell stages to give rise
25.9% of cases were provisionally diagnosed as a radicular to the enamel organ from which each tooth develops. The
cyst or periapical granuloma, and 27.8% as a lateral perio- tooth germ is attached to the overlying oral epithelium by
dontal cyst. However, a small majority (31.5%) were still an intact dental lamina, until the late bell stage after which
diagnosed correctly as keratocysts. it begins to disintegrate. Early studies of fetal tissues
From this discussion it can be seen that a preoperative (Stoelinga and Peters 1973; Moskow and Bloom 1983) have
diagnosis of large multilocular odontogenic keratocysts shown that as the dental lamina breaks down, multiple
can often be made because of their characteristic fusiform small epithelial islands, often with microcystic change, can
growth pattern and multilocular ‘soap-­bubble’ appearance. be found overlying the developing tooth. Moreillon and
Similarly, a large unilocular lesion in the ramus, not associ- Schroeder (1982) showed that these ‘microkeratocysts’ are
ated with a tooth, is most likely to be a keratocyst. The most frequently found in the gingivae or alveolus of infants and
problematic lesions to accurately diagnose are small uni- may give rise to gingival cysts of infants (see Chapter 9 and
locular lesions in the tooth-­bearing areas of the jaws that Figure 9.7). It is thought that after tooth development and
may be indistinguishable from common periapical lesions eruption are complete, most of the residues of dental lam-
(Ali and Baughman  2003; MacDonald et  al.  2013; ina involute, but some rest cells of dental lamina are
MacDonald  2016; Slusarenko da Silva et  al.  2019a). undoubtedly retained in the mucosae and in the alveolar
MacDonald et  al. (2013) showed that lesions that were bone adjacent to the teeth.
diagnosed as keratocysts before surgery were significantly A number of observations from these studies are perti-
less likely to recur than lesions that had a provisional diag- nent to the following discussion. First, the epithelial rests
nosis of another cyst type. Ironically, because larger multi- and small microcysts are often found to be continuous
locular cysts were more likely to get an accurate preoperative and fused with the overlying oral epithelium (Stoelinga
diagnosis, the authors suggested that small, simple uniloc- and Peters 1973; Moreillon and Schroeder 1982; Moskow
ular odontogenic keratocysts are more likely to recur and Bloom  1983). Secondly, the remnants of the dental
(MacDonald et al. 2013; MacDonald 2016). Factors associ- lamina typically form strands that lie within the guber-
ated with recurrence of the odontogenic keratocyst are dis- nacular canal (also referred to as the gubernaculum den-
cussed at the end of this chapter. tis, or gubernacular tract). The gubernacular canal is a
bony channel that lies between the lamina propria of the
oral mucosa and the dental follicle of the developing suc-
Pathogenesis cessional (permanent) teeth. The canal contains fibrous
tissue (the gubernacular cord) that provides a pathway for
It is now widely accepted that the odontogenic keratocyst is the erupting tooth. Significantly, a gubernacular canal (or
associated with activation of the HH signalling pathway, most tract) may also aid eruption of the permanent molars, and
often as a result of alterations in the PTCH1 gene (reviewed by may persist into adult life. CT imaging has shown that it
Gomes et  al.  2009; Li  2011; Gomes et  al.  2017; Diniz forms a continuity between odontogenic lesions (amelo-
et al. 2017). Downstream targets of the HH pathway drive cell blastoma, odontogenic keratocyst, and dentigerous cyst)
proliferation and growth, and this almost certainly acts to ini- and the lamina propria of the oral mucosa (Oda
tiate cyst formation in odontogenic epithelial rest cells. et al. 2018). The gubernacular canal and its contents (rest
cells of dental lamina) have been implicated in the patho-
genesis of the adenomatoid odontogenic tumour
Source of Epithelium
(Philipsen et  al.  1992; Ide et  al.  2011) and odontomas
Most authors agree that the odontogenic keratocyst arises (Oda et al. 2016), but have not been discussed in the con-
from the odontogenic cell rests of the dental lamina, but text of the odontogenic keratocyst. Of significance is that
Pathogenesis  107

Oda et al. (2018) showed that patent gubernacular tracts From these studies, Stoelinga (2001, 2005) proposed that
were present in 46 of 50 (92%) odontogenic keratocysts odontogenic keratocysts, particularly in the ramus of the
studied, and were frequently seen just distal to the third mandible, may not arise from dental lamina rests, but from
molars, forming a continuity between the fibrous capsule epithelial islands derived as offshoots of the basal layer of
of the cyst and the lamina propria of the retromolar the oral epithelium, which are referred to as ‘basal cell
mucosa. They also showed that in about 25% of cases the hamartias’. Furthermore, he suggested that these islands
gubernacular canal was expanded to form a wider com- derived from the oral epithelium are a major cause of
munication through the anterior cortical plate of the recurrences, and advocated that when keratocysts are enu-
ramus. These communications are rarely seen on plane cleated, the overlying mucosa should also be excised.
panoramic radiographs, but when they have been noted, Stoelinga has also discussed the recurrence of odontogenic
have probably been interpreted as cortical perforation due keratocysts in bone grafts that had been used to repair the
to cyst growth. surgical defects after marginal or segmental resection.
Stoelinga and Slusarenko da Silva (2021) reviewed the lit-
Epithelial Islands Derived from Oral Epithelium erature and found seven well-­documented cases where a
Stoelinga and colleagues, in a number of studies keratocyst had recurred in a bone graft. The authors sug-
(Stoelinga  1976,  2001,  2005; Stoelinga and Peters  1973; gest that the source of the recurrence must be from the
Stoelinga et  al.  1975), have suggested that odontogenic overlying mucosa and cite such recurrences as further evi-
keratocysts may also arise from a proliferation of basal cells dence for the role of mucosal epithelial islands in the
of the oral epithelium, which results in islands ‘dropping pathogenesis of the odontogenic keratocyst.
down’ into the underlying lamina propria. In their early Most oral pathologists will have encountered proliferat-
studies (Stoelinga and Peters  1973; Stoelinga  1976), they ing islands of epithelium within the oral mucosa, espe-
undertook histological examination of fetal mandibles and cially in the region of the retromolar area or within the
demonstrated that the dental lamina extended distally to follicles associated with unerupted or impacted teeth. On
the second primary molars, suggesting that it became ‘sub- occasion these may be quite prominent and the term basal
merged’ into the deeper connective tissues where the tooth hamartia has been used (Ide and Kusama  2002b). An
germs of the successional teeth were formed. They pro- example of epithelial islands, similar to those described by
posed that dental lamina rests may be found in the gingivae Stoelinga and overlying a keratocyst, is shown in
of dentate areas of the jaws, but that distal to the third Figure  7.12. Many pathologists, however, would regard
molars, the dental lamina is adherent to the tooth follicle these as proliferating islands of odontogenic epithelium. In
and cannot give rise to keratocysts in the ascending ramus. a recent study of 167 biopsies taken from the edentulous
They also examined six keratocysts enucleated from the alveolus or retromolar area, Almazyad et al. (2020) found
third molar region (Stoelinga and Peters 1973). In all cases, that 11 (6.6%) contained odontogenic epithelial rests as a
the cysts were attached to the oral mucosa overlying the chance finding. Conversely, Diniz et  al. (2009) provided
anterior aspect of the ascending ramus. These authors some support for Stoelinga’s hypothesis. They investigated
attributed this attachment to the presence of a fenestration PTCH1 splice variants (three isoforms: exons 1b, 1d, and
through the cortical plate of the ascending ramus. 1e) and found that exon 1b was expressed in 90% of odon-
Histological examination showed epithelial islands and togenic keratocysts and in 83% of samples of oral mucosa
occasional microcysts within the connective tissues of the adjacent to keratocysts. This suggests that the mucosa adja-
oral mucosa overlying the cysts. No islands were found in cent to a keratocyst may harbour similar alterations in the
areas of the cyst walls away from the mucosal aspect. The PTCH gene and may therefore have the potential to form
islands were described as odontogenic or basal cell like, new lesions. However, Diniz et al. (2009) were not able to
although some appeared to be simple squamous epithe- undertake histological examination of their mucosal sam-
lium. In a later study (Stoelinga 2001), the author reported ples, so it remains possible that the source of the exon 1b
the treatment of 82 keratocysts over a period of 25 years. In was remnants of dental lamina in the mucosa rather than
44 cases, the oral mucosa overlying the cysts was excised oral epithelium.
and examined, and in 23 (52.3%) he found clusters of epi- Heikinheimo et al. (2007, 2015) investigated genomic
thelial islands or microcysts in the region where the cyst aberrations in sporadic odontogenic keratocysts using
wall was attached to the mucosa. Occasionally the islands cDNA expression arrays, comparative genomic hybridi-
or microcysts were fused with the oral epithelium. He sation, or genome wide expression analysis, and found
showed that the attachment of the cyst wall to the oral significant expression of genes associated with tooth
mucosa was via a perforation or fenestration in the bone development supporting an origin from dental lamina.
just posterior to the third molars. They also found overexpression of genes associated with
108 Odontogenic Keratocyst

If this is the case, the advice from Stoelinga that it is wise


to remove the attached overlying oral mucosa after enu-
cleation of a keratocyst is still valid and applicable. Such a
procedure is often used and may have an effect on reducing
recurrence (see later in this chapter).

Summary and Conclusions
Although it is possible that epithelial islands from the oral
mucosa may give rise to odontogenic cysts, this hypothesis
has not gained widespread support and the findings have
not been repeated or described by other workers. Overall,
the accumulated evidence supports the concept that the
odontogenic keratocyst arises from cell rests of the dental
lamina. Initiation of cyst formation is probably early in the
development of the teeth, involving alterations in the
PTCH1 gene (see below) before the dental lamina begins to
disintegrate at the bell stage of tooth morphogenesis. In
Figure 7.12  Islands and cords of epithelium in the oral mucosa
overlying an odontogenic keratocyst. In places these merge with
this way, the keratocyst lining derives from a clone of
the oral epithelium and have been referred to as ‘basal cell genetically altered epithelial remnants that give rise to the
hamartias’ (see text). Some islands show peripheral palisaded cyst, but may also explain the frequent presence of satellite
cells (arrows), typical of odontogenic epithelial rests. Source: cysts and also the propensity for recurrence if residual epi-
Courtesy of Dr Adalberto Mosqueda.
thelial islands remain after surgery (Browne 1975; Browne
and Smith 1991).
keratin 6B (Heikinheimo et al. 2007), as well as enrich-
ment of genes associated with the HH signalling path-
Hedgehog Signalling Pathway, PTCH Gene
way and with squamous epithelial differentiation
Alterations, and Odontogenic Keratocyst
(Heikinheimo et al. 2015), suggesting that the keratocyst
arises from remnants of dental lamina that have not dif- Hedgehog (HH) signalling is a fundamental feature of nor-
ferentiated towards formation of the enamel organ. They mal development, with crucial roles in cell fate, differentia-
found that genes enriched in ameloblastoma were asso- tion, and patterning. In normal odontogenesis, HH
ciated with later stages of tooth development, including activation through binding of the SHH ligand regulates the
genes of the WNT pathway. This supports the concept development of the dental lamina and is responsible for
that odontogenic cysts and tumours may arise from cells tooth morphogenesis and patterning. It is likely therefore
at different stages of tooth development (reviewed by that constitutive or aberrant activation of HH signalling in
Diniz et al. 2017). odontogenic epithelium initiates the formation of kerato-
An origin from dental lamina does not necessarily con- cysts. An understanding of tooth development and the role
tradict the findings of Stoelinga. The more recent demon- of the dental lamina is helpful for understanding cyst
strations of a patent gubernacular canal overlying pathogenesis, and readers are referred to specialist texts or
odontogenic keratocysts in adults (Oda et  al.  2018) are reviews for up-­to-­date and detailed accounts of odontogen-
quite in keeping with Stoelinga’s descriptions of fenestra- esis and the role of HH signalling (Nanci  2017; Diniz
tions in the anterior cortical plate in the retromolar region et  al.  2017; Seppala et  al.  2017; Hovorakova et  al.  2018;
of the ascending ramus (Stoelinga and Peters  1973; Sasai et al. 2019).
Stoelinga  1976,  2005). The gubernacular canal contains A simplified description of the HH signalling pathway is
remnants of the dental lamina, which are also seen extend- illustrated in Figure 7.13. The key regulator of the pathway
ing into the overlying oral mucosa (Moskow and is the PTCH protein, which is a transmembrane protein that
Bloom  1983; Philipsen et  al.  1992; Ide et  al.  2011; acts to suppress or inhibit SMO (smoothened) at the cell
Nanci  2017). It is possible therefore that the epithelial surface. PTCH is a receptor for SHH and under normal con-
islands found on the deep aspect of oral mucosa overlying ditions ligand binding releases SMO and results in con-
primary and recurrent keratocysts arise not from basal cell trolled and regulated activation of the HH signalling
buds ‘dropping off’, but from residual rest cells of dental pathway (Figure 7.13b). The downstream outcomes of sig-
lamina lying superficial to the opening of the gubernacu- nalling are tissue specific, but in odontogenesis HH signal-
lar canals. ling regulates epithelial–mesenchymal interactions, cell
Pathogenesis  109

(a)

H
SH

(b) (c)

SHH
SMO
X SMO
X SMO

PTCH 1 PTCH 1 PTCH 1


Cell membrane Cell membrane Cell membrane

SU
SUFU

SU
FU
GLi

FU
GLi GLi

Gene expression off Gene expression on Gene expression on


Nucleus Nucleus Nucleus

Figure 7.13  The hedgehog signalling pathway. (a) In the absence of ligand binding, the PTCH protein suppresses SMO at the cell
membrane and signalling and gene expression are switched off. (b) Sonic hedgehog (SHH) is the ligand for the PTCH receptor and,
when bound, PTCH inhibition of SMO is released and signalling is switched on in a controlled manner (dashed green arrow). Gli
proteins are destabilised by release from SUFU and are translocated to the nucleus where gene expression is switched on. (c)
Reduction or loss of PTCH protein results in ligand-­independent activation of the HH signalling pathway (solid green arrow). This may
occur as a result of mutation or loss of heterozygosity (LOH) of PTCH1.

proliferation, and cell fate in the dental lamina and tooth major cause of basal cell carcinomas and other neoplasms
germ, and is crucial for normal tooth morphogenesis. Gene in these patients. Loss of the second allele may be by a sec-
expression is regulated through the Gli transcription fac- ond (somatic) mutation or, more commonly, due to loss of
tors. In the absence of SHH ligand binding, Gli proteins are heterozygosity (LOH). Mutation or loss of both copies of a
inactive and bound to the protein SUFU (Figure 7.13a), but gene is referred to as the ‘two-­hit hypothesis’ of carcino-
on activation of SMO, SUFU is released and Gli migrate into genesis, first described by Knudson in 1971 (see
the nucleus (Figure 7.13b). The downstream target genes of Knudson  1996; Tomlinson et  al.  2001), but it should be
the pathway include the transcription factor SOX2 (impor- noted that two hits may not always cause neoplasia.
tant in odontogenesis), the anti-­apoptotic protein bcl-­2, and Alterations in the PTCH gene have also been demon-
cell cycle regulatory proteins (including cyclin D1). strated in sporadic odontogenic keratocysts, but in most
Alterations of the PTCH gene cause reduced expression or cases this has only involved one copy of the gene, resulting
loss of PTCH protein at the cell surface and results in ligand-­ in haploinsufficiency and reduced expression of PTCH
independent, constitutive activation of the HH signalling protein at the cell surface, with activation of the HH path-
pathway (Figure 7.13c). As discussed previously, PTCH muta- way. Alterations of the PTCH gene may be by somatic
tions are the cause of the NBCCS and are also involved in the mutations, LOH, or due to gene silencing by DNA methyla-
pathogenesis of odontogenic keratocysts  – both syndromic tion (Gomes et al. 2009). Table 7.6 summarises a number of
and sporadic. The majority of causative mutations are studies that have determined the frequency of PTCH alter-
frameshift, nonsense, or missense mutations in the PTCH1 ations in NBCCS and sporadic cysts. In general, the num-
gene (Guo et al. 2013; Reinders et al. 2018), but mutations or ber of cysts analysed is small, but the overall findings are
alterations in PTCH2, SUFU, and SMO have been implicated similar and consistent: 80% or more of NBCCS and spo-
in NBCCS and in sporadic keratocysts (Rui et al. 2014). radic cysts show mutations or LOH on chromosome 9q at
In NBCCS, germline mutations are found in one allele of the PTCH1 gene locus. Two things can be noted from these
PTCH and are inherited in an autosomal-­dominant man- data. First, not all NBCCS cysts show a mutation, despite
ner. Thus NBCCS patients will have reduced expression the fact that in the syndrome the mutations are found in
(haploinsufficiency) of PTCH protein, causing activation the germline. This is in part due to technical factors
of the HH pathway with the resulting developmental whereby mutations may not always be detected, but
defects. Loss of one PTCH allele predisposes patients to also because small numbers of cases may harbour muta-
neoplasia and loss of the second allele is thought to be the tions not yet found, or may involve mutations in other
110 Odontogenic Keratocyst

Table 7.6  Frequency of PTCH1 gene alterations (mutations or loss of heterozygosity, LOH) in naevoid basal cell carcinoma syndrome
(NBCCS) and sporadic odontogenic keratocysts. Based on Myoung et al. (2001) and Kahraman et al. (2018).

NBCCS cysts Sporadic cysts

PTCH1 alterations PTCH1 alterations

References n Mutations (%) LOH (%) n Mutations (%) LOH (%) Notes

Levanat et al. (1996) 6 – 83.3 14 – 28.6 3 (50%) NBCCS cysts showed


evidence of biallelic loss
Lench et al. (1996) – – – 6 – 50.0
Lench et al. (1997) 16 31.3 0 – – –
Barreto et al. (2000) 3 66.6 – 3 33.3 –
Agaram et al. (2004) – – – 10 – 60.0
Ohki et al. (2004) 4 0 – 9 77.8 –
Gu et al. (2006) 2 100 – 10 30.0 –
Sun et al. (2008) 10 80.0 – 20 25.0 – 2 (20%) NBCCS and 3 (15%) sporadic
cysts showed evidence of biallelic loss
Pan et al. (2010) 15 86.7 46.7 29 31.0 27.6 8 (53%) NBCC and 5 (17%) sporadic
cysts showed evidence of biallelic loss
Guo et al. (2013) 14 78.6 – 29 44.8 –
Shimada et al. (2013) 16 56.3 42.0 20 0 15.0 25% of sporadic cysts showed LOH at
PTCH2 and 15% at SUFU.
Qu et al. (2015) – – – 19 84.2 –
Stojanov et al. (2020) – – – 44 93.2 34.1 35 (80%) showed evidence of biallelic
loss

n, number of cysts analysed.

HH-­related genes, including PTCH2, SUFU, or SMO (dis- GLI1, and GLI2, as well high levels of the downstream tar-
cussed by Li 2011). This may also apply to sporadic cysts get SOX2.
(Rui et al. 2014), so the actual number of cysts with genetic Ohki et al. (2004) examined 18 sporadic and 4 NBCCS-­
alterations may be higher than so far discovered. Secondly, associated keratocysts and found expression of SHH,
the frequency of mutations found in cysts has increased in PTCH, SMO, and Gli1  mRNA and protein in all cases.
more recent studies. This is due to more advanced tech- Expression of PTCH and SMO was similar in the epithelial
niques (e.g. next-­generation sequencing), and also because lining and in the fibrous cyst wall, but expression of SHH
some more recent studies have used microdissection to iso- and Gli1 was higher in the epithelium, suggesting an ele-
late keratocyst epithelium and thus remove stromal con- ment of epithelial–mesenchymal cross-­talk in the signal-
tamination (Qu et al. 2015; Stojanov et al. 2020). Ohki et al. ling process. Hoyos Cadavid et al. (2019) confirmed these
(2004) and Shimada et  al. (2013) found no mutations in findings and showed immunohistochemical expression of
NBCCS and sporadic cysts, respectively, but this is unusual SHH, PTCH1, PTCH2, SMO, Gli1, Gli2, and Gli3 in both
and the reasons are uncertain. sporadic and NBCCS-­associated cysts.
A number of studies have confirmed that the HH signal- In a similar study, Vered et al. (2009) showed PTCH and
ling pathway is activated by examining the expression of Gli1 protein expression in all keratocysts studied (27 spo-
HH-­associated genes or proteins. Gurgel et al. (2014) ana- radic and 6 NBCCS associated), with intermediate to high
lysed 23 keratocysts and showed up-­regulation of the HH staining in all layers of the epithelial lining. Expression of
pathway–associated genes SMO, PTCH1, and GLI1, as well SMO was found mainly in the basal and parabasal layers
as overexpression of the downstream target genes for cyc- in 69% of primary sporadic keratocysts, 54% of recurrent
lin D1 and bcl-­2. In their gene expression analysis, cysts, and 50% of NBCCS-­associated cysts. However, this
Heikinheimo et  al. (2015) also found up-­regulation of expression was not specific to odontogenic keratocysts,
genes of the HH pathway, including PTCH1, PTCH2, SMO, since a similar pattern of PTCH and Gli1 staining was
Pathogenesis  111

found in ameloblastomas and also in orthokeratinised of the PTCH gene. Three of seven (43%) dentigerous cysts
odontogenic cysts, dentigerous cysts, and radicular cysts. and four of seven (57%) keratocysts showed evidence of
SMO expression was seen in 22% of ameloblastomas, 58% LOH. LOH was not detected in any radicular cysts. In a
of unicystic ameloblastomas, 33% of orthokeratinised further study, the same group (Pavelić et  al.  2001) con-
odontogenic cysts, and 40% of dentigerous cysts. SMO firmed these findings and found LOH in the same region in
was not expressed in radicular cysts. Vered et  al. (2009) five of ten (50%) dentigerous cysts, but not in radicular
also examined expression of the anti-­apoptoticprotein cysts. They also used reverse transcription polymerase
bcl-­2, which is known to be a downstream target of the chain reaction (RT-­PCR) in two cysts to show that the
HH pathway. Bcl-­2  was strongly expressed in the basal PTCH gene was still expressed despite evidence of LOH,
layers of all the odontogenic keratocysts, but only weakly suggesting that reduced expression of the PTCH protein
expressed in ameloblastoma. Orthokeratinised odonto- may still lead to activation of the HH signalling pathway.
genic cysts, dentigerous cysts, and radicular cysts did not These studies confirm that aberrations in the PTCH gene
express bcl-­2. are not specific to keratocysts, but may represent an initiat-
These findings confirm previous studies showing that ing event in the formation of developmental odontogenic
PTCH protein is expressed in a wide range of odontogenic cysts, possibly in a progenitor epithelial cell that then gives
lesions, including radicular cysts and dentigerous cysts rise to the entire epithelial lining. The resultant activation
(Pavelić et al. 2001; Barreto et al. 2002). It is probable that of the HH signalling pathway may then drive growth and
PTCH is constitutively expressed on the cell surface of the expansion of the cyst. Such a scenario could explain a role
epithelial linings, but there is an apparent anomaly in that for PTCH in dentigerous cysts, but does not exclude a fur-
PTCH protein is still expressed in cysts, even when there is ther role for specific PTCH mutations in keratocysts.
evidence of a mutation or LOH. This can be explained by Gomes and Gomez (2011) supported this view and sug-
the fact that usually only one copy of the gene is affected gested that PTCH1 may act as a gatekeeper gene for many
(haploinsufficiency), so the protein is reduced in amount types of odontogenic lesions, and that further genetic
but is still expressed and detectable. It is also possible that events drive the formation of different cysts or tumours.
the mutant protein, although inactive, still has the appro- However, more research is needed, since the studies show-
priate epitopes for immunohistochemical detection. ing PTCH alterations in dentigerous cysts (Levanat
Although these data show that PTCH gene alterations et al. 2000; Pavelić et al. 2001) have not yet been repeated
are important in keratocysts, they do not seem to be spe- and sequencing studies to investigate mutations of PTCH
cific, and there is evidence that mutations or LOH of the in dentigerous cysts have not been performed.
PTCH gene or activation of the HH signalling pathway may
be involved in other odontogenic lesions (Gomes and
Is the Odontogenic Keratocyst a Neoplasm?
Gomez 2011), including orthokeratinised odontogenic cyst
(Diniz et  al.  2011), calcifying epithelial odontogenic It has long been thought that the odontogenic keratocyst
tumour (Peacock et al. 2010), glandular odontogenic cyst may be a benign cystic neoplasm, rather than a develop-
(Zhang et  al.  2010), and dentigerous cyst (Levanat mental cyst. Paul Toller (1967) first suggested that the
et al. 2000; Pavelić et al. 2001; Barreto et al. 2002; Zhang keratocyst had ‘some of the potentialities of a benign neo-
et  al.  2010). It should also be noted that LOH of other plasm’, but the debate was fully initiated by Ahlfors et al.
tumour-­suppressor genes, including p16, p53, MCC, (1984), who examined 319  keratocysts from 255 patients
TSLC1, LTAS2, and FHIT (Agaram et  al.  2004; Henley and found one or more recurrences in 69 (27%). They also
et al. 2005), has been found in keratocysts, but the signifi- demonstrated variable histological appearances of the cyst
cance of these findings remains uncertain, since these linings with evidence of a proliferative activity in some
changes may also be seen in radicular and dentigerous cysts, including budding of the basal layer in 25%, epithe-
cysts (Malcić et al. 2008). Overall, however, these data sug- lial islands in the cyst wall (23%), satellite cysts (7%), and
gest that LOH is not uncommon and, although it may pro- mural proliferations (8%). However, these features were
mote cell proliferation and growth, it cannot be regarded as not associated with recurrence and were seen most fre-
a specific pathogenetic event. However, these are tumour-­ quently in cases of multiple cysts or in cysts associated
suppressor genes and their loss is also associated with with NBCCS. Nevertheless, based on these observations
many types of neoplasms. they proposed a mechanism of growth involving down-
Of particular interest are studies that have shown LOH growth of the epithelial lining into the cyst capsule, and
at the PTCH1 locus in dentigerous cysts. Levanat et  al. suggested that the lesion should be regarded as a benign
(2000) examined seven each of dentigerous cysts, radicular cystic neoplasm. These early papers cited a high recurrence
cysts, and odontogenic keratocysts for LOH in two regions rate, an unusual growth pattern, and increased cell
112 Odontogenic Keratocyst

proliferation as evidence of neoplastic origin. Subsequently, is widely accepted that the key driver for formation of the
the neoplastic nature of the odontogenic keratocyst has odontogenic keratocyst is activation of the HH signalling
been much debated in the literature and has been reviewed pathway and that in the vast majority of cases this is due to
in the previous edition of this book and by others ligand-­independent activation following alterations in the
(Shear  2002a,b,c; Gomes and Gomez  2007; Gomes levels of PTCH protein receptor secondary to a genetic
et al. 2009; Li 2011; Bhargava et al. 2012). aberration. It is also discussed that alterations of the PTCH
Most textbooks still define neoplasia in the terms first gene may not be a simple mutation, but can be due to a
used by Willis in 1960, as ‘an abnormal mass of tissue, the number of genetic events. In NBCCS there is a germline
growth of which exceeds and is uncoordinated with that of mutation in one allele of PTCH, which gives rise to the
normal tissues, and persists in the same excessive manner developmental anomalies and also to NBCCS-­associated
after cessation of the stimuli that evoked the change’. This keratocysts. Single mutations have been found in sporadic
implies autonomous and uncontrolled growth, and on this keratocysts, but sporadic cases may also be associated with
basis the odontogenic keratocyst would not meet the crite- LOH at the PTCH locus (Table 7.6). In most cases there is
ria for a neoplasm. The reasons for the high recurrence evidence of change in only one allele of PTCH (‘one hit’),
potential are discussed later in this chapter, and although presumably resulting in haploinsufficiency and a reduced-­
this is suggestive of an element of autonomy, it appears level of PTCH receptor protein sufficient to activate the HH
insufficient to categorise the behaviour of all keratocysts as pathway. The important point is that LOH or haploinsuffi-
neoplastic. A further strong challenge to the concept of ciency of PTCH and HH pathway activation is not specific
neoplasia is the now commonly reported finding of a good to the odontogenic keratocyst but, as discussed previously,
response to marsupialisation, with regression of the typical may be seen in developmental cysts, including orthokerati-
keratocyst lining to an epithelium indistinguishable from nised odontogenic cyst and dentigerous cyst (Levanat
that seen in normal oral mucosa (Marker et  al.  1996; et al. 2000; Pavelić et al. 2001; Barreto et al. 2002; Gomes
Ninomiya et  al.  2002; August et  al.  2003; Pogrel  2003b; and Gomez 2011; Diniz et al. 2011). It is also the case that
Pogrel and Jordan 2004; Zhang et al. 2014). Pogrel (2003b) single-­point mutations cannot define neoplasia, since a
treated 13 patients with large or surgically problematic number of disorders thought to be non-­neoplastic or devel-
keratocysts with marsupialisation. The cyst cavities were opmental are known to harbour specific gene mutations,
kept open and cleaned for follow-­up periods ranging from including fibrous dysplasia (GNAS1; Idowu et  al.  2007),
1.9 to 6.9 years. In this and in a follow-­up paper (Pogrel and Cherubism (SH3BP2; Ueki et  al.  2001), and giant cell
Jordan  2004), the authors found that all the keratocysts lesions of the jaws (KRAS, TRPV4, FGFR1; Gomes
resolved and histological examination of the retained cyst et al. 2020).
wall at the base of the healed cavities showed normal epi- Notwithstanding this argument, there remains good evi-
thelium only, with no signs of cystic remnants, daughter dence that at least a subset of odontogenic keratocysts may
cysts, or budding of the basal layer. The role of marsupiali- be neoplastic. The most compelling evidence for a neoplas-
sation in the management of the odontogenic keratocyst is tic origin is that a number of studies have shown that
discussed later in this chapter (see ‘Treatment and keratocysts may harbour biallelic loss (‘two hits’) of both
Recurrence of the Odontogenic Keratocyst’), but it is often copies of the PTCH gene (Table  7.6; Levanat et  al.  1996;
cited as the main argument against a neoplastic origin. Sun et  al.  2008; Pan et  al.  2010; Stojanov et  al.  2020).
On the other hand, it is known that neoplasia is accom- According to Knudson’s two-­hit hypothesis, loss of both
panied by (often multiple) genetic aberrations and it is alleles is a crucial step towards neoplasia and in some cases
increasingly apparent that neoplasia is being defined, may be sufficient for neoplastic change. The concept was
rightly or wrongly, on the basis of the findings of genetic first developed to explain the onset of childhood retino-
changes. An important driver of neoplasia is thought to be blastoma. The first hit was a germline (inherited) mutation
loss of both copies of a tumour-­suppressor gene following in the retinoblastoma gene (RB1), followed by a second
the ‘two-­hit hypothesis’ of Knudson (Knudson  1996). In somatic mutation resulting in lesions in early childhood.
the case of the odontogenic keratocyst, alterations in the Occasional retinoblastomas may arise by two somatic ‘hits’,
PTCH gene discussed in the previous section have been usually a mutation followed by LOH. RB1 acts as a tumour-­
taken as unequivocal evidence that the lesion is a neo- suppressor gene and loss of one allele due to a germline
plasm. However, as discussed in a number of excellent mutation results in an inherited predisposition to cancer.
reviews (Gomes and Gomez  2007; Gomes et  al.  2009; The second ‘hit’ may involve truncating mutations, LOH,
Li  2011), the situation is more nuanced and evidence of or DNA silencing by methylation. Although two hits may
PTCH alterations cannot be taken as unconditional evi- initiate neoplasia, multiple genetic events are usually
dence of neoplasia. As discussed in the previous section, it needed for lesion development. Normal expression of the
Pathogenesis  113

PTCH protein receptor supresses activation of the HH sig- Summary and Conclusions


nalling pathway, so PTCH acts as a tumour-­suppressor The accumulated evidence from these studies shows that
gene, and the pathway to neoplasia in the odontogenic activation of the HH signalling pathway appears to under-
keratocyst may follow the two-­hit process, analogous to pin the initiation and growth of the odontogenic keratocyst
retinoblastoma. and that in the vast majority of cases this is associated with
In the very first paper to show PTCH alterations in odon- alterations in the PTCH1 gene. However, the line of demar-
togenic keratocysts, Levanat et al. (1996) also provided the cation between aberrant development and neoplasia is
first molecular evidence of a two-­hit mechanism for the blurred and it seems that the keratocyst may sit on a spec-
pathogenesis of these cysts. They examined six keratocysts trum between a developmental cyst and a benign cystic
from four NBCCS patients and fourteen sporadic cysts for neoplasm. The PTCH gene alterations may be brought
polymorphisms in the PTCH region on chromosome 9. Five about by a one-­hit or a two-­hit mechanism involving muta-
of the six (83.3%) syndrome cysts and four (28.6%) sporadic tions or LOH. In NBCCS all cysts are associated with at
cysts showed LOH at two or more loci. Furthermore, three least one germline mutation, and the majority of sporadic
cysts from two NBCCS siblings showed evidence of two hits cysts show one alteration via a mutation or LOH in one
in the form of a germline mutation in one allele and LOH in allele. This suggests that reduced PTCH receptor protein
the second allele. Subsequently, these findings were con- expression (due to haploinsufficiency) is sufficient to acti-
firmed in two papers from T-­J Li’s research group (Sun vate the HH pathway and is an important pathogenic
et al. 2008; Pan et al. 2010). These authors showed evidence mechanism in many cysts. As noted previously, this is not
of biallelic loss in up to about 50% of NBCCS cysts and 17% specific to keratocysts, since LOH and HH activation are
of sporadic cysts (Table 7.6). Sun et al. (2008) found that two also seen in dentigerous cysts and other non-­neoplastic
patients with NBCCS and two sporadic cases showed bial- lesions.
lelic mutations, while the third sporadic case showed a In addition, however, a number of keratocysts are associ-
mutation in one allele and LOH at the second allele. In their ated with two hits on the PTCH1 gene, strongly suggesting
second study (Pan et al. 2010), the group found alterations that at least a subset of odontogenic keratocysts are truly
in PTCH1 in 27 of 44 (61.4%) keratocysts, with evidence of neoplastic. In these cases, loss of PTCH protein leads to
biallelic loss in 13 cases (8 NBCCS and 5 sporadic cysts), of constitutive activation of the HH pathway and this may be
which 10 showed inactivation of one allele by mutation and associated with a different phenotype or with different
the second by LOH, and 3 contained two inactivating muta- clinical behaviour. Because patients with NBCCS have a
tions. Overall, 53.3% of NBCCS cysts and 17.2% of sporadic PTCH germline mutation, they are much more likely to
cysts were found to harbour two hits, while 33.3% and have a second hit and keratocysts in syndrome patients are
31.0%, respectively, showed one hit. It must be noted that 2 probably more likely to be truly neoplastic than sporadic
(13.3%) NBCCS cysts and 51.7% of sporadic cysts showed no cysts. It remains to be determined, preferably in controlled
PTCH1 alterations at all. In these cases, aberrations in other prospective studies, if there are clinicopathological differ-
genes associated with the HH pathway may be involved, ences between cysts showing different genetic aberrations.
including PTCH2, SMO, and SUFU (Shimada et  al.  2013; Retrospective studies have suggested that this is the case,
Rui et al. 2014). since there are a number of histological differences
A more recent study has suggested that as many as 80% between NBCCS and sporadic cysts and there is evidence
of sporadic odontogenic keratocysts may show biallelic loss that the genotype may affect rates of cell proliferation, his-
of PTCH1 (Stojanov et  al.  2020). Using next-­generation tological features, and recurrence (Yagyuu et al. 2008; Pan
sequencing, these authors found PTCH1-­inactivating et al. 2010; Shimada et al. 2013). This will be discussed later
mutations in 93% of 44 cases and LOH in the PTCH1 locus in this chapter in ‘Histopathology’ and ‘Recurrence of the
in 34%. Furthermore, 20 cases showed two biallelic PTCH1 Odontogenic Keratocyst’.
mutations and 15 cases showed a single mutation and LOH
in the second allele. Overall, therefore, 35 of 44 cases
Growth and Enlargement of the
(79.5%) showed evidence of two hits on the PTCH1 gene.
Odontogenic Keratocyst
The authors interpreted their findings to support the clas-
sification of keratocysts as a benign cystic neoplasm. They The odontogenic keratocyst has a different pattern of
attributed their high yield of positive cases to dissection of growth to most other odontogenic cysts. In particular, the
the cyst epithelium, thus removing contamination by stro- two most common cysts, the radicular and dentigerous
mal DNA. It is worth noting, therefore, that they found no cysts, grow in a centripetal or ‘ballooning’ pattern (dis-
evidence of mutations or LOH in any other HH pathway cussed in Chapters  3 and  5) to produce a characteristic
genes (SMO, SUFU, GLI1, GLI2). spherical radiolucency. It is thought that this pattern of
114 Odontogenic Keratocyst

growth is mediated primarily by increased intracystic pres- over 90% of Ki-­67–positive cells found in the suprabasal
sure due to osmosis across the cyst wall. In contrast, the layers (Figure 7.14) and few in the basal layers. In contrast,
odontogenic keratocyst appears to grow in a mesio-­distal about 80% of Ki-­67–positive cells in dentigerous cysts and
direction, with minimal expansion of the cortical plates. radicular cysts were located in the basal layer. Furthermore,
The characteristic appearance on plane radiographs or CT there was no significant difference in the Ki-­67 count
is of a fusiform expansion with large lesions extending between sporadic and recurrent keratocysts, but the num-
through the mandible, but with minimal bucco-­lingual ber of Ki-­67–positive cells in NBCCS-­associated lesions
expansion. This pattern of growth is thought to be mediated (n = 9; 91.8 ± 35.6 cells/mm) was significantly greater than
by increased cell proliferation of the epithelial lining, with in the other groups (Li et al. 1995).
growth of the cyst through the cancellous bone in an irregu- In a more recent discussion of these data, Li (2011) con-
lar lobulated or locular pattern (see ‘Radiological Features’). cluded that their finding of no significant differences in
Many papers have shown that proliferation in the epithe- cell proliferation between sporadic and recurrent kerato-
lial lining of the odontogenic keratocyst is greater than in cysts suggested that incomplete removal, rather than
dentigerous or radicular cysts, but is similar to that seen in intrinsic growth, was likely to be responsible for the
ameloblastoma. The early literature up to 2002  has been reported high rates of recurrence. On the other hand, the
reviewed by Shear (2002a,b). The very first study was by finding that Ki-­67–positive counts in NBCCS-­associated
Toller (1971), who estimated mitotic activity in an autoradio- cysts were almost twice the level of those in sporadic cysts
graphic study following the in vitro incubation of cyst linings indicated a greater level of proliferative activity in syndro-
with tritiated thymidine. He showed mean labelling indices mic lesions. This was reflected in the multiplicity of cysts
of 13.0% for a series of six odontogenic keratocysts, com- and the increased numbers of satellite cysts and epithelial
pared with 1.7% for five other odontogenic cysts and 7.0% for islands in the cysts of NBCCS patients. This suggested
normal oral mucosa. Some of the earliest, but still most com- increased cell proliferation may be related to PTCH altera-
prehensive, immunohistochemical studies were undertaken tions and activation of the HH signalling pathway. This was
by Li and colleagues (Li et  al.  1994,  1995). These authors confirmed in a later study from the same group (Pan and
investigated the expression of proliferating cell nuclear anti-
gen (PCNA) and Ki-­67 in odontogenic keratocysts, dentiger- (a)
ous cysts, and radicular cysts and found that expression was
consistently higher in keratocysts and was also suprabasal.
In their first study (Li et al. 1994), they found that the epithe-
lial linings of keratocysts (n  =  11) contained the highest
number of PCNA-­positive cells, most of which were in the
suprabasal layers, with fewer than 5% in the basal layer (e.g.
see Figure  7.14). The total PCNA count in the keratocysts
had a mean value of 94.4 ± 22.7 cells/mm, and was signifi-
cantly higher than in dentigerous cysts (n = 10; 5.1 ± 3.0 cells/
mm) or radicular cysts (n = 10; 11.0 ± 4.1 cells/mm). These
differences were entirely due to the higher number of PCNA-­
positive cells in the suprabasal layers of the keratocysts,
because there were no significant differences in positive cells
in the basal layers. These authors also found that PCNA (b)
expression in the keratocysts was similar to that seen in nor-
mal oral epithelium.
In their second study, Li et  al. (1995) investigated the
expression of Ki-­67 in 27 odontogenic keratocysts (10 spo-
radic, 8 recurrent, 9 associated with NBCCS), 5 dentigerous
cysts, 5 radicular cysts, and 7 samples of normal oral
mucosa. The number of Ki-­67–positive cells in the sporadic
keratocysts (53.1 ± 17.8 cells/mm) was similar to normal
epithelium (42.5 ± 12.7 cells/mm), but was significantly
greater than in dentigerous cysts (3.9 ± 1.3 cells/mm) or
Figure 7.14  (a) Ki-­67 and (b) proliferating cell nuclear antigen
radicular cysts (6.7 ± 4.8 cells/mm). This study also found (PCNA) expression in odontogenic keratocysts. Most positive
that the pattern of staining was the same as for PCNA, with cells are found in the suprabasal layers.
Pathogenesis  115

Li 2009), when it was shown that Ki-­67 labelling in kerato- significantly greater in keratocysts and were more often
cysts with PTCH1 mutations (n = 29; 103.29 ± 35.47 cells/ found in the suprabasal layers. Bcl-­2 was expressed only
mm) was significantly higher than in keratocysts with no in the basal layers, but was significantly greater in kerato-
mutations (n = 33; 69.82 ± 24.28 cells/mm). The study also cysts (99.6 ± 0.5% of basal cells) than in dentigerous cysts
found that the labelling index was higher in lesions with (3.4 ± 2.2%). This confirmed the findings of Lo Muzio
truncating mutations compared to non-­truncating muta- et  al. (1999) that bcl-­2  was almost exclusively found in
tions, and was highest in NBCCS-­associated cysts (n = 20; the basal cell layer of keratocysts. The authors suggested
115.56 ± 32.33 cells/mm). that these data explain why keratocysts grow as cystic
Many papers have confirmed these findings and have lesions rather than a solid mass. They concluded that
consistently shown that cell proliferation-­associated mark- apoptosis occurs at a high rate in the superficial layers of
ers (including Ki-­67, PCNA, p53, and p73) are expressed the epithelial lining, while at the same time bcl-­2 inhib-
suprabasally, and more strongly in keratocysts compared to ited apoptosis to facilitate cellular proliferation in the
other cysts (Ogden et  al.  1992; Thosaporn et  al.  2004; Lo basal layer. By such a mechanism, the higher rate of cell
Muzio et al. 2005; Kolář et al. 2006; de Oliveira et al. 2011; proliferation was regulated by the apoptotic activity, so
Soluk Tekkeşın et al. 2012a; Hammad et al. 2020) and are that these lesions maintain a cystic lumen and do not
also more highly expressed in NBCCS-­associated cysts (EI form solid masses (Kichi et al. 2005).
Murtadi et al. 1996; Lo Muzio et al. 1999). Cyclin D1 has These studies show that there is a higher rate of cell pro-
also been shown to be up-­regulated in keratocysts, with liferation in odontogenic keratocysts than in other cyst
greater expression in NBCCS cysts and in sporadic cysts types that show a typical centripetal or spherical growth
with PTCH gene alterations (Lo Muzio et al. 1999; Shimada pattern (dentigerous and radicular cysts), and support the
et al. 2013). Studies have further shown that the rate of pro- concept that expansion of the keratocyst is primarily driven
liferation in keratocysts is similar to that found in amelo- by mural growth due to proliferation of the epithelial lin-
blastomas (Thosaporn et  al.  2004; Soluk Tekkeşın ing. Furthermore, it is suggested that a balance between
et al. 2012a; Hammad et al. 2020), but the significance of cell proliferation in the basal and immediate suprabasal
this is uncertain, since the rate may also be similar to pro- layers is counterbalanced by apoptosis in the superficial
liferation in normal oral epithelium (Li et al. 1994, 1995). layers, maintaining the integrity of the cyst lumen (Kichi
Bcl-­2 is an anti-­apoptotic protein that promotes growth et al. 2005).
and is transcriptionally regulated by the HH signalling Although cell proliferation may explain growth of the
pathway. A number of studies have shown increased cyst lining, it may still be insufficient to entirely explain the
expression of bcl-­2 in odontogenic keratocysts compared to expansion or enlargement of the cyst, and there is evidence
other cysts (Lo Muzio et al. 1999; Kolář et al. 2006; Vered that intraluminal pressure may still play a role. In early
et al. 2009; Soluk Tekkeşın et al. 2012a; Shimada et al. 2013) studies, Toller (1970b) considered the part played by intra-
and have also found that expression is associated with luminal pressure and the osmolality of the cyst fluid in
NBCCS-­associated cysts and with PTCH alterations enlargement of odontogenic keratocysts. He showed that
(Shimada et  al.  2013). More recently, Zhang et  al. (2018) the mean cyst fluid osmolality of odontogenic keratocysts
found increased Ki-­67, PCNA, and bcl-­2  in keratocysts (296 ± 15.16 mOsm) was similar to that of dentigerous cysts
compared to dentigerous cysts, and further showed that (291 ± 14.42 mOsm) and radicular cysts (290 ± 14.93 mOsm)
increased proliferative and anti-­apoptotic activity corre- and was higher than the mean serum osmolality
lated to expression of the AP-­1 signalling pathway tran- (282 ± 14.75 mOsm). In view of the low total soluble pro-
scription factors Fra-­1, c-­Jun, and c-­Fos. tein level in keratocysts (Toller  1970a), he suggested that
To investigate the pattern of growth of the keratocyst, osmotic differences between sera and cyst fluids may be
Kichi et  al. (2005) investigated Ki-­67, p53, and bcl-­2 the result of the liberation of the products of cell lysis at the
expression in 20 odontogenic keratocysts and 20 denti- luminal surface. He believed strongly that the raised osmo-
gerous cysts and compared expression to the location of lality has an important role in the expansive growth of all
apoptotic cells, using the TUNEL method (Terminal jaw cysts, including keratocysts.
deoxynucleotidyl Transferase Biotin-­dUTP Nick End Main (1970), on the other hand, felt that mural growth in
Labelling). They showed that apoptotic cells were located the form of epithelial proliferation was the essential pro-
exclusively in the surface layer of the lining epithelium cess involved in the enlargement of the odontogenic
of both keratocysts and dentigerous cysts, but were far keratocyst and that the evidence for osmotic diffusion was
more common in keratocysts (25.5 ± 2.1% of surface inconclusive. This view was supported by Browne
cells) than in dentigerous cysts (5.5 ± 1.8%). Numbers of (1970,  1975) and Kramer (1974). They believed that the
proliferating cells (Ki-­67 and p53 positive) were multilocular and loculated outlines exhibited by many
116 Odontogenic Keratocyst

keratocysts were difficult to interpret on the basis of uni- immunohistochemical study to determine the frequency of
centric hydrostatic expansion alone. This form of cyst out- stromal myofibroblasts (α smooth muscle actin [αSMA]–
line suggested a multicentric pattern of cyst growth brought positive cells) in a range of odontogenic cysts and tumours,
about by the proliferation of local groups of epithelial cells including 8 odontogenic keratocysts, 9 orthokeratinised
against the semi-­solid cyst contents. odontogenic cysts, and 7 dentigerous cysts. Of the odonto-
Kubota et al. (2004), however, have confirmed the find- genic cysts, keratocysts had the highest number of αSMA-­
ings of Toller and found similar intracystic fluid pressures positive cells (25.7 ± 11.4 cells per high-­power field),
in keratocysts, dentigerous cysts, and radicular cysts. followed by orthokeratinised odontogenic cysts
Furthermore, they showed that the intracystic pressure in (12.4 ± 12.3), while the dentigerous cysts had the lowest
all cyst types reduced as the cyst increased in size. They (8.7 ± 11.6). The myofibroblasts were located in the superfi-
concluded that increased pressure played a pivotal role in cial cyst wall, orientated parallel to the basement mem-
the initiation of cysts and early growth, but was less impor- brane. The number of myofibroblasts found in keratocysts
tant as the cyst got bigger. was similar to that in ameloblastomas (n  =  7; 29.0 ± 7.0)
Previous studies (Kubota et  al.  2000; Ninomiya and to squamous carcinomas (n = 5; 21.3 ± 5.3), but only
et al. 2002) had shown that interleukin (IL)-­1α was strongly small numbers were found in unicystic ameloblastomas
expressed in the epithelial cells of keratocysts, and that the and ameloblastic fibromas. The authors suggested a posi-
expression of IL-­1α and cell proliferation were reduced tive link between the occurrence of large numbers of
proportionally by marsupialisation. The authors suggested myofibroblasts in the stroma, and the more “aggressive”
therefore that increased intracystic pressure might stimu- behaviour of the keratocyst and ameloblastomas compared
late IL-­1α and prostaglandin secretion in the cyst walls and to the other lesions studied. However, the number of
play a crucial part in the growth of odontogenic jaw cysts. myofibroblasts was variable, and their density was not cor-
Since IL-­1 is also involved in the stimulation of bone related to histological features or to behaviour.
resorption, maintenance of a positive pressure may further More recent studies have confirmed these findings.
facilitate cyst enlargement. Syamala et  al. (2016) also showed that the number of
A later study by this group found further evidence for the myofibroblasts in odontogenic keratocysts (n  =  20;
role of intracystic pressure in facilitating growth and 22.9 ± 6.5 cells per high-­power field) was greater than in
enlargement. Oka et  al. (2005) investigated the effects of dentigerous cysts (n = 20; 12.3 ± 4.1), but was not signifi-
positive pressure on the expression of IL-­1α, matrix metal- cantly different to that in ameloblastoma (n  =  20;
loproteinases (MMPs), and prostaglandin E2 (PGE2). They 24.8 ± 3.9). However, although the mean numbers were
found that positive pressure stimulated secretion of IL-­1α high, they showed that a small number of keratocysts (10%)
and MMPs in a co-­culture of keratocyst epithelial cells and did not express αSMA at all. They did not correlate the den-
fibroblasts. They concluded that positive pressure may up-­ sity or presence of myofibroblasts to histological features.
regulate expression of IL-­1α and MMPs in the wall of odon- Kouhsoltani et  al. (2016) also found a higher density of
togenic keratocysts, and that this may stimulate myofibroblasts in keratocysts than in dentigerous and
osteoclastogenesis and promote connective tissue break- radicular cysts, but showed that expression was variable, in
down and bone resorption. These studies support early that 2 of 15 (13.3%) keratocysts did not express αSMA at all,
work by Meghji et  al. (1992), who showed that levels of and 60% of cases showed only weak expression. Only 4
IL-­1α were significantly higher in the fluids of keratocysts cases (26.7%) showed strong expression compared to 33.3%
than in dentigerous or radicular cysts and that IL-­1α medi- of dentigerous cysts.
ated the secretion and activation of MMP-­9. Myofibroblasts are derived from fibroblasts under the
Mechanisms for the degradation of the connective tis- influence of TGF-­β1, and secretion of TGF-­β1 by the epithe-
sues and bone resorption in odontogenic cysts and the role lial lining of odontogenic keratocysts (Piattelli et al. 2004)
of cytokines, including IL-­1, and MMPs are discussed in suggests a mechanism whereby myofibroblastic differentia-
detail in the pathogenesis of radicular cysts in Chapter 3. tion is driven directly by the cyst epithelium. However,
Although inflammation plays a minimal role in the growth expression may be variable and the true significance of
of keratocysts, the basic mechanisms of cytokine-­mediated myofibroblasts remains uncertain. Further work is needed
resorption are similar. to correlate myofibroblasts to histological features and to
A number of studies have investigated the presence of clinical behaviour. Intriguingly, it has recently been shown
myofibroblasts in the walls of odontogenic cysts, on the that myofibroblastic activation of lung fibroblasts may be
basis that these cells are important in healing and extracel- under the control of SHH and the HH signalling pathway
lular matrix turnover and are found to be prominent in the (Cao et  al.  2020), suggesting another possible avenue for
stroma of tumours. Vered et  al. (2005) undertook an investigations of the role of myofibroblasts in keratocysts.
­Histopatholog  117

A number of studies have suggested that podoplanin podoplanin, and mediates the functions of the actin
may be important in keratocyst growth. Podoplanin is a cytoskeleton, including its role in cell migration. This sug-
transmembrane glycoprotein normally expressed on lym- gests a possible role for podoplanin in mediating growth
phatic endothelial cells and was thought to be a specific and cell migration in odontogenic lesions, including the
marker of lymphatic vessels, but it is now known to be odontogenic keratocyst.
expressed in a range of tissues and in pathological condi- A further study investigated podoplanin expression in 16
tions, including squamous cell carcinoma and ameloblas- sporadic odontogenic keratocysts before and after decom-
toma. The exact function of podoplanin is unknown, but it pression surgery (marsupialisation; Zhang et  al.  2014).
is thought to be involved in cell migration and develop- Samples obtained at the time of decompression surgery
ment, and has been implicated in tumour invasion. showed strong podoplanin expression in the basal layers in
However, it is also expressed in tooth germs and may have 15 (93.8%) cases. In the samples obtained at subsequent enu-
a role in development, and has been found in the pulp tis- cleation, 3 showed similar expression, but in 13 cases podo-
sue, odontoblasts, and ameloblasts in odontomas planin expression was significantly decreased, including 1
(González-­Alva et  al.  2011). Okamoto et  al. (2010) exam- case in which expression was lost entirely. The linings also
ined podoplanin expression in 46 odontogenic keratocysts, lost the typical features of a keratocyst and took the form of
11 orthokeratinised odontogenic cysts, and 15 dentigerous hyperplastic epithelium with inflammation. The authors
cysts. Podoplanin was expressed in 42 (91.3%) of the kerato- suggested that decompression reduces the processes of cell
cysts but in only 11 (73.3%) dentigerous cysts and 3 (27.2%) proliferative and local growth, a suggestion that is in keeping
orthokeratinised odontogenic cysts. Expression in the with the possible role of intracystic pressure in growth of the
keratocysts was consistently strong and was located in the keratocyst, discussed earlier in this section.
basal and suprabasal layers of the epithelial lining; it was
also seen in areas of basal budding, and in epithelial islands
and satellite cysts in the wall. In contrast, expression in ­Histopathology
orthokeratinised and dentigerous cysts was weak and usu-
ally only in the basal layer, and was often associated with Odontogenic keratocysts are usually removed conserva-
areas of inflammation. Okamoto et  al. interpreted these tively by enucleation and/or curettage, so the pathologist
findings to suggest that podoplanin contributed to the local will often receive multiple fragments of cyst wall
‘invasiveness and neoplastic nature’ of the odontogenic (Figure  7.15). The lining is usually thin and folded, and
keratocyst. The same group, however, in their study of may be friable with detachment of the epithelium. The
odontomas, interpreted podoplanin expression to be asso- specimen may also include the cyst contents, which are
ciated with epithelial–mesenchymal interactions and cell composed of ‘cheesy’, ‘fatty’ or ‘buttery’ keratinaceous and
differentiation (González-­Alva et al. 2011). Its exact role in necrotic material that is cream or yellow coloured, with a
these lesions remains to be determined. characteristically unpleasant odour. Similar contents may
Caetano Ados et  al. (2013) studied podoplanin expres- be found in orthokeratinised odontogenic cysts, but will
sion in a range of odontogenic cysts and tumours, includ- also be familiar to many pathologists as a characteristic fea-
ing 20  keratocysts. They found a similar pattern of ture of epidermal cysts of the skin.
expression to the previous studies (Okamoto et  al.  2010; Smaller cysts may be enucleated intact, but are usually
González-­Alva et  al.  2011), with strong expression in the collapsed with a thin wall, although the irregular growth
peripheral or basal layers of the epithelium and in odonto- pattern that is responsible for the scalloped radiographic
blasts. They found strong expression in the epithelium of margins may be apparent (see Figure  7.4). On dissection
odontogenic tumours, including ameloblastoma, adenom- the cyst may contain a thick keratinaceous material as well
atoid odontogenic tumour, and calcifying epithelial odon- as a thin, watery straw-­coloured fluid. Thickenings of the
togenic tumour. In odontogenic keratocysts they found wall or multicystic lesions are rare, but when seen may sug-
strong expression in the basal and suprabasal layers and in gest a cyst associated with NBCCS (Figure 7.15b).
satellite cysts in the wall. These results have been further The histological features of the typical odontogenic
confirmed by Oliveira et al. (2014), who found strong podo- keratocyst are so characteristic as to be regarded as diag-
planin expression in the basal and suprabasal cells of all nostic (Figures  7.16 and  7.17; Box  7.3). The cyst wall is
(n = 18) keratocysts studied. They also found a significant loosely collagenous and is lined by a thin regular, par-
correlation with ezrin expression, which was strong in the akeratinised stratified squamous epithelium that is usually
cytoplasm of basal cells in over 80% of the keratocysts. about 5–8 cell layers thick and without rete ridges
Ezrin (also called cytovillin) is a cytoplasmic protein that (Figures 7.16a, b and 7.17). The basal layer is well defined
interacts with a range of cell surface molecules, including and is composed of palisaded cuboidal to columnar cells,
118 Odontogenic Keratocyst

(a) (b)

Figure 7.15  Low-­power scans of two odontogenic keratocysts. The lining of the cyst is thin and friable, so specimens are often
fragmented. (a) A sporadic cyst. Even at this low power it can be seen that the epithelial lining is thin and regular, the wall is
uninflamed, and there are no satellite cysts or islands. (b) A cyst associated with naevoid basal cell carcinoma syndrome (NBCCS).
Many islands and satellite cysts can be seen in the wall (arrows).

(a) (b)

(c) (d)

Figure 7.16  Characteristic features of the odontogenic keratocyst. (a) The lining is thin and regular, 5–8 cell layers thick. The basal
layer is palisaded with focal areas showing reversal of nuclear polarity (arrows). Areas of hyalinisation are seen in the connective
tissue wall. (b) The parakeratinised surface layer is corrugated. (c) The lining shows prominent folding. (d) The epithelium is separated
from the underlying fibrous wall.
­Histopatholog  119

sections must be examined. Another characteristic feature,


seen in between 50 and 95% of cases, is separation of the
epithelial lining from the underlying connective tissue
(Figure 7.16c, d; Ahlfors et al. 1984; Brannon 1977; Cottom
et al. 2012). Occasionally the epithelium splits suprabasally
to leave an intact layer of basal cells on the cyst wall
(Brannon 1977; Ahlfors et al. 1984; Cottom et al. 2012).
These classic features of the odontogenic keratocyst are
diagnostic and are seen in at least a portion of all lesions.
They are not seen in any other cyst type and serve to distin-
guish the odontogenic keratocyst from other cysts that may
show keratinisation (Box 7.3).
The high proliferation potential of the lining of the odon-
Figure 7.17  A high-­power view shows reversal of nuclear togenic keratocyst has been discussed previously, but on
polarity in the epithelial basal layers and a small cluster of
suprabasal mitotic figures (arrows).
histological sections this is reflected by the consistent find-
ing of mitotic figures (Figure  7.17; Browne  1971a;
Brannon  1977; Woolgar et  al.  1987a; Cottom et  al.  2012).
Box 7.3  Odontogenic Keratocyst: Histopathology and Woolgar et  al. (1987a) found mitotic figures in 100% of
Diagnostic Criteria cysts, but also noted that they were more prominent and
more often suprabasal in NBCCS cysts. Cottom et al. (2012)
●● Lined by a thin, regular stratified squamous epithe- examined 110  keratocysts and found mitotic figures in
lium, 5–8 cell layers thick 93.6%. In 16 cases (14.5%) they were located only in the
●● Always parakeratinised basal layers, but suprabasal mitoses were seen in 79.1% of
●● 90% show a corrugated surface the cysts and in 56 cases (50.9%) they were found only in
●● Well-­defined and palisaded basal layer of cuboidal or the suprabasal layers.
columnar cells About 65% of all odontogenic keratocysts are simple
●● Focal areas of reversal of nuclear polarity cysts, with no changes in the cyst wall (e.g. Figure 7.15a;
●● The lining is irregular and folded, with characteristic Brannon 1977; Woolgar et al. 1987a). However, a number
‘hairpin’ bends of additional histological features may be seen with varia-
●● Up to 25% show satellite cysts in the wall ble frequency (Table  7.7). Most striking is the finding of
●● Up to 25% show solid epithelial islands or odonto- satellite cysts or epithelial islands within the cyst wall,
genic rests in the wall which are seen in up to 50% of cases (Figure 7.18; Table 7.7;
●● Up to 20% show budding of the epithelial basal layers Myoung et al. 2001; Kahraman et al. 2018), and have been
●● Small focal areas of orthokeratin may be seen shown to be more common in NBCCS-­associated lesions
●● Over 60% may be inflamed (Table  7.8; Ahlfors et  al.  1984; Woolgar et  al.  1987a;
●● Focal areas of inflammation bulging into the lumen Dominguez and Keszler  1988). The variable reporting of
are characteristic the frequency of these features (Table  7.7) appears to be
●● In areas of inflammation the lining is lost or may be due to differences in definition. Review of the papers shows
simple non-­keratinised epithelium that some authors do not distinguish between solid epithe-
lial islands and odontogenic epithelial rests, and in some
cases satellite cysts are only included if they have an obvi-
with focal areas where the nuclei are orientated away from ous lumen, whereas cysts filled with keratin are counted as
the basement membrane (reversal of nuclear polarity; solid islands.
Figure 7.16a arrows; Figure 7.17). In the majority of cases Nevertheless, satellite cysts are regarded as a typical fea-
the nuclei are hyperchromatic. In over 90% of cases the epi- ture of the odontogenic keratocyst (Figure  7.18) and are
thelial lining shows a characteristic corrugated surface pat- seen in up to 25% of sporadic cysts and 50% of NBCCS-­
tern (Figure  7.16b) and is folded to form sharp ‘hairpin associated cysts (Tables 7.7 and 7.8). These may show simi-
bends’ (Figures  7.15a and  7.16c; Brannon  1977; Cottom lar histological features to the main cyst and have a thin,
et  al.  2012). The irregular and folded lining may give an parakeratinised epithelial lining. Often, however, the wall
erroneous impression of a multicystic lesion in histological shows areas of thickening and the lumen may be small or
sections, and care must be taken not to misinterpret cross-­ filled with parakeratin (Figure  7.18a, b). The distinction
cut folds as satellite cysts. If necessary, multiple serial between a satellite cyst and a solid epithelial island is
Table 7.7  Frequency (%) of histological features reported in odontogenic keratocysts in selected case series.

n Satellite cysts Epithelial islands Odontogenic rests Basal budding Inflammation Cholesterol Calcifications Focal OK Hyaline bodies

Brannon (1977)a 266 24.4 ND 33.3 17.2 66.9 12.5 17.0 8.3 11.2
Ahlfors et al. (1984)b 319 7.2 8.2 22.9 24.8 NR NR NR NR NR
Woolgar et al. (1987a) 164 10.7 7.9 25.0 14.0 57.9 11.6 8.5 32.3 9.8
Azevedo et al. (2012)b 177 7.9 ND 4.5 12.4 67.8 5.6 7.9 2.8 2.8
Bello (2016)b 95 23.3 ND 23.3 8.4 57.9 8.4 8.4 NR 5.3
Cottom et al. (2012)b 110 21.8 9.1 21.8 26.4 90.0 9.1 51.8 1.8 17.3

n, number of cysts; ND – not defined, epithelial and odontogenic rests not distinguished; NR, not reported; OK, orthokeratin.
a
 Brannon reported 312 cysts; data are corrected to remove orthokeratinised and syndromic cysts.
b
 These data include a small number of syndromic cysts (<10%).
­Histopatholog  121

(a) (b)

(c) (d) (e)

Figure 7.18  Satellite cysts and epithelial islands in the wall of odontogenic keratocysts. The satellite cysts usually resemble the
main cyst, but are often filled with parakeratin (a, b) and may become solid (b, arrows). Epithelial islands (c–e) often show peripheral
palisaded cells and resemble odontogenic cell rests. (d) The islands may proliferate and form branching islands. This pattern has been
referred to as ‘ameloblastomatoid’ (see text).

therefore not always apparent. A cyst may be filled with (1977) interpreted these islands as transformation to
parakeratin, and an island may occasionally show micro- ameloblastoma, but Ahlfors et al. (1984) and Woolgar et al.
cystic change (Figure  7.18b, arrows). It is probable that (1987a) did not think they met the criteria for ameloblas-
there is a spectrum of change, with islands becoming toma and interpreted them as islands of proliferating odon-
increasingly cystic as they grow. Brannon (1977) is proba- togenic epithelium (Figure  7.18). Review of the
bly correct in his proposal that satellite cysts arise by cystic photomicrographs in all three papers show solid islands or
change in epithelial islands or odontogenic rests within the proliferating strands, in all cases consistent with quiescent
wall. He also noted that satellite cysts were most common or proliferating odontogenic rests and resembling dental
in keratocysts found at the angle and ramus of the mandi- lamina (Figure 7.18 c-e). Similar islands have been reported
ble, supporting the concept that the satellite cysts develop in more recent papers and have been interpreted as islands
from rests of dental lamina that are most frequently of odontogenic epithelium, with no suggestions of an asso-
encountered at this site. The odontogenic rests may prolif- ciation with ameloblastoma (Myoung et al. 2001; Cottom
erate and form branching islands, which resemble et al. 2012; Cunha et al. 2016; Naruse et al. 2017).
ameloblastoma. Woolgar et  al. (1987a) reported ‘amelo- Many authors assume that all the epithelial islands and
blastomatoid’ islands in 7 of 164 (4.3%) NBCCS-­associated satellite cysts in the wall arise from odontogenic rests, but
cysts, and Brannon (1977) and Ahlfors et  al. (1984) some may be a result of budding of the basal layers of the
described them in 0.6% and 1.5%, respectively. Brannon cyst lining (Ahlfors et al. 1984). Budding of the basal layer
122 Odontogenic Keratocyst

Table 7.8  Frequency (%) of clinical and histological features stratified squamous epithelium (Figure  7.20c). This is
reported in naevoid basal cell carcinoma syndrome (NBCCS) indistinguishable from the epithelium of the radicular cyst
and sporadic odontogenic keratocysts.
or other inflamed odontogenic cysts and in small biopsies
of an inflamed odontogenic keratocyst it may not be possi-
NBCCS Sporadic
n = 164 n = 164
ble to make a diagnosis.
Other reactive changes, more often associated with
Clinical features inflammatory cysts, may also be seen. Cholesterol clefts
Age (mean) 26.2 40.4 and hyaline (Rushton) bodies are seen in about 10% of
Males 45 62 cases (Figure 7.20d). Other changes that may be seen are
mucous cells in the superficial epithelial layers in about 2%
Site: Mandible 66.0 74.0
of cases (Brannon 1977; Cottom et al. 2012) and a report of
Posterior mandible 44.0 60.0
sebaceous cells in three cases (Brannon 1977).
Posterior maxilla 21.0 11.0 A rare finding is the presence of epithelial dysplasia,
Histological features which has been noted in less than 1% of cases
Satellite cysts 54.3 10.7 (Brannon  1977; Ahlfors et  al.  1984; Cottom et  al.  2012).
Solid epithelial islands 33.5 7.9 Basal cell hyperplasia and bulbous rete pegs are a feature of
Odontogenic rests 40.2 25.0 dysplasia in the oral mucosa, but budding and bulbous pro-
Basal budding 14.0 14.0 liferations in the wall of a keratocyst (Figure 7.19) should
not be interpreted as dysplasia unless associated with
Mitotic figures 100.0 100.0
cytological atypia. When dysplasia does occur, it appears
Inflammation 71.3 57.9
as basal cell crowding and loss of stratification, with
Cholesterol 8.5 11.6
cell and nuclear pleomorphism, hyperchromatism, and
Calcifications 19.5 8.5 dyskeratosis.
‘Ameloblastomatoid’ islands 4.3 0 The lumen of odontogenic keratocysts may be full of
Bold – significantly different from sporadic cysts. Source: Data from keratin, but the frequency is uncertain since the luminal
Woolgar et al. (1987a,b) and Rippin and Woolgar (1991). contents are often lost during removal and processing.
Nevertheless, it has been reported that at least 40% may
contain variable amounts of desquamated keratin
occurs in up to 25% of cases (Table 7.7) and is most often (Brannon 1977; Cottom et al. 2012). Keratocysts have also
seen as a focal area of multiple small buds pushing into the been shown to contain a thin straw-­coloured fluid at opera-
superficial connective tissue of the wall (Figure  7.19). tion, but may appear empty on macroscopic or microscopic
Occasionally, however, large buds resembling bulbous rete examination.
pegs may be seen and sometimes these appear to give rise The fibrous cyst wall is usually thin and composed of col-
to satellite cysts or separate islands (Figure 7.19d; Ahlfors lagenous connective tissue, with few distinctive features.
et al. 1984; Myoung et al. 2001; Cunha et al. 2016). Hyalinisation of the connective tissue, usually in a subepi-
Occasionally, satellite cysts and epithelial islands may be thelial band, has been described in up to about 50% of cases
so prominent that the lesion takes on a more solid multi- (Figure  7.16a; Browne  1971a; Brannon  1977; Cottom
cystic appearance. When this occurs, the term solid odonto- et al. 2012). Often the epithelium is missing in areas of hya-
genic keratocyst has been used. This lesion will be discussed linisation and it has been suggested that this may be caused
later in this section. by alterations in the basement membrane, resulting in loss
Over half of odontogenic keratocysts, and in some series of adhesion and separation of the epithelial lining (Cottom
the vast majority, show evidence of inflammation et  al.  2012). Occasional cases show evidence of keratin
(Table  7.7). In some cases this may be due to trauma or within the fibrous wall, with an associated granulomatous
continuity with the oral cavity through cortical and or foreign-­body giant cell reaction. Focal areas of calcifica-
mucosal perforation and infection, but often the cause is tion are occasionally seen. Sometimes these appear to be
not apparent. If the cyst is large and traumatised or infected dystrophic, but often small ‘cementicle’-­like calcifications
then the inflammation may be widespread, but a character- may be seen in association with odontogenic epithe-
istic appearance is of focal areas of inflamed fibrous or lial rests.
granulation tissue pushing into the cyst lumen There have been a number of reports of cartilage within
(Figure 7.20a, b). In areas of inflammation the typical par- the wall of odontogenic keratocysts. The first report was by
akeratinised pattern of the epithelium is lost and the cyst Arwill and Kahnberg (1977), who noted a keratocyst in the
lining is composed of variably hyperplastic, non-­keratinised anterior mandible with an associated ‘chondroma’. A
­Histopatholog  123

(a) (b)

(c) (d)

Figure 7.19  (a–d) Hyperplasia of the epithelial lining and budding of the basal layer in odontogenic keratocysts. Sometimes,
epithelial islands appear to ‘drop off’ from the lining to form islands or satellite cysts (d).

further four cases were reported by Kratochvil and Brannon occasionally found in the anterior regions of the jaws, so
(1993). These showed the typical features of odontogenic this may explain some of the cases, especially that of Yih
keratocysts, but with focal areas of quite well-­formed carti- and Krump (2005), which was located in the nasopalatine
lage lying just below the epithelial lining. Three of the four canal. Other cases are most probably metaplastic in nature.
lesions were located in the anterior region of the jaws, two Early descriptions of the odontogenic keratocyst included
in the mandible, and one in the ‘globulomaxillary’ region. an orthokeratinised variant, but it is now appreciated that
The fourth case was located in the premolar region. the orthokeratinised odontogenic cyst should be classified
Subsequently, a further five cases have been reported as a separate entity (Speight and Neville 2022); this is dis-
(Mosqueda-­Taylor et  al.  1998; Fornatura et  al.  2001; cussed in Chapter 12. Nevertheless, most large series have
Vicente-­Barrero et  al.  2004; Yih and Krump  2005; Ide described a small number of cysts with focal areas of
et al. 2009). All showed similar features of a typical kerato- orthokeratin (Table 7.7). In Brannon’s (1977) early series of
cyst but with focal areas of cartilage formation, usually just 312 odontogenic keratocysts, he found that 9.7% would
below the epithelium. Interestingly, all the cases arose in meet the criteria for an orthokeratinised odontogenic cyst,
the anterior regions of the jaws, three in the mandible and but that 22 parakeratinised cysts (8.3%) showed focal areas
two in the maxilla. One of the maxillary lesions arose in the of orthokeratin. Because the orthokeratin was focal, he
nasopalatine canal (Yih and Krump  2005). The origin of regarded these lesions as odontogenic keratocysts. Crowley
the cartilage is open to speculation, but the authors have et al. (1992) also found a small number of lesions with both
suggested that the cartilage might be vestigial rests, or para-­ and orthokeratin (7 of 449 cases; 1.6%) and also con-
metaplastic, or a result of unusual inductive change from sidered these to be keratocysts. Woolgar et  al. (1987a,c),
the cyst epithelium. In all cases the cartilage has been however, found that up to 32% of odontogenic keratocysts
bland and essentially normal, so a coincidental neoplastic may contain areas of orthokeratosis. The reason for this
process seems unlikely. Vestigial rests of cartilage are high figure is uncertain, but Woolgar et  al. (1987a)
124 Odontogenic Keratocyst

suggested that they interpreted keratin squames within the


Box 7.4  Features Suggestive of a Diagnosis of
lumen as indicating orthokeratinisation. This suggests that
Naevoid Basal Cell Carcinoma Syndrome (NBCCS) That
their figure of 32% is too high. There are no clear guidelines
Warrant Further Clinical Investigations or Careful
for how much orthokeratin must be present to determine a
Follow-­up
diagnosis of orthokeratinised odontogenic cyst. The WHO
definition states that the lesion is entirely or predominantly ●● A diagnosis of odontogenic keratocyst (even solitary)
lined by orthokeratinised stratified squamous epithelium in a patient under 20 years of age
(Speight and Neville 2022). We would interpret these data ●● Multiple keratocysts, especially involving multiple
to mean that an odontogenic keratocyst may show small quadrants
focal areas of orthokeratin, and that an orthokeratinised ●● Lesions in the posterior maxilla, especially in
odontogenic cyst may show focal areas of parakeratin. younger people
However, if the lining shows obvious areas of typical par- ●● Prominent satellite cysts, epithelial islands, or odon-
akeratinised epithelium (Figure 7.16; Box 7.3), then a diag- togenic rests
nosis of odontogenic keratocyst should be given ●● Numerous satellite cysts, often in groups of varied size
●● High number of mitotic figures, especially in the
suprabasal layers
Odontogenic Keratocysts Associated
with Naevoid Basal Cell Carcinoma Syndrome Caution:
NBCCS cysts constitute less than 10% of the total num-
The pathogenesis of the odontogenic keratocyst and its
ber of keratocysts that may be encountered (Table 7.2),
relationship to NBCCS has been discussed in detail earlier
so care must be taken not to overinterpret the histo-
in this chapter. It was proposed that the keratocyst may sit
logical findings, since satellites and islands may also be
on a spectrum between a developmental lesion and a neo-
seen in solitary cysts (Table  7.7). Each cyst should be
plasm, with variable degrees of genetic aberrations
evaluated on the clinical, radiological, and histological
(Table  7.6), but involving activation of the HH signalling
findings.
pathway secondary to alterations in the PTCH1 gene. This
would suggest that keratocysts with different degrees of
genetic change may show different phenotypes, and there that NBCCS patients may be more susceptible to prolif-
is evidence that this may be the case. The differences in the erations of odontogenic epithelium.
clinical presentation of sporadic and NBCCS-­associated Surprisingly, there were no differences between the cysts
cysts has already been discussed (see Box 7.1) and are sum- with regard to budding of the epithelial basal layers or to
marised again in Table 7.8 and Box 7.4. the presence of mitoses. Mitoses were seen in all cysts
There is also good evidence that keratocysts associated examined, although there were significantly more in syn-
with NBCCS show different and characteristic histologi- dromic cysts than in sporadic cysts. Dominguez and
cal features and that there may be some genotype–phe- Keszler (1988) compared keratocysts from NBCCS patients
notype correlations. The most comprehensive studies of and sporadic cases and also found that significantly more
NBCCS cysts were carried out by Woolgar and colleagues NBCCS cysts contained satellite cysts or islands (36.4%;
(Woolgar et  al.  1987a,b; Rippin and Woolgar  1991) and n = 33) than sporadic cysts (6.1%; n = 33).
their main findings are summarised in Table 7.8. NBCCS The finding of a greater frequency and number of satel-
cysts had a higher frequency of satellite cysts, epithelial lite cysts and islands in NBCCS-­associated keratocysts is in
islands, and odontogenic rests in the wall than sporadic keeping with the concept that in these lesions there is a
cysts, and overall 67% of NBCCS cysts showed one or germline mutation in the PTCH1 gene that constitutively
more of these features compared to only 34% of sporadic activates the HH signalling pathway, resulting in a clone of
cysts. NBCCS cysts were also more likely to be inflamed genetically altered dental lamina rests that subsequently
and to show evidence of dystrophic calcifications in the proliferate to form cysts with a high frequency of satellite
wall. Woolgar et al. (1987a) also found that satellite cysts cysts and proliferating islands. This provides some expla-
in NBCCS cases were more numerous than in solitary nation for how the genetic changes may influence the his-
cysts, and often arose in groups of varied sizes, giving an tological phenotype.
impression of ‘several generations of cysts’. Islands of In keeping with the findings of increased mitotic figures,
proliferative epithelium resembling ameloblastoma Li et al. (1995) showed that cell proliferation (Ki-­67–posi-
(‘ameloblastomatoid’ islands) were only seen in syndro- tive cells) was significantly greater in NBCCS-­associated
mic cysts. These authors found no evidence that these cysts than in sporadic cysts. In a later study, the same group
islands develop into true ameloblastomas, and suggested (Pan and Li 2009) found that Ki-­67 labelling in keratocysts
­Histopatholog  125

with PTCH1 mutations (n = 29; 103.29 ± 35.47 cells/mm) As discussed previously, the most common single site for
was significantly higher than in keratocysts with no muta- odontogenic keratocyst is the angle and ramus of the man-
tions (n = 33; 69.82 ± 24.28 cells/mm). Furthermore, they dible, and at this site there are three lesions that may have
also found that the labelling index was higher in lesions a similar radiological appearance and may be considered in
with truncating mutations compared to non-­truncating the clinical differential diagnosis – dentigerous cyst, kerato-
mutations in both NBCCS cysts (138.04 ± 24.07 cells/mm cyst, and ameloblastoma. Usually these are easy to diag-
vs 99.93 ± 28.18) and sporadic cysts (92.16 ± 22.50 vs nose histologically, but in a small incisional biopsy,
67.82 ± 23.78). The authors concluded that PTCH1 muta- especially if a lesion is inflamed, it may not be possible (see
tions, especially truncating mutations that affect protein below). If a diagnosis cannot be made, then a second biopsy
function, may define a subset of keratocysts with a higher or conservative excision (enucleation) may be necessary. A
proliferative potential. cystic ameloblastoma or a unicystic ameloblastoma may on
Shimada et  al. (2013) investigated the genotype of occasion show a keratinised surface or eosinophilic surface
16  NBCCS and 20 sporadic cysts and found LOH at the epithelial cells that resemble parakeratin. However, promi-
PTCH1, PTCH2, or SUFU locus in 79% of NBCCS cysts and nent basal columnar cells resembling ameloblasts and the
42% of sporadic cysts, with PTCH1 being the most com- presence of suprabasal stellate reticulum–like areas should
monly affected (see Table  7.6). NBCCS cysts showed the enable a correct diagnosis to be made.
highest frequency of budding of the epithelial basal layers A particular problem may arise if an odontogenic kerato-
and of satellite cysts, but across all cyst types LOH at cyst becomes heavily inflamed through trauma or infec-
PTCH1 and SUFU showed a significant correlation with tion. In areas of inflammation, the typical features are lost
epithelial budding. Epithelial budding was also associated and the lining reverts to a simple non-­keratinised stratified
with recurrence. Satellite cysts and islands were not associ- squamous epithelium (Figure 7.20), which may show vari-
ated with LOH or recurrence. able degrees of hyperplasia and rete peg formation.
These studies further demonstrate that odontogenic Histologically such a lining is indistinguishable from radic-
keratocysts are genetically heterogeneous, and provide ular cyst or other inflamed odontogenic cysts. If a patholo-
early evidence that there are genotypic subsets with differ- gist is faced with such an appearance, and the clinical and
ent clinical or histological features. This may explain the radiological features suggest a keratocyst, then multiple
range of behaviours and histological features seen, but fur- blocks and sections must be examined to search for areas of
ther research is needed to determine whether the kerato- typical keratocyst lining. Usually these will be present and
cyst can be divided into truly distinctive entities. a correct diagnosis can be made. Occasionally a cyst may be
entirely inflamed and show a non-­descript epithelial lin-
ing, and even widespread loss of the lining. In these cases a
Histological Differential Diagnosis
definitive diagnosis is not possible, and a judgement of the
The classic histological features of the odontogenic kerato- most likely diagnosis must be made based on the clinical,
cyst are diagnostic (Figures  7.16 and  7.17; Box  7.3) and radiological, and surgical findings. It should be noted that
when seen the diagnosis should not be in doubt and few immunohistochemistry is not helpful in these cases,
alternative lesions need to be considered. Occasionally the because any biomarkers (usually cytokeratins) considered
typical parakeratinised epithelium may not be prominent typical of parakeratinised keratocyst epithelium are not
and may only comprise a small portion of the cyst lining. expressed in the simple epithelium of the inflamed cyst.
This may occur in inflamed cysts or in cysts that show A further diagnostic problem arises when a lesion with
areas of orthokeratinisation. Differentiation from ortho­ cystic spaces typical of a keratocyst appears solid or
keratinised odontogenic cyst has been discussed previously multicystic.
and is considered in detail in Chapter  12. We have sug-
gested that if the lining shows areas of a typical parakerati-
Solid Odontogenic Keratocyst
nised odontogenic keratocyst, then a diagnosis of
odontogenic keratocyst should be given. Occasionally There are a number of case reports of ‘solid’ odontogenic
other odontogenic cysts, especially radicular and dentiger- keratocysts. All appear to be unusual and show such vari-
ous cysts, may show keratinisation through a process of able features that the true nature of this lesion remains
metaplasia. This is rare and when it does occur it usually uncertain. It is important, however, because the authors of
affects only a small portion of the cyst lining and is typi- all the reports have suggested that the solid odontogenic
cally orthokeratin. Even if parakeratin is seen, it does not keratocyst is the ‘link’ that demonstrates the true neoplas-
resemble a typical keratocyst and the overall clinical and tic nature of the lesion. As an analogy, a number of authors
histological features will enable a correct diagnosis. (Vered et  al.  2004; Daley et  al.  2007; Shuster et  al.  2012;
126 Odontogenic Keratocyst

(a) (b)

(c) (d)

Figure 7.20  Inflammatory changes in the odontogenic keratocyst. A characteristic feature is focal areas of inflammation pushing into
the cyst lumen (a, b). In areas of inflammation the epithelial lining may be lost (a, b) or may revert to simple non-­keratinised stratified
squamous epithelium (c), with a sharp transition from the more typical parakeratinised surface (c, arrow). (d) Focal accumulations of
hyaline bodies are occasionally seen.

Table 7.9  Reported cases of solid odontogenic keratocyst.

Age Sex Location Radiology

Omura et al. (1997) 70 F Mandible angle/ramus Multilocular


Vered et al. (2004) 52 M Maxilla posterior Multilocular
Daley et al. (2007) 52 M Mandible canine/premolar Unilocular
Iezzi et al. (2011) 52 F Mandible premolar Unilocular
Shuster et al. (2012) 47 M Mandible premolar Unilocular
Kahraman et al. (2018) 42 M Mandible posterior Multilocular
30 F Mandible posterior Multilocular

Kahraman et al. 2018) have suggested that the solid kerato- reports in Table 7.9 are regarded as acceptable because all
cyst is equivalent to the dentinogenic ghost cell tumour, were primary lesions and were well documented with radi-
representing a ‘solid variant’ of calcifying odontogenic cyst ology, histology showed that at least some cysts were lined
that is a true neoplasm. by a parakeratinised epithelium typical of odontogenic
Table  7.9 summarises the features of seven well-­ keratocyst, and none reported features suggestive of amelo-
documented cases. The clinical and radiological presenta- blastoma (see also Box 7.5). The lesions were described as
tion is similar to a conventional keratocyst (Table  7.1), ‘solid’ at surgery or on macroscopic examination, but all
although the age of presentation may be slightly older. The were actually composed of multiple, variably sized cysts
­Histopatholog  127

aggressive behaviour typical of a neoplasm. Others, how-


ever, found that the lesions were well demarcated with no
bone involvement and suggested that there is no evidence
that the solid variant should be regarded as more aggres-
sive than a conventional cystic odontogenic keratocyst
(Daley et  al.  2007; Iezzi et  al.  2011; Shuster et  al.  2012;
Kahraman et  al.  2018). None of the lesions shown in
Table 7.9 recurred.
These reports demonstrate that the features of the solid
odontogenic keratocyst are quite variable, but also suggest
that there are too few cases reported to draw any firm con-
clusions regarding their true nature. Because the solid
lesions show variably sized multiple cysts, the line of dis-
tinction between a ‘solid’ keratocyst and a conventional
keratocyst with multiple satellite cysts and islands in the
Figure 7.21  A solid odontogenic keratocyst that arose in the wall is not clear. Kahraman et  al. (2018) examined
maxilla (see Figure 7.9). The lesion is actually composed of 204  keratocysts and found that 149 (73.0%) were simple
multiple irregular and variably sized cystic spaces and small
cysts with no satellite cysts or epithelial islands, 43 (21.1%)
islands in a dense fibrous stroma.
had fewer than 10 mural satellites or islands, and only 12
cases (5.9%) had more than 10 satellite cysts or islands. Only
and solid islands in a dense fibrous stroma (Figure 7.21). In 2 of these 12 cases were regarded as solid (Table 7.9). These
the first report, Omura et  al. (1997) described numerous were very similar to the lesion shown in Figure 7.21 and
variably sized keratocysts lined by parakeratinised epithe- described as showing multiple, randomly distributed small
lium and often filled with desquamated keratin and cysts and islands within a dense fibrous connective tissue,
necrotic material. Some cysts were lined by non-­keratinised and presumably did not have a single large or dominant cyst.
epithelium, and some of the larger cysts were occasionally Overall, it seems that the histological features of the
accompanied by smaller satellite cysts. Of note is that they keratocyst may sit on a spectrum from simple cysts with no
showed that many small cysts were located within the can- mural changes, to cysts with scattered satellite cysts and
cellous bone. Subsequent reports have shown similar fea- islands, to the more solid multicystic lesions described
tures (Vered et al. 2004), but others have also found many here. Although there is evidence of an association between
solid islands and sheets or cords of epithelium (Daley genetic changes and histology, and satellite cysts are more
et al. 2007; Shuster et al. 2012). The case reported by Daley common in NBCCS-­associated cysts (Table 7.8), there have
et  al. (2007) showed a solid tumour mass with multiple been no studies to determine the status of the PTCH gene
keratocysts, but also with solid islands and branching cords in ‘solid’ lesions. None of the lesions in Table 7.9 or reported
of epithelium. The cysts were lined by parakeratinised epi- as solid in the literature has been associated with NBCCS. As
thelium and many were filled with desquamated keratin discussed previously, further studies are needed to under-
and inflammatory cells. Similarly, the lesions described by stand the genetic changes involved in cysts with different
Shuster et  al. (2012) and Kahraman et  al. (2018) showed features and to determine if there are distinctive subsets of
multiple keratocysts, but also multiple small solid islands. odontogenic keratocyst.
Many of these resemble odontogenic epithelium and some A further area of difficulty is the differential diagnosis of
showed microcyst formation. In some cases the keratinised the solid lesions. Those described have been composed of
islands have been described as keratin pearls (Kahraman multiple cystic spaces as well as solid islands and cords of
et  al.  2018) and feature a flattened epithelium without a epithelium, many of which show slits or microcystic change,
prominent basal layer, lining a cystic space filled with kera- filled with keratin. Although typical parakeratinised areas
tin or keratinaceous material with macrophages. should be present, many cystic spaces are filled with lamel-
Examination of some of the published histological images lae of keratin and resemble keratin pearls. These lesions
shows a striking similarity to well-­differentiated squamous often resemble a well-­differentiated squamous cell carci-
cell carcinoma (Daley et  al.  2007; Shuster et  al.  2012; noma. In particular, they may resemble carcinoma cunicula-
Kahraman et  al.  2018). A number of cases reported (and tum (Fonseca et al. 2013; Barrett et al. 2020; Janardhanan et
illustrated) islands of epithelium, or small cysts within the al. 2021), which has been shown to permeate the bone
adjacent cancellous bone (Omura et  al.  1997; Vered and  form branching crypts and cyst-­like spaces lined
et  al.  2004), and this was interpreted as indicating by  keratinised epithelium or filled with keratin squames.
128 Odontogenic Keratocyst

Fonseca et al. (2013) reviewed 10 cases that had perme- appears to represent a follicular ameloblastoma with exten-
ated the jaw bones and found that 2  had been misdiag- sive keratinisation and cystic change, and is virtually
nosed as odontogenic keratocyst. More recently, Barrett ­indistinguishable from acanthomatous ameloblastoma.
et al. (2020) described a series of 12 cystic squamous cell Confusion with odontogenic keratocyst arises when con-
carcinomas within the jaws and highlighted the diagnos- fronted with types iii and iv. These may show areas of cystic
tic pitfalls, including misdiagnosis as odontogenic change lined by typical keratocyst epithelium, but with
keratocyst. One lesion in particular appeared to be intra- minimal areas resembling ameloblastoma. The case illus-
osseous and had cysts resembling keratocyst, with a par- trated by Whitt et  al. (2007) showed a complex plexiform
akeratinised surface and palisaded basal cells. Review of pattern with islands and strands of epithelium in a dense
the figures in these papers (Fonseca et al. 2013; Barrett collagenous stroma. Cystic spaces lined by keratocyst-­like
et al. 2020) will show a striking resemblance to a num- parakeratinised epithelium were seen and lamellated sheets
ber of cases reported as solid odontogenic keratocyst. of parakeratin were prominent, both within the cysts and
The key feature of a squamous carcinoma or carcinoma extruding into the stroma. There were only small areas of
cuniculatum is that it ‘burrows’ into the bone from the peripheral palisading resembling ameloblastoma, but the
surface mucosa and usually there is evidence of a surface authors did not describe or illustrate ameloblastomatous
lesion. Kawano et  al. (2013) reported a case of a solid areas. Whitt et  al. (2007) used the term keratoameloblas-
variant of odontogenic keratocyst that showed unusual toma in the title of their paper, but they were uncertain of
features that raise doubt about the diagnosis. This lesion its true nature. They speculated that it may be an odonto-
presented as a moth-­eaten radiolucency in an area of the genic keratocyst with focal areas resembling ameloblas-
mandible where molar teeth had been previously toma, or a hybrid lesion. Their illustrations, however, are
extracted, but the reason for extractions were not noted. very similar to other reports describing lesions as solid
Clearly the lesion had continuity with the oral mucosa odontogenic keratocyst. Geng et al. (2012) reported a very
(via the extraction sites) and histologically resembled a similar case that showed some features typical of kerato-
well-­differentiated carcinoma or carcinoma cunicula- cyst, but also areas of ameloblastomatous change, and
tum. Clear cells and mucous cells were also noted, and called it a solid odontogenic keratocyst with ameloblasto-
although the authors suggest the possibility of odonto- matous transformation. This case is not included in
genic origin, the true nature of the lesion could not be Table 7.9 since it more closely meets the criteria for a type of
determined. In diagnosing a solid keratocyst, care should keratoameloblastoma.
be taken to ensure that the lesion is truly intraosseous It is clear, however, that the keratoameloblastoma and
and does not have an origin from surface (or gingival) odontogenic keratocyst are related, at least by a similar his-
epithelium. tology, and some have proposed a histogenetic relationship
A further lesion that may resemble a solid or multicystic whereby a keratocyst may give rise to an ameloblastoma, or
odontogenic keratocyst is the keratoameloblastoma. These a solid keratocyst may evolve from keratoameloblastoma
two lesions show an almost identical morphological archi- (Ide et al. 2012). Ide et al. (2003) are credited with one of
tecture and only subtle differences in histological appear- the earliest reports of a solid odontogenic keratocyst, but
ance. Both are multicystic, with cysts lined by keratinising their case had originally shown features of keratoamelo-
epithelium. The keratoameloblastoma, however, is blastoma and had recurred or persisted over a period of
thought to be a variant of ameloblastoma and shows basal 25 years, finally showing an unusual histology of an infil-
ameloblast-­like cells and typical areas of ameloblastoma- trative lesion composed of cords and islands of epithelium
tous epithelium with stellate reticulum. Nevertheless, the and irregular cysts filled with parakeratin. Ide and col-
lesions are so similar that it is still debated whether the leagues (Ide et  al.  2003,  2012) and others have cited this
keratoameloblastoma is a multicystic keratinising amelo- case as evidence of transition from (kerato)ameloblastoma
blastoma, or an odontogenic keratocyst undergoing amelo- to keratocyst, but because of its unusual features its true
blastomatous change. nature remains uncertain.
Whitt et  al. (2007) reported a case of keratoameloblas- Because of the known ability of odontogenic epithelium
toma and reviewed 12 cases reported up to 2007. They sug- to show multiple routes of differentiation and to give rise to
gested that these lesions may show a range of features that a plethora of cysts and tumours, it is perfectly possible for
fall into four broad categories: (i) papilliferous, (ii) simple these lesions to be histogenetically related, with resulting
histology, (iii) simple histology with keratocyst-­like areas, combinations of features when a lesion is examined histo-
and (iv) a complex histology. The papilliferous type is logically at a fixed point in time. As a result, the literature
regarded as a separate entity and would not be confused contains many case reports of very similar lesions under
with an odontogenic keratocyst. Similarly, the simple type different names, often depending on the past experiences
­Histopatholog  129

of the pathologist or their preferred interpretation. Many Cytology and Aspiration Biopsy


of these lesions probably represent unusual tumours aris-
Toller (1970a) and Kramer and Toller (1973) were the first to
ing from odontogenic epithelium but defying categorisa-
suggest that a preoperative diagnosis of odontogenic kerato-
tion, and should be regarded as oddities rather than
cyst might be made by the examination of aspirated cyst
entities. Nevertheless, there is little doubt that an odonto-
fluid. Toller (1970a) showed that fluids from keratinising
genic keratocyst may show multiple satellite cysts and epi-
cysts had soluble protein levels below 3.5 g per 100 mL (mean
thelial islands, and the evidence suggests that on occasion
2.2 g per 100 mL), whereas the values for non-­keratinising
these may be so prolific as to produce a lesion that appears
cysts were in the range 5.0–11.0 g per 100 mL, with a mean of
solid (see Figures 7.9 and 7.21). Further reports and genetic
7.1 g per 100 mL. Electrophoretic studies corroborated the
studies may, in the future, clarify the true nature of this
finding that keratinising cysts were very low in soluble pro-
lesion. To ensure consistency and to assist future studies,
teins, and Toller postulated that a protein level of less than
we suggest that strict criteria are applied for a diagnosis of
4.8 g per 100 mL indicated a diagnosis of odontogenic kerato-
a solid or multicystic odontogenic keratocyst (Box 7.5).
cyst. A value of over 5.0 g per 100 mL, however, would suggest
We have previously mentioned a multicystic lesion,
a radicular or dentigerous cyst, or even an ameloblastoma.
keratocystoma, with features resembling odontogenic
He suggested that fully keratinised epithelium was impervi-
keratocyst that has been described in the parotid gland
ous to proteins, whereas the non-­keratinised linings of
(Seifert et al. 1999; reviewed by Aresta et al. 2019; Komatsu
radicular or dentigerous cysts would at least slowly transmit
et al. 2020). Histologically the keratocystoma is multicystic
smaller proteins. For a time, it was quite common for special-
and is almost indistinguishable from lesions described as
ist oral pathology laboratories to examine cyst fluid by elec-
solid odontogenic keratocyst. However, the keratocystoma
trophoresis for protein levels (reviewed by Smith 1991), but
arises only in the parotid gland and, despite the histological
with the advent of larger, automated, and highly regulated
similarities, it is not related to the odontogenic keratocyst.
laboratories, infrequently used specialist techniques are now
Lesions located in the parotid gland should be diagnosed as
rarely applied and few if any laboratories examine cyst fluids
keratocystoma, and not as soft-­tissue or solid keratocyst.
for protein content as a diagnostic test.
The keratocystoma is discussed further in Chapter 15.

Box 7.5  Solid Odontogenic Keratocyst: Suggested Criteria for Diagnosis


●● The lesion is histologically multicystic
●● The cysts may be irregular and vary in size, but no single large or dominant cyst should be present
●● At least some of the cysts and in particular larger cysts must be lined by parakeratinised epithelium typical of
odontogenic keratocyst (Figure 7.16)
●● Palisading of the basal cells and reversal of nuclear polarity may be seen
●● Solid epithelial islands and odontogenic rests may be present
●● Branching strands and cords of epithelium may be seen, but these often resemble collapsed cystic structures and
may contain ‘slits’ filled with parakeratin
●● The lesion should be intraosseous
Caution: If these features are present, consider an alternative diagnosis:
●● A single large typical keratocyst surrounded by smaller cysts and islands. This is probably a conventional odonto-
genic keratocyst with prominent satellites. Also consider the possibility of naevoid basal cell carcinoma syn-
drome (NBCCS)
●● Ameloblasts or stellate reticulum. This excludes the diagnosis of keratocyst. Consider a keratinising type of amelo-
blastoma (acanthomatous ameloblastoma) or keratoameloblastoma
●● Prominent burrowing strands, islands, or crypts of keratinising epithelium. Consider carcinoma cuniculatum
●● Prominent keratin pearls or any evidence of cytological atypia. Consider well-­differentiated squamous cell carci-
noma, either primary or a metastasis
●● Any evidence of continuity with the overlying oral mucosa or with gingival epithelium. If perforation of the cortical
plate by an intraosseous lesion can be excluded, consider mucosal or gingival well-­differentiated squamous cell
carcinoma
130 Odontogenic Keratocyst

Cytological examination of cyst aspirates, however, is In a similar study, Vargas et al. (2007) examined aspirates
included in the repertoire of routinely applied techniques from eight odontogenic keratocysts and found that
in most centres, and aspiration cytology may be a useful aid Papanicolaou stain showed positive staining (orange/red)
to preoperative diagnosis. Kramer and Toller (1973) were for keratinised squames in all cases. Cells were also posi-
the first to report the use of aspiration cytology for the pre- tive for pan-­cytokeratin (AE1/AE3) and CK19 (Figure 7.22),
operative diagnosis of keratocysts. They examined a total but the specificity of the cytokeratin expression was not
of 53 cystic lesions and subsequent histological examina- determined by comparison with other non-­keratinising
tion showed that 21  were odontogenic keratocysts and cysts. The authors also prepared cell blocks from the aspi-
32 were non-­keratinised cysts. Microscopic examination of rates and were able to show lamellae of desquamated kera-
smears of the aspirates showed keratinising epithelial cells tin that expressed cytokeratins (Figure 7.22b). Cell blocks
(squames) in 17 of the 21 (81.0%) keratocysts, but in only 3 are prepared by centrifugation of the aspirate to produce a
(9.4%) of the 32  non-­keratinising cysts, giving an overall cell pellet that can then be embedded and sectioned using
correct diagnosis in 46 of the 53 cysts (86.8%). routine histological techniques. The advantage is that cells
August et  al. (2000) examined fine needle aspiration are concentrated and some of the architectural arrange-
biopsies from 18 odontogenic cysts and compared the pre- ment of the tissue may be preserved. Rivero et  al. (2014)
operative cytology findings to the gold-­standard histologi- prepared cell blocks from aspirates of 135 cystic jaw lesions.
cal diagnosis. During aspiration, they deliberately contacted Blocks were sectioned and stained with haematoxylin and
and sampled the wall of the cyst to ensure a yield of surface eosin (H&E) and the findings were compared to the final
epithelial cells. Smears were stained with ‘Diff-­Quik’ diagnosis made on examination of surgical biopsies.
(Romanowsky stain) and the Papanicolaou method, and Examination of the cell blocks showed keratin in 20 cases,
using immunocytochemistry for CK10. The epithelial cells of which 19  were odontogenic keratocysts and 1  was an
of 10 cysts were strongly positive for CK10 and all 10 were orthokeratinised odontogenic cyst. Of the 115 cases that
subsequently confirmed to be odontogenic keratocysts, did not show evidence of keratin, none was diagnosed as a
with strong positivity for CK10 in the superficial parakera- keratocyst after examination of the surgical specimen. As a
tin layers. Four dentigerous cysts and four radicular cysts diagnostic test for odontogenic keratocyst, these data sug-
were negative for CK10 in the aspirates and in the histo- gest a sensitivity of 100%, specificity of 99.1%, and positive
logical sections. The authors suggested that expression of and negative predictive values of 95% and 100%, respec-
CK10 is specific for the odontogenic keratocyst and allows tively. The images in this paper (Rivero et al. 2014) show
keratinised epithelial cells to be easily distinguished from clearly visible desquamated epithelial cells, evidence of
non-­keratinised cells. parakeratin, and lamellae of keratin, even in cases that also

(a) (b)

Figure 7.22  Sections from a cell block preparation of an aspirate from an inflamed odontogenic keratocyst. (a) Lamellae of keratin
can clearly be seen among the inflammatory cells. (b) Pan-­cytokeratin (AE1/AE3) highlights the keratin. Source: Courtesy of Prof.
Pablo Vargas.
­Histopatholog  131

showed inflammation (see also Figure  7.22a). A more investigated CK19. These showed either no differences
recent study (Patidar et al. 2015) supported these findings between the different cyst types, or wide variability within
and showed that epithelial cells could be found in aspirates or between the studies. This degree of variability suggests
from 14 of 15 (93.3%) keratocysts compared to only 3 of 15 that the use of these cytokeratins as a diagnostic test on a
(20.0%) non-­keratinised odontogenic cysts. single isolated case would be inappropriate and grossly
These studies demonstrate that aspiration cytology, espe- inaccurate.
cially with examination of cell blocks, may provide a useful A further limitation is that if the clinical, radiological,
and accurate method for a preoperative diagnosis of the and histological features of the individual cyst types are
odontogenic keratocyst. However, among all radiolucent characteristic and if strict diagnostic criteria (Boxes  7.2
lesions of the jaws, keratocysts are relatively rare and and  7.3) are followed, a histological diagnosis of odonto-
inflammatory cysts are more common. If aspirates are used genic keratocyst is rarely a problem and immunocyto-
indiscriminately, the vast majority will show inflammatory chemical studies are not needed. It is also notable that in
cells and some epithelial cells, but will be non-­diagnostic. most studies comparing different cyst types, the specimens
A diagnosis of keratocyst can only be made if keratinising are chosen to show the typical histological features, regard-
cells or lamellae of keratin are seen. Aspiration biopsies less of the fact that if the features are typical, then there is
should only be used when a preoperative diagnosis is clini- no requirement for staining beyond a good H&E-­stained
cally indicated and a conventional biopsy may be difficult. section to make the diagnosis.
It is most beneficial for large lesions, especially in the pos- Despite these limitations, most studies have shown that
terior mandible, where there may be a wide differential odontogenic keratocysts consistently express cytokeratins
diagnosis, and large lesions in the posterior maxilla, where that are associated with a cornified epithelium, in par-
surgical access may be limited. ticular CK1, CK10, and CK11 (Shear 2002c), while these
are not seen in non-­keratinised cyst types. This is of
course consistent with the histological finding of keratin
Immunohistochemistry and Biomarker Studies
in keratocysts, and since this can be observed in routine
There are many studies investigating the expression of sections, then the role of these markers is limited.
cytokeratins and other biomarkers in odontogenic kerato- However, as discussed previously, expression of CK10 was
cysts and comparing expression to other cyst types. Most shown to be a useful diagnostic aid in cytological prepara-
studies have tried to determine whether particular patterns tions of cyst aspirates (August et al. 2000) and expression
of expression could provide accurate diagnostic markers of pan-­cytokeratin can also help identify keratin in aspi-
for the common odontogenic cysts. Other studies have rates of inflamed cysts (Vargas et  al.  2007; Figure  7.22).
used markers to explore the pathogenesis and mechanisms Although useful to differentiate keratocysts from denti-
of growth of the keratocyst. Studies of HH signalling path- gerous cysts, radicular cysts, and even ameloblastomas,
way–associated proteins and proliferation markers have similar cytokeratin expression may be seen in an
already been discussed in ‘Pathogenesis’. orthokeratinised odontogenic cyst (Aragaki et al. 2010). A
With regard to cytokeratins, Shear (2002c) and the previ- more recent study (Tsuji et  al.  2014), however, found
ous edition of this book reviewed more than 16 studies CK10 expression in the superficial layers of only 3 of 25
published between 1987 and 2005 and discussed their odontogenic keratocysts, compared to all of 15 orthokerati-
potential utility as diagnostic markers. The overall conclu- nised cysts. Conversely, they found that all 25 keratocysts
sion was that the diagnostic potential appeared to be lim- showed moderate to strong expression of CK17, but all
ited, due in part to the variability of results from different other cyst types, including orthokeratinised, dentigerous,
laboratories and a lack of standardised methodology. In and radicular cysts, were negative. Expression of
particular, many early studies used antibodies that had lim- CK17 has been shown to be associated with neoplastic or
ited availability, could only be used on fresh frozen tissues, proliferative epithelium, and a number of studies have
and produced results that were widely variable and could suggested that it is strongly expressed in keratocysts
not be compared (Smith and Matthews  1991). More (Meara et al. 2000; Stoll et al. 2005; Cserni et al. 2020b).
recently, Bhakhar et al. (2016) investigated the expression Nevertheless, it is not specific, since it may be weakly
of CK18 and CK19  in dentigerous cysts, odontogenic expressed in up to one-­third of dentigerous and radicular
keratocysts, and radicular cysts, and found that while more cysts (Meara et al. 2000; Stoll et al. 2005). These studies
dentigerous cysts than keratocysts expressed both demonstrate that, on average, keratocysts may show
cytokeratins, expression in radicular cysts was the same as strong expression of CK17, and this is consistent with the
in dentigerous cysts. These authors also reviewed 5 studies higher rate of proliferation, but for a single case CK17
that had investigated CK18 and 17 studies that had cannot exclude other cyst types.
132 Odontogenic Keratocyst

A particular diagnostic challenge may arise when a specific for ameloblastomas. In a similar study, De Villiers
pathologist is faced with a small biopsy from an inflamed et al. (2008) compared 19 ameloblastomas (including 3 uni-
cyst. Few studies have been able to identify markers that cystic cases) with 17 odontogenic keratocysts and found
can differentiate between the histologically identical fea- that all ameloblastomas showed calretinin expression, but
tures of an inflammatory cyst and an inflamed odonto- all keratocysts were negative. These authors and Altini
genic keratocyst. Cserni et al. (2020a) addressed this by et al. (2000) also found that calretinin was more strongly
comparing inflamed and uninflamed areas of 11 kerato- expressed in ameloblastomas with areas of squamous
cysts for expression of bcl2, CK17, CK10, and CK19. In metaplasia. Subsequently, two further studies have con-
typical uninflamed keratocyst epithelium, all markers firmed that odontogenic cysts, including keratocysts, are
were positive. Bcl2 was expressed in the basal and supra- negative for calretinin, but that ameloblastomas (including
basal layers, CK17 and CK19 showed strong expression unicystic ameloblastoma) are positive in up to 100% of
through the whole epithelium, and CK10 was expressed cases (De Villiers et  al.  2008; Jeyaraj  2019; Rudraraju
in the superficial layers, but was often patchy with focal et al. 2019).
unstained areas. In areas of inflammation the epithelium These studies suggest that calretinin is useful to distin-
was non-­keratinised and proliferative, but all showed guish ameloblastoma from odontogenic keratocyst, but
loss of bcl2 and complete (four cases) or weak and patchy none has investigated expression in inflamed lesions. Also,
(six cases) loss of CK17. Similarly, there was complete there are no studies that have investigated the utility of cal-
loss (three cases) or markedly reduced expression (eight retinin, or any markers, in distinguishing between a solid
cases) of CK10. CK19  was reduced in six cases. These keratocyst and keratoameloblastoma.
data show that the characteristic keratin profile of the
odontogenic keratocyst is lost when inflamed, suggesting
Malignant Change in Odontogenic Keratocysts
that immunocytochemistry for cytokeratin is of little
diagnostic value in inflamed lesions. The authors also Intraosseous or odontogenic squamous cell carcinoma is a
noted that inflammation and expression of cytokeratins central carcinoma of the jaws that arises from odontogenic
may be patchy, indicating that a diagnosis of keratocyst epithelium (Koutlas and Sloan 2022). Although they may
cannot be excluded if only a small sample is examined. arise from odontogenic epithelial rests, the majority prob-
Their finding of focal and patchy expression for ably arise from the epithelium of odontogenic cysts.
CK10 may explain the variable results for CK10 expres- Although about 70% of primary intraosseous carcinomas
sion in studies on histological sections (Aragaki arise from cysts, it has been estimated that less than 0.2% of
et al. 2010; Tsuji et al. 2014), but consistent positivity in cysts may show malignant change (Stoelinga and Bronkhorst
aspirates (August et al. 2000). 1988; Bodner et  al.  2011; Borrás-­Ferreres et  al.  2016).
In the posterior mandible, the two most common lesions Because the odontogenic keratocyst shows an increased
that must be considered in the differential diagnosis are rate of cell proliferation and an increased number of
dentigerous cyst and ameloblastoma, and a preoperative mitotic figures, early papers suggested that they have a
diagnosis is always desirable before definitive surgery, greater tendency to malignant change than other cyst types
especially for large lesions. At this site an aspiration biopsy (Toller  1967). Others have drawn attention to the occa-
may be useful, but often a small incisional biopsy is taken sional finding of epithelial dysplasia in keratocysts, but this
in the retromolar area. For typical lesions a biopsy should is actually very rare and has not been associated with
be diagnostic, but occasionally a cystic or unicystic amelo- malignant change (Brannon  1977; Ahlfors et  al.  1984;
blastoma may show features similar to a keratocyst, espe- Cottom et al. 2012).
cially if there is an element of inflammation. In this Ye et al. (2020) presented 5 cases of carcinomas arising
context, immunohistochemistry for calretinin may be use- in odontogenic keratocysts and reviewed a further
ful, since it has been shown to be expressed in a high pro- 29 well-­documented cases up to 2020. The average age of
portion of solid, unicystic, and multicystic ameloblastomas presentation was 48.8 years (range 15–81) and 58.6% arose
(Altini et al. 2000; Coleman et al. 2001). Altini et al. (2000) in males. The majority (69.0%) were in the mandible and
showed that calretinin was expressed in 22 of 27 (81.5%) 37.9% arose in recurrent cysts. All the cases were squa-
unicystic ameloblastomas and in 29 of 31 (93.5%) solid/ mous cell carcinomas, although one was also described as
multicystic ameloblastomas. In a subsequent study, the verrucous. Only three patients (8.8%) developed cervical
same authors (Coleman et  al.  2001) studied a series of lymph node metastases, but a further three developed dis-
22 keratocysts, 26 residual cysts, and 20 dentigerous cysts, tant metastases and the overall five-­year survival was only
and found that all were negative for calretinin, suggesting 53.2%. Ye et  al. (2020) did not comment on the criteria
that, in the context of odontogenic lesions, calretinin is applied for diagnosis in the reviewed cases, but in all of
Recurrence of Odontogenic Keratocyst  133

their own cases they demonstrated evidence of a typical Overall, the accumulated data suggest that malignant
parakeratinised keratocyst lining. In a recent systematic change in the odontogenic keratocyst is very rare and is no
review, Kumchai et al. (2021) found 34 papers reporting more common than in other cyst types. When it does occur
37 carcinomas arising in odontogenic keratocyst. it presents at a slightly later age than a conventional kerato-
However, 9 cases were recorded as arising in an cyst, but the sex and site distribution are similar.
orthokeratinised ‘variant’, suggesting that these were Histologically, the lesion shows areas of conventional
associated with orthokeratinised odontogenic cyst. The mean squamous cell carcinoma arising in continuity with a typi-
age was 45.1 years (range 8–81 years). These authors cal keratocyst epithelial lining (Figure 7.23).
found that the majority of cases (59.5%) were diagnosed
when there was histological evidence of carcinoma in
continuity with a typical cyst lining (e.g. Figure 7.23). Recurrence of Odontogenic
In the remaining cases (40.5%) the diagnosis was made on Keratocyst
the basis of the carcinoma arising at a site of a previously
diagnosed odontogenic keratocyst. It is well established that the odontogenic keratocyst has a
With regard to the relative frequency of malignant particular tendency to recur after surgical treatment. Many
change in keratocysts, Bodner et al. (2011) reviewed the authors and textbooks state that the keratocyst has the
literature and found reports of 116 intraosseous carcino- highest recurrence of all the odontogenic cysts, and cite
mas that had arisen in odontogenic cysts between 1938 this as evidence of its different and more aggressive behav-
and 2010. Only 16 (13.8%) arose in keratocysts compared iour, but there is so much variability in reported recurrence
to 19 (16%) in dentigerous cysts and 70 (60%) in radicular rates that it is difficult to provide a definitive probability of
cysts. Of the lesions, 85% were well or moderately differ- recurrence for any individual lesion. It should also be noted
entiated squamous cell carcinomas, and there were no that the overall recurrence rate for the odontogenic kerato-
differences in lesions from different cyst types. Although cyst is about 20% (Table  7.10), and this is similar to that
these data suggest that malignant change is far more com- reported for glandular odontogenic cyst and botryoid odon-
mon in radicular cysts, they do not take account of the togenic cyst, both of which are also characterised by a mul-
relative frequency of each cyst type. Since radicular cysts tilocular morphology and have recurrence rates of between
are about six times more common than odontogenic 20 and 30% (see Chapters 8 and 10).
keratocysts, then the frequency of reports of carcinomas The earliest reports of recurrence suggested rates of
arising in odontogenic cysts is proportionate, suggesting between 50 and 60% (Pindborg and Hansen  1963;
that the actual rate of malignant change in each cyst type Hansen 1967; Toller 1967), but most later reports and larger
is similar. series have found lower rates. In the previous edition of
this book, we tabulated 32 reports that documented recur-
rence rates of between 3% (Voorsmit et al. 1981) and 62%
(Pindborg and Hansen  1963). In a similar, more recent
review, Mendes et al. (2010) reviewed 31 studies that had
correlated treatment and recurrence and found rates of
between 0 and 100%. They showed wide variability in
recurrence rates depending on treatment methods, but also
reported studies with 0% or 100% recurrence where the
number of cases included were three or fewer. Other
reviews have reported similar wide ranges of recurrence
(Kaczmarzyk et al. 2012; Diaz-­Belenguer et al. 2016), but
careful reading of the papers shows that recurrence rates of
50% or even 100% have been taken from studies where the
number of cases was only two.
There are many reviews and small case series that report
widely variable recurrence rates and great care is needed in
interpreting the data. Studies that have investigated clinical
and radiological factors associated with recurrence are
Figure 7.23  Malignant change in odontogenic keratocyst.
almost all retrospective analyses that compare ‘recurrent’
Islands of squamous cell carcinoma arise in continuity with
the keratocyst lining (left side). Source: Courtesy of Dr Lisette and ‘non-­recurrent’ cases. However, few authors define the
Collins. meaning of ‘recurrent’ and it is not always clear if the data
134 Odontogenic Keratocyst

Table 7.10  Summary of five systematic reviews that have evaluated the effectiveness of treatments and recurrence rates for the
odontogenic keratocyst.

De Castro
Blanas Johnson Al-­Moraissi Chrcanovic and et al. (2018)
et al. (2000) et al. (2013) et al. (2017) Gomez (2017) P = 29;
P = 15; N = 578 P = 8; N = 362 P = 35; N = 2287 P = 94; N = 6427 N = 1321

n R (%) n R (%) n R (%) n R (%) n R (%)

Enucleation and/or curettage 413 28.1 156 25.6 1269 19.9 2639 23.3 1049 20.8
Enucleation and Carnoy’s 60 1.6 63 7.9 549 11.5 319 6.6 – –
Enucleation and cryotherapy 16 31.3 – – 137 14.5 91 23.1 – –
Marsupialisationa 45 24.4 21 4.8 63 32.3 226 24.8 126 18.3
Marsupialisation and – – 76 15.8 141 14.6 102 14.7 146 13.7
cystectomy
Resection 38 0 16 6.3 92 8.4 159 2.5 – –
All treatments 23.2 18.7 16.6 22.8 19.8

N, total number of cysts included; n, number of cysts in each treatment group; P, number of papers included in the review; R (%), proportion (%)
that recurred.
a
 Some studies include decompression as well as marsupialisation.

presented are for primary lesions that have subsequently Clinical and Radiological Factors
recurred, or for the actual recurrent lesions. When consult- A number of clinical factors have been shown to be associ-
ing the literature, readers are cautioned not to cite the sum- ated with recurrence, but there is great variability between
mary findings presented in abstracts without consulting studies. For example, Woolgar et al. (1987c) explored fac-
definitions and the detailed data in the text of the paper. tors associated with recurrence by comparing 228 solitary
non-­syndromic keratocysts, followed for five or more years,
with 44 recurrent cysts for which data were available for
Factors Associated with Recurrence
both the primary lesion and the recurrence. They found no
Woolgar et al. (1987c) suggested that the three main causes differences in age distribution, sex, or location between the
of recurrence are incomplete removal of the cyst lining; non-­recurrent and recurrent cysts. In similar studies, Zhao
growth of a new cyst from satellite cysts or epithelial et al. (2002), Berge et al. (2016), and Naruse et al. (2017)
islands left behind at the time of surgical removal; and an studied recurrences in 255, 92, and 65 cases, respectively.
apparent recurrence associated with a new cyst developing They reported overall recurrence rates of 12%, 29%, and
at or adjacent to a site of a previous removal. This latter 20%, but found no associations with any clinical or radio-
association is most often seen in patients with NBCCS, logical parameters. Myoung et al. (2001), however, found
when a new cyst may be interpreted as a recurrence. that the highest recurrence rates (82.4%) were found in
With regard to the first two causes, these can be miti- patients in the fifth decade and for cysts located in the
gated against by using treatment methods that aim to mandibular molar region (75%). Chrcanovic and Gomez
ensure complete removal of the cyst as well as any satel- (2017) reviewed 94 publications reporting 6427 keratocysts
lite cysts or epithelial islands. It is not surprising, there- and found an overall recurrence of 22.8%. Meta-­analyses
fore, that the accumulated evidence from large case showed that clinical or radiological factors significantly
series and from systematic reviews suggests that the rate associated with an increased risk of recurrence were an
of recurrence is primarily associated with the type of association with NBCCS (34.3% recurred; n  =  329) and
treatment; this is discussed at the end of this chapter. multilocular lesions (25.7% recurred; n  =  447) compared
However, the success of treatment may also be affected with unilocular lesions (14.3% recurred; n  =  1027).
by a number of clinicopathological factors, including the Recurrence was not associated with age, sex, or location
location and size of the lesion, association with teeth, (mandible or maxilla).
cortical perforation, the presence or number of satellite MacDonald et  al. (2013) followed patients for a mini-
cysts or islands, association with NBCCS, and the length mum of five years and found that those with cysts that
of the follow-­up period. recurred were significantly older (43.4 years; n = 18) than
Recurrence of Odontogenic Keratocyst  135

patients with non-­recurrent lesions (28.0 years; n = 11), but year, meaning that the true recurrence rates remain uncer-
there were no differences in sex. There were no significant tain and it is difficult to compare studies.
associations with size of the cysts, expansion, cortication of These studies show great variability in clinical and radio-
the margins, multilocularity, association with teeth, root logical factors that might predict recurrence of the odonto-
resorption, or involvement of the maxillary antrum. Of genic keratocyst, and suggest that they may not be helpful for
interest, however, is that they did find that keratocysts in making management decisions about individual lesions.
the posterior mandible and lesions with a preoperative Although a number of studies suggest that larger and multi-
diagnosis of odontogenic keratocyst were less likely to recur. locular lesions, especially in the posterior mandible, may be
As discussed previously (see ‘Radiological Features’), more likely to recur, this is contradicted by others who have
MacDonald (MacDonald et al. 2013; MacDonald 2016) has suggested that smaller lesions in dentate areas show the
suggested that large and multilocular lesions are more highest recurrence rates (MacDonald et  al.  2013;
likely to have a preoperative diagnosis of odontogenic MacDonald 2016; Slusarenko da Silva et al. 2019a). The dif-
keratocyst and to be treated less conservatively, and thus to ferent findings probably reflect different treatment
be less likely to recur. Conversely, small unilocular radiolu- approaches, since it is suggested that the greater risk of recur-
cencies in the dentate areas of the jaws may be indistin- rence for smaller lesions is a lack of appreciation that the
guishable from common periapical lesions and may be lesion may be a keratocyst, resulting in more conservative
treated more conservatively, with a higher chance of recur- removal and retention of the teeth. A number of studies have
rence (MacDonald et al. 2013; MacDonald 2016; Slusarenko shown that preservation of involved teeth (Chirapathomsakul
da Silva et al. 2019a). In support of this, MacDonald et al. et al. 2006; Cunha et al. 2016; Fidele et al. 2019) is associated
(2013) found that small unilocular lesions located in the with a high risk of recurrence, and suggest that this is due to
premolar region were only seen in the group of keratocysts the retention of residual fragments of cyst lining or epithelial
that recurred. These data are supported by a number of islands that reside adjacent to the tooth roots and are not
other studies that have shown that recurrence may be asso- removed during conservative enucleation.
ciated with conservative enucleation with retention of
involved teeth (Chirapathomsakul et  al.  2006; Cunha Histological Factors
et al. 2016; Fidele et al. 2019). In their study, Cunha et al. A number of papers have investigated histological factors
(2016) treated 18 keratocysts that involved the roots of the that may be associated with recurrence, but the findings
teeth by enucleation. In 9 cases the teeth were removed have been variable. In a comparison of the histology of 44
and none recurred, but 6 of 9 (66.7%) cases where the teeth cases each of primary, recurrent, and non-­recurrent cysts,
were retained recurred. Fidele et  al. (2019) reviewed Woolgar et  al. (1987c) found no histological factors that
274 keratocysts that had been enucleated. In 56 cases teeth might predict recurrence. The only significant finding was
involved with the cyst were preserved and 42 (75%) of these less inflammation in the recurrent lesions compared with
recurred, compared to only 6 recurrences (2.8%) among the primary cysts. Of note, however, is that satellite cysts,
218 cases where teeth were extracted or not involved. solid islands, odontogenic rests, or budding did not show
Furthermore, they found that 19 of the 56 patients with any association with recurrence. Others have confirmed
involved teeth also had cortical perforations and all 19 these findings and have also found no relationship between
(100%) recurred, compared to only 23 (62.2%) of the 37 recurrence and the presence of satellite cysts or islands in
cases without evidence of cortical perforation. the wall (Brannon  1977; Yagyuu et  al.  2008; Cottom
Another reason for the considerable variation in recur- et  al.  2012; Shimada et  al.  2013; Naruse et  al.  2017;
rence rates reported by different workers may be the wide Augustine et al. 2021).
variability in the reported follow-­up periods. The majority Nevertheless, others have shown an increased rate of
of recurrences occur within the first 5 years, but many recurrence of keratocysts that had satellite cysts or epithelial
keratocysts have recurred 10 years or more after surgery, islands. Myoung et al. (2001) found that about 75% of cysts
with occasional reports of recurrent lesions after 25 that recurred had satellite cysts and this had a statistically
(Stoelinga  2001) or even 40 years (Crowley et  al.  1992; significant association with a higher rate of recurrence. De
Fidele et al. 2019). Berge et al. (2016) followed patients for Franca et al. (2020) compared 22 non-­recurrent cysts with 18
up to 25 years and found that although the mean time to that recurred and found that only 1 (4.5%) of the non-­
recurrence was 53 months, the range was between 9 months recurrent group had satellite cysts, compared to 50% of the
and 11.5 years. The risk of recurrence increased up to 7 cysts that recurred (P = 0.002). There were no differences,
years after diagnosis and then remained constant at about however, between the primary lesions and the recurrence.
40%. The majority of studies have reported follow-­up peri- In contrast to Woolgar et al. (1987c), they also found signifi-
ods of between 1 and 5 years, and occasionally less than 1 cantly greater inflammation in recurrent cysts. Similarly,
136 Odontogenic Keratocyst

Cunha et al. (2016) found that epithelial islands or budding has been the subject of many hundreds of papers since the
were associated with a higher recurrence rate. Cottom et al. keratocyst was first described in 1956, but more recently there
(2012) found no differences between recurrent and non-­ have been a number of systematic reviews that have attempted
recurrent lesions with regard to satellite cysts or epithelial to explore this relationship (Blanas et al. 2000; Kaczmarzyk
islands, but did find that recurrence was significantly associ- et al. 2012; Johnson et al. 2013; Antonoglou et al. 2014; Sharif
ated with subepithelial hyalinisation, separation of the epi- et al. 2015; Dias et al. 2016; Diaz-­Belenguer et al. 2016; Al-­
thelium, and the number of basally located mitoses. More Moraissi et  al.  2016a,b,  2017; Chrcanovic and Gomez  2017;
recently, Augustine et al. (2021) also showed that subepithe- Tabrizi et al. 2019; Slusarenko da Silva et al. 2019b).
lial hyalinisation was associated with recurrence. Before considering treatment options, it needs to be stated
We have previously discussed the work of Stoelinga and that overall, the evidence for or against any method is weak.
colleagues (Stoelinga  1976,  2001,  2005; Stoelinga and This is because of the wide variability or heterogeneity
Peters 1973; Stoelinga et al. 1975; see ‘Pathogenesis’), who between studies and a complete lack of any randomised
have described epithelial islands and microcysts in the controlled trials. In a Cochrane systematic review, where
walls of odontogenic keratocysts and have suggested that the primary inclusion criterion was randomised controlled
these are located almost exclusively in the superficial por- trials, no studies were identified (Sharif et al. 2015), and the
tions of the cyst wall and are often seen in areas where the authors had to conclude that they were unable to assess
cyst is continuous with the oral mucosa through fenestra- the effectiveness of any interventions for the treatment
tions in the overlying bone. In an early study, seven odon- of  the odontogenic keratocyst. Subsequent reviews have
togenic keratocysts were examined histologically in serial also failed to identify any randomised trials and all authors
sections, and microcysts and islands were only found in the have highlighted the need for properly controlled prospec-
oral mucosa above the cysts (Stoelinga and Peters  1973). tive studies. Nevertheless, systematic reviews have
No islands or microcysts were found in other regions of the attempted to assimilate the evidence from case series and
cyst wall. Subsequently, Stoelinga (2001,  2005) has sug- retrospective cohort studies, and some have attempted
gested that there is no evidence that microcysts can be meta-­analyses. Overall, however, they have shown great
found in the bone surrounding keratocysts and that, for variability between studies and most comparisons of treat-
this reason, surgical resection of keratocysts is not justified. ment methods have been inconclusive. The major causes of
Furthermore, he proposes that microcysts and islands heterogeneity have included small numbers of subjects;
overlying the cysts are a major cause of recurrence and inadequate follow-­up period; poor definition of recurrence;
advocates that when keratocysts are enucleated, the overly- inclusion of orthokeratinised ‘variants’; lack of histological
ing mucosa should also be excised. As further evidence of verification; poor definition of treatment methods; and no
this, he has discussed the issue of recurrences in bone controls for clinicopathological factors such as site or size of
grafts used to repair surgical defects after resection the lesions, involvement of teeth, or association with
(Stoelinga and Slusarenko da Silva 2021). The authors sug- NBCCS. For these reasons, a number of the reviews
gest that the source of the recurrences must be found in the included very few studies (Kaczmarzyk et al. 2012; Sharif
overlying mucosa that is replaced after surgery. et al. 2015; Al-­Moraissi et al. 2016b; Dias et al. 2016) or were
Studies of cell proliferation markers have been discussed unable to reach any conclusions (Antonoglou et  al.  2014;
previously, but it is of note that Li et al. (1995) and, more Diaz-­Belenguer et  al.  2016). Despite this, a number of
recently, Naruse et al. (2017) found no relationship between authors have reviewed large numbers of keratocysts and
the number of Ki-­67–positive cells and recurrence. Both have produced qualitative analyses that suggest differences
authors have proposed that recurrence is not associated between treatments. These are summarised in Table 7.10.
with increased proliferation, but is primarily a result of
inappropriate surgery or inadequate removal (Li et al. 1995;
Treatment Methods
Li 2011; Naruse et al. 2017).
The most common treatment is enucleation, but this has
been applied either alone or with other adjunctive meth-
Treatment and Recurrence ods, and in some cases is poorly defined. The treatment
of Odontogenic Keratocyst methods used in the management of the odontogenic
keratocyst are briefly described.
Treatment and recurrence are considered together because all
studies, including systematic reviews, that have investigated Enucleation and Curettage
the effectiveness of treatment have used recurrence as the pri- In early studies enucleation and curettage were considered
mary outcome measure. Evaluation of treatment methods as two separate methods of surgical management (Blanas
Treatment and Recurrence of Odontogenic Keratocyst  137

et  al.  2000). Curettage was defined as surgical scraping (n = 35) of their patients over a 12-­year period were treated
(curetting) of the cyst cavity with removal of the contents, with resections. All cases were in the mandible and the
and usually resulted in piecemeal removal of the cyst in majority (77%) involved the molar–ramus area. Most
multiple fragments. Enucleation was defined as removal of (88.6%) were treated with segmental resection. Only one
the lesion by shelling out, so the cyst was removed intact. case recurred. The authors acknowledged that resections
However, the lining of the odontogenic keratocyst is thin are rarely indicated, but suggested a management protocol
and friable, so true enucleation is rarely achieved. whereby a resection should be considered for large lesions
Simple cyst enucleation without curettage is no longer with evidence of cortical perforations or extension into
advocated, and enucleation and curettage should be adjacent tissues.
regarded as components of the same method and are often
used synonymously. The cyst is enucleated, intact or piece- Adjunctive Methods
meal, and the underlying bone is curetted. Some authors A number of additional methods have been used to accom-
regard curettage as an adjunctive method to remove super- pany the primary treatment with the aim of ensuring com-
ficial bone in the exposed cavity, with the aim of ensuring plete removal of the cyst along with any satellite cysts or
removal of residual lining or any satellite cysts or epithelial epithelial islands. Adjunctive methods are used in combi-
islands. In this respect curettage has occasionally been nation with enucleation. The most commonly used adjunc-
used as meaning peripheral ostectomy (see ‘Adjunctive tive methods are the application of Carnoy’s solution,
Methods’). cryotherapy, peripheral ostectomy, and excision of the
overlying mucosa.
Marsupialisation and Decompression Carnoy’s solution is a tissue fixative that can be applied to
These two terms are often used synonymously, since the the cyst cavity to ‘fix’ or render non-­viable any residual cyst
objective of both methods is to ‘decompress’ the cyst by lining or islands. The solution is made up of alcohol, glacial
exposing the lumen and releasing the intracystic pressure. acetic acid, ferric chloride, and chloroform. In recent years,
However, the procedures are quite different. Marsupiali­ however, there has been a ban on the use of chloroform in
sation involves removing a wide area of superficial cyst some countries (primarily in the USA) and some surgeons
wall, including overlying bone, and suturing the cyst wall have used Carnoy’s without chloroform, but the compara-
to the adjacent mucosa. This creates a pouch exposed to the tive effectiveness of each variant is not known (Ecker
oral cavity that is left open, allowing the cyst to reduce in et  al.  2016). Carnoy’s solution can be applied to the cyst
size as the cavity heals. The pouch may be left open for up lining before it is enucleated, but most often the solution is
to 24 months and must be cleansed by the patient on a reg- applied to the bone cavity after the cyst has been enucle-
ular basis. Sometimes the lesion resolves with marsupiali- ated (Stoelinga 2001, 2005). The solution is applied for up
sation alone, but often the residual cyst lining is enucleated to 15 minutes and may penetrate about 1.5 mm into the
after the lesion has reduced in size. Marsupialisation is bone. Care is needed, because Carnoy’s solution will also
becoming increasingly used and is particularly useful for damage normal tissues and it should not be applied to
larger lesions, where reduction in the size of the cyst allows exposed tooth roots or nerves, or when there is cortical per-
a more conservative enucleation with less risk to vital foration or involvement of the maxillary sinus.
structures. Cryotherapy serves a similar function to Carnoy’s solution,
Decompression is similar to marsupialisation in that the but uses a liquid nitrogen cryoprobe to freeze the bone cavity
objective is to relieve the intracystic pressure, but in this with the purpose of killing any residual lining or epithelial
case the cyst lumen is exposed through a small hole and islands. Usually the cavity is frozen two or three times for one
kept patent with a tube or gauze. Decompression is always minute each. Schmidt and Pogrel (2001) used a combination
followed by enucleation after an appropriate period. of enucleation and liquid nitrogen cryotherapy, and found
that only 3 of 26 (11.5%) cysts recurred over a 10-­year period.
Resection Peripheral ostectomy is used after enucleation to remove
The term resection is used in the conventional sense to a margin of bone at the periphery of the cyst. The purpose
mean surgical removal of the whole lesion with a surround is to remove between 1 and 2 mm of bone with the aim of
of up to 1 cm of adjacent normal bone. Resection may be eliminating any residual lining or islands. Ostectomy may
segmental, whereby a whole segment of the jaw (usually be carried out by firm sharp scraping or curettage of the
the mandible) containing the cyst is removed, or marginal, bone cavity, but more often is done using a round bone bur.
where a rim of bone is removed with the cyst, but the lower Sometimes this has been facilitated by dying the surface of
or posterior border of the mandible is left intact. In one of the bone cavity (for example with toluidine blue) and
the largest studies, Fidele et al. (2019) reported that 6.19% removing the dye to ensure complete surgical cleansing.
138 Odontogenic Keratocyst

Excision of the overlying mucosa has been championed by Another study examined the effectiveness of marsupialisa-
Stoelinga (2001,  2005) on the basis that the superficial tion alone (Zecha et  al.  2010). These authors treated
aspect of the cyst wall, in continuity with the overlying 68 keratocysts, 58 with enucleation alone and 10 with mar-
mucosa, is the site of most of the microcysts and epithelial supialisation alone, with no further cystectomy. The recur-
islands. The process involves removal of the overlying rence rates were 20.7% and 40%, respectively.
mucosa in continuity with the enucleated cyst. In the pos- These data support the findings summarised in
terior regions this means removal of the mucosa in the ret- Table 7.10, and suggest that marsupialisation alone is not
romolar region just posterior to the last standing molar, or an effective treatment and should be followed by enuclea-
removal of the mucosa of the maxillary tuberosity. If a cyst tion of the cyst lining with or without application of
is being marsupialised or decompressed, then access to the Carnoy’s solution or cryotherapy.
cyst lumen should involve removal of the appropriate area Al-­Moraissi et al. (2016b) undertook a systematic review
of mucosa. to determine the effectiveness of excision of the overlying
mucosa. They identified two papers (Voorsmit et al. 1981;
Stoelinga 2001), which had reported 90 keratocysts treated
Evaluation of Treatment Methods
by enucleation alone and 82 treated by enucleation with
Table 7.10 summarises five of the larger systematic reviews excision of the overlying mucosa. The recurrence rates in
that have evaluated the most common treatments for the the two groups were 14.4% and 4.8%, respectively, suggest-
odontogenic keratocyst. As already mentioned, it is impor- ing that removing the mucosa reduced recurrence.
tant to note that none of these reviews has been able to However, close examination of the data shows that 78
identify any randomised controlled trials and that hetero- (95%) of the cysts in the group that had mucosa excised
geneity across the studies means that quantitative analyses were also treated with Carnoy’s solution, compared with
are difficult, with very few overall significant differences. only 5 (5.6%) in the enucleation-­only group. It seems there-
Despite this, it can be seen that the most common treat- fore that the treatment benefit was also due to the use of
ment is enucleation and that in all analyses this results in a Carnoy’s solution and the authors concluded that there is
recurrence rate of between about 20 and 30%, which is no evidence to support the beneficial effect of removing the
similar to or higher than the overall recurrence rate for all overlying mucosa. They did however recommend that exci-
treatments of about 20%. A further consistent finding is sion of the mucosa should be performed, since it is easy to
that enucleation followed by the application of Carnoy’s do and because there is good evidence that epithelial
solution shows the lowest recurrence rates, from as low as islands may reside in the mucosa overlying the cyst
1.6% to 11.5%. In most studies, marsupialisation alone has (Stoelinga 2001, 2005).
a similar recurrence rate to enucleation, but if this is fol-
lowed by cystectomy (enucleation of the cyst) then the rate Summary and Conclusions
drops to about 15%. As would be expected, resection of the The accumulated evidence from studies of different treat-
lesion is associated with a very low recurrence rate. ments shows that resection of the odontogenic keratocyst
These reviews also suggest that marsupialisation before is the most effective for the avoidance of recurrence.
enucleation may have a better outcome than enucleation However, there is a general consensus in the literature that,
alone. Slusarenko da Silva et  al. (2019b) considered this in most cases, resection is an unnecessarily aggressive
issue specifically and reviewed six studies that had reported treatment for a benign cystic lesion. The most effective
202 keratocysts. The recurrence rate for cysts that had been treatment therefore appears to be enucleation with or with-
enucleated was 28.9% (n  =  135), compared to 16.4% out peripheral ostectomy, followed by treatment with
(n  =  67) when enucleation was preceded by up to Carnoy’s solution. This may result in a recurrence rate of
23.5 months of marsupialisation. A meta-­analysis, how- less than 2% (Table 7.10).
ever, showed that although this difference was consistent Stoelinga (2001, 2005) proposed a treatment strategy for
across all studies, it was not significant. In a similar study, the odontogenic keratocyst and Chapelle et  al. (2004)
Tabrizi et  al. (2019) reviewed 192  keratocysts treated by developed these ideas into decision trees for diagnosis and
marsupialisation alone (n  =  118) or by marsupialisation treatment of cystic lesions of the jaws. They suggested that
followed by cystectomy (n = 64) and found recurrences of any unilocular cystic lesion in the mandible or maxilla,
27.1% and 10.9%, respectively. In a study by Zhao et  al. except for those in the mandibular third molar or ramus
(2002), 255 patients were treated by enucleation (n = 163), region, should be enucleated and submitted for histological
enucleation with Carnoy’s solution (n = 29), marsupialisation examination. If a keratocyst is subsequently diagnosed, a
followed by enucleation (n = 11), or resection (n = 52). The ‘wait and see’ policy, with strict follow-­up, can be applied,
recurrence rates were 17.8%, 6.7%, 0%, and 0%, respectively. since any recurrences at these sites are easy to treat.
Treatment and Recurrence of Odontogenic Keratocyst  139

Unilocular lesions that are not dentigerous, in the man- (Table  7.10; Zhao et  al.  2002; Zecha et  al.  2010; Tabrizi
dibular third molar region or in the ascending ramus, are et al. 2019; Slusarenko da Silva et al. 2019b).
likely to be keratocysts and should be treated with careful A further consideration is the management of involved
enucleation, including excision of the overlying mucosa, teeth. A number of studies discussed previously
followed by application of Carnoy’s solution or cryotherapy. (Chirapathomsakul et  al.  2006; Cunha et  al.  2016; Fidele
If histological examination confirms a keratocyst, then et al. 2019) have shown that preservation of involved teeth
careful follow-­up is indicated. If the lesion proves to be a is associated with a risk of recurrence, suggesting that teeth
solid ameloblastoma, then further surgery may be indicated. should be removed. However, this is not straightforward,
Multilocular lesions are more difficult to manage and since the diagnosis of an unsuspected odontogenic kerato-
because of the potential for a wider differential diagnosis, cyst will not be made until after the lesion has been enucle-
an incisional or aspiration biopsy is always indicated. If an ated. Thus, a second operation for removal of the teeth may
odontogenic keratocyst is diagnosed, the cyst should be not be justified and, as discussed above, it has been sug-
enucleated with excision of the overlying mucosa and gested that small lesions, even if thought to be a keratocyst,
application of Carnoy’s solution or cryotherapy. can be treated conservatively with a ‘wait and see’ policy,
Although marsupialisation has become a more com- with removal of recurrences as they arise (Stoelinga
monly used treatment, the recurrence rates are variable 2001, 2005; Chapelle et al. 2004). This appears to suggest a
and there is little objective evidence to support its effective- policy of ‘managed recurrence’. Pogrel (2003a) also noted
ness. Marsupialisation may be particularly useful for the this approach in his historical review and quoted a surgeon
management of larger lesions, when shrinkage may enable as suggesting that repeated removal of recurrences was
subsequent enucleation with less risk to vital structures. ‘part of the normal management of jaw cysts’. Such an
Marsupialisation followed by enucleation may have recur- approach may in part explain the higher rates of recur-
rence rates similar to enucleation with Carnoy’s solution rence in the older literature.
140

Lateral Periodontal Cyst and Botryoid Odontogenic Cyst

CHAPTER MENU
Lateral Periodontal Cyst, 141
●● Clinical Features,  141
–– Frequency, 141
–– Age, 141
–– Sex, 141
–– Site, 142
–– Clinical Presentation,  142
●● Radiological Features,  142
–– Radiological Differential Diagnosis,  143
●● ­Pathogenesis,  144
–– Phase of Initiation and Source of Epithelium,  144
–– Reduced Enamel Epithelium,  144
–– Cell Rests of Malassez,  144
–– Cell Rests of Dental Lamina,  145
–– Phases of Cyst Formation and Growth and Enlargement,  146
●● ­Histopathology,  146
●● Treatment, 150
Botryoid Odontogenic Cyst, 150
●● Clinical Features,  150
–– Frequency, 150
–– Age, 150
–– Site, 151
–– Clinical Presentation,  151
●● Radiological Features,  151
●● Recurrence of the Botryoid Odontogenic Cyst,  152
●● Pathogenesis, 152
●● Histopathology, 152
●● Differential Diagnosis,  154
●● Treatment, 154

There has been a great deal of confusion about the relation- a generic sense and included any cyst that may have
ship between the gingival cyst of adults, lateral periodontal arisen adjacent to a tooth as a ‘lateral periodontal cyst’
cyst, and botryoid odontogenic cyst, and much of the early (Fantasia 1979; Eliasson et al. 1989). Thus, some cysts in
literature described them together as variants of the same the lateral periodontal position are really odontogenic
lesion. This appears to have arisen because they all have a keratocysts, while others may be lateral radicular cysts or
predilection for occurrence in the canine and premolar inflammatory collateral cysts.
region of the mandible and maxilla, and the pathogenesis, The relationship between the gingival cyst, lateral perio-
particularly with regard to the cells of origin, is similar dontal cyst, and botryoid odontogenic cyst was discussed
(Browne 1991b). The confusion is further complicated by by Wysocki et  al. in  1980, and their observations and
the fact that many early papers used ‘lateral periodontal’ in thoughtful discussion have not been bettered in the

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
­Lateral Periodontal Cys  141

intervening four decades. They postulated, on the basis of are summarised in Table 8.1. Most of the publications on
histological similarities, that they have a common his- the subject have been short case reports. Siponen et  al.
togenesis but show distinct clinicopathological differences. (2011) and de Andrade et al. (2012) reviewed the literature
They regarded the lateral periodontal cyst and gingival cyst and recorded about 270 cases up to 2012. Chrcanovic and
of adults as intraosseous and extraosseous variants of the Gomez (2019b) have undertaken a systematic review that
same lesion. Others agree with this interpretation, and also included reports of 213 lateral periodontal cysts.
emphasise the distinct differences in clinical presentation
and management (Shear and Pindborg  1975; Altini and Frequency
Shear  1992; Siponen et  al.  2011; de Andrade et  al.  2012; Lateral periodontal cysts are rare and represent less than
Chrcanovic and Gomez 2019a). 1% of all odontogenic cysts. In Shear’s series from the
The botryoid odontogenic cyst is regarded as a variant University of the Witwatersrand, only 24 lateral periodon-
of the lateral periodontal cyst, with a similar pathogene- tal cysts were encountered over a 46-­year period, represent-
sis but with distinctive, and important, differences in ing 0.7% of the total (Table 1.1). In the Sheffield series of
clinical and pathological features (Wysocki et  al.  1980; 7121 odontogenic cysts, only 28 (0.4%) lateral periodontal
Altini and Shear  1992; Chrcanovic and Gomez  2019b; cysts were diagnosed (Table 1.2; Jones et al., 2006). Of the
Vargas and White 2022). studies shown in Table 1.3, 11 included data for lateral per-
In this chapter, we will discuss the lateral periodontal iodontal cysts, with a range from 0.1% (Soluk Tekkeşin
cyst and botryoid odontogenic cyst as related lesions, but et al. 2012b) to 1.5% (Daley et al. 1994). Daley et al. (1994)
with distinctive clinicopathological features. Gingival cysts is the only study to record a frequency of greater than 1%.
of adults and infants, which have similar histogenesis and
presentations, will be considered together in Chapter 9. Age
Lateral periodontal cysts affect adults, with a peak in the
fifth or sixth decades and an overall mean age of about
­Lateral Periodontal Cyst 47 years (Table 8.1). They are almost unknown in children,
with studies showing an age range from 19 to 85 years
The lateral periodontal cyst is defined as a developmental (Wysocki et  al.  1980; Cohen et  al.  1984; Rasmusson
odontogenic cyst that is located at the lateral aspect of a et al. 1991; Carter et al. 1996; Jones et al. 2006). A system-
tooth or between the roots of erupted teeth (Vargas and atic review of 213 cases recorded a mean age of 46.8 years
White 2022). The cyst has a distinctive histology and other (range 12–81 years), with the greatest number of cases
cysts that may arise in a lateral periodontal position, (40%) in the sixth decade (Chrcanovic and Gomez 2019b).
including inflammatory cysts and odontogenic keratocyst, Figure 8.1 illustrates the age distribution of 114 patients,
must be excluded (Shear and Pindborg 1975; Browne 1991b). including the 24 from Shear’s series pooled with those of
Cohen et  al. (1984), Rasmusson et  al. (1991), and Carter
et al. (1996).
Clinical Features
Only a small number of case series have been published Sex
(Wysocki et  al.  1980; Cohen et  al.  1984; Rasmusson Wysocki et al. (1980) and Rasmusson et al. (1991) observed
et al. 1991; Carter et al. 1996; Jones et al. 2006) and these that about 70% of cases arose in males, but this was

Table 8.1  Age, sex, and site distribution from selected series of lateral periodontal cysts and from the systematic review of
Based on Chrcanovic and Gomez (2019b).

References n Mean age Peak decade Male (%) Mandible (%)

Wysocki et al. (1980) 39 50.0 6th 70.2 66.6


Cohen et al. (1984) 37 54.0 5th 48.6 78.3
Rasmusson et al. (1991) 31 55.0 6th 68.0 88.0
Carter et al. (1996) 25 49.4 5th 52.2 87.5
Jones et al. (2006) 28 48.2 6th 55.5 NR
Chrcanovic and Gomez (2019b) 213 46.8 6th 52.6 69.8

NR, not reported.


142 Lateral Periodontal Cyst and Botryoid Odontogenic Cyst

45 Figure 8.1  Age distribution of 114


41 patients with developmental lateral
40 periodontal cysts. Source: Based on Cohen
et al. (1984), Rasmusson et al. (1991), and
Carter et al. (1996).
35

30 28
25
No. of cases

25

20

15

10
6 7
4
5 2
1
0
0
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89
Age

unusual, since most other studies have reported more region. The other three were associated with lateral inci-
equal sex distribution (Table 8.1; Cohen et al. 1984; Carter sors. Cohen et al. (1984) recorded 3 cases associated with
et  al.  1996; Jones et  al.  2006). In their systematic review, mandibular incisors, 1 of which was between the central
Chrcanovic and Gomez (2019b) found a male to female incisors.
ratio of 1.1 (52.6% males : 47.4% females). Some studies
(Carter et al. 1996) have noted that the cysts may occur at a Clinical Presentation
significantly younger age in females (mean 40.5 years) than Lateral periodontal cysts are usually symptomless and are
in males (mean 58.4 years), but pooled data from 197 stud- typically discovered fortuitously during routine radiologi-
ies where these data were available showed no difference cal examination of the teeth. Only 12% show symptoms,
in age between the sexes (Chrcanovic and Gomez 2019b). although up to 50% may be associated with bone expansion
or erosion of the cortical plate (Chrcanovic and
Site Gomez 2019b). Lesions always occur on the buccal or labial
Overall, 70% of lateral periodontal cysts are found in the aspect of the alveolus and may present as a gingival swell-
mandible (Chrcanovic and Gomez 2019b), although there ing, which if the bone is perforated may be blueish and
is some variation between the larger series (Table 8.1). All fluctuant. Swelling is never a prominent feature, however,
reported cases have been found anterior to the first molars. since the cyst rarely exceeds 10 mm in diameter. The associ-
There are no reported cases arising in association with ated teeth will be vital, unless affected by another unre-
molars, except for a small number that are sited distal to lated pathology.
the root of a second premolar (Cohen et al. 1984; Rasmusson
et al. 1991; Carter et al. 1996).
Radiological Features
The most frequent location of a lateral periodontal cyst is
the mandibular canine/premolar area, followed by the The clinical and radiological features are typical and virtu-
anterior region of the maxilla. Wysocki et al. (1980) found ally diagnostic (Box 8.1). The cysts are found on the lateral
that 26 of their 39 cases (67%) occurred in the premolar– aspect of a tooth, or between the roots of two teeth, as a
canine–incisor region of the mandible and 7 of these were round or oval, well-­circumscribed radiolucent area, usually
located between the first and second premolars. In the with a corticated, sclerotic margin (Figure 8.2). If the teeth
studies of Cohen et al. (1984) and Rasmusson et al. (1991), are displaced, the lesion may become tear-­or pear-­shaped.
all the mandibular cases were in the canine/premolar area. The cysts lay somewhere between the apex and the cervical
Carter et al. (1996) found that 21 of 24 cases occurred in the margin of the tooth, but usually in the middle third. If a
mandible and 18 of these were in the canine/premolar large radiolucency fills the space between divergent teeth,
­Lateral Periodontal Cys  143

year. In their systematic review, Chrcanovic and Gomez


Box 8.1  Lateral Periodontal Cyst: Clinical
(2019b) found 144 cases where the size had been measured
and Radiological Features
and recorded a mean diameter of 10 mm (standard devia-
●● They are rare – less than 1% of odontogenic cysts tion [SD] ±6 mm; range 2–30 mm).
●● Only seen in adults, mean age about 50 years. Peak in As the cysts grow, they always expand towards the buccal
the sixth decade or labial (facial) aspect of the jaw, and the corticated mar-
●● Slightly more common in males gin becomes superimposed over the adjacent teeth
●● 70% are found in the mandible. Most common in the (Figure  8.2). A review of radiographs published in the
canine/premolar area many case reports shows that, almost without exception,
●● Always arise anterior to the second premolars the periodontal space is normal, and the lamina dura sur-
●● Round,well-­defined radiolucency with corticated margin rounding the tooth is intact (Figure 8.2, arrow). This is an
●● Very rarely greater than 10 mm in diameter important sign that serves to differentiate this developmen-
●● The periodontal space is normal and the lamina dura tal cyst from a lateral radicular cyst that may have arisen at
is intact the opening of a lateral root canal. The teeth may be dis-
placed in up to 40% of cases, but resorption of the roots is
rarely seen (Chrcanovic and Gomez 2019b).
Occasionally lateral periodontal cysts may be multifocal.
Siponen et al. (2011) described four patients with multiple
lesions. In two cases up to four separate lesions were seen
at different sites and all showed histological features of a
lateral periodontal cyst. A further two patients showed two
cysts at the same site. In the first patient, one cyst showed
a well-­demarcated round radiolucency, while the second
was more apically placed and was multilocular. In the sec-
ond case the two cysts abutted onto each other. Although
the lesions showed features of lateral periodontal cysts, the
authors speculated that fusion of multiple cysts may give
rise to a multicystic botryoid odontogenic cyst. Multiple
cysts are rare, however, and in their review Siponen et al.
Figure 8.2  Radiograph of a lateral periodontal cyst lying
between the mandibular premolar teeth. The margins are well
(2011) could find only three previous reports.
demarcated and corticated, indicative of slow enlargement. Note
that the periodontal space is normal, the lamina dura is intact, Radiological Differential Diagnosis
and the cyst is superimposed over the root of the tooth (arrow). A final diagnosis can only be confirmed on histological
Source: Courtesy of the late Professor J.J. Pindborg.
examination, but the radiographic features described above
are almost diagnostic. However, caution is needed, because
then an alternative diagnosis should be considered. In this a number of other lesions may arise at the lateral aspect of
respect the most important lesion to consider is a botryoid a tooth or between the tooth roots. The most commonly
odontogenic cyst, but odontogenic keratocysts may also encountered are a lateral radicular cyst, odontogenic
present at this site. The old term ‘globulomaxillary cyst’ keratocyst, botryoid odontogenic cyst, and glandular odon-
described an inverted pear-­shaped lesion that filled the togenic cyst. Fantasia (1979) examined 46 cysts that had
space between a divergent maxillary canine and second arisen at the lateral aspect of a tooth and found that only 13
incisor. If small they may be a lateral periodontal cyst, but (28.3%) met the histological criteria for lateral periodontal
many larger lesions are odontogenic keratocysts. The term cyst. The remainder were laterally placed radicular cysts
‘globulomaxillary’ should be used as a descriptive term (20 cases, 43.5%) and 8 (17.4%) odontogenic keratocysts.
only, or not at all (Dammer et al. 2014). Five cysts could not be categorised. In the series of 20 cases
Lateral periodontal cysts are rarely more than 10 mm in reported by Altini and Shear (1992), a reappraisal shows
diameter. In the series reported by Rasmusson et al. (1991) that 8 were probably botryoid odontogenic cysts and 3 were
and Carter et  al. (1996), only four and two cysts, respec- glandular odontogenic cysts.
tively (about 10%) measured more than 10 mm on radio- The key to diagnosis is attention to detail on the radio-
graphs. In four cases Rasmusson et al. (1991) were able to graphs. The lateral periodontal cyst is almost always round,
compare serial radiographs over a period of up to 14 years less than 10 mm in diameter, and does not disrupt the lam-
and estimated that the mean growth rate was 0.7 mm per ina dura. Radicular cysts arise from rest cells of Malassez
144 Lateral Periodontal Cyst and Botryoid Odontogenic Cyst

within the periodontal ligament, and therefore the perio- the fully erupted tooth. To explain the development of a
dontal space is expanded and the corticated margin of the multicystic variant or botryoid odontogenic cyst, they pro-
cyst is continuous with the lamina dura (see Chapter  3). pose that epithelial plaques may bud off from the cyst lin-
Botryoid odontogenic cysts, odontogenic keratocysts, and ing and form daughter cysts, which then expand to form a
glandular odontogenic cysts are rarely round in shape and multicystic lesion (Altini and Shear 1992).
may be multilocular or lobulated. They also grow and In support of this mechanism, they argued that the lin-
exceed 10 mm in diameter. Glandular odontogenic cysts ing of the lateral periodontal and botryoid odontogenic
often affect the anterior mandible, but usually reach a large cyst is composed for the most part of thin, non-­keratinised
size and extend across the midline. Any cyst found lateral to epithelium that resembles reduced enamel epithelium.
a tooth, which is greater than 10 mm in diameter or is mul- They also suggested that lateral periodontal cysts tend to
tilocular, is almost certainly not a lateral periodontal cyst. occur in areas where dentigerous cysts are likely to be asso-
ciated with vertically impacted teeth such as mandibular
premolars and maxillary incisors and canines, and that epi-
Pathogenesis
thelial plaques similar to those seen in the lateral periodon-
Three elements are needed for a cyst to develop: a source of tal cyst are occasionally found in dentigerous cysts.
epithelium, a stimulus for proliferation, and a mechanism This hypothesis is unsatisfactory in a number of respects.
for growth and bone resorption. While these are well First, if the enlarged follicle is displaced laterally as the
described for radicular cysts (see Chapter 3), the phases of tooth erupts, then it will develop within the crypt of the
the pathogenesis of developmental cysts are poorly under- developing tooth and will come to lie within the lamina
stood. Although we can speculate on the source of epithe- dura and will expand the periodontal space. However, as
lium, the initiating factors or the stimulus for proliferation described above, the lateral periodontal cyst invariably lies
are largely unknown. outside of the periodontal ligament, the periodontal space
It is widely accepted that the lateral periodontal cyst and is not expanded, and the lamina dura is intact. Second,
botryoid odontogenic cyst have similar histological fea- the hypothesis suggests that lateral periodontal cysts are
tures and are almost certainly derived from the same associated with those teeth that are also most likely to be
source of epithelium. In this section the pathogenesis of associated with dentigerous cysts that develop over verti-
these two cyst types will be considered together and we will cally impacted teeth. This is not in fact the case. Lateral
attempt to explain how each cyst might develop the distinc- periodontal cysts are most often found in the mandibular
tive morphological features. canine/first premolar region, which is a site rarely affected
by dentigerous (or eruption) cysts. Furthermore, such an
Phase of Initiation and Source of Epithelium association cannot explain why molar teeth, in particular
There has been considerable debate about which odonto- mandibular third molars, are never affected by lateral peri-
genic epithelium the lateral periodontal cyst and botryoid odontal cysts. Finally, it is suggested that the epithelial
odontogenic cyst arise from. Proof of origin from any par- plaques and thickenings that are typical of the lateral peri-
ticular source is lacking and any hypothesis must therefore odontal cyst may be seen in dentigerous cysts, and that this
be based on presumptive evidence. There seem to be three implies an origin from reduced enamel epithelium. In fact,
possibilities: reduced enamel epithelium, cell rests of epithelial thickening is rarely seen in dentigerous cysts and
Malassez, and remnants of dental lamina. The arguments the typical whorled or clear cell plaques of lateral perio-
for and against these three possibilities have been debated dontal cysts are not a feature of the lining of dentiger-
extensively in the literature and in previous editions of this ous cysts.
book. They will be briefly reviewed.
Cell Rests of Malassez
Reduced Enamel Epithelium The second possibility is an origin from the cell rests of
Shear and Pindborg (1975), Altini and Shear (1992), and Malassez. The rests of Malassez occur in the periodontium
previous editions of this book have argued that the reduced and they are well positioned for a lateral periodontal cyst,
enamel epithelium is the most likely source of the lining of but the support for this theory of origin is scanty. In an
the lateral periodontal cyst. They suggested a mechanism early paper and in the previous edition of this book, we
whereby the follicle overlying an erupting tooth may reported a case (Buckley et al. 1989) in which two separate
expand and develop into an early dentigerous cyst. developmental odontogenic cysts were associated with an
Subsequently, the cyst is displaced laterally (for example, unerupted lower third molar tooth. We suggested that
see Figures  5.7 and 5.10) and is then ‘left behind’ as the these were a lateral periodontal cyst and a dentigerous cyst,
tooth erupts, resulting in a cyst lying lateral to the root of and contended that this provided evidence that the
­Lateral Periodontal Cys  145

periodontal cyst may have an origin from the cell rests of The mechanism they propose is that cyst formation is
Malassez and not reduced enamel epithelium. This paper initiated by cystic degeneration in rests of dental lamina to
has been cited a number of times as evidence for an origin form a small microcyst that then develops into a unilocular
from the rest cells of Malassez. However, with the benefit cyst. Dental lamina rests are commonly found in the alveo-
of experience and a reappraisal, we now refute this idea. lar bone and gingival tissues (Hodson  1962) and often
Careful review of the images in Buckley et al. (1989) shows show a clear cell appearance, similar to the cells seen in the
a laterally (mesial) placed dentigerous cyst and a second lining of lateral periodontal cysts. In their paper, Wysocki
well-­defined cyst overlying the distal root of a mesio-­ et al. (1980) show that the walls of the lateral periodontal
angular displaced third molar. This second cyst is superim- cyst contain variable numbers of epithelial islands resem-
posed over the middle third of the root, but the periodontal bling rests of dental lamina, a feature that has been consist-
space is normal and the lamina dura is intact. Thus, it does ently described in these lesions (see “Histopathology”).
not appear to have an origin within the periodontal liga- They further suggested that a unicystic lateral periodontal
ment from the rest cells of Malassez. The origin is most cyst (or gingival cyst) arises through cystic change in a single
likely to be rests of dental lamina, which are known to be dental lamina rest and that a multicystic, botryoid odonto-
prominent at this site. Furthermore, this cyst is associated genic cyst develops if simultaneous changes occur in several
with a third molar tooth, which is very unusual for a lateral adjacent cell rests. As noted above, Altini and Shear (1992)
periodontal cyst. Review of the histology shows typical suggested that multicystic lesions arose from a budding or
whorled plaques, but also mucous cells, small duct-­like ‘pinching’ off of the epithelial plaques to produce mural
structures, and superficial columnar cells that would be islands that then undergo cystic change. Others (Redman
very unusual for a lateral periodontal cyst, but are now et al. 1990; Siponen et al. 2011) have proposed that some bot-
known to be typical features of a glandular odontogenic ryoid odontogenic cysts may arise by fusion of multiple lat-
cyst (Fowler et al. 2011; Speight and Rautava 2022). eral periodontal cysts at the same site (see discussion under
‘Radiological Features’). These alternative mechanisms for
Cell Rests of Dental Lamina the formation of a multicystic lesion are possible, but are still
The third potential source of the epithelium is the cell consistent with an origin from dental lamina rests.
rests of the dental lamina. This hypothesis was first put An origin from rest cells of the dental lamina is now con-
forward by Wysocki et al. (1980), who provided strong cir- sidered to be the most logical and likely explanation for the
cumstantial evidence and a logical description of how origin of these cysts (Wysocki et  al.  1980; Rasmusson
such a mechanism might work. They suggested that the et al. 1991; Siponen et al. 2011). This would explain why the
lateral periodontal cyst and gingival cyst of adults are both cysts lie adjacent to teeth, but outside of the periodontal liga-
derived from rests of dental lamina, and represent intraos- ment, and supports a unifying mechanism for the pathogen-
seous and extraosseous variants of the same entity. esis of unilocular and multilocular lesions (Figure 8.3).

Figure 8.3  A unifying mechanism for


the formation of unicystic and multicystic
lesions. The cysts arise by cystic
degeneration in rest cells of dental
lamina (top). A unicystic lesion arises
from cyst formation in a single rest, but if
multiple rests are involved a multicystic
lesion may be formed. Multicystic lesions,
whether enclosed in a single fibrous
capsule (unilocular) or multilocular,
should be diagnosed as botryoid
odontogenic cysts (see text).

Unicystic, unilocular Multicystic, unilocular Multicystic, multilocular

Lateral periodontal cyst Botryoid odontogenic cyst


146 Lateral Periodontal Cyst and Botryoid Odontogenic Cyst

However, if dental lamina rests are the source of the lat- cyst growth, after which the osmotic gradient equalises and
eral periodontal cyst, this does not easily explain why they intraluminal pressure reduces (Ward et  al.  2004; Kubota
are never found associated with third molars, where dental et al. 2004). In other cyst types proliferation of the epithe-
lamina rests are abundant. Also, although odontogenic lial lining may promote further growth, but that does not
keratocysts may arise at similar sites in the anterior aspects seem to the case with lateral periodontal and gingival cysts.
of the jaws, they are found more frequently at the angle of In summary, these considerations would support the
the mandible and, despite a similar origin from dental lam- view that the lateral periodontal cyst arises from the rest
ina, they show a quite different histology and pattern of cells of the dental lamina and that cyst growth is driven by
behaviour. Genetic factors are almost certainly involved, a high luminal osmotic pressure.
but Wysocki et al. (1980) also suggested that lateral perio-
dontal cysts arise in older age groups from postfunctional
Histopathology
cells of the dental lamina, whereas the odontogenic kerato-
cyst arises in younger individuals from dental lamina still Lateral periodontal cysts are small, with a thin and fragile
possessing marked growth potential. lining. They are usually removed conservatively by enu-
cleation with or without curettage. The pathologist often
Phases of Cyst Formation and Growth receives multiple fragments of a collapsed cyst wall.
and Enlargement Sectioning of a collapsed or fragmented cyst may cause a
These processes are poorly understood and have not been false impression of a multicystic or multilocular lesion
described for lateral periodontal cysts. There is little evi- microscopically. If there is doubt, multiple blocks and sec-
dence of inflammation, so the inflammatory processes tions should be examined and radiographs should be
described for other cyst types, including radicular cyst and reviewed to confirm a simple unilocular lesion. If a lesion
inflamed dentigerous cyst, are not involved. However, cyst is truly multicystic, then the correct diagnosis is probably
formation may still be promoted by accumulation of cell botryoid odontogenic cyst (see below).
debris within the degenerating cell rests or microcysts, On histological examination, lateral periodontal cysts
resulting in an increased luminal osmotic pressure that are composed of a fibrous wall lined by a thin, non-­
then drives cyst expansion and growth. Bone resorption keratinising squamous epithelium usually two to five cell
follows as a result of pressure on the surrounding tissues. layers thick (Figure 8.4). The epithelial cells are typically
Such a process is supported by the observation that the lat- flattened, but may be cuboidal. Quite often the wall
eral periodontal cyst and gingival cyst are spherical, sug- shows dense hyalinisation in the immediate subepithe-
gesting a process where intraluminal pressure causes lial area and we have noted that this is usually associated
centripetal growth, similar to that described for radicular with an exceptionally thin lining, only one cell thick, that
cysts (Chapter 3). Furthermore, the cysts are never large, is friable and separates from the connective tissue
suggesting a limited growth potential and consistent with (Figure 8.5)
the findings that osmotic pressure only plays a role in early

Figure 8.4  Lateral periodontal cyst is


lined by thin non-­keratinising epithelium.
Epithelial thickening can be seen (left)
and the wall often contains small islands
of epithelium (centre).
­Lateral Periodontal Cys  147

et al. 1980). Altini and Shear (1992) disputed this, however,


and suggested that the islands may arise by ‘pinching-­off’
of extensions of the epithelial plaques (Figure  8.7). Both
mechanisms may apply, but similar islands are seen in
other cyst types and in the alveolar mucosa, and are widely
regarded as residues of the dental lamina. Occasionally
these islands show clear cell change and/or microcystic
degeneration (see later Figure  8.9). The epithelial linings
may be tenuous and may separate from the fibrous cyst
wall, especially in areas of juxta-­epithelial hyalinised col-
lagen (Figure 8.5).
Although plaques and thickenings in the epithelial lin-
ings are a key diagnostic feature of the lateral periodontal
cyst (Box 8.2), they are not specific. They are also seen in
Figure 8.5  The wall of the lateral periodontal cyst may show
hyalinisation, and this is often associated with a very thin botryoid odontogenic cyst, gingival cyst of adults, and glan-
attenuated epithelium that separates from the underlying dular odontogenic cyst. It is not known how or why these
connective tissue. localised epithelial proliferations are formed. However,
they do seem to be a spontaneous process that occurs in the
A characteristic feature that is seen in all lateral perio- development of these cysts, although similar whorls can be
dontal cysts and botryoid odontogenic cysts is the presence seen in some odontogenic tumours. It is possible that the
of localised plaques or thickenings of the epithelial lining thickenings represent another of the many examples of
(Figure  8.6). Sometimes only one or two plaques may be odontogenic epithelium recapitulating stages of tooth
seen, but most cysts contain multiple plaques. The plaques development under pathological conditions. In this
show a number of different patterns (Figure 8.6). They may instance, the process seems to be similar to that which
be small and flattened, but others are larger and extend takes place during the early stages of tooth development,
into the surrounding cyst wall or produce mural bulges when there is thickening of the oral epithelium to form the
that protrude into the cyst lumen. Most plaques are com- dental lamina.
posed of flattened or cuboidal cells that may form central Examination of histological material at the light micro-
whorls (Figure 8.6a, b), but clear cell change is common. scope level, and particularly study of serial sections, has
The clear cells may be seen in up to one-­third of cases suggested a possible mode of formation of these plaques
(Wysocki et al. 1980; Altini and Shear 1992) and are found (Shear and Pindborg  1975; Figure  8.7). The sequence
in the thin lining as well as in the plaques (Figure 8.6c, d). appears to be proliferation of the flat basal cells, which
They contain abundant glycogen and are therefore positive produces a slight localised thickening of the epithelium
with periodic acid–Schiff (PAS), and are diastase labile. (Figure 8.7b). At this stage, the thickened epithelium con-
Glycogen may also be seen in the superficial layers of the sists predominantly of darkly staining fusiform basal cells.
squamous and cuboidal cells of the lining epithelium. This early plaque may extend into the fibrous wall of the
These will also be PAS positive, and can be digested with cyst or bulge into the lumen (Figure 8.7c, d). The plaque
diastase. It should be noted that mucous cells are not a fea- increases in size both by further basal cell proliferation
ture of lateral periodontal cysts. If mucous cells are sus- and by swelling of these epithelial cells. At this stage there
pected (PAS positive, diastase resistant), these should be is a more pronounced bulging into the lumen and also
confirmed using other stains and an alternative diagnosis into the wall (Figure  8.7e). The bulbous nature of the
should be considered, in particular the possibility of glan- thickening into the wall sometimes leads to undermining
dular odontogenic cyst. of the adjacent cyst lining. Complex convolutions of the
Occasionally the epithelial thickenings are markedly epithelium may be seen in the larger plaques (Figures 8.6e
prominent and may be folded and convoluted (Figure 8.6e). and 8.7f).
The wall of the lateral periodontal cyst is composed of Altini and Shear (1992) described the histology of 20
loose collagenous connective tissue and is usually unin- cases of ‘lateral periodontal cyst’. A review of their paper
flamed, although occasional inflammatory cells may be now suggests that 3 of the lesions they described were actu-
seen. The wall often contains small islands of odontogenic ally glandular odontogenic cysts. Of the remaining 17,
epithelium resembling rests of dental lamina (Figure 8.4), however, they found that 9 were unicystic and 8 were mul-
and it was this finding that was originally cited as strong ticystic. Six of the multicystic group comprised two or more
evidence for an origin from dental lamina rests (Wysocki cystic spaces contained within a single round or ovoid
148 Lateral Periodontal Cyst and Botryoid Odontogenic Cyst

(a) (b)

(c) (d)

(e) (f)

Figure 8.6  Epithelial thickenings and plaques are seen in lateral periodontal and botryoid odontogenic cysts. These may be flattened
(a) or may produce prominent bulges into the lumen or the wall (c, d). Whorling (a, b) and clear cell change are common (c, d). In
botryoid odontogenic cysts, the plaques may be particularly prominent and may become folded (e) or may form papillary processes (f).
­Lateral Periodontal Cys  149

Box 8.2  Lateral Periodontal Cyst: Histopathology and Diagnostic Criteria


●● A simple cyst lined by non-­keratinised epithelium two to five cells thick
●● Localised plaques and thickenings are seen in virtually all cases
●● Clear cell change with abundant glycogen (periodic acid–Schiff/diastase positive)
●● Epithelial lining is friable and often separates from the underlying connective tissue
●● The cyst wall is loosely collagenous and is uninflamed. Areas of subepithelial hyalinisation may be seen
●● The wall contains variable numbers of epithelial islands resembling rests of dental lamina. These may show clear
cell and microcystic change.
Caution: if any of the following features are noted, consider an alternative diagnosis:
●● If the lesion is multicystic, consider a botryoid or glandular odontogenic cyst
●● If luminal columnar cells, duct-­like structures, or mucous cells are present, consider a glandular odontogenic cyst
●● If there is evidence of keratinisation, consider an odontogenic keratocyst

Figure 8.7  Diagram illustrating the possible Ep


mode of formation of epithelial plaques by
localised proliferation of cells. (a) Cyst lined by
thin epithelium. (b) Early epithelial thickening Lu
by basal cell proliferation. (c) Basal cells
continue to proliferate. Superficial cells swell by
accumulation of intracellular fluid and glycogen (a) (b)
(c, d) and the plaques protrude into the cyst
cavity and wall (d, e). The epithelial plaques can
become folded and form convolutions (f, see
also Figure 8.6e). Protrusions into the cyst wall
(c–f) may be ‘pinched off’ and develop into
daughter cysts (see text), leading to the
formation of the botryoid odontogenic cyst.
Source: Shear and Pindborg (1975).
(d)
(c)

(e) (f)

fibrous capsule, whereas two were larger, with an irregular type they believed was a botryoid odontogenic cyst, while
lobulated fibrous wall. The latter had a distinctly botryoid the first and second types were variants of lateral periodon-
appearance, consisting of several cystic spaces of varying tal cysts.
size, separated by fibrous tissue. From these observations, This proposal therefore suggested that the lateral peri-
they suggested that there were three variants of the lateral odontal cyst may be unicystic or multicystic. However,
periodontal cyst (see Figure 8.3): a unicystic variant com- we do not now think this is the case. The pathology of
posed of a single simple cyst, a multicystic variant com- the botryoid odontogenic cyst is discussed in more detail
posed of multiple simple cysts within a single fibrous in the next section, but originally the diagnosis depended
capsule, and a multicystic variant composed of multiple on the gross specimen showing a multilocular (botryoid)
irregular cysts, each separated by fibrous tissue. This third morphology, as well as being multicystic on histology
150 Lateral Periodontal Cyst and Botryoid Odontogenic Cyst

(Weathers and Waldron  1973). A botryoid morphology multilocularity is characteristic, it has been shown that
may be seen during surgery, or at macroscopic examina- some unilocular lesions are multicystic and, conversely,
tion if the cyst has been removed intact. In practice this that some radiologically and grossly multilocular lesions
rarely occurs and the diagnosing pathologist can rarely are unicystic on histological examination (Greer and
verify a macroscopic botryoid pattern. Furthermore, it is Johnson  1988; Siponen et  al.  2011). In their systematic
reported that less than half of botryoid odontogenic review, Chrcanovic and Gomez (2019b) found that only
cysts are multilocular on radiographs (Chrcanovic and 36.5% of lesions diagnosed as botryoid odontogenic cyst
Gomez 2019b). were found to be multilocular. Of the rest, 42.7% were uni-
Therefore, we believe that the diagnostic criteria (see locular and in 15.6% the locularity was unknown. This sug-
Box  8.2) for lateral periodontal cyst should include the gests that many pathologists will diagnose a lesion as
finding of a single cyst as described above within a thin botryoid based on a multicystic histology regardless of
fibrous capsule, although the wall may contain islands of locularity.
epithelium. A lesion that is multicystic, whether within a We would support the view that the term botryoid odon-
single capsule (unilocular) or separated by fibrous tissue togenic cyst should be used for a cyst in a lateral periodontal
(multilocular), should be diagnosed as a botryoid odonto- position that is multicystic on gross or microscopic exami-
genic cyst (Figure 8.3; see next section; Siponen et al. 2011). nation (Siponen et  al.  2011; Vargas and White  2022).
Although a variant of lateral periodontal cyst, it is impor-
tant that botryoid odontogenic cyst is used as a diagnostic
Treatment term because the cyst has a greater potential for growth
The lateral periodontal cyst is treated by surgical enuclea- and recurrence.
tion with or without curettage of the underlying bone, but
care must be taken not to damage the associated tooth. Clinical Features
Occasional recurrences have been reported, but this is
unusual. In their systematic review, Chrcanovic and Frequency
Gomez (2019b) found only two reports of a cyst recur- Botryoid odontogenic cyst is extremely rare. In most series
ring (2.4%). of odontogenic cysts, it has been included as a variant of
lateral periodontal cyst and therefore the exact frequency is
uncertain. In the series reported in Table 1.3 (Chapter 1),
only two included data for botryoid odontogenic cyst and
­Botryoid Odontogenic Cyst these were 0.07% (Grossmann et al. 2007) and 0.03% (Soluk
Tekkeşin et al. 2012b). In the larger series of lateral perio-
The botryoid odontogenic cyst is regarded as being a dontal cysts shown in Table  8.1, most included botryoid
multicystic variant of lateral periodontal cyst (Vargas odontogenic cysts, but the numbers reported were only 3 of
and White 2022). This is because they present at a similar 39 (Wysocki et al. 1980), 6 of 27 (Cohen et al. 1984), 6 of 32
site and the epithelial linings show the same histological (Rasmusson et al. 1991), and 2 of 25 (Carter et al. 1996).
features. However, the botryoid odontogenic cyst has This represents only about 10–20% of lateral periodontal
distinctive radiological, morphological, and behavioural cysts and suggests an overall frequency of less than 0.1% of
characteristics that justify it being regarded as a sepa- all odontogenic cysts.
rate entity. In their systematic review, Chrcanovic and Gomez (2019b)
It was first described by Weathers and Waldron (1973), found only 96 reported cases of botryoid odontogenic cyst
who reported two examples of a cystic lesion of the jaws and Cunha et  al. (2019) found 98. However, both these
that had a multilocular radiological appearance and, on reviews included a number of cases that have subsequently
examination of the gross specimen, had the appearance been shown to be glandular odontogenic cysts.
of a small bunch of grapes. They therefore suggested the
term botryoid, meaning ‘like a bunch of grapes’, and Age
derived from the ancient Greek word for grape: botrus The literature review published by Ramer and Valauri (2005)
(βοτρυς). identified 66 cases of botryoid odontogenic cyst, including
Subsequently there has been confusion over the precise the large series of 33 cases of Gurol et al. (1995). The age
definition, with some demanding that the lesion must be distribution of these 66 patients was very similar to lateral
grossly ‘grape-­like’ or multiloculated, while others have periodontal cyst and is shown in Figure  8.8. The lesion
used the term for any multicystic lesion with an epithelial occurs in adults and has a peak incidence in the sixth and
lining similar to the lateral periodontal cyst. Although seventh decades. In the two largest reviews, Chrcanovic
­Botryoid Odontogenic Cys  151

Figure 8.8  Age distribution of 66 patients with 20


botryoid odontogenic cysts. Source: Data from 18 18
Ramer and Valauri (2005). 18

16

14

12

No. of cases
10
10

8 7
6
6 5

4
2
2
0 0 0
0
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89 90-99
Age

and  Gomez (2019b) and Cunha et  al. (2019) both found a Ramer and Valauri (2005) included the data of Gurol
mean age of 54.0 years (ranges 15–85 and 20–85, respec- et al. and identified 59 cases with information on clinical
tively) and a peak in the sixth and seventh decades. presentation. Of these cases, 30 (50.1%) had evidence of
swelling, but only 7 (11.9%) complained of other symp-
Site toms, including 3 with pain, 2 with paraesthesia and 2 with
About 85% of botryoid odontogenic cysts are found in the a discharge. In a more recent systematic review, Chrcanovic
mandible (Ramer and Valauri  2005; Chrcanovic and and Gomez (2019b) reviewed 96 cases of botryoid odonto-
Gomez 2019b; Cunha et al. 2019). Of the 10 lesions docu- genic cyst and in those where data were available, 56.9%
mented by Greer and Johnson (1988) and by Santos et al. showed swelling or bone expansion, but only 15 patients
(2011b), 8 involved the mandible, predominantly the ante- (15.6%) recorded other symptoms.
rior region. The distribution appears to be very similar to
the lateral periodontal cyst, with most lesions arising in the
Radiological Features
mandibular canine/premolar area (Gurol et  al.  1995;
Méndez et al. 2007). Occasional cases have been reported The botryoid odontogenic cyst shows radiological features
in the midline (Kaugars 1986). The lesion very rarely occurs similar to lateral periodontal cyst. It is located at the lateral
posterior to the premolar teeth. In the largest series of 33 aspect of a tooth root or between the roots of two teeth. The
cases, only 1 was found to be associated with a molar tooth lesion is usually well demarcated with a corticated outline
(Gurol et al. 1995). Santos et al. (2011b) reported 10 cases, and the periodontal space and lamina dura are typically
2 of which were in the mandibular molar region and 1 in intact. Botryoid odontogenic cysts may show two charac-
the maxillary molar region. Chrcanovic and Gomez (2019b) teristic features not seen in lateral periodontal cysts: they
reported the precise location of 92  lesions and recorded may be multilocular and larger.
only 6 (6.5%) in the molar regions, but also 6 that were Although multilocularity is thought of as typical, only
extensive and involved an entire mandibular quadrant, 46.1% were found to be multilocular (Chrcanovic and
including one that extended from the mandibular premo- Gomez  2019b). All 3  lesions reported by Kaugars (1986)
lars to the ramus, and enveloped an unerupted third molar showed multilocular radiolucencies, but in the series of
(Vidaković et al. 2016). Greer and Johnson (1988), 8 of 10  were unilocular, and
Gurol et al. (1995) found that 15 of 16 cysts with informa-
Clinical Presentation tion available were unilocular. In their review, Méndez
The literature suggests that about 55% of patients present et al. (2007) found 45 cases with information on the radio-
with a swelling or expansion of the bone (Gurol et al. 1995; graphic appearance and reported that only 40% were mul-
Ramer and Valauri 2005; Chrcanovic and Gomez 2019b), tilocular. Cunha et al. (2019) identified 78 cases where data
but other symptoms are rare. were available and reported that 52.6% were multilocular.
152 Lateral Periodontal Cyst and Botryoid Odontogenic Cyst

Whereas a lateral periodontal cyst rarely exceeds 10 mm proportion, because those that were not followed up may
in diameter, the botryoid cyst may grow to larger sizes. In simply not have returned because they had no further
Greer and Johnson’s (1988) series the size ranged from 4 to lesions. Also, Siponen et  al. (2011) have pointed out that
45 mm, and Altini and Shear (1992) reported that one of some of the earlier reports (for example Phelan et al. 1988;
their botryoid cases measured 50 mm in diameter. Santos Heikinheimo et al. 1989; Öçok et al. 2005) have included
et al. (2011b) reported 10 cases, all of which were greater glandular odontogenic cysts, which may have a higher
then 10 mm in size, with a range of 15–80 mm. In their sys- recurrence rate.
tematic review, Chrcanovic and Gomez (2019b) found 49 Nevertheless, the data show that the botryoid odonto-
cases where the size had been measured and recorded a genic cyst may have a recurrence rate of up to 30% and that
mean diameter of 28 mm (SD ±28 mm. range 3–150 mm). multilocular or larger lesions may have an increased risk of
It has been reported that multilocular lesions may be recurrence. Patients treated for a botryoid odontogenic cyst
larger than unilocular lesions, although the range is quite should be followed periodically.
large. In their review, Cunha et  al. (2019) identified
34  lesions where size was reported and showed that the
mean size for unilocular lesions was 15.2 mm (range Pathogenesis
4–45 mm), while multilocular lesions showed a mean of
The pathogenesis of the botryoid odontogenic cyst has
27.9 mm (range 2–75 mm).
been discussed along with the lateral periodontal cyst. A
unifying mechanism is suggested, in which the lateral peri-
Recurrence of the Botryoid Odontogenic Cyst odontal and botryoid odontogenic cysts arise from rest cells
of dental lamina (Figure 8.3). It is proposed that the botry-
A key feature of the botryoid odontogenic cyst is that it has
oid cyst arises due to simultaneous cystic degeneration of
a tendency for recurrence. Almost certainly this is due to
multiple dental lamina rests to produce a multicystic lesion
its multilocular or multicystic nature, making it more
(Wysocki et  al.,  1980). Altini and Shear (1992) proposed
likely that remnants of cyst lining may be left behind after
that a multicystic lesion is formed by ‘pinching-­off’ of the
enucleation. Overall, it is reported that 21.7% recur, com-
epithelial plaques to form mural islands, which then
pared to only 2.4% of lateral periodontal cysts (Chrcanovic
undergo cystic change. As this process is repeated, a multi-
and Gomez 2019b).
cystic lesion develops. A further mechanism was proposed
Of the 10 cases documented by Greer and Johnson
by Redman et  al. (1990) and Siponen et  al. (2011), who
(1988), 3 recurred between 8 and 10 years after surgery.
described multiple lateral periodontal cysts, some of which
Follow-­up information on recurrences was available for 11
were closely apposed and multilocular. They suggested
cases of the 33 reported by Gurol et al. (1995), of which 2
that some botryoid odontogenic cysts may arise by fusion
recurred. Further documentation of the tendency for the
of multiple lateral periodontal cysts. These mechanisms
botryoid odontogenic cyst to recur has been provided by
are not mutually exclusive and some or all may apply in
Ramer and Valauri (2005).
individual cases. Figure 8.3 illustrates a unifying hypothe-
Méndez et al. (2007) have reviewed these reports and oth-
sis for formation of unicystic and multicystic lesions, and
ers and have identified factors associated with recurrence.
suggests correct terminology.
They found 37 cases where follow-­up had been recorded
and of these, 12 (32.4%) recurred. They showed that 81.8%
of recurrent cysts were multilocular compared with only
Histopathology
23.8% of non-­recurrent cysts. Recurrent cysts were also
larger, with a mean size of 31 mm compared to 9.6 mm in The botryoid odontogenic cyst was so named because, on
non-­recurrent cases. Recurrent cases also occurred in a macroscopic examination, it resembled a small bunch of
younger age group: 47.8 years compared to 54.9 years. grapes (Weathers and Waldron 1973), and some regarded
Cunha et al. (2019) identified 12 recurrent cases, 11 (92%) of this as an essential appearance to establish the diagnosis.
which were multilocular and, as noted above, found that However, this is rarely seen, because most lesions collapse
multilocular lesions were on average larger. Similar find- or fragment at surgery and the diagnosing pathologist
ings have been reported by others (Vidaković et al. 2016). receives a folded lesion or multiple fragments. Furthermore,
A note of caution is needed. A review of many of these some lesions are multicystic but not multilocular, since
reports shows that very few botryoid odontogenic cysts the cysts are contained within a single fibrous capsule
have actually been followed up and the number of those (Figure  8.3). A correct diagnosis can often only be made
that recurred is recorded as a proportion of those where after a full consideration of the clinical, radiological, and
follow-­up data are available. This may overestimate the histological features (Box 8.3).
­Botryoid Odontogenic Cys  153

Box 8.3  Botryoid Odontogenic Cyst: Key Facts and Diagnostic Criteria
●● They are rare – less than 0.1% of odontogenic cysts
●● Only seen in adults, mean age about 50 years. Peak in sixth decade
●● Always arise anterior to the second premolars
●● 70% are found in the mandible. Most common in the canine/premolar area
●● Well-­defined radiolucency with corticated margin
●● About 50% are multilocular
●● Often greater than 10 mm diameter
●● Histology shows multiple cysts enclosed in a single fibrous capsule or may be multilocular
●● Each cyst is lined by non-­keratinised epithelium with plaques or thickenings similar to lateral periodontal cyst
●● Between 20% and 30% may recur
Caution: if any of the following features are noted, consider an alternative diagnosis:
●● If the radiolucency is large, or crosses the midline, or is posterior to premolars, possibilities include glandular odontogenic
cyst or odontogenic keratocyst
●● If luminal columnar cells, duct-­like structures, or mucous cells are present, consider a glandular odontogenic cyst

Figure 8.9  A botryoid odontogenic cyst. The


lesion is multicystic, with thin fibrous walls lined
by epithelium two to five cell layers thick. Many
epithelial thickenings and plaques can be seen.
Inset: a small island shows clear cell and
microcystic change. Source: Martin and Speight
2017. Reproduced with permission from Elsevier.

The defining feature of the botryoid odontogenic cyst is The cyst cavities are of varying size, but each one is lined
that it is multicystic (Figure 8.9), with most reports show- by epithelium identical to that described previously for the
ing between two and about six cystic cavities, although lateral periodontal cyst (Figures 8.6 and 8.10). The lining is
additional small microcysts and epithelial islands may be a thin non-­keratinised epithelium comprising, for the most
seen. Occasionally the cysts are rounded and ballooning part, one to three layers of flattened cells but with plaque-­
(Weathers and Waldron 1973; Rasmusson et al. 1991), con- like thickenings, showing whorling and clear cell change
sistent with the idea that they expand by hydrostatic pres- (Figure  8.6). The plaques and thickenings are seen in all
sure, but most often they are collapsed with folded irregular cases, and may be more prominent in botryoid lesions than
outlines, producing a multilocular appearance. in unicystic lateral periodontal cysts, and occasionally
154 Lateral Periodontal Cyst and Botryoid Odontogenic Cyst

Differential Diagnosis
The diagnostic features of botryoid odontogenic cyst are
quite clear and misdiagnoses can be avoided if care is taken
to correlate the clinical, radiographic, and histological find-
ings (Box 8.3). However, one lesion in particular – glandular
odontogenic cyst – may share some features with botryoid
odontogenic cyst and care should be taken to avoid this
pitfall. Both may arise at the same site (anterior mandible),
both may be radiologically multilocular, and both are
multicystic. On radiological examination, botryoid odonto-
genic cysts are located between two teeth, rarely reach a
large size, and are rare in the midline. In contrast, glandu-
Figure 8.10  Botryoid odontogenic cyst with numerous small lar odontogenic cysts are often apical to the teeth or
cysts showing a thin lining with epithelial plaques. Epithelial embrace multiple teeth, may reach a very large size, and, in
islands with clear cell and microcystic change can also be seen
the mandible, have frequently been shown to cross the
(bottom left).
midline (see Figures 10.3 and 10.4). Histologically, glandu-
lar odontogenic cysts are often multicystic and do show
tangential sectioning may result in the appearance of pap- epithelial plaques and thickenings, but the lining is thicker
illary projections (Figure 8.6f). The fibrous cyst wall is not and is characterised by luminal cuboidal or columnar
usually inflamed and in 70% of cases may show hyalinisa- cells, intraepithelial microcysts or duct-­like structures,
tion immediately below the epithelium (Gurol et al. 1995; and mucous cells (Fowler et al. 2011; Chapter 10). These
Santos et  al.  2011b; see Figure  8.5). Epithelial islands, features are not seen in lateral periodontal or botryoid
resembling rests of dental lamina, are seen in about 50% of odontogenic cysts. Note that mucous cells are only seen in
cases and often show clear cell change or cystic degenera- about 70% of glandular odontogenic cysts, so an absence of
tion (Figure 8.9, inset). mucous does not exclude the diagnosis (Kaplan et al. 2008;
Altini and Shear (1992) described three morphological Fowler et  al.  2011). A number of papers have reported
variants of the lateral periodontal cyst (see Figure  8.3). unusual lateral periodontal or botryoid odontogenic cysts,
They suggested that the lateral periodontal cyst may be which on review are in fact glandular odontogenic cysts
unicystic or multicystic, but both types are contained (Phelan et  al.  1988; Heikinheimo et  al.  1989; Buckley
within a single fibrous capsule. The third variant was et al. 1989; Öçok et al. 2005).
composed of an irregular or lobulated thin-­walled multi-
cystic structure, which they felt was consistent with a
Treatment
botryoid odontogenic cyst. As discussed previously, it is
often difficult to establish multilocularity on gross or his- Botryoid odontogenic cysts are usually treated conserva-
tological examination, and many multicystic lesions are tively by enucleation, but because they are multicystic and
unilocular on radiographic examination. We believe many are multilocular, care is needed to ensure that all
therefore that botryoid odontogenic cysts can be diag- tissue is removed. Curettage to underlying clean bone is
nosed when a lesion is found to be multicystic and that recommended. As discussed above, the cyst has a propen-
multilocularity cannot be used as a defining criterion for sity for recurrence so cases should be carefully followed
diagnosis. Lateral periodontal cysts are unilocular and for a number of years, and complete healing should be con-
unicystic. firmed radiologically.
155

Gingival Cysts

CHAPTER MENU
­Gingival Cyst of Adults, 155
•• Clinical Features,  156
–– Frequency, 156
–– Age, 156
–– Sex, 156
–– Site, 156
–– Clinical Presentation,  157
•• Radiological Features,  158
•• Pathogenesis, 158
•• Histopathology, 158
–– Differential Diagnosis,  159
–– Microkeratocysts, 159
•• Treatment, 160
­Mucosal Cysts in Infants and Neonates, 160
•• G
­ ingival Cyst of Infants,  160
–– Clinical Features,  161
◦ Frequency and Prevalence, 161
◦ Age and Sex,  161
◦ Site, 161
◦ Clinical Presentation,  161
–– Pathogenesis, 161
–– Histopathology, 162
–– Treatment, 162
•• ­Epstein Pearls – Midpalatal Raphe Cyst,  162
–– Clinical Features,  162
◦ Frequency and Prevalence,  163
◦ Age and Sex,  163
◦ Clinical Presentation,  163
–– Pathogenesis,  163
–– Histopathology,  163
–– Treatment,  163

Gingival cysts are discussed together as a group of lesions ­Gingival Cyst of Adults
that are extraosseous and arise within the soft tissues. The
lesions to be discussed in this chapter are the gingival cyst The relationship between the gingival cyst of adults and the
of adults and gingival cyst of infants, which arise in the lateral periodontal cyst has been discussed in the previous
tooth-­bearing areas and are of odontogenic origin, and chapter (Chapter 8). It is believed that they have a similar
the midpalatal raphe cyst, which arises in the palate of pathogenesis and represent intraosseous and extraosseous
infants. variants of the same lesion (Wysocki et al. 1980). Although

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
156 Gingival Cysts

this may be the case, there are distinct differences in clini- (1968), 26 by Buchner and Hansen (1979), and 16 from
cal presentation and management that deserve separate Jones et  al. (2006). This shows only 3 cases occurring
consideration (Shear and Pindborg  1975; Altini and before the age of 30 and a peak frequency in the sixth dec-
Shear  1992; Siponen et  al.  2011; de Andrade et  al.  2012; ade, with the majority of patients (71.4%) in the fifth and
Chrcanovic and Gomez 2019a). sixth decades.
Wagner et  al. (2015) reviewed 47 reports and found an
overall mean age of 49.10 years, and a more recent system-
Clinical Features
atic review (Chrcanovic and Gomez  2019a) recorded 157
Up to 2019 there have been about 50 papers reporting 157 gingival cysts of adults and found a mean age of 48.6 years
cases of gingival cyst (Wagner et al. 2015; Chrcanovic and (range 7–79), with a peak in the sixth decade.
Gomez 2019a), but most of these have been reports of one Ironically, despite the name, a small number of gingival
or two cases. There have been few case series, but those of cysts of adults are found in children (Buchner and
reasonable size are summarised in Table  9.1. Of the 33 Hansen 1979; Giunta 2002; Richman and Johnston 2020).
cases reported by Buchner and Hansen (1979), 7  were The criteria for diagnosis used by these authors was of a
found on histological examination to be keratinising cysts cyst on the gingivae associated with erupted teeth, in con-
and are best excluded if a critical assessment of the gingival trast to gingival cysts of infants that arise on the edentulous
cyst of adults is to be performed. alveolus. Richman and Johnston (2020) reported details of
a case in a 5-­year-­old boy. It presented as a cystic swelling
Frequency on the gingiva between a mandibular deciduous second
Gingival cysts are rare and comprise less than 0.5% of all incisor and the canine. The lesion was lined by a thin non-­
odontogenic cysts (Tables 1.1–1.3). In the studies summa- keratinised stratified squamous epithelium.
rised in Table 1.3, 8 included data on gingival cyst of adults
and showed a frequency of between 0.03% (Grossmann Sex
et al. 2007) and 0.48% (Daley et al. 1994). In their study of Data accumulated from all published cases (n = 157) shows
7121 odontogenic cysts over a period of 30 years, Jones that females are slightly more often affected than males,
et  al. (2006) found 16 (0.2%) gingival cysts of adults. with a male : female ratio of 1 : 1.44 (41% male : 59% female)
Viveiros et al. (2019) found 20 gingival cysts in a series of (Wagner et  al.  2015; Chrcanovic and Gomez  2019a).
7023 odontogenic cysts (0.3%). However, the ratio varies considerably between series
(Table 9.1), with the proportion of males ranging from as
Age low as 12.5% to a high of 55.6%.
The mean age of presentation of gingival cyst of adults is
about 50 years, with a peak in the fifth or sixth decade Site
(Table 9.1). The age distribution of 56 cases is illustrated in Gingival cysts of adults occur much more frequently in the
Figure 9.1. These data include 10 from Shear (University mandible than in the maxilla (Table  9.1), with the large
of the Witwatersrand), 4 reported by Reeve and Levy reviews showing 76% in the mandible with almost all cases

Table 9.1  Age, sex, and site distribution from selected case series of gingival cyst of adults, and from the systematic review of
Chrcanovic and Gomez (2019a).

References n Mean age Peak decade Male (%) Mandible (%)

Buchner and Hansen (1979) 26 48.2 5th and 6th 42.0 73.0
Wysocki et al. (1980) 10 50.7 5th 55.6 70.0
Nxumalo and Shear (1992) 14 50.0 6th 50.0 57.1
Bell et al. (1997) 8 51.0 NR 12.5 87.5
Giunta (2002) 22 52.0 5th 28.5 88.8
Jones et al. (2006) 16 52.9 6th 25.0 NR
Viveiros et al. (2019) 20 53.5 6th 35.0 60.0
Chrcanovic and Gomez (2019a) 157 48.6 6th 41.6 76.5

NR, not reported.


­Gingival Cyst of Adults 157

Figure 9.1  Age distribution of 56 30


patients with gingival cyst of the adult.

25 24

20

No. of cases
16
15

10
7
5
5

1 2
1
0 0
0
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89
Age

arising anterior to the molar teeth. Overall, about 70% are course of histological examination of large numbers of gin-
found in the anterior mandible or canine/premolar region, gival biopsies (Moskow  1966). Ritchey and Orban (1953)
although the single most common site is the mandibular discovered 6 such cysts in 350 gingival biopsies.
canine/premolar area with 35% of cases (Wagner et al. 2015; In their systematic review, Chrcanovic and Gomez
Chrcanovic and Gomez  2019a). Chrcanovic and Gomez (2019a) found 56 cases where symptoms had been
(2019a) found reports of the precise location in 112 cases recorded, but only 8 (14.3%) reported symptoms. In their
and reported that only 4 (3.5%) were found posterior to the series of 20 cases, Viveiros et  al. (2019) found 12  with
second premolars. evidence of a swelling or ‘vesicle’, but only 2 cases pre-
sented with symptoms. In 12 cases where size had been
Clinical Presentation recorded, 3 were over 10 mm in diameter, but the remain-
The gingival cyst of adults is usually symptomless, although ing 9  were less than 5 mm. Giunta (2002) recorded an
the patient may give a history of a slowly enlarging, pain- average dimension of 6 mm and his largest lesion was
less swelling (Figure 9.2). Many are only found fortuitously 12 mm in diameter. Chrcanovic and Gomez (2019a)
during an oral examination or have been detected in the found 93 cases where size had been recorded and found a
mean diameter of 6 mm (standard deviation [SD] ±3 mm;
range 2–20 mm).
The cysts are round to oval, well-­circumscribed, usually
less than 10 mm in diameter, and may occur in the
attached gingiva or the interdental papilla, on the buccal
or labial aspect (Figure 9.2). The surface is smooth and is
usually the colour of normal gingiva, but occasional
lesions are translucent or blueish (Viveiros et al. 2019). If
they are traumatised, they may have an ulcerated surface
or may be haemorrhagic. The lesions are soft and fluctu-
ant and the adjacent teeth are vital. During surgical explo-
ration, slight erosion or saucerisation of the surface of the
bone may be observed, without extension into the perio-
dontium. Very rarely, more than one gingival cyst may be
Figure 9.2  Gingival cyst of an adult. The lesion is a sessile found in a patient (Shade et al. 1987; Giunta 2002; Wagner
swelling of normal colour, about 10 mm in diameter (arrows). et al. 2015).
158 Gingival Cysts

close to the junctional epithelium. They suggested a mech-


anism similar to that proposed for lateral periodontal cyst
(discussed in Chapter  8), whereby both cysts are derived
from reduced enamel epithelium (Nxumalo and
Shear 1992). This proposes that an early dentigerous cyst
forms on an erupting tooth and becomes displaced later-
ally, as the tooth emerges into the mouth, to form a lateral
cyst. A gingival cyst may form from junctional epithelium
that is derived from ‘postfunctional’ reduced enamel epi-
thelium. It is possible that a developmental cyst may derive
from a down-­growth of junctional epithelium, but this
mechanism has not gained favour for gingival cyst of
adults.
Figure 9.3  Radiograph of a gingival cyst in an adult. There is a The rest cells of the dental lamina are now considered to
faint radiographic shadow (marked with arrows), indicative of be the source of gingival cyst of adults and provide a logical
superficial bone erosion.
explanation for their location in the gingival tissues and
the histological similarity to lateral periodontal cyst. It  is
now accepted that the gingival cyst of adults, lateral perio-
Radiological Features
dontal cyst, and botryoid odontogenic cyst are derived from
Gingival cyst of adults is a soft tissue lesion, so radiology the same source of epithelium, and this was discussed in
may not be helpful in diagnosis. However, a cyst originat- detail in Chapter 8.
ing in the gingival soft tissues may enlarge sufficiently to There is no clear explanation as to what the stimulus
cause superficial erosion of the cortical plate, with radio- for the proliferation of these rests and their subsequent
logical evidence of a faint round shadow (Figure  9.3). cystic breakdown might be. It is certainly not an inflam-
Chrcanovic and Gomez (2019a) found 75 cases where radi- matory stimulus, which produces well-­recognised effects
ological features had been recorded, and 38 (50.7%) showed on odontogenic epithelium in radicular cysts, but is prob-
evidence of bone erosion. In their review of 157 cases, ably similar to that described previously for lateral peri-
Wagner et  al. (2015) found that 35 (22.3%) showed evi- odontal cyst (Chapter 8). This suggests that cyst formation
dence of bone erosion, and described a case of their own and growth are stimulated by accumulation of cell debris
where surgery revealed exposure of a tooth root. However, and increased luminal osmotic pressure that drives cyst
only 22 cases (14%) recorded radiographic evidence of bone expansion. A detailed discussion of the role of osmotic
resorption. pressure in cyst growth can be found in Chapter 3.

Pathogenesis Histopathology
Although a number of suggestions have been made about The pathologist will often receive an excisional biopsy (a
the pathogenesis of the gingival cyst of adults, the most ‘gingivectomy’ specimen) composed of mucosa with the
favoured theory of origin is from odontogenic epithelial cyst on the deep aspect. Gingival cysts are small and fragile
cell rests derived from the dental lamina (Wysocki et  al. and may be spherical, but are usually collapsed and folded,
1980). Early studies have shown that dental lamina rests and may have a thin translucent wall.
are commonly found in the alveolar bone and gingival tis- Gingival cysts are lined by thin non-­keratinised epithe-
sues and often show evidence of microcystic change (see lium, comprising one to five layers of flat to cuboidal cells
Figure  9.7; Hodson  1962; Moskow and Bloom  1983). containing darkly staining nuclei (Figures  9.4-9.6).
Wysocki et  al. (1980) suggested that lateral periodontal Localised areas of plaques or thickened epithelium are
cysts and gingival cysts had the same origin from rest cells often seen (Figure 9.4, arrows). Occasionally these may be
of dental lamina, but from intraosseous and extraosseous whorled or nodular, similar to those seen in a lateral peri-
rests, respectively. odontal cyst (see Figure 8.6), but are usually not as promi-
Others have suggested an origin from junctional epithe- nent and may be smaller and flattened (Figure  9.6).
lium. In material studied by Nxumalo and Shear (1992), Nevertheless, the similarities support the commonly held
some cases were observed in which the epithelial lining view that the two cysts originate from the same epithelium.
resembled reduced enamel epithelium and sometimes The attachment of the epithelium to the underlying con-
extended to the deep aspect of the specimen, where it lay nective tissue is tenuous and it easily peels off, leaving
­Gingival Cyst of Adults 159

epithelial discontinuities (Figure 9.4, asterisks; Figure 9.6).


The fibrous connective tissue wall is usually uninflamed,
except close to the junctional epithelium where a chronic
inflammatory cell infiltrate may occur, or if the lesion has
been traumatised. The wall may contain small epithelial
islands (Figure  9.6). The cyst is almost always unicystic,
but rare multicystic lesions have been reported (Wysocki
et al. 1980; Nxumalo and Shear 1992).

Differential Diagnosis
In most cases the diagnosis should be straightforward
based on the characteristic clinical and histological fea-
tures and radiological evidence that the lesion is extraosse-
ous (Box  9.1). The main lesions to exclude are lateral
periodontal cyst or botryoid odontogenic cyst that may
have perforated the cortical plate. These may be suspected
if the plaques or thickenings are especially prominent or if
the lesion is multicystic. However, these lesions have an
intraosseous origin and the radiographs should be exam-
ined to confirm the site of the lesion. Peripheral odonto-
genic keratocyst may have an identical clinical presentation, Figure 9.4  Gingival cyst of the adult. The cyst lies just below
but this is rare and is defined histologically as having a par- the gingival epithelium (left) and is lined by thin, regular
akeratinised lining (see Chapter 7). non-­keratinised epithelium. Epithelial thickenings can be seen
(arrows), as well as areas of fragility where the epithelium is
separated from the cyst wall (*). Source: Martin and Speight
Microkeratocysts (2015), reproduced with permission of Elsevier.
Buchner and Hansen (1979) described gingival cysts of adults
with an ortho-­ or parakeratinised lining, and suggested that
these should be diagnosed as epidermoid cysts or keratocysts,
respectively. If keratin is seen, then a diagnosis of odonto-
genic keratocyst must be considered, and radiographs should
be examined to ensure that the lesion is not a peripheral
extension of a central cyst. Extraosseous or peripheral odon-
togenic keratocysts have been reported and many of these

Box 9.1  Gingival Cyst of Adults: Key Features


●● They are rare – less than 0.5% of odontogenic cysts
●● Seen in adults, mean age about 50 years. Peak in
sixth decade
●● Slightly more common in females Figure 9.5  The epithelial lining of a gingival cyst of the adult
●● Always arise anterior to the second premolars is thin and regular, two to five cells thick.
●● 75% are found in the mandible. Most common in the
canine/premolar area
●● Round, well-­circumscribed swelling on the attached show features similar to gingival cyst, but with a parakerati-
gingiva nised lining. Most oral pathologists will have, on occasion,
●● Usually normal colour and symptomless encountered islands of odontogenic epithelium in mucosal
●● Very rarely more than 10 mm diameter biopsies, especially from the retromolar region. Sometimes
●● 50% may show erosion of surface bone with slight these show cystic degeneration to form small microcysts
radiolucency (Figure  9.7), which are similar to the microkeratocysts
●● Lined by thin regular epithelium with areas of small described by Moreillon and Schroeder (1982). These arose
plaques or thickenings from dental lamina rests in the alveolus of fetuses and neo-
nates, and are the origin of gingival cysts of infants (see next
160 Gingival Cysts

cysts that affect the oral cavity in newborn infants. The


first descriptions of these lesions were by Bohn in 1866 and
Epstein in 1880, but a contemporary classification was first
proposed by Fromm (1967), who undertook an extensive
study of oral lesions in the mouths of newborn infants. He
pointed out that Bohn originally described mucosal cysts
derived from mucous glands, which occurred on the palate
or on the buccal or lingual aspects of the alveolar ridges.
Based on his own observations, Fromm (1967) proposed
that there were three cyst types (Table  9.2). Despite this,
some or all of these terms are used synonymously to
describe any cyst with the characteristic features in a new-
born infant. In particular, the term Bohn’s nodule is fre-
Figure 9.6  Gingival cyst of the adult, showing a thin epithelial quently used as a clinical term to refer to any cystic nodule
lining with a central small, flattened plaque. Small islands of on the alveolar ridges, regardless of origin or histology. The
epithelium are seen in the wall, and an area of separation is also lesions are rarely biopsied, so a histological diagnosis is not
seen (left).
made and the close proximity of the crest of the alveolus to
the lateral aspects means that a precise anatomical position
is infrequently recorded.
Since most lesions are only diagnosed on their clinical
appearance and treatment is rarely needed, we agree with
Van Heerden and Van Zyl (2010) that a pragmatic approach
to classification is appropriate and that two types of cysts
should be identified. In this section we will categorise them
into two types and discuss the gingival cyst of infants and
the midpalatal raphe cyst.

Table 9.2  A summary of the characteristics of the three types


of developmental cysts found in neonates.
Figure 9.7  A mucosal biopsy from the retromolar area of an
adult contains islands of dental lamina rests that have
undergone cystic change to form ‘microkeratocysts’ (see text). Source of
Site epithelium Other names
section). It is possible that similar ­‘dental lamina cysts’ may be
Epstein Midline palate Epithelial Midpalatal
found in the gingivae of adults. These may be extremely rare, pearls inclusions raphe cyst
or may be common and, regarded as inconsequential, and Palatal cyst
therefore rarely reported. Nevertheless, they are probably
Bohn’s Lateral aspect ?Mucous Gingival cyst
oddities rather than entities and care should be taken not to nodules of alveolus minor salivary of infants
overinterpret them as fully formed odontogenic keratocysts, glands
since this may result in overtreatment. Dental Crest of the Rest cells of Gingival cyst
lamina cyst alveolar ridges the dental of infants
lamina
Treatment
Source: Adapted from Fromm (1967).
The gingival cyst is removed by local surgical excision and
in the majority of cases there is no tendency for recurrence.
In reports of 157 cases, only 1  was recorded as recurring
(Chrcanovic and Gomez 2019a). ­Gingival Cyst of Infants

Here, we define the gingival cyst of infants as an odonto-


­Mucosal Cysts in Infants and Neonates genic cyst found in the edentulous alveolar mucosa of new-
born infants (Vargas and Gomez  2022). Other names for
There is much confusion and misunderstanding around this lesion include dental lamina cyst, alveolar cyst, and
the classification and terminology of the developmental Bohn’s nodules, but we regard these as synonymous.
Gingival Cyst of Infants 161

Clinical Features Clinical Presentation


The lesions present as small, white or cream-­coloured nod-
Frequency and Prevalence
ules from 1 to 3 mm in diameter (Figure 9.8). They appear
Gingival cysts of infants are rarely biopsied and therefore
to be symptomless and will disappear of their own accord,
their frequency among odontogenic cysts has not been
usually before 3 months of age. They are usually multiple,
recorded. However, studies of oral lesions in newborns
with up to 12 lesions seen in clusters and at more than one
suggests that they are very common and should probably
site. In Perez-­Aguirre et al.’s (2018) study, the average num-
be regarded as a normal occurrence rather than a patho-
ber of lesions per child was 5.4. When found on the crest of
logical change. In his original paper, Fromm (1967) exam-
the ridge, they resemble natal teeth, but the nodules are
ined 1367  newborn infants and found that 75.9% had
not as hard and may be mobile.
evidence of an oral cyst on the alveolus or palate. A high
frequency was also found in a Taiwanese study, in which
420 neonates were examined and palatal or gingival cysts Pathogenesis
were found in 94% of the infants (Liu and Huang  2004).
There is general agreement that gingival cysts in infants
More recently, in a cross-­sectional study of 2216 newborns
arise from remnants of the dental lamina. The epithelial
in Mexico, Perez-­Aguirre et al. (2018) found that 79% had
cell rests of the dental lamina, the so-­called glands of Serres,
gingival cysts, with a total of 11 862 cysts between them.
have the capacity, from as early a stage in development as
Zen et al. (2019) found an overall prevalence of 60.3%, but
10 weeks in utero, to proliferate, keratinise, and form small
also showed that the cysts were significantly more com-
cysts. Moskow and Bloom (1983) noted that as tooth devel-
mon in full-­term than in preterm (born at less than
opment progressed, but prior to separation of the tooth
36 weeks) infants.
germ from the oral epithelium, the dental lamina may pro-
liferate, with the formation of multiple areas of distinct
Age and Sex
microcysts with keratin production. In the late bell stage of
The cysts arise in newborn infants. There is good evidence
tooth development, according to Moskow and Bloom, disin-
from early studies that they begin to develop in early fetal
tegration of the dental lamina occurs and numerous islands
life and are well developed by 22 weeks of gestation
and strands of odontogenic epithelium are seen in the
(Moreillon and Schroeder  1982). After this, they seem to
mucosa just below the oral epithelium (Figure  9.9).
begin to involute and after birth they rapidly rupture or
Moreillon and Schroeder (1982) showed that dental lam-
merge with the overlying mucosa. Most are therefore seen
ina–derived microcysts increased in number between the
in the first few weeks of life and are rare after 3 months.
12th and 22nd weeks of gestation. They called these ‘micro-
Males and females seem to be affected equally (Perez-­
keratocysts’, with histological features very similar to those
Aguirre et al. 2018; Zen et al. 2019).
shown in Figure 9.7. The cysts were seen to fuse with the
overlying epithelium and discharge their keratin contents
Site
into the oral cavity. Just before and after birth, most of these
More than 85% of gingival cysts are encountered in the
inclusions and microcysts atrophy and become resorbed,
maxilla, and usually towards the buccal or labial aspect,
but some persist to produce clinically detectable cysts.
with a slight majority towards the posterior regions. A
number of studies have distinguished between lesions
classified as Bohn’s nodules (on the buccal, palatal, or lin-
gual aspects of the alveolus) and dental lamina cysts (on
the crest of the alveolus). In the most detailed analysis,
Perez-­Aguirre et al. (2018) identified a total of 11 862 cysts
on the alveoli of 2216  newborn infants. They classified
80.4% (9542) as Bohn’s nodules, of which 3516 (84.7%)
were in the maxilla and over 95% were on the buccal
aspect, but evenly distributed between anterior and poste-
rior regions. In the mandible, 62.4% were located buccally
and 37.6% were lingual. Of the 19.6% (2320) located on
the crest of the alveolar ridge (classified as dental lamina
cysts), 91.5% were located on the maxilla and 85.3% of
these were in the posterior region. Only 198 cysts were Figure 9.8  Gingival cysts in an infant. Multiple lesions on the
buccal aspect of the alveolus of a neonate. One lesion is white
found in the mandible, but most of these (84.3%) were and prominent (arrowhead), while others are pale and relatively
anterior. inconspicuous (arrows). Source: Courtesy of Prof. Ian Brook.
162 Gingival Cysts

Figure 9.9  A mucosal biopsy from an infant shows a Figure 9.10  Gingival cyst in an infant. The cyst lies just below
fragmenting dental lamina with small residual islands scattered the surface epithelium, and is filled with whorls of keratin and
below the surface epithelium. debris. Source: Courtesy of Prof. John Wright.

It has been suggested that lesions on the lateral aspects of may fuse with the overlying epithelium and occasionally a
the alveolus (Bohn’s nodules) are derived from degeneration punctum is seen. Although it is suggested that Bohn’s nod-
of developing glandular tissue, but this does not appear to ules have an origin from glandular tissue, there are no
have been confirmed histologically, and minor salivary glands reports of cysts lined by glandular epithelium.
are rarely found on the attached mucosa of the alveolar ridges.
It seems more likely therefore that the primary source of the
epithelium at all sites on the alveolus is the dental lamina. Treatment
There is no indication for any treatment of gingival cysts
Histopathology of infants. Once their contents are expelled, they atrophy and
disappear. It has been suggested that massage or ­‘teething
Most gingival cysts of infants resolve spontaneously very early rings’ may help them resolve, but this is not usually necessary.
in life and it is unusual for a pathologist to receive a specimen
for diagnosis, which is typically made clinically (Box  9.2).
Most reports of the histological features have come from ­ pstein Pearls – Midpalatal
E
studies of fetal or infant tissues, or cysts encountered by Raphe Cyst
chance. The cysts are lined by a thin, regular parakeratinised
stratified squamous epithelium, similar to those described Epstein pearls is the widely accepted term for the small
above (Figure 9.7), and are often filled with whorls of keratin cysts that arise in the centre of the palate in newborn
and debris (Figure 9.10). Occasionally keratin from the cysts infants. They arise from epithelial inclusions within the
may extrude into the adjacent connective tissues, causing mucosa along the palatal raphe  – the line of fusion of
inflammation and a foreign-­body giant cell reaction. They the  epithelium of the palatal shelves along the midline
of the palate. The midpalatal raphe cyst must not be con-
Box 9.2  Gingival Cyst of Infants: Key Features fused with the so-­called median palatal cyst, which has
been described as an intraosseous cyst arising at the fusion
●● They are common and may affect up to 90% of
of the palatal shelves. The existence of such median cysts is
newborns
questionable and is discussed in Chapter 13. Epstein pearls
●● Only seen in newborns and are rare after 3 months
is the term widely used in paediatric dentistry and is well
of age
understood. Since the diagnosis is almost always made
●● 85% or more are found in the maxilla
clinically, we suggest that this common term should con-
●● Round, well-­circumscribed nodules 1–3 mm in
tinue to be used.
diameter
●● Usually in clusters of 2–12 lesions
●● Usually white or yellowish and may resemble natal Clinical Features
teeth, but are softer and mobile
Epstein pearls are so common that they are usually regarded
●● Lined by thin keratinised epithelium
as a normal occurrence in the mouths of neonates. Although
­Epstein Pearls – Midpalatal Raphe Cyst 163

a number of papers record their prevalence, few studies epithelial inclusions or down-­growths into the superficial
have reported in detail their clinicopathological features. lamina propria. They were often fused with the overlying
epithelium with a punctum or duct-­like morphology, and
Frequency and Prevalence the authors suggested that they may represent abortive
Numerous studies have recorded the prevalence of oral glandular differentiation. However, this seems unlikely,
lesions in neonates and most report that more than 80% of since glands are rarely found at this site. More likely, they
newborns have small nodules, or cysts, in the oral mucosa represent remnants of the surface epithelium of the fused
(Fromm  1967; Liu and Huang  2004; Perez-­Aguirre et  al. palatal shelves.
2018; Zen et al. 2019). Most reports have found that Epstein In a meticulous study on serial sections of 32 human
pearls are the most commonly encountered anomaly. In heads, approximately 8–22 weeks of fetal age, Moreillon
their examination of 2216  newborns, Perez-­Aguirre et  al. and Schroeder (1982) showed epithelial rests of palatal
(2018) found Epstein pearls in 1463 (66.0%). Other studies fusion from 8 to 12 weeks of gestation, with formation of
have shown a prevalence of between 38.3% (Niranjan midpalatal keratinising microcysts between 10 and
et al. 2020) and 70.2% (Moosavi and Hosseini 2006). 22 weeks. The number of cysts in any one individual var-
ied from 1 to 20 and appeared to increase up to 22 weeks
Age and Sex and then decline. The cysts were heavily keratinised and
Lesions are found in newborn infants and are rarely were often fused with the surface epithelium. The authors
encountered after 3 months of age. The largest studies have noted that cysts were found in all fetuses and that the
not shown any sex predilection (Moosavi and Hosseini 2006; number of cysts (15–20 at 22 weeks) contrasted sharply
Monteagudo et  al.  2012; Perez-­Aguirre et  al.  2018; Zen with the number of cysts encountered in newborns or
et al. 2019). adults. They suggested that as the cysts develop, they fuse
with the oral epithelium, discharge their contents, and
Clinical Presentation resolve, so that at birth and shortly after the cysts have dis-
Midpalatal raphe cysts are found along the midline of the appeared. Hence, they are rarely seen in infants over
hard palate, often towards the posterior aspect, but not 3 months of age.
involving the soft palate. They are small, white or yellowish
nodules usually less than 3 mm in diameter and, true to
Histopathology
their name, resemble small pearls. They often arise in clus-
ters of two to five lesions. There are very few reports of the histopathology of Epstein
pearls in infants, but studies of their development show
that the cysts are round or ovoid, with a thin lining of strat-
Pathogenesis
ified squamous epithelium and a para-­or orthokeratinised
Cysts along the midpalatal raphe arise from epithelial surface, and keratin fills the cyst cavity, usually in concen-
inclusions at the line of fusion of the palatal shelves. tric laminations containing flattened cell nuclei (Burke
As for many lesions of developmental origin, our under- et al. 1966; Moreillon and Schroeder 1982). The basal cells
standing of their pathogenesis often comes from early are flat, unlike those in the keratocyst. Epithelial-­lined
studies of human fetal tissues that are not likely or needed clefts may develop between the cyst and the surface oral
to be repeated. To appreciate the tissues involved, readers epithelium. The features have been described as ‘micro-
are encouraged to consult an up-­to-­date text on oral anat- keratocysts’ (Moreillon and Schroeder 1982) and are very
omy and embryology of the head and neck. The origin similar to those described above for gingival cysts of infants
of Epstein pearls has been well described in the early stud- and the small dental lamina cysts occasionally encoun-
ies of Burke et al. (1966) and of Moreillon and Schroeder tered in the oral mucosa (Figures 9.7 and 9.10).
(1982). Burke et  al. (1966) examined the palates of
32 human fetuses and found cysts in 31. They were mostly
Treatment
confined to the posterior half of the hard palate and were
most numerus between 3 to 6 months of gestation. After There is no indication for any treatment of midpalatal
this, the cysts gradually reduced in number. The cysts raphe cysts in infants. Once their contents are expelled,
were found in the palatal midline, associated with they atrophy and disappear.
164

10

Glandular Odontogenic Cyst

CHAPTER MENU
Clinical Features, 165
●● Frequency, 165
●● Age, 165
●● Sex, 166
●● Site, 166
●● Clinical Presentation,  166
Radiological Features, 167
●● Radiological Differential Diagnosis,  168
Recurrence of the Glandular Odontogenic Cyst, 169
Pathogenesis, 169
Histopathology, 170
●● Diagnostic Criteria,  173

●● Histological Differential Diagnosis,  175


–– Central Mucoepidermoid Carcinoma,  176
–– Immunohistochemistry and Molecular Studies,  178
Treatment, 181

Glandular odontogenic cyst is a developmental cyst in The lesion has evoked a considerable amount of interest
which the epithelium resembles salivary or glandular tis- because of its tendency to recur and its resemblance to, and
sue (Speight and Rautava 2022). It was first mentioned by possible relationship with, the central mucoepidermoid
Padayachee and Van Wyk (1987), who described two mul- carcinoma of the jaws. There have been few case series of
ticystic lesions with features of both botryoid odontogenic glandular odontogenic cyst, but numerous reports of single
cyst and ‘central mucoepidermoid tumour’. They named or relatively few cases had been published between its rec-
the lesion sialo-­odontogenic cyst to emphasise the resem- ognition in 1992 and the fourth edition of this book in 2007
blance to salivary tissue. Soon after, Gardner et al. (1988) (van Heerden et al. 1992; Semba et al. 1994; Economopoulou
reported eight cases and introduced the name ­glandular and Patrikiou 1995; Savage et al. 1996; de Sousa et al. 1997;
odontogenic cyst, and this was subsequently adopted as a Magnusson et  al.  1997; Ramer et  al.  1997; Koppang
new entity in the second edition of the World Health et al. 1998; Manojlović et al. 1998; Chavez and Richter 1999;
Organization (WHO) classification of odontogenic Jose et al. 2000; Lin et al. 2000; Tosios et al. 2000; Barreto
tumours (Kramer et al. 1992). The characteristic features of et  al.  2001; Noffke and Raubenheimer  2002; Ertaş et  al.
the cyst were that it was intrabony and multilocular radio- 2003; Osny et al. 2004; Pires et al. 2004; Tran et al. 2004;
logically; that it could recur if not adequately excised; and Abu-­Id et al. 2005; Qin et al. 2005; Shen et al. 2006). Since
that it was composed of multiple cystic spaces lined by a 2006 there have been a further 72 case reports. Mascitti
non-­keratinised epithelium with thickenings or plaques. et al. (2014) reviewed 110 reported cases, and Chrcanovic
Mucous and cylindrical cells also formed an integral part and Gomez (2018b) reviewed the literature up to 2018 and
of the epithelial component. found 169 fully documented cases.

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
Clinical Features 165

­Clinical Features is some variation globally. Table 10.2 shows that the reported
frequencies range from as low as 0.03% up to 1.16%.
Most publications on glandular odontogenic cyst have
been single case reports or small case series. The largest
Age
series was by Fowler et al. (2011), who described 46 cases.
The few reports of more than 10 cases are summarised in Glandular odontogenic cyst presents at an average age of
Table 10.1, along with data from the review of 169 cases by about 50 years, with a peak in the sixth decade (Table 10.1).
Chrcanovic and Gomez (2018b). Figure 10.1 illustrates the age and sex distribution of 105
glandular odontogenic cysts that we have pooled from the
review of Koppang et al. (1998) and from subsequent pub-
Frequency
lications that have recorded age and sex by decade
The glandular odontogenic cyst is a rare lesion and consti- (Economopoulou and Patrikiou  1995; Savage et  al.  1996;
tutes less than 1% of odontogenic cysts. Our data from the Magnusson et  al.  1997; Ramer et  al.  1997; de Sousa
archives of the University of the Witwatersrand for the et al. 1997; Manojlović et al. 1998; Chavez and Richter 1999;
period 1992–2004  indicated only six cases in a series of Lin et al. 2000; Tosios et al. 2000; Barreto et al. 2001; Noffke
3498  jaw cysts (0.2%) (Table  1.1). The Sheffield data were and Raubenheimer  2002; Ertaş et  al.  2003; Abu-­Id
similar, with only 11 cases (0.2%) in a sample of 7121 odon- et  al.  2005; Kaplan et  al.  2005b; Qin et  al.  2005; Jones
togenic cysts over a 30-­year period (Jones et al. 2006). Most et  al.  2006). There was a distinct peak frequency in the
other studies agree with this low frequency, although there sixth decade, particularly in males.

Table 10.1  Age, sex, and site distribution from selected reports of glandular odontogenic cysts (where n ≥ 10), and from the review of
169 cases by Chrcanovic and Gomez (2018b).

References n Mean age Male (%) Mandible (%)

Kaplan et al. (2005a) 11 49.0 100 72.7


Jones et al. (2006) 11 48.5 80.0 63.6
Fowler et al. (2011) 46 51.0 48.9 80.0
Soluk Tekkeşin et al. (2012b) 23 47.5 82.6 65.2
Tamiolakis et al. (2019) 19 51.1 68.4 44.4
Chrcanovic and Gomez (2018b) 169 48.1 53.6 73.2

Table 10.2  The frequency of glandular odontogenic cyst reported in selected studies from around the world.

Country Total odontogenic cysts Glandular odontogenic cysts

References n n %

Daley et al. (1994) Canada 6847 3 0.04


Mosqueda-­Taylor et al. (2002) Mexico 856 2 0.10
Meningaud et al. (2006) France 695 2 0.20
Jones et al. (2006) UK 7121 11 0.20
Shen et al. (2006) Japan 7023 12 0.17
Ochsenius et al. (2007) Chile 2944 1 0.03
Grossmann et al. (2007) Brazil 2812 2 0.07
Sharifian and Khalili (2011) Iran 1227 4 0.30
Soluk Tekkeşin et al. (2012b) Turkey 5003 23 0.45
Tamiolakis et al. (2019) Greece 5165 19 0.40
Aquilanti et al. (2021) Italy 2150 25 1.16
166 Glandular Odontogenic Cyst

30

25 Females
25
Males

20 11
No. of cases 17 17
15
15
6
12
7 11
10 9
5
1 7
14 1
5 11
8 8 7
6 6 2
0 1
0
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89 90+
Age

Figure 10.1  Age and sex distribution of 105 patients with glandular odontogenic cysts.

Table  10.1 summarises data from a number of studies, mandible often cross the midline in a characteristic sym-
including the aggregated data from 169 cases reviewed by metrical pattern (Figures  10.3 and  10.4). In the first 10
Chrcanovic and Gomez (2018b). Although there is some reported cases, 6 showed this appearance (Padayachee and
variation in age, there have been no cases reported in the Van Wyk 1987; Gardner et al. 1988). In the series of Noffke
first decade and very few over the age of 79. The youngest and Raubenheimer (2002), 6 of 9 cases involved the mandi-
reported cases have been in 11-­year-­olds (Noffke and ble and 4 of these crossed the midline. Koppang et  al.
Raubenheimer 2002; Faisal et al. 2015). (1998) listed the individual locations of 40 cases in both
mandible and maxilla. Most of these (34 cases: 85.0%) were
in the anterior mandible and 6 were in the maxilla. Some
Sex
involved the mandible from incisor or canine to the molar
The data in Figure  10.1 shows 61  males (58.1%) and 44 regions. One was described as involving the entire mandi-
females (41.9%). Most studies show a male predilection ble. Of the 7 cases reported in the study by Kaplan et  al.
(Table  10.1) and although this is variable, the overall sex (2005b), 4  were in the maxilla, 3  with extensive involve-
difference is not statistically significant (Chrcanovic and ment including the maxillary sinus.
Gomez 2018b).
Clinical Presentation
Site
Most cases are symptomless and are discovered by chance
Glandular odontogenic cyst is found most frequently in the on radiographs, or present as a slowly expanding, painless
mandible, with an overall proportion of 73.2% (Chrcanovic swelling. Only about 24% may show symptoms, although
and Gomez (2018b); Table  10.1). In their review of 110 73% had evidence of swelling (Chrcanovic and
cases, Mascitti et al. (2014) found that 74.8% arose in the Gomez 2018b). All 9 patients of Noffke and Raubenheimer
mandible, with 33.3% in the anterior mandible and a (2002) presented with swelling of the jaws, as did the 7
­further 25.2% extending from the anterior to the posterior patients reported by Kaplan et  al. (2005b), although 6 of
region. Chrcanovic and Gomez (2018b) found 60 cases the 7 were painless. One of their maxillary cases showed
(35.5%) that affected the anterior/premolar region of the facial and nasal swelling with infra-­orbital paraesthesia.
mandible. Lesions can grow to a large size, and they Fowler et  al. (2011) presented 46 cases of which 30  had
recorded 14 cases that filled the body of the mandible, 6 information on signs and symptoms; 20 (66.6%) showed
cases that extended across the whole maxilla, and 8 that bone expansion, but only 8 had other symptoms, including
extended from the mandibular body to the angle or ramus. 5 with pain, 2 with evidence of infection, and 1 with par-
Our experience has shown that lesions in the anterior aesthesia. The clinical features are summarised in Box 10.1.
­Radiological Features 167

borders, which were sclerotic in 8% and scalloped in 13%.


Information on cortical plate integrity was available in only
24 cases, half of which reported perforation and 17% ero-
sion or thinning of the cortical plates. Root resorption was
reported in 22% of patients and tooth displacement in 24%.
The size of the lesions ranged from 5 to 120 mm, but uni-
locular lesions were smaller on average (39 mm × 28 mm)
than multilocular lesions (63 mm × 30 mm).
Noffke and Raubenheimer (2002) reported a radiological
study of nine cases. All showed cortical expansion and
those with a diameter of more than 60 mm had perforated
the cortical plate. Seven (77.8%) were unilocular and well
circumscribed: five with smooth borders and two with
irregular borders. Eight cases showed tooth displacement.
The two multilocular lesions showed irregular or scalloped
borders and perforations. Their measurements ranged
from 32 × 20 mm in a 14-­year-­old boy to a multilocular
Figure 10.2  Radiograph of a glandular odontogenic cyst in the lesion of 165 × 40 mm in a 53-­year-­old woman. Unlike the
maxilla. There is a large unilocular radiolucent area with a
survey by Manor et al. (2003), no significant root resorption
smooth but slightly scalloped corticated margin (arrows). The
histology of this lesion is shown in Figure 10.6a. of involved teeth was observed.
These authors suggested that multilocularity might be
a size-­related phenomenon, since lesions over 60 mm
­Radiological Features tended to be multilocular and showed bone expansion
and perforation (Noffke and Raubenheimer 2002; Manor
Glandular odontogenic cysts are well-­defined radiolucen- et al. 2003).
cies with a corticated margin (Figure 10.2-10.4). Although In their review of 169 cases, Chrcanovic and Gomez (2018b)
they may be multicystic and are often regarded as charac- found that 26% showed evidence of perforation of the cortical
teristically multilocular, in fact up to 60% are unilocular plate, 31% were associated with displaced or unerupted teeth,
(Manor et  al.  2003; Mascitti et  al.  2014; Chrcanovic and but only 13.9% showed evidence of tooth resorption.
Gomez 2018b). Occasional cases have been reported to occur in a denti-
Manor et  al. (2003) reviewed the radiology of 56 cases gerous relationship with unerupted teeth. Fowler et  al.
and found that 52% had been described as unilocular and (2011) reported that 8 of their 46 cases were in a dentiger-
48% as multilocular. Almost all (94.5%) had well-­defined ous relationship radiographically, but no details were

Figure 10.3  Radiograph of an extensive multilocular glandular odontogenic cyst. The lesions crosses the midline in an almost
symmetrical pattern. Courtesy of Prof. Christoffel Nortjé.
168 Glandular Odontogenic Cyst

include a wide range of benign cysts of the jaws as well as


Box 10.1  Glandular Odontogenic Cyst: Clinical
other lesions, including giant cell granulomas. For multi-
and Radiological Features
locular lesions the differential diagnosis may be more lim-
●● They are rare – less than 1% of odontogenic cysts ited. We have noted a number of large glandular
●● Seen in adults, mean age about 50 years. Peak in odontogenic cysts that cross the midline of the mandible
sixth decade with a characteristic, almost symmetrical pattern
●● Slightly more common in males (Figures 10.3 and 10.4). Other lesions, in particular amelo-
●● Over 70% occur in the mandible. Most common in the blastoma, may arise at this site and show similar multi-
anterior/premolar area locular features. However, the glandular odontogenic cyst
●● Usually symptomless, but 70% show swelling/bony seems to show relatively little bucco-­lingual expansion
expansion relative to the lateral growth, whereas ameloblastomas
●● Well-­defined radiolucency with corticated margins tend to show a ballooning growth pattern with a similar
●● About one-­third are multilocular degree of bucco-­lingual and lateral expansion. For com-
●● May grow to a large size and be extensive parison, an example of a large ameloblastoma is shown in
●● Crossing the midline in a symmetrical pattern is Figure 10.5.
characteristic Odontogenic keratocyst may show a very similar growth
●● Recurrence rate is about 22% pattern to glandular odontogenic cyst, but a location in the
anterior mandible is unusual.
A further consideration in the differential diagnosis is lat-
given. Ferreira et al. (2019) reported a case in a dentiger- eral periodontal cyst or botryoid odontogenic cyst (see dis-
ous relationship with a mesially impacted mandibular cussion in Chapter 8), both of which are also found in the
third molar, and reviewed a further 22 cases reported since anterior regions of the jaws. All three lesions may arise in a
1996. Of 13 cases where site was specified, 5 were associ- lateral periodontal position with a predilection for the man-
ated with mandibular third molars and 3 with mandibular dible. Glandular odontogenic cyst and botryoid odonto-
canines. genic cyst may also be multilocular. On radiology alone, it
may not be possible to differentiate between these lesions.
However, lateral periodontal cysts are usually round or oval
Radiological Differential Diagnosis
and are rarely greater than 10 mm in diameter, while botry-
Generally, the radiological features of glandular odonto- oid odontogenic cysts have a mean diameter of 15 mm and
genic cyst are not specific. The overall clinical and are rarely greater than 50 mm. Therefore, a glandular odon-
­radiological features suggest a benign process and, for sim- togenic cyst should always be suspected if a lesion embraces
ple unilocular lesions, the differential diagnosis may multiple teeth or is larger than 50 mm (Figure 10.2).

Figure 10.4  Another example similar to Figure 10.3. The cyst crosses the midline from second molar to second molar, but there is
relatively little expansion of the mandible. Source: Prof. Paul Speight (Previously published: El-­Naggar AK, Chan JKC, Grandis JR, Takata
T, Slootweg PJ. World Health Organization Classification of Tumours of Head and Neck. IARC, Lyon, 2017. Courtesy of IARC.)
Pathogenesis 169

(a) (b)

Figure 10.5  A radiograph (a) and an axial computed tomography (CT) scan (b) of a large ameloblastoma. In comparison to the
glandular odontogenic cyst (Figures 10.3 and 10.4), the lesion is ‘ballooning’, with considerable expansion of the cortical plates.

­ ecurrence of the Glandular


R reported that 9 (19.6%) cases were known to have recurred
Odontogenic Cyst with an average time interval of 8 years (range 3–13 years);
6 cases recurred twice. They found no significant differ-
The glandular odontogenic cyst has a reputation for ences in the histological features between recurrent and
recurrence and many publications tend to label it as non-­recurrent cysts.
‘aggressive’. The lesion may certainly reach a large size, In their analysis of recurrences after treatment, Kaplan
but growth appears to be slow, as evidenced by the corti- et al. (2005b) reviewed 48 patients, including 7 treated in
cated or even sclerotic margins. Recurrence is probably a their own department. Pooling together patient data that
consequence of the size of the lesion and its tendency to recorded location, size, radiographic features, different
be multilocular and multicystic, making it more likely modalities of primary treatment of the lesions, and length
that fragments of cyst are left behind after surgery. of follow-­up, they found that recurrences occurred in 13
The overall recurrence rate from 58 studies reporting cases (27.1%); 2 patients had three recurrences each within
97 lesions where data was available was 21.6% (Chrcanovic 2 years. Citing a series of publications, they found recur-
and Gomez 2018b). These authors also undertook an anal- rence rates ranging from 21 to 55%. They also compared
ysis of factors associated with recurrence. They showed recurrent and non-­recurrent lesions and showed that large
that lesions that were curetted were less likely to recur than lesions recur (85.6%) more often than small lesions (14.4%)
those with enucleation alone (16.7% vs 25.4%). Other fac- and that recurrent lesions were more likely to be multiloc-
tors associated with a higher recurrence rate were bone ular (64.3 versus 41.2%) or associated with perforation or
expansion, cortical bone perforation, and multilocularity. thinning of the cortical plates (71.4 versus 47.1%; Kaplan
Histological factors associated with recurrence included et al. 2005b, 2008).
multicystic lesions and the presence of mucous cells or These studies suggest that about 22% of glandular odonto-
cilia. However, it must be noted that none of these factors genic cysts may recur, and that recurrence is associated with
was shown to have a statistically significant association larger multilocular or multicystic lesions with evidence of
with the recurrence rate. bone resorption and perforation of the cortical plates.
In their review of 45 cases, Koppang et al. (1998) found
information about recurrence in 38, of which 8 (21%)
recurred; 6 of these recurred after 2–8 years, and 2 cysts ­Pathogenesis
recurred twice after 2 and 5 years, and 3 and 5 years. Of 6
recurrent lesions where information was available, 5 were Very little has been written on the pathogenesis of the glan-
multilocular. In their series of 46 cases, Fowler et al. (2011) dular odontogenic cyst. Although most authors agree that
170 Glandular Odontogenic Cyst

it is of odontogenic origin and probably arises from rest homogeneous expression of the hedgehog pathway–related
cells of the dental lamina, there is still much speculation. proteins SHH, PTCH, SMO, and Gli1 in the epithelial lin-
The very first reports described the close similarities ing of all lesions. They also found similar expression in all
between the lining of the glandular odontogenic cyst and cases (n = 20) of dentigerous cysts examined.
those of the lateral periodontal and botryoid odontogenic
cysts (Padayachee and Van Wyk 1987; Gardner et al. 1988).
These authors were particularly struck by the similar ­Histopathology
­epithelial thickenings and plaques, and the multicystic or
botryoid nature of the lesions. This was so striking that Lesions are usually enucleated with or without curettage,
many early authors regarded the glandular odontogenic so the pathologist often receives a fragmented or collapsed
cyst as a variant of the botryoid odontogenic cyst specimen, making it difficult to always appreciate the true
(Browne 1991b; Noffke and Raubenheimer 2002) and the morphology of the lesion. Lesions may be multicystic and
two lesions are still often confused (see discussion on dif- this may not be clearly apparent on a fragmented speci-
ferential diagnosis below and in Chapter  8). Occasional men. Larger lesions may occasionally be removed with a
cases have been associated with other odontogenic lesions, localised block resection and examination of the intact
including dentigerous cysts and ameloblastomas, and specimen may show a botryoid, bosselated, or lobular cyst
some have contained hyaline bodies (Gardner et al. 1988; that, when bisected, has a multicystic appearance on the
Ide et al. 1996; Cousin et al. 2017), which has been taken as cut surface. Examination of the cyst wall will often show
further e­ vidence that confirms an odontogenic origin. thickenings and papillary projections and the lumen may
Although the lesion shows features of glandular or even occasionally contain mucus.
‘salivary’ differentiation, the possibility of an origin from The first reports of the glandular odontogenic cyst pre-
intraosseous entrapped salivary tissue has not gained any sented comprehensive and detailed descriptions of the his-
support. The glandular changes are regarded as metaplastic tological features (Gardner et al. 1988; Padayachee and Van
in nature and an example of the known pluripotentiality of Wyk 1987) that have not been bettered and still provide the
odontogenic epithelium. benchmark for diagnosis. The cyst shows a number of
The possibility that glandular odontogenic cyst is a ­different features, few of which are specific, but when con-
benign neoplasm cannot be dismissed without further sidered in combination may be diagnostic. Gardner et al.
research. The relationship between the cyst and the intra- (1988) listed more than seven features that may be seen in
osseous or central mucoepidermoid carcinoma is discussed the glandular odontogenic cyst, and subsequent authors
in the next section. These two lesions show a number of (Kaplan et al. 2005a; Fowler et al. 2011) have used these to
histological similarities (Kaplan et  al.  2008; Fowler develop diagnostic criteria to help differentiate it from
et  al.  2011; Reddy et  al.  2019) and some authors have other lesions. Table 10.3 lists the features that may be found
shown evidence of similar MAML2 rearrangements, sug- in glandular odontogenic cysts, with their relative fre-
gesting that glandular odontogenic cyst may be a precursor quency as reported in the series of 46 cases of Fowler et al.
to central mucoepidermoid carcinoma (Greer et  al.  2018; (2011) and the review of 169 cases by Chrcanovic and
Nagasaki et al. 2018). Gomez (2018b).
The role of the PTCH gene and of the hedgehog signal- Glandular odontogenic cysts are characteristically multi-
ling pathway in the pathogenesis of the keratocyst is dis- locular on radiography or multicystic on histology
cussed in detail in Chapter  7. Hedgehog signalling is (Figure 10.6), although overall, only about 60–70% of cases
activated by interaction between the Sonic hedgehog have been described as multicystic or botryoid (Fowler
(SHH) protein and its receptor PTCH on the cell surface, et al. 2011; Chrcanovic and Gomez 2018b).
although constitutive ligand-­independent activation may The cyst may be lined in parts by a non-­keratinised squa-
also occur as a result of truncating mutations or loss of het- mous epithelium of variable thickness, which lies on a flat
erozygosity (LOH) of the PTCH gene. Activation of the basement membrane. In places the lining may be only two
pathway results in downstream expression of factors medi- layers thick, with a cuboidal or flattened basal layer and a
ating cell growth, proliferation, and differentiation, and columnar surface layer (Figure 10.7). The surface layer may
there is now evidence that SHH/hedgehog signalling is not contain occasional mucous cells or show apocrine ‘snout-
unique to keratocysts, but may drive growth and prolifera- ing’ or decapitation secretion. We have seen one case
tion of a number of odontogenic cysts and tumours (Gomes (Figure 10.7) in which most of the lining had this appear-
and Gomez  2011), including glandular odontogenic cyst ance. However, most cases show epithelial thickenings or
(Barreto et al. 2002; Zhang et al. 2010). Zhang et al. (2010) plaques that may be present in the thin epithelium or in the
examined 12 glandular odontogenic cysts and found stratified squamous epithelium (Figure 10.8). The plaques,
­Histopathology 171

(a) (b)

Figure 10.6  Two examples of multicystic glandular odontogenic cysts. Even at this low magnification it can be seen that the linings
are mostly thin, but show multiple plaque-­like thickenings and areas with papillary projections (b, lower right). (a) The histology of the
cyst in Figure 10.2. Source: Martin and Speight (2017), reproduced with permission from Elsevier.

Figure 10.7  Glandular odontogenic cyst may have a thin regular lining only two to three cell layers thick. The superficial cells are
cuboidal or columnar and may show apocrine secretion (arrows).

when present, are similar morphologically to those in the A characteristic feature, seen in almost all cases, is glan-
gingival cyst of adults, the lateral periodontal cyst, and the dular or pseudo-­glandular structures within the epithelial
botryoid odontogenic cyst, and may show spherules or lining. These form microcysts or pools that are lined by
­whorling (Figure  10.8b). Occasionally these plaques cuboidal or columnar cells, to give an appearance of small
­protrude into the cyst cavity to form short bulbous ­papillary ducts (Figure  10.9). Occasionally these are also lined by
projections, that may be numerous and prominent mucous cells (Figure 10.9, arrow). In certain planes of sec-
(Figure 10.8a, d). tion, these microcysts may be seen to open onto the surface
In almost all cases (Table  10.3; Fowler et  al.  2011; of the epithelium through openings or crypts, giving the
Chrcanovic and Gomez 2018b), the epithelial lining has a epithelium a papillary or corrugated surface, described by
superficial layer of columnar or cuboidal cells (Figure 10.8c, Fowler et al. (2011) as ‘tufting’. The crypts are sometimes
d), sometimes referred to as ‘hobnail’ cells, which often empty and sometimes contain mucus (positive for periodic
show cilia or apocrine snouts (Figure  10.9). Superficial acid–Schiff [PAS] or mucicarmine).
hobnail cells may be seen throughout the ­lining, but may Mucous cells are also characteristic, but it must be noted
be most prominent overlying areas of thickening. that these are not essential for the diagnosis: up to 30% of
172 Glandular Odontogenic Cyst

(a) (b)

(c) (d)

Figure 10.8  Epithelial thickenings and plaques in glandular odontogenic cysts. Sometimes these are prominent and form papillary
projections (a, b, d). Superficial cuboidal or columnar (‘hobnail’) cells can clearly be seen (c, d).

cases may not contain mucous cells and in others they may Figure 10.6, but some of these may be due to the plane of
be very few in number (Table  10.3; Fowler et  al.  2011; section. Irregular calcifications may also be present in the
Chrcanovic and Gomez 2018b). When present, they most connective tissue wall, but inflammation is unusual, except
often appear as superficial goblet cells that may be seen in large lesions that may have been traumatised or perfo-
alone as single cells or in groups (Figure  10.10). In some rated the cortical plate. In one case we have seen small
cases focal accumulations of plump mucous cells have focal areas of flattened epithelium, with a parakeratinised
been seen (Figure 10.11), and these may be admixed with surface (Figure 10.12).
squamous cells in a pattern similar to that seen in mucoep- There have been reports of glandular odontogenic cysts
idermoid carcinoma (see below). Most cysts also show scat- associated with ameloblastoma. Cousin et  al. (2017)
tered or focal accumulations of clear or vacuolated cells. reported a single case and reviewed four previous reports.
Occasional clear cells can be seen in the cyst linings shown These suggested ameloblastomatous epithelium within the
in Figures 10.7–10.10 and 10.12. lining of the cyst, or islands of ameloblastoma within the
Islands of odontogenic epithelium and even microcysts wall of an otherwise typical glandular odontogenic cyst.
may be present in the connective tissue wall of the cyst. However, on review we feel that two of these reports do not
Fowler et  al. (2011) described three cases with islands in provide sufficient evidence that the lesions meet the crite-
the wall as ‘mucoepidermoid carcinoma-­like’. In two cases ria for a diagnosis of glandular odontogenic cyst, and one
these islands were found within bone. Small cysts and report is so unusual as to be difficult to diagnose. Thus
islands can be seen in the walls of the cysts shown in there are only two reports that are sufficiently well
­Histopathology 173

Figure 10.9  Part of the lining of the cyst shown in Figure 10.6b. An area of a thickened lining with microcysts, lined by cuboidal cells
to give an appearance like small ducts. Mucous cells are also noted (arrow). Inset: duct-­like appearance and cilia.

Table 10.3  Histological features of the glandular odontogenic unicystic ameloblastoma. These lesions appear to be unu-
cyst and their frequency (%). From the series of Fowler et al. (2011) sual oddities and, based on only two cases, the clinical sig-
and the review of Chrcanovic and Gomez (2018b).
nificance of these findings remain uncertain.

Fowler Chrcanovic and


et al. (2011) Gomez (2018b) Diagnostic Criteria

n = 46 cases n = 169 cases (review) It can be seen that there are many and varied histological
features that may be encountered in the glandular odonto-
Superficial 100.0 95.0 genic cyst. Each individual cyst may present some but not
cuboidal cells all of the features and diagnosis may be difficult if key fea-
Microcysts/ 95.7 97.5 tures are absent in any given biopsy. Whenever possible the
duct-­like full specimen should be examined and, with large lesions, it
structures
is necessary to examine multiple blocks. Box 10.2 summa-
Apocrine snouting 91.3 40.4
rises the diagnostic criteria and emphasises the need to also
Clear cells 89.1 45.3 consider the clinical and radiological findings (Box 10.1).
Variable thickness 89.1 96.3 Kaplan et al. (2005a) divided the histological features into
Papillary 84.8 77.6 major and minor categories and suggested that each of the
projections major features must be present for a diagnosis of glandular
(tufting)
odontogenic cyst, while the presence of any of the minor fea-
Mucous cells 71.7 72.0 tures was supportive. The five major features were (i) squa-
Epithelial spheres 67.4 72.7 mous epithelial lining; (ii) variations in thickness of the lining
Multicystic 63.0 70.2 with or without epithelial ‘spheres’ or ‘whorls’ or focal lumi-
Ciliated cells 21.7 61.5 nal proliferation; (iii) cuboidal eosinophilic cells or ‘hobnail’
cells; (iv) mucous (goblet) cells with intraepithelial mucus
pools with or without crypts lined by mucus-­producing cells;
documented to suggest that ameloblastoma and glandular and (v) glandular microcystic or duct-­like structures. The four
odontogenic cyst may co-­exist (Gardner et al. 1988; Cousin minor features were (i) papillary proliferation; (ii) ciliated
et al. 2017). Both cases arose in the premolar/molar region cells; (iii) multicystic or multiluminal architecture; and (iv)
of the mandible and showed islands of quite typical follicu- clear or vacuolated cells in the basal or spinous layer.
lar ameloblastoma in the wall of a glandular odontogenic A significant problem with these criteria is that if a major
cyst. Cousin et  al.’s (2017) case also showed transition of feature is missing, then the diagnosis cannot be made. In
the lining from cyst to ameloblastoma. The authors sug- their own study, Kaplan et al. (2005a) reviewed 29 reports
gested that the features were analogous to a mural type of of glandular odontogenic cyst and showed that no reports
174 Glandular Odontogenic Cyst

Figure 10.10  A periodic acid–Schiff (PAS)-­stained


section of part of the lining of the cyst shown in
Figure 10.6a. Mucous cells were not prominent, but
were seen in the superficial layers, either as single
cells or in small groups.

Figure 10.11  An example of a glandular


odontogenic cyst with prominent mucous cells in
focal accumulations. Plaque-­like thickenings,
whorling, and superficial columnar cells can also
be seen.

Figure 10.12  A portion of a cyst wall from the


lesion illustrated in Figure 10.6a. Note small area
of parakeratinisation (arrows). Clear cells are also
noted in the basal layers.
­Histopathology 175

Box 10.2  Glandular Odontogenic Cyst: Diagnostic Criteria and Differential Diagnosis


●● Over 70% occur in the mandible. Most common in the anterior/premolar area
●● Well-­defined radiolucency with corticated margin. About one-­third are multilocular
●● Histology typically shows multicystic lesion, superficial cuboidal/columnar (hobnail) cells, microcysts, mucous
cells, cilia, and plaque-­like thickenings
●● If 6 of 10 features are present (Table 10.3), the diagnosis is almost certain
●● Immunocytochemistry with cytokeratin (CK)19 and CK18 may help. Glandular odontogenic cyst is always CK19+,
but only 30% express CK18
●● Do not show MAML2 rearrangement
Caution: features that suggest an alternative diagnosis:
●● Multicystic lesion with plaques or thickenings, but no hobnail or mucous cells or microcysts: consider botryoid odon-
togenic cyst
●● Unilocular lesion with plaques or thickenings, but no hobnail or mucous cells or microcysts: consider lateral perio-
dontal cyst
●● Cyst with mucous cells, but no microcysts, apocrine secretion, cilia, or thickenings: consider odontogenic cyst with
mucous metaplasia, especially radicular or dentigerous cyst
Mucoepidermoid carcinoma (Table 10.4) shows:
●● Evidence of invasion of bone or the cyst wall
●● Admixture of epidermoid or intermediate cells and mucous cells
●● Solid areas
●● Immunocytochemistry with CK19 and CK18 may help. Mucoepidermoid carcinoma is always CK18+, but only 50%
express CK19
●● Fluorescence in situ hybridisation (FISH) shows MAML2 rearrangement in 50% of cases

recorded the presence of all five features listed as major cri- multicystic morphology were statistically more likely to be
teria. For example, only 65.5% recorded the presence of found in the glandular odontogenic cyst. Furthermore, they
mucous cells or hobnail cells and only 58.6% reported showed that if microcysts are present, then a multicystic
microcysts. It is not clear, therefore, why the authors chose morphology is most associated with the diagnosis.
to make these features mandatory. These analyses suggest that great care needs to be taken
Fowler et al. (2011) analysed 46 cases and also showed that in diagnosing these lesions. Although the features are char-
not all features were present in all cysts (Table 10.3), and this acteristic, each is not specific and the diagnosis is made on
was confirmed in a review of 169 cases by Chrcanovic and consideration of a combination of the features. If 7 of the 10
Gomez (2018b). It can be seen, for example, that almost all features described in Table 10.3 are present, then this is vir-
cysts contain superficial columnar cells and microcysts, but tually diagnostic of glandular odontogenic cyst. However,
mucous cells are absent from about 30% of lesions. Therefore, in our experience this number of features may not always
a common diagnostic error is to exclude a diagnosis of glan- be present. The features most commonly seen are superfi-
dular odontogenic cyst if mucous cells are not seen. Fowler cial cuboidal (‘hobnail’) cells, and a combination of these
et al. (2011) thus adopted a more pragmatic approach to diag- with microcysts, epithelial plaques or spheres, mucous
nosis and suggested that the presence of six or more of the cells, and a multicystic morphology is usually sufficient to
features listed in Table 10.3 was highly predictive of a diagno- render a diagnosis of glandular odontogenic cyst. Special
sis of glandular odontogenic cyst, and if there were five or care must be taken when examining small incisional biop-
fewer then this diagnosis was less likely. They showed that all sies, because multiple features are rarely present. The histo-
46 cysts had six features or more and that 43 (93.5%) had logical features should always be interpreted along with the
seven or more. Only two cysts not diagnosed as glandular clinical and radiological features (Boxes 10.1 and 10.2).
odontogenic cyst had more than seven features, but both
were unusual and the precise diagnosis was not certain.
Histological Differential Diagnosis
When compared to 21 lesions with similar features to glan-
dular odontogenic cyst (‘mimickers’), these authors found As discussed above, a diagnosis of glandular odontogenic
that microcysts, epithelial spheres, clear cells, and a cyst is made on the recognition of a combination of a
176 Glandular Odontogenic Cyst

number of histological features. However, these varied mucous cells, especially in dentigerous cysts, to arrive at a
appearances may still cause considerable diagnostic diffi- misdiagnosis of glandular odontogenic cyst. Takeda et al.
culty and this cyst is probably the most commonly missed (2005) examined 130 dentigerous cysts and found mucous
or misdiagnosed of all the odontogenic cysts. As discussed cells and cilia in 23.8% and 10.8%, respectively. Many cases
in the next section and in Chapter  8, the most common also showed intraepithelial mucus pools that resemble
errors of diagnosis are confusion with central mucoepider- microcysts or duct-­like structures. Fowler et  al. (2011)
moid carcinoma and botryoid odontogenic cyst. examined 13 selected dentigerous cysts that showed evi-
In early reports, glandular odontogenic cyst was regarded dence of metaplastic changes and found evidence of some
as a variant of botryoid odontogenic cyst (Browne 1991b; overlap with features seen in glandular odontogenic cyst.
Noffke and Raubenheimer 2002) and a number of papers The changes seen in dentigerous cysts included superficial
reported unusual lateral periodontal or botryoid odonto- cuboidal cells, microcysts, mucous cells, apocrine snout-
genic cysts, which on review are in fact glandular odonto- ing, ‘tufting’, and clear cells, but none showed a combina-
genic cysts (Phelan et  al.  1988; Buckley et  al.  1989; tion of features that would lead to a diagnosis of glandular
Heikinheimo et  al.  1989; Öçok et  al.  2005). Glandular odontogenic cyst. These authors also pointed out that the
odontogenic cyst and botryoid odontogenic cyst may be ‘microcysts’ seen in dentigerous cysts are usually simple
multilocular on radiographs and multicystic on histology mucus pools and not true microcysts. We would agree with
and both show typical epithelial thickenings or plaques, this and would suggest that similar pools are also often
which they also share with lateral periodontal cyst (com- seen in radicular cysts with mucous metaplasia. The pools
pare Figures  8.6 and  10.8). However, glandular odonto- are small, intraepithelial, and lined by flattened epithelial
genic cyst also shows microcysts, superficial columnar cells, which appear to have been ‘pushed aside’ by the
cells, apocrine secretion, and mucous cells, which are not accumulation of mucus (Figure  10.13). In contrast, the
features of lateral periodontal or botryoid cysts. Also, we microcysts in glandular odontogenic cyst are often lined by
have noted that the epithelial plaques are often more prom- cuboidal or columnar cells to invoke the appearance of
inent in glandular odontogenic cyst and may form bulbous ductal structures, some of which may open into the cyst
papillary processes (e.g. Figure 10.8a, d), but these are rare lumen (Figure 10.9). Although cases have been reported, it
in lateral periodontal and botryoid cysts. Furthermore, lat- should also be noted that it is not common for glandular
eral periodontal and botryoid odontogenic cysts lie lateral odontogenic cysts to be found in a dentigerous relationship
to the teeth and are rarely greater than 10 mm or 40 mm in and therefore a pericoronal radiolucency argues against
diameter, respectively. In the differential diagnosis, there- the diagnosis.
fore, if lesions are large and microcysts, hobnail cells, or
mucous cells are seen, then a diagnosis of botryoid odonto- Central Mucoepidermoid Carcinoma
genic cyst is probably incorrect and glandular odontogenic Most histopathologists, faced with histological sections of a
cyst should be considered. glandular odontogenic cyst, will have noted the similarities
All pathologists are aware that many types of odonto- to the central mucoepidermoid carcinoma. The resem-
genic cyst may show evidence of metaplasia with mucous blance between these two lesions was noted in the first
cells, but a common error is the overinterpretation of reported cases by Padayachee and Van Wyk (1987), who

Figure 10.13  A dentigerous cyst with mucous


metaplasia. There are mucous cells, but also mucus
pools lined by flattened epithelial cells (compare
with Figure 10.9).
­Histopathology 177

described their lesions as having features of both botryoid there is much speculation in the literature about their rela-
odontogenic cyst and ‘central mucoepidermoid tumour’. tionship, in particular that mucoepidermoid carcinomas
As early as 1956, however, Hodson had drawn attention to may arise from glandular odontogenic cysts, or that glan-
mucous cells in odontogenic cysts and reported a series of dular odontogenic cyst is at the benign end of a spectrum
cases as ‘muco-­epidermoid odontogenic cysts of the jaws’ of mucoepidermoid carcinoma (Waldron and Koh 1990).
(Hodson 1956). Most of these showed mucous metaplasia If care is taken and the diagnostic criteria discussed
in radicular or dentigerous cysts, but his illustrations sug- above are followed, diagnosis should rarely be a problem
gest that at least one case could have been a glandular (Box 10.2). However, about 50% of intraosseous mucoepi-
odontogenic cyst, and he drew attention to the similarities dermoid carcinomas are low grade, and may be cytologi-
with ‘muco-­epidermoid salivary tumours’. Since then, cen- cally bland and multicystic, leading to potential confusion.
tral (intraosseous) mucoepidermoid carcinoma has been Mucoepidermoid carcinomas are almost always composed
shown to be the most frequently encountered central sali- of multiple variably sized cystic spaces, whereas glandular
vary gland tumour (Bouquot et  al.  2000; Li et  al.  2008), odontogenic cysts are multicystic in only 60–70% of cases
with 147 cases reported up to 2018 (de Souza et al. 2018). (Table 10.3). Both lesions, however, may contain mucous
There has been much speculation about the origin of this cells, duct-­like structures, and clear cells. In glandular
lesion, but possible sources include intraosseous salivary odontogenic cyst these features are seen within the lining
tissue entrapped during development, glandular differen- of the cyst, whereas in mucoepidermoid carcinoma more
tiation of odontogenic epithelial rests, or metaplasia in solid islands may be seen, or sheets of tumour may infil-
odontogenic cysts (Eversole et al. 1975; Bouquot et al. 2000; trate from the cyst lining into the underlying connective
Li et al. 2008). Most authors favour an odontogenic origin, tissue (Figure  10.14) or adjacent bone (Figure  10.15).
since central mucoepidermoid carcinomas have been Mucoepidermoid carcinomas also show a characteristic
shown to arise in association with odontogenic cysts admixture of epidermoid or intermediate cells, with
(Eversole et al. 1975; Chaisuparat et al. 2012). mucous or clear cells (Figure 10.14b), and may show areas
Because of the histological similarities between glandu- of keratinisation or keratin pearls (Figure  10.15, inset).
lar odontogenic cyst and mucoepidermoid carcinoma, Conversely, glandular odontogenic cysts may show ciliated

(a) (b)

Figure 10.14  Two examples of central mucoepidermoid carcinoma. (a) Islands of tumour invade the connective tissues. (b) Solid
islands of tumour are seen in the cyst wall. These show a characteristic admixture of epidermoid or intermediate cells with clear cells
and mucous cells (inset).
178 Glandular Odontogenic Cyst

Figure 10.15  Central mucoepidermoid carcinoma (same lesion as Figure 10.14a). The lesion is multicystic and small cysts are
invading into the adjacent bone. The cysts are lined by epidermoid and mucous cells (inset) and a small island can be seen with
central keratinisation (inset, arrowhead).

cells and apocrine secretion, and be characterised by super- and tumours. Central (n = 6) and salivary (n = 23) mucoep-
ficial hobnail cells, plaque-­like thickenings, whorling, and idermoid carcinomas showed very similar cytokeratin pro-
epithelial spheres and papillary projections, all features files, with consistent expression of CK5, CK7, CK8, CK14,
that are rarely or never seen in mucoepidermoid carcino- and CK18. Glandular odontogenic cysts (n  =  10) shared
mas. Invasion into adjacent bone is also a feature of expression of CK5, CK7, CK8, and CK14 with mucoepider-
mucoepidermoid carcinoma, but has been rarely described moid carcinoma, but only 30% expressed CK18. Conversely,
in glandular odontogenic cyst (Figure  10.15; Fowler 100% of glandular odontogenic cysts showed CK19 expres-
et  al.  2011). Table  10.4 summarises the histological simi- sion compared to only 50% of central mucoepidermoid car-
larities and differences between glandular odontogenic cinoma. However, CK19 expression was higher in salivary
cyst and mucoepidermoid carcinoma. tumours, with up to 100% of intraoral lesions (palate and
retromolar) being positive. CK19 expression was also seen
Immunohistochemistry and Molecular Studies in the majority of other odontogenic lesions.
A number of studies have been undertaken to examine the These results would support the view that glandular
potential role of immunocytochemistry in the diagnosis of odontogenic cyst and central mucoepidermoid carcinoma
glandular odontogenic cyst, especially expression of are distinct entities, characterised in particular by high
cytokeratins (de Sousa et  al.  1997; Koppang et  al.  1998; expression of CK19 and CK18, respectively. When faced
Pires et al. 2004; Shen et al. 2006). However, most studies with an individual lesion, immunocytochemistry for a
have only used one or two cases and have not compared combination of CK19 and CK18 may be a helpful diagnos-
expression to any potential alternative lesions. Overall they tic adjunct. It must be noted that although CK 19 is sensi-
have shown that glandular odontogenic cysts tend to show tive for glandular odontogenic cyst, it is not specific, and a
patchy and variable expression of simple cytokeratins, but positive result is not diagnostic. However, since 100% of
in particular most cases have been positive for CK7 and glandular odontogenic cysts are positive for CK19, a nega-
CK19 and are negative for CK20 (Shen et al. 2006). Pires tive result may exclude this diagnosis. The converse is true
et al. (2004) specifically addressed the issue of differential for CK18: a negative result would mitigate against mucoep-
diagnosis and investigated expression of 11 cytokeratins in idermoid carcinoma. Thus the immunophenotypes that
glandular odontogenic cyst and central mucoepidermoid would support a diagnosis are CK19+/CK18– for glandular
carcinoma, as well as in a number of salivary mucoepider- odontogenic cyst and CK19–/CK18+ for central mucoepi-
moid carcinomas and a range of other odontogenic cysts dermoid carcinoma (Pires et al. 2004; Table 10.4).
­Histopathology 179

Table 10.4  Features that may assist in the differential diagnosis of glandular odontogenic cyst and central mucoepidermoid
carcinoma.

Histological features Glandular odontogenic cyst Central mucoepidermoid carcinoma

Multilocular radiolucency Y Y
Multicystic Y Y
Perforates cortical plate Y Y
Mucous cells Y Y
Duct-­like structures Y Y
Plaques and thickenings Y N
Surface columnar/cuboidal cells Y N
Ciliated cells Y N
Apocrine secretion Y N
Commonly anterior mandible Y N
CK19+/CK18−a Y N
Solid areas infiltrating cyst wall N Y
Admixture of epidermoid and mucous cells N Y
Invasion into cyst wall and bone N Y
Commonly posterior mandible N Y
CK18+/CK19−a N Y
Maspin+ mucous cells N Y
MAML2 translocation N Y

CK, cytokeratin; N, not seen or very rare; Y, characteristic feature often present.
a
 See text for detailed explanation. Source: Based on Pires et al. (2004).

Other studies have investigated proliferation markers as These papers demonstrate some differences in prolifera-
potential diagnostic aids. Tosios et  al. (2000) compared tion markers, but given the great range of expression, vari-
expression of bcl-­2 protein, Ki-­67 antigen, and p53 protein ation between lesions, and the overall low proportion of
in three glandular odontogenic cysts with expression in six positive cells, they do not seem promising as diagnostic
dentigerous cysts. The glandular odontogenic cysts showed aids in differentiating the glandular odontogenic cyst from
homogeneous reactivity for bcl-­2 protein, but the dentiger- the central mucoepidermoid carcinoma.
ous cysts were negative or had faint basal cell staining. Vered et  al. (2010) explored expression of maspin as a
Ki-­67  was slightly lower in glandular odontogenic cysts, potential discriminatory marker. They compared glandular
but there were no differences in p53 staining. The authors odontogenic cyst (n = 8) with low-­grade mucoepidermoid
speculated that the increased expression of the anti-­ carcinoma of salivary origin (not central; n = 6), and found
apoptotic bcl-­2 may contribute to growth of the cyst; cell that maspin expression was similar in the epithelial cells.
proliferation and p53 status did not seem to play a signifi- However, they found a significantly greater expression of
cant role. maspin in mucous cells of mucoepidermoid carcinoma
Kaplan et al. (2005a) performed a similar study, compar- (16.5 ± 10.8%) than in glandular odontogenic cyst
ing p53, Ki-­67, and proliferating cell nuclear antigen (1.5 ± 2%), and suggested that this may be diagnostically
(PCNA) expression in a series of 11 glandular odontogenic helpful, especially in small biopsies.
cysts, 9 mucoepidermoid carcinomas (only 2 were central), More recently there has been a focus on rearrangements
and 15 radicular cysts showing mucous metaplasia. Only of the MAML2 gene as a potential molecular marker
the Ki-­67 results showed a difference between glandular to  ­distinguish glandular odontogenic cyst from central
odontogenic cyst (mean ± SD: 4.4 ± 4.7%) and the mucoepi- mucoepidermoid carcinoma. It is well established that
dermoid carcinomas (0.7 ± 1.6%). Interestingly, Ki-­67 mucoepidermoid carcinomas are characterised by a
expression in radicular cysts (3.7 ± 6.7%) was similar to t(11;19) gene rearrangement and associated CRTC1/3/
glandular odontogenic cyst and was lowest in the MAML2 fusion transcript. The MAML2 rearrangement can
carcinomas. be detected by reverse transcription polymerase chain
180 Glandular Odontogenic Cyst

reaction (RT-­PCR) or, more usually, by fluorescence in situ Nagasaki et al. (2018) reported a similar case where a typi-
hybridisation (FISH); a positive result is useful for the diag- cal glandular odontogenic cyst, showing 7 of the 10 criteria
nosis of mucoepidermoid carcinoma and may also assist in (Table  10.3), recurred after nine years. The primary lesion
tumour grading. Up to 80% of salivary and about 50% of was negative for the MAML2 rearrangement, but PCR
central mucoepidermoid carcinomas have been shown to showed a CRTC3/MAML2 fusion in the recurrent lesion,
be positive for the MAML2 rearrangement (Bell et al. 2016; confirming a mucoepidermoid carcinoma. Review of the
Cipriani et al. 2019; Reddy et al. 2019). histology illustrated in their paper shows that the recurrent
Bishop et  al. (2014) used analysis of the MAML2 lesion has features quite unlike glandular odontogenic cyst,
­rearrangement to help clarify the relationship between but is more typical of a multicystic and infiltrative mucoepi-
glandular odontogenic cyst and central mucoepidermoid dermoid carcinoma. The authors interpreted these findings
carcinoma. They used FISH to seek evidence of a MAML2 as confirming that mucoepidermoid carcinoma can arise
rearrangement in 21 glandular odontogenic cysts and 5 from a glandular odontogenic cyst. An alternative explana-
central mucoepidermoid carcinomas. All 5 central mucoep- tion, however, is that the primary lesion was also a mucoepi-
idermoid carcinomas were positive for the MAML2 rear- dermoid carcinoma, but indistinguishable from a glandular
rangement, whereas all 21 glandular odontogenic cysts odontogenic cyst, similar to the cases of Greer et al. (2018).
were negative. They noted that FISH demonstrated the Argyris et  al. (2015b) investigated the presence of
rearrangement in solid areas of mucoepidermoid carcino- MAML2 rearrangements in odontogenic cysts with evi-
mas, but also in cystic areas that histologically resembled dence of mucous cells. Their study was based on the prem-
glandular odontogenic cyst. The authors concluded that ise that many central mucoepidermoid carcinomas have
glandular odontogenic cyst is a separate entity to central been shown to arise in odontogenic cysts, and that the
mucoepidermoid carcinoma and should not be regarded as CRTC1/MAML2 fusion transcript may be an early molecu-
a ‘benign’ form or precursor of mucoepidermoid carci- lar event in the development of mucoepidermoid carcino-
noma. Barrett et  al. (2016) described three cystic lesions mas (Bell et  al.  2010). They hypothesised therefore that
where the differential diagnosis was between mucoepider- some cysts with mucous metaplasia may be precursor
moid carcinoma and glandular odontogenic cyst, and lesions and may harbour the MAML2 rearrangement. They
where the demonstration of MAML2 rearrangements was used FISH to determine the MAML2 status of eight denti-
able to resolve the dilemma. gerous and one radicular cyst that contained mucous cells,
Subsequently, three papers have suggested that MAML2 and found that the radicular cyst and four dentigerous
may not be so specific and may be found in glandular odon- cysts had the MAML2 rearrangement. The authors
togenic and other odontogenic cysts (Argyris et al. 2015b; described this as a pilot study and the findings have not yet
Greer et al. 2018; Nagasaki et al. 2018). Because of the topi- been independently repeated, but they are interesting, and
cal nature of this argument, these papers will be consid- they do raise the possibility that odontogenic cysts with
ered in some detail. mucous metaplasia may harbour early molecular events
Greer et al. (2018) deliberately selected a subset of 11 glan- that precede progression to mucoepidermoid carcinoma.
dular odontogenic cysts that had recurred (and that they Taken together, these studies provide some evidence to
labelled as ‘biologically aggressive’) for investigation of support the concept that glandular odontogenic cyst may
MAML2 rearrangements. All 11 cases were histologically be a precursor lesion to central mucoepidermoid carci-
diagnosed as glandular odontogenic cyst and showed at least noma, and that the possibility that glandular odontogenic
6 of the 10 features described by Fowler et  al. (2011) cyst is itself a benign neoplasm cannot yet be dismissed. A
(Table 10.3). One case showed the MAML2 rearrangement. further interpretation is that, in rare cases, the histological
The text of the published paper suggests that the rearrange- features of glandular odontogenic cyst may be indistin-
ment was found in the recurrent cyst and that the primary guishable from a low-­grade central mucoepidermoid carci-
lesion was not available. All the clinical and histological fea- noma. Nevertheless, the work of Bishop et al. (2014) still
tures suggest that this recurrent lesion was a typical glandu- provides strong evidence that glandular odontogenic cysts
lar odontogenic cyst. The only unique features were the do not have MAML2 rearrangements. In cases of doubt, the
finding of prominent goblet cells and cilia, almost involving finding of MAML2 should be regarded as diagnostic of cen-
the entire luminal surface. This case raises two possibilities: tral mucoepidermoid carcinoma. Conversely, although a
first, that glandular odontogenic cyst, even with typical negative result suggests glandular odontogenic cyst, it is
­histology, may show the MAML2 gene rearrangement; not diagnostic. Faced with a single lesion, pathologists
or  secondly, that a low-­grade (MAML2-­positive) central must consider all the clinical, radiological, and histological
mucoepidermoid carcinoma may be clinically and histologi- features in reaching a diagnosis (Boxes 10.1 and 10.2). The
cally indistinguishable from a glandular odontogenic cyst. differentiating features are summarised in Table 10.4.
­Treatment 181

­Treatment should be followed for at least three years and preferably as


long as seven years.
Glandular odontogenic cyst is usually treated conserva- Large unilocular or multilocular lesions should be biop-
tively, by enucleation with or without curettage. In their sied before treatment is planned. Kaplan et al. recommended
review of 169 cases, Chrcanovic and Gomez (2018b) that as unilocular lesions had a lower risk of recurrence than
reported that the majority had been treated by enucleation multilocular ones, even large lesions can be enucleated with
(66.4%) or curettage (22.1%) and only 10% had a marginal preservation of vital structures. Peripheral ostectomy in con-
or segmental resection. junction with enucleation can reduce recurrence and should
Kaplan et al. (2005b) carried out an extensive survey of be performed whenever possible. Marsupialisation is an
the treatment and follow-­up of a sample of 56 patients with option if the lesions approach vital structures. If marsupiali-
glandular odontogenic cysts, including 3 of their own sation is performed, curettage and peripheral ostectomy are
cases. The mandible was involved in 41 cases (73%) and the advised for second-­phase surgery.
maxilla in 15 (27%). Of the lesions, 11 were located in the For large multilocular lesions, major treatment modali-
anterior region, 9 in the posterior, and 36 overlapped both. ties are indicated. These include peripheral ostectomy,
The authors suggested that the protocol for treatment marginal resection, or partial jaw resection, depending on
should take into account that small unilocular lesions are the size of the lesion, integrity of the jaw borders, and prox-
usually enucleated before a definitive histological diagno- imity to vital structures. Kaplan et  al. suggest that if the
sis has been made, and, if they are completely enucleated, lesion lies close to the sinus, pterygoid, or nasal cavity, mar-
further surgery is not indicated because recurrence is supialisation with second-­phase enucleation after reduc-
unlikely. They recommend, however, that these patients tion of the lesion can be undertaken.
182

11

Calcifying Odontogenic Cyst

CHAPTER MENU
­Classification and Terminology of the Calcifying Odontogenic Cyst, 182
­Clinical Features, 185
●● Frequency, 185
●● Age, 186
●● Sex, 186
●● Site, 186
●● Clinical Presentation,  187
–– Peripheral (Extraosseous) Calcifying Odontogenic Cyst,  188
­Radiological Features, 188
●● Radiological Differential Diagnosis,  190
­Pathogenesis, 190
●● Ghost Cells and the β-­Catenin Gene (CTNNB1), 191
­Histopathology, 193
●● Histological Differential Diagnosis,  199
●● Ghost Cell Odontogenic Carcinoma and Malignant Progression of Calcifying Odontogenic Cyst,  199
­Treatment, 200

Calcifying odontogenic cyst is an odontogenic cyst, lined considerable debate in the literature about the classification
by an ameloblastoma-­like epithelium, and characterised by and terminology of the odontogenic ghost cell lesions.
focal accumulations of ghost cells that may calcify. It was
first described and named by Gorlin et  al. (1962,  1964),
who were impressed by the presence of the ghost cells ­ lassification and Terminology of the
C
and the cyst’s histological resemblance to the cutaneous cal- Calcifying Odontogenic Cyst
cifying epithelioma of Malherbe (pilomatrixoma). The
lesions they described were simple cysts and, because When Gorlin et  al. (1962,  1964) first described the lesion,
­dystrophic calcification in the ghost cells was prominent, they recognised its characteristic features and named it the
they named them calcifying odontogenic cyst. Since this early calcifying odontogenic cyst. It should be noted that the term
description, however, it has become appreciated that lesions ‘calcifying’ was used primarily because of the prominent
with similar features can show considerable histomorpho- dystrophic calcification of the ghost cells, although dentine-­
logical variation, including solid lesions that may be neo- like material may also be seen. The lesion became well rec-
plastic. It is now recognised that calcifying odontogenic cyst ognised and was included in the 1971  World Health
is one of a family of odontogenic ghost cell lesions that Organization (WHO) classification of odontogenic tumours
includes dentinogenic ghost cell tumour and ghost cell odonto- and was defined as a ‘non-­neoplastic cystic lesion’ (Pindborg
genic carcinoma (Ledesma-­Montes et  al. 2008; Wright and and Kramer  1971). For reasons that are uncertain, it was
Soluk ­Tekkeşin 2022; WHO 2022a). Nevertheless, over the listed under the benign ‘neoplasms and other tumours’
six decades since Gorlin et al.’s description, there has been rather than under the developmental odontogenic cysts.

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
­Classification and Terminology of the Calcifying Odontogenic Cys 183

This categorisation and a number of misunderstandings and a solid neoplasm. It is a source of some bemusement
(discussed below) seem to have started decades of confusion that for many years the neoplastic lesion was often referred
about the nature of this lesion. It does seem that several to as the ‘solid calcifying odontogenic cyst’. This first clas-
authors have noted that the cyst is classified under ‘tumours’ sification is summarised in Table 11.1.
and have considered this to be synonymous with a neo- In 1991, two groups reviewed the classification of the
plasm, often ignoring its designation as ‘non-­neoplastic’. ghost cell lesions and agreed with the dualistic concept that
Subsequent to the 1971 classification, it became apparent they fell into a non-­neoplastic cystic group and a solid neo-
that the nature of the lesion was more varied and that a num- plasm (Buchner 1991; Hong et al. 1991). Both continued to
ber of odontogenic lesions may contain ghost cells. This recognise that the cyst had a number of variants, including
included a ‘solid variant’ of the calcifying odontogenic  cyst an association with odontomas and proliferative types.
that was variously called the ‘calcifying ghost cell odontogenic They also drew attention to the fact that peripheral lesions
tumour’ (Fejerskov and Krogh 1972), the  ‘cystic calcifying may be encountered. Buchner (1991) furthermore included
odontogenic tumor’ (Freedman et al. 1975), and the ‘epithelial a malignant variant in his classification – the lesion that is
odontogenic ghost cell tumor’ (Ellis and Shmookler 1986). now known as ghost cell odontogenic carcinoma.
Gorlin and his colleagues revisited the nature of the The next milestone in the story of terminology and classifi-
lesion in 1981 (Prætorius et  al.  1981) and identified two cation came with the publication of the second edition of the
types. Type 1  was a simple unicystic lesion with the fea- WHO classification of odontogenic tumours (Kramer
tures first described by Gorlin et al. in 1962. Type 2 was a et al. 1992). The terminology used and the subsequent mis-
solid lesion with a growth pattern like ameloblastoma, but understanding led to a further period of confusion. In the
with ghost cells and prominent formation of dentinoid 1992 classification, the cyst was still (incorrectly in our view)
material. They regarded this solid lesion as a neoplasm and included under the benign odontogenic ‘neoplasms and
proposed the name dentinogenic ghost cell tumour. For the other tumours’, but the authors seemed uncertain about its
cystic lesion they retained the name calcifying odontogenic nature – they used an almost identical definition to 1971, but
cyst, but also identified three variants (Table 11.1). Type 1A removed ‘non-­neoplastic’ and defined it as a ‘cystic lesion’.
was a simple calcifying odontogenic cyst as described pre- This implied that the calcifying odontogenic cyst should be
viously. Type 1B showed features of a typical calcifying defined as a neoplasm, although in the text they stated clearly
odontogenic cyst, but with epithelial islands in the wall that the cyst was ‘non-­neoplastic’, but that a more solid vari-
and with an associated odontoma. Type 1C was similar ant was neoplastic, and used the term ‘dentinogenic ghost
to  1A, but with ameloblastoma-­like proliferations in the cell tumour’ that had been suggested by Prætorius et al. (1981).
lumen and in the wall. This became the first attempt to In 1998, Toida attempted a further clarification of the
subdivide the ghost cell lesion into categories and became nature of the calcifying odontogenic cyst and its classifica-
known as the ‘dualistic’ concept: that the calcifying odon- tion. He reviewed the previous classifications described
togenic cyst represented two entities, a non-­neoplastic cyst above and discussed the two concepts of a ‘monoistic’ clas-
sification, which regarded all calcifying odontogenic cysts
Table 11.1  The first detailed classification of the odontogenic as neoplastic, and a ‘dualistic’ classification, which recog-
ghost cell lesions. nised a (non-­neoplastic) cyst and a neoplasm. Toida (1998)
interpreted the 1992 WHO classification as being ‘monois-
Calcifying tic’ and as meaning that all calcifying odontogenic cysts
Type 1 odontogenic cyst Key features
‘both cystic and solid are neoplastic in nature’. To resolve
1A Simple cyst Ameloblastoma-­like lining. the issue, he proposed a new classification that he called
Ghost cells. Calcifications ‘simple and basic’, but it included three types and at least
1B Simple cyst, with As 1A, but with multiple nine variants, compared to two types and three variants
odontoma forming in tooth-­like structures proposed by Prætorius et al. (1981) (Table 11.1). Toida also
the wall preferred the term ‘calcifying ghost cell odontogenic
1C Simple cyst, but with As 1A, but with luminal tumour’ and proposed that it included cystic and solid neo-
ameloblastoma-­like and mural proliferations plastic variants. If a non-­neoplastic cyst existed, he favoured
proliferations in the wall and prominent dentinoid
and lumen
the term ‘calcifying ghost cell odontogenic cyst’.
Subsequent authors have continued to interpret the
Type 2 Dentinogenic ghost Solid neoplasm with
cell tumour ameloblastoma-­like growth 1992  WHO classification as indicating that the calcifying
pattern (may be odontogenic cyst may be a neoplasm with a cystic and
multicystic). Prominent solid  variant, but have still continued to describe a non-­
ghost cells and dentinoid neoplastic (simple cystic) variant under a number of differ-
Source: Based on Prætorius et al. (1981). ent names. Li and Yu (2003) and Reichart and Philipsen
184 Calcifying Odontogenic Cyst

(2004) proposed similar classifications that included a non-­ cystic lesions should be classified as a developmental cyst,
neoplastic (simple) cyst, a neoplasm that may be solid or which arises alone or in association with odontomas
cystic, and a malignant neoplasm. They also recognised (Ledesma-­Montes et al. 2008).
subtypes that may be associated with odontomas, and Thus, in 2017, the fourth edition of the WHO classifi-
peripheral variants. Reichart and Philipsen (2004) pre- cation (El-­Naggar et al. 2017) reverted to the original ter-
ferred the terms calcifying ghost cell odontogenic cyst and minology and classified the odontogenic ghost cell
calcifying ghost cell odontogenic tumour. lesions as calcifying odontogenic cyst, dentinogenic ghost
For reasons that are still not clear, the third edition of the cell tumour, and ghost cell odontogenic carcinoma.
WHO classification (Barnes et al. 2005) completely dispensed Furthermore, the 2017 classification included the odonto-
with the term ‘calcifying odontogenic cyst’ and with the dual- genic cysts and the calcifying odontogenic cyst was there-
istic concept, and redefined the ghost cell lesions into three fore classified as a developmental odontogenic cyst
types, all of which were neoplasms: the calcifying cystic odon- (Speight et al. 2017a). The latest edition of the WHO clas-
togenic tumour (CCOT), the dentinogenic ghost cell tumour, sification also includes the odontogenic cysts and has
and the ghost cell odontogenic carcinoma. The 2005 classifica- retained these definitions and terminology (WHO 2022a;
tion did not include a classification of cysts, and so from this Wright and Soluk ­Tekkeşin 2022).
time onwards the calcifying odontogenic cyst became known It can be seen from this discussion that there has been
as the CCOT and was classified as a neoplasm. considerable debate and confusion over the terminology
A further reappraisal was undertaken in 2008 by a large and classification of the ghost cell lesions over a period of
multicentre group that included the authors of the WHO more than five decades. We have been able to identify at
classification (Ledesma-­Montes et  al.  2008). They under- least 11 different proposed classifications and numerous
took a detailed review of 122 ghost cell lesions and their different names for the various lesions, often with complex,
terminology. They categorised the lesions into three groups confusing, and similar acronyms, and sometimes with
using the 2005 WHO terminology, and divided the calcify- excessive subclassifications. Details of some of the classifi-
ing cystic odontogenic tumour into four types: cations have been reviewed and published by Toida (1998)
and Robinson and Vincent (2012), and also by Reichart and
●● CCOT Type 1: simple cystic
Philipsen (2004), who proposed a further unified classifica-
●● CCOT Type 2: CCOT associated with an odontoma
tion. Ide and colleagues have also published one of their
●● CCOT Type 3: with ameloblastomatous-­like proliferation
interesting historical reviews (Ide et al. 2012) cataloguing
●● CCOT Type 4: associated with other benign odontogenic
the early history of the odontogenic ghost cell lesions.
tumours other than odontoma
Although Gorlin et  al. (1962,  1964) are credited with the
They found that 87% of their cases were simple cysts, first definitive description of the calcifying odontogenic
either alone (Type 1: 65%) or associated with odontomas cyst, Ide et al. (2012) were able to identify 32 cases of jaw
(Type 2: 22%). Of the 113  lesions in this category, only 6 lesions that would meet the criteria for calcifying odonto-
(5.3%) recurred. Only 3 cases showed ameloblastomatous genic cyst or dentinogenic ghost cell tumour published
proliferations and only 4 (3.3%) were associated with other between 1917 and 1962, and credit Rywkind, a Russian
odontogenic lesions. There were 6 cases (5%) that were solid author, with the first description in 1931. They note that
and could be regarded as true neoplastic dentinogenic ghost ghost cell lesions regularly featured in Thoma’s textbooks
cell tumours. Although the authors used the term CCOT, between 1917 and 1960, but were categorised as odonto-
they seemed uncertain about its nature. They defined the mas, as variants of ameloblastoma, or as variants of other
CCOT as a ‘cystic lesion’ or ‘cystic tumour’, whereas they tumours or cysts. Ide et  al. (2012) also identified 40 case
clearly defined the solid dentinogenic ghost cell tumour as reports of jaw lesion that included ghost cells going as far
a ‘neoplastic tumour’ or ‘solid neoplastic growth’. back as 1838.
These findings agreed with the previous studies of A feature of all the classifications is that a proportion
Buchner (1991) and Hong et al. (1991), who also showed of  calcifying odontogenic cysts are associated with other
that simple cystic lesions rarely recur and have a com- odontogenic lesions, most often odontomas. As will be
pletely benign course. As discussed above, most authors up noted from other chapters in this book, it is not unusual
to 2005 classified this lesion (the CCOT) as a simple cyst for  odontogenic cysts to occasionally occur with other
and only regarded the solid lesion as a true neoplasm. odontogenic lesions, either as hybrid lesions or due to
There seems, therefore, to be good evidence that all the ­co-­development of two lesions. With regard to calcifying
early papers were correct (Pindborg and Kramer  1971; odontogenic cyst this will be discussed in later sections, but
Prætorius et al. 1981; Buchner 1991; Hong et al. 1991; Li associations with ameloblastomatous proliferations and
and Yu 2003; Reichart and Philipsen 2004) and that simple occasional odontogenic tumours are probably unusual
­Clinical Feature 185

occurrences that should be regarded as oddities that do that itemise each histological variant. Each of the three
not warrant a separate categorisation (e.g. CCOT Types 3 types may, on occasion, present as peripheral or extraosse-
and 4). However, in relation to the association between cal- ous lesions. This classification and terminology will be fol-
cifying odontogenic cyst and odontoma, this appears to be lowed in this chapter. Only the calcifying odontogenic cyst
a common occurrence seen in up to 25% of cases. So dis- will be considered in detail.
tinctive is this association that some have suggested it
should be regarded as a separate entity and have proposed
the term odontocalcifying odontogenic cyst (Hirshberg
et al. 1994). This will be discussed in a later section, but it
­Clinical Features
is such a frequent occurrence that it may be justified to
There are almost 250 reports of calcifying odontogenic cyst
classify calcifying odontogenic cyst associated with odon-
in the literature, most of which are single reports of unusual
toma as a distinctive clinicopathological variant.
or interesting lesions. There have also been a number of case
Based on the latest WHO classification (WHO  2022a)
series, but many early reports include ‘cystic’ and ‘solid’
and from a consideration of the issues discussed above, a
lesions, making it difficult to interpret data specifically for
simpler classification of odontogenic ghost cell lesions is
calcifying odontogenic cyst. There have been two substan-
proposed (Table 11.2) that removes complex subcategories
tial multicentre case series, of 113 cases by Ledesma-­Montes
Table 11.2  A simple classification of odontogenic ghost cell et  al. (2008) and 268 cases by de Arruda et  al. (2018b).
lesions that will be followed in this chapter. Buchner et al. (1991) reported a detailed review of 215 cases
and de Arruda et al. (2018a) undertook a systematic review
Key features of 367 cases reported up to 2018. These reviews and selected
case series that have provided clinical data specifically for
Calcifying calcifying odontogenic cyst are summarised in Table 11.3.
odontogenic cyst
●● Simple cyst Typical cystic lesion, ameloblastoma-­like
lining with ghost cells. Includes occasional
Frequency
variants with proliferating epithelium
●● Cyst associated Cyst associated with an odontoma in the Calcifying odontogenic cysts are rare and constitute less
with odontoma wall than 1% of odontogenic cysts. Over a 46-­year period
Dentinogenic Solid lesion with ameloblastomatous 1958–2004, only 28 examples were recorded in the archives
ghost cell tumour epithelium, ghost cells, and dentinoid of the University of the Witwatersrand, representing 0.8%
Ghost cell Malignant lesion, ameloblastoma-­like of 3498  jaw cysts (Table  1.1). In the large series from
odontogenic epithelium, ghost cells, and dentinoid Sheffield, Jones et al. (2006) found 21 cases over a 30-­year
carcinoma
period, representing 0.3% of odontogenic cysts (Table 1.2).

Table 11.3  Age, sex, and site distribution and association with odontomas from selected series of calcifying odontogenic cyst, and
from the reviews of Based on Buchner (1991) and de Arruda et al. (2018a).

References Country n Mean age Peak decade Male (%) Mandible (%) Associated with odontoma (%)

Hong et al. (1991) USA 79 33.0 2nd 60.3 55.2 20.3


Yoshida et al. (2001) Japan 16 23.0 2nd 56.3 31.3 75.0
Li and Yu (2003) China 16 31.0 2nd 56.3 31.3 31.3
Ledesma-­Montes et al. (2008) Multicentre 113 26.0 2nd 55.5 44.8 23.9
Gomes da Silva et al. (2014) Brazil 21 27.1 NR 52.5 38.1 19.0
Irani and Foroughi (2017) Iran 43 26.2 2nd 55.8 62.8 37.2
Rosa et al. (2019) Brazil 30 37.1 2nd 50.0 50.0 26.7
Nel et al. (2021) S Africa 27 29.0 2nd 55.5 59.3 22.2
Buchner (1991) Review 215a 30.3 2nd 48.8 48.4 26.5
de Arruda et al. (2018a) SR 367a 30.7 2nd 51.8 51.7 23.4

NR, not reported; SR, systematic review.


a
 Total cases in review, but n for each parameter varies depending on number of reports.
186 Calcifying Odontogenic Cyst

Five of the reports summarised in Table 1.3 included calci- Buchner (1991) showed that lesions associated with
fying odontogenic cyst and reported frequencies of 1.0% odontomas occurred in a younger age group than cystic
(Grossmann et al. 2007), 0.7% (Soluk Tekkeşin et al. 2012b), lesions alone. The mean age of patients with calcifying
0.4% (Tamiolakis et al. 2019), 0.8% (Bhat et al. 2019), and odontogenic cyst was 34 years, but patients with odontoma-­
1.48% (Kammer et al. 2020). associated lesions had a mean age of 17 years. Of 52
With regard to the subtypes of calcifying odontogenic odontoma-­associated lesions, 92.3% were below the age of
cyst, simple cystic lesions and cysts associated with odon- 30 years, compared to only 53.5% of conventional cystic
tomas represent about 90% of all cases (Ledesma-­Montes lesions. Hong et al. (1991) found a similar age difference.
et al. 2008). Of these, simple cystic calcifying odontogenic In their series the average age of all calcifying odontogenic
cysts were the most common, constituting 65% of the total, cyst patients was 33 years, but patients with odontoma-­
while lesions associated with odontoma represented 22%. associated lesions had an average of 14.7 years.
Overall, about 25% of cysts are associated with odontomas
(Buchner 1991; de Arruda et al. 2018a; Table 11.3).
Sex
All studies show an almost equal sex distribution of calcify-
Age
ing odontogenic cyst, with only a small male predilection
The two largest reviews of calcifying odontogenic cyst have in some series (Table 11.3). In the two largest reviews, there
shown an average age of 30 years (Buchner 1991; de Arruda was no significance difference between males and females
et al. 2018a), with a peak in the second decade (Table 11.3). (Buchner  1991; de Arruda et  al.  2018a). Nel et  al. (2021)
Figure 11.1 shows the age distribution of 708 cases. These found an equal sex distribution for simple cystic calcifying
comprise 141 cases reported in the previous edition of this odontogenic cysts (n  =  20), but a male predilection for
book (Prætorius et al. 1981; Shamaskin et al. 1989; Yoshida other types, with 71.4% males (n = 7). Furthermore, 4 of
et  al.  2001; Moleri et  al.  2002; Fregnani et  al.  2003; Li the 6 cases associated with odontomas were found in males.
and  Yu  2003) and a further 567 cases accumulated from
Buchner (1991), Ledesma-­Montes et  al. (2008), and de
Site
Arruda et  al. (2018b). The lesion occurs over a wide age
range, with the youngest recorded patient at 1 year old and Most studies have shown a slight predilection for the max-
the oldest 82 years. There is however a significant peak in illa (Table 11.3), with as few as 31% arising in the mandi-
the second decade, and 60% of all cases were encountered ble (Yoshida et  al.  2001; Li and Yu  2003). The larger
before the age of 30 years. reviews however show an almost equal distribution
between mandible and maxilla (Buchner 1991; de Arruda

300 Figure 11.1  Age distribution of 708


patients with calcifying odontogenic
263 cysts (see text for details).
250

200
No. of cases

150

119

100
85

57 57
52
50 37
33

5 0
0
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89 90-99
Age
­Clinical Feature 187

Figure 11.2  Site distribution of calcifying


odontogenic cyst. Data are the percentage of Maxilla
lesions in each sextant of the jaws. Source: Data
from Buchner (1991) and Ledesma-­Montes et al.
(2008) (n = 255). Mandible

Percent of cases
37.6

13.3
3.1

10.2 13.7
22.0

Incisor/Canine Premolar Molar

et al. 2018a). Figure 11.2 illustrates the site distribution of


Box 11.1  Calcifying Odontogenic Cyst: Clinical
255 cases where the site has been recorded by sextant: 179
and Radiological Features
cases from Buchner (1991) and 76 from Ledesma-­Montes
et al. (2008). Of these, 54% were located in the maxilla, and ●● They are rare – less than 1% of odontogenic cysts
the majority of lesions (60%) were located in the anterior ●● Average age 30 years. Peak in the second decade
regions of the jaws, with the single most common site the ●● Occur equally in males and females
maxillary incisor/canine region (96 cases: 37.6%). The pos- ●● Slightly more common in the maxilla
terior mandible is the second most frequent site, but ●● Single most common site (37%) is the maxillary inci-
lesions in the posterior maxilla are rare. In the series of sor/canine region
Nel et al. (2021), simple cystic lesions showed an equal dis- ●● Usually symptomless, but 80% show swelling/bony
tribution between mandible and maxilla, but 5 of 6 lesions expansion
(83.3%) associated with odontomas were found in the ●● Well-­defined radiolucency with corticated margin
mandible. ●● Most are unilocular. Up to 18% may be multilocular
Multiple or bilateral calcifying odontogenic cysts appear ●● 60% contain areas of calcification
to be very rare and none of the series summarised in ●● 25% are associated with an odontoma
Table  11.3 recorded multiple lesions. There is only one ●● Average size is 30 mm. Only about 25% are greater
fully documented report of bilateral calcifying odontogenic than 40 mm
cysts. Gadipelly et al. (2015) reported bilateral lesions in an ●● Recurrence is rare. Overall recurrence rate is less than 4%
8-­year-­old child, arising in the posterior mandible. Both
lesions occurred in association with unerupted second
molars, and extended from the canine region to the ante- Over 80% of lesions cause bony expansion (de Arruda
rior ramus. et al. 2018a), so the swelling is often bony hard and is usu-
ally on the buccal or labial aspect of the jaws. Occasional
lesions may perforate the cortical plate and extend into the
Clinical Presentation
soft tissues.
The majority of lesions present as a painless swelling. In The calcifying odontogenic cyst rarely recurs. Buchner
their systematic review, de Arruda et al. (2018a) identified (1991) reviewed 215 cases and found only 8 cases that had
249 cases in which the clinical signs and symptoms had recurred (3.7%). Of their 113 cases, Ledesma-­Montes et al.
been reported, and found that 76.7% (191) presented with (2008) recorded only 5 recurrences of intraosseous lesions
swelling alone and a further 20 (8.0%) had swelling and (4.6%), Hong et  al. (1991) found only 1 recurrence in 79
pain. Only 7 (2.9%) reported pain alone, and 20 cases (8.0%) cases (1.3%), and de Arruda et  al. (2018b) found only 10
were symptomless incidental findings. Occasional cases (3.7%) recurrent cases out of 268. In their systematic review
presented with unerupted teeth (3.2%) or with tooth dis- they identified 202 cases where follow-­up had been
placement (1 case; 0.4%), and up to 20% may be associated recorded and found that only 8 (4.0%) had recurred (de
with tooth mobility (Nel et al. 2021). Arruda et al. 2018a).
188 Calcifying Odontogenic Cyst

Peripheral (Extraosseous) Calcifying margin (Figure 11.3). Up to 60% of cases contain variable


Odontogenic Cyst amounts of radiopaque material (Buchner 1991; de Arruda
Peripheral lesions of calcifying odontogenic cyst are well et al. 2018a; Nel et al. 2021), which vary from occasional
documented, but only represent a small proportion of the small flecks to dense radiopaque masses. These calcifica-
total reported cases. In their large series Ledesma-­Montes tions may represent foci of dystrophic calcification in the
et al. (2008) reported that 5 of their 113 cases (4.4%) were ghost cells, accumulations of dentinoid, or may have the
peripheral, and de Arruda et al. (2018b) recorded 7 out of density of enamel and dentine if the cyst is associated with
268 cases (2.6%). Hong et  al. (1991), in their series of 92 an odontoma (Figures  11.4 and  11.5). Occasional cases
‘calcifying odontogenic cysts’, found 8 cases that were may be multilocular. Buchner (1991) found that 12  cases
peripheral, but all were of the ‘solid’ variety. Buchner et al. (7.1%) were multilocular, but in their systematic review de
(1991) reviewed 45 cases of peripheral calcifying odonto- Arruda et  al. (2018a) found that 21 of 116 cases (18.1%)
genic cyst, but 36% were described as ‘solid’ and repre- where locularity had been described were multilocular. Nel
sented dentinogenic ghost cell tumours. Chrcanovic and et al. (2021) reviewed the radiological features of 27 cases
Gomez (2016) undertook a systematic review of peripheral and found that 19 (70.4%) were unilocular and 6 (22.2%)
ghost cell lesions and found reports of 55 calcifying odon- were multilocular; 2 cases (7.4%) were unilocular, but with
togenic cysts and 50 dentinogenic ghost cell tumours. a scalloped margin.
The age data in Figure 11.1 include a small number of Buchner (1991) reported 68 cases (32.2%) as being associ-
peripheral lesions – for example, Ledesma-­Montes et al. ated with unerupted teeth, but did not describe the rela-
(2008) and de Arruda et  al. (2018b) included 5 and 7 tionship in detail, so how many may be seen in a radiological
cases, respectively – but these are unlikely to affect the dentigerous relationship is uncertain. The most commonly
overall distribution. There is however some evidence involved tooth in both jaws was a canine, but in the mandi-
that peripheral lesions may arise in an older age group ble second and third molars were also involved. Review of
than intraosseous calcifying odontogenic cysts. Kaugars case reports suggests that lesions do arise in a dentigerous
et al. (1989a) reviewed 29 extraosseous cases, and found relationship, but displacement of unerupted teeth is also a
that 16 patients were in their sixth decade or older. In frequent feature (Figure  11.5). De Arruda et  al. (2018a)
their study of 45 peripheral lesions, Buchner et al. (1991) reported that 57% of lesions were associated with unerupted
observed a bimodal age distribution with a peak in the teeth and 39.5% caused tooth displacement or divergence
second decade, but a larger peak (24.4% of cases) in the of the roots (e.g. see Figures 11.4 and 11.5).
sixth decade. In a more recent systematic review, Iida et al. (2006a) gave a detailed description of the radio-
Chrcanovic and Gomez (2016) identified 55 peripheral logical features of 11 cases and showed that 5 enveloped
calcifying odontogenic cysts and found an average age of the crowns of unerupted teeth and that all 5 arose in the
41.8 years.
The series of extraosseous cases reported by Kaugars
et al. (1989a) and Buchner et al. (1991), and the systematic
review of Chrcanovic and Gomez (2016), all showed an
equal sex distribution.
When they do arise in an extraosseous location, calcify-
ing odontogenic cysts appear to arise slightly more often in
the mandible (59.6%; Chrcanovic and Gomez  2016), but
have a predilection for the incisor/canine region of both
jaws (Buchner et  al.  1991; Chrcanovic and Gomez 2016).
They tend to be pink to red, smooth, circumscribed ele-
vated masses on the gingivae or alveolar mucosa. They
measure an average diameter of 11 mm (range: 5–30 mm;
Chrcanovic and Gomez 2016), and there is only one report
of a lesion exceeding 30 mm (Buchner et al. 1991).

­Radiological Features Figure 11.3  Calcifying odontogenic cyst. The lesion is well


demarcated and corticated, with a slightly loculated outline.
Note resorption of the roots of the second pre molar first molar.
The calcifying odontogenic cyst most often presents as Source: Prof Paul Speight (Previously published: El-Naggar AK
a  unilocular radiolucency with a well-­defined corticated 2017 / Courtesy of IARC).
­Radiological Feature 189

about 40% of cases (de Arruda et al. 2018a; Figure 11.3). In


the series of Nel et al. (2021), 23 cases were found in den-
tate areas of the jaws and of these, 19 (82.6%) showed root
resorption, 14 (60.9%) were associated with tooth displace-
ment, and 8 (34.8%) caused tooth impaction.
Many lesions are spherical or rounded in shape and
cause expansion of the cortical plates, usually towards the
buccal or labial aspect. De Arruda et al. (2018a) found that
81.9% of cases showed bone expansion and 61.6% had per-
forated the cortical plate. Computed tomography (CT) is
particularly useful for demonstrating bucco-­lingual corti-
cal expansion and radiopacities (Figure  11.6). Uchiyama
et  al. (2012) analysed nine cases by conventional radio-
graphs and by CT, and found that CT showed two cases
with radiopacities and three cases with odontomas that
were not apparent on plane radiographs. They also noted
that when present (in eight cases), radiopaque calcifica-
tions tended to be orientated around the margins of the
lesions, confirming a feature described previously as a
‘peripheral band of intracystic calcification’ that was
regarded as a characteristic finding of calcifying odonto-
genic cyst on CT (Rushton and Horner 1997).
With regard to the size of calcifying odontogenic cysts,
Buchner (1991) found data for 58 cases and showed a range
from 5 to 120 mm. However, the average size was 33 mm
Figure 11.4  Calcifying odontogenic cyst of the maxilla. and 76% of cases were less than 40 mm. De Arruda et al.
Radiograph shows a well-­demarcated margin and calcifications
(2018a) found data for 109 cases and showed an average
suggestive of an odontoma. The adjacent teeth are displaced.
Source: Courtesy of Prof. J.E. Seeliger. size of 29.5 mm (range 2–90 mm).
Radiology does not have a role in the diagnosis of
posterior regions, most often (3 cases) associated with peripheral calcifying odontogenic cysts, except to exclude
mandibular second or third molars. They also found that 5 peripheral extension of an intraosseous lesion. However,
cases (45.5%) caused tooth displacement, and 4 (36.4%) peripheral lesions may show superficial bony erosion and
showed evidence of root resorption. Resorption of the roots saucerisation of the cortical plate in about 17% of cases
of adjacent teeth is a frequent finding, and may be seen in (Chrcanovic and Gomez 2016).

Figure 11.5  Radiograph of a large


calcifying odontogenic cyst with
well-­demarcated margins extending from
the right canine to the left premolar
region of the mandible. Numerous
calcifications are present, some
suggestive of small denticles or a
compound odontoma. Note displacement
of the developing canine to the lower
border of the mandible.
190 Calcifying Odontogenic Cyst

Figure 11.6  Computed tomography (CT) and a three-­dimensional CT reconstruction of a calcifying odontogenic cyst in the mandible.
The extent of the lesion is well demonstrated by CT, which shows the prominent buccal expansion and evidence of perforation. This
case is in a 4-­year-­old child.

Radiological Differential Diagnosis reduced enamel epithelium. Gorlin et  al. (1962) and
Prætorius et  al. (1981) in their early descriptions showed
The radiological features of calcifying odontogenic cyst are
lesions that enveloped the crown of unerupted teeth and
not diagnostic and overall the lesion is so rare that an initial
where the lining was in continuity with the ‘epithelium of
diagnosis of calcifying odontogenic cyst is unlikely to be
a developing or unerupted tooth’. They speculated that the
made on clinical and radiological features alone. The charac-
lesion thus arose from reduced enamel epithelium or from
teristic feature of a unilocular radiolucency containing calci-
rest cells of dental lamina in the dental follicle, bone, or
fications may be seen in a number of odontogenic tumours,
gingival tissues. This issue has never been resolved, but ori-
including adenomatoid odontogenic tumour, calcifying epi-
gins from different sources of epithelium are not mutually
thelial odontogenic tumour, and dentinogenic ghost cell
exclusive. What remains unknown are the factors that ini-
tumour. Rushton and Horner (1997), however, showed that a
tiate cyst formation and growth.
peripheral band of calcification at the margins of the radiolu-
Previously we have discussed the long-­standing debate
cency was characteristic of calcifying odontogenic cyst on CT
regarding the terminology and classification of the calcify-
examination, and this was confirmed by Uchiyama et  al.
ing odontogenic cyst. This reflects the uncertainty regard-
(2012). For lesions that are heavily calcified, the differential
ing the nature of the lesion, and although the calcifying
diagnosis will include a conventional odontoma.
odontogenic cyst has found a place in the classification
When calcifying odontogenic cyst presents as a simple
under the developmental cysts (WHO  2022a), this does
radiolucency without calcifications, the differential diag-
not  exclude the possibility that it is a neoplasm or that a
nosis may include any of the cystic jaw lesions, and a final
neoplastic variant (e.g. the ‘calcifying cystic odontogenic
diagnosis may only be possible on histological examina-
tumour’) exists (see discussion below on β-­catenin).
tion. However, lesions do have characteristic clinical and
Elucidation of the pathogenesis is further complicated
radiological features in terms of age, sex, and site that can
by the fact that the epithelial lining of a calcifying odonto-
assist in making a differential diagnosis (Box 11.1). Buchner
genic cyst appears to have the ability to induce the forma-
(1991) reported that the most frequent pre-­biopsy diag-
tion of dental hard tissues in the adjacent connective tissue
nosis was dentigerous cyst, followed by radicular cyst,
wall; and that other odontogenic tumours  – especially
ameloblastoma, and residual cyst. Occasionally lesions are
odontomas, but also ameloblastoma, ameloblastic fibroma,
suspected to be odontogenic keratocysts or myxomas, espe-
and adenomatoid odontogenic tumour – may be associ-
cially if they are lobulated or multilocular.
ated with it (Prætorius et  al.  1981; Hirshberg et  al.  1994;
Lin et  al.  2004; Ledesma-­Montes et  al.  2008; de Arruda
­Pathogenesis et al. 2018a).
The apparent ability of the cyst lining to induce dentine
The literature is unanimous that the calcifying odonto- formation and the association with odontomas have led to
genic cyst is of odontogenic origin and probably arises suggestions that the calcifying odontogenic cyst is itself a
from the rest cells of the dental lamina, or possibly from hamartoma and that lesions will progressively undergo
­Pathogenesi 191

formation of dental hard tissues to develop into an odon- (2004) reported three cases of ameloblastic fibroma with
toma. This concept has been generally discounted on the calcifying odontogenic cyst and presented details of three
basis that calcifying odontogenic cysts associated with previous cases. Five of these six presented at a site and an
odontoma arise in a younger age group than calcifying age that would be consistent with the lesion being an early
odontogenic cyst alone, and there is no evidence of pro- stage of odontoma formation within the wall of the cyst.
gression over time, since hard tissue is less likely to be seen A further area of confusion is the association of
in older patients. Hirshberg et  al. (1994) discussed this ­ameloblastoma and calcifying odontogenic cyst.
issue at length and suggested that the cyst associated with Ameloblastomatous proliferations of the cyst lining should
an odontoma is so distinctive that it should be regarded as not be confused with, or misinterpreted as, an ameloblas-
a separate entity. They proposed the name odontocalcifying toma forming in the cyst wall. Although this is possible, it
odontogenic cyst and suggested that it should be classified does seem to be an extremely rare event, if it has occurred at
as a benign mixed odontogenic tumour, although they all (Hong et al. 1991; Ledesma-­Montes et al. 2008). Nel et al.
were undecided as to whether it is a neoplasm or a hamar- (2021), however, in their series of 27 cases, had one with
toma. Alternatively, Hong et al. (1991) felt that the calci- ameloblastomatous proliferations and found that this case
fying odontogenic cyst may develop secondary to the showed aggressive behaviour, being highly expansive and
odontoma, similar to development of a dentigerous cyst. featuring destruction of cortical bone. This does suggest that
This would explain the association, and the younger age such lesions are possible, but should be regarded as a hybrid
group affected by this lesion. of ­calcifying odontogenic cyst and ameloblastoma, and that
Others have proposed that odontoma and ameloblasto- management should be for the most aggressive component.
matous proliferations in the wall of the cyst – correspond-
ing to Types 1B and 1C, Table 11.1; or CCOT types 2 and 3
Ghost Cells and the β-­Catenin Gene (CTNNB1)
described by Ledesma-­Montes et  al. (2008)  – are integral
components of the lesion that may arise in a small number A characteristic feature of the calcifying odontogenic cyst
of cases and are due to inductive change or to excessive is the presence of ghost cells (sometimes referred to as
proliferation of the lining. Odontoma formation is seen in shadow cells). These are not specific to calcifying odonto-
about 25% of cases and may be regarded as a clinicopatho- genic cyst and are seen in a number lesions that recent evi-
logical variant, but luminal or mural ameloblastomatous dence suggests may be related by a unifying mechanism
proliferation of the cyst lining, although well described, is involving  mutations in the β-­catenin gene, CTNNB1
seen in less than 3.0% of cases (Ledesma-­Montes et al. 2008; (Gomes et al. 2019). The lesions are odontogenic ghost cell
Nel et  al.  2021). Hong et  al. (1991) suggested that those lesions, the cutaneous pilomatrixoma (calcifying epitheli-
with mural proliferations should be diagnosed as a sepa- oma of Malherbe), and craniopharyngioma (specifically
rate ‘ameloblastomatous’ variant, but Ledesma-­Montes the adamantinomatous variant), all of which are character-
et  al. (2008) pointed out that these lesions behave as a ised by accumulations of ghost cells.
benign cyst and do not show any of the features of amelo- In 2003, two concurrent publications reported β-­catenin
blastoma, and should be regarded as histological variants expression or mutations in calcifying odontogenic cysts.
with proliferation of the lining. First, Hassanein et  al. (2003) showed strong cytoplasmic
Occasionally, calcifying odontogenic cysts have been and nuclear expression of β-­catenin in the basaloid cells of
described in association with other odontogenic tumours. calcifying odontogenic cyst, pilomatrixoma, and crani-
Ledesma-­Montes et al. (2008) found 4 cases (3.3%) in their opharyngioma. This was unusual, because normally
series, associated with ameloblastic fibroma, ameloblastic β- catenin is expressed at the cell membrane. In all cases,
fibro-­odontoma, adenomatoid odontogenic tumour, and the ghost cells were negative. In the second study, Sekine
odontoameloblastoma, and de Arruda et al. (2018a), in an et  al. (2003) showed that 9 out of 10 cases of calcifying
analysis of 367 cases, found 14 (3.85%) associated with odontogenic cyst harboured a somatic mutation in the β-­
ameloblastic fibroma (5), ameloblastic fibro-­odontoma (2), catenin gene (CTNNB1). All lesions also showed cytoplas-
adenomatoid odontogenic tumour (3), ameloblastoma (2), mic and nuclear expression of β-­catenin by
odontogenic keratocyst (1), and orthokeratinised odonto- immunohistochemistry. A further 20 ameloblastomas (10
genic cyst (1). Many of these probably represent hybrid follicular and 10 plexiform) were also studied and only
lesions or unusual oddities. However, it should be noted one case showed a CTNNB1 mutation. All 10 follicular
that ameloblastic fibroma and ameloblastic fibro-­odontoma ameloblastomas showed cytoplasmic and nuclear stain-
are indistinguishable from early developing odontomas, ing, but the plexiform lesions were negative. The authors
and these may represent the early stages of formation of an interpreted the results to show that calcifying odontogenic
odontoma associated with a calcifying odontogenic cyst, cyst and ameloblastoma may show similar histology, but
rather than a separate tumour arising in the wall. Lin et al. are genetically distinct, and that CTNNB1 mutations and
192 Calcifying Odontogenic Cyst

intracellular accumulation of β-­catenin are important for et al. 2005) and found positively stained ghost cells in 66.7%
the histogenesis of calcifying odontogenic cyst. of cases (78.8% of compound odontomas and 29.4% of
These results were confirmed by others (Ahn et al. 2008) complex odontomas). Ghost cells were mostly observed in
and suggest a role for β-­catenin and the WNT signalling odontogenic epithelium adjacent to immature enamel in
pathway in the pathogenesis of calcifying odontogenic developing lesions. In more mature lesions the ghost cells
cyst. β-­catenin and LEF-­1 act as transcriptional co-­factors became calcified, but still expressed the keratin proteins.
that regulate the WNT signalling pathway, which is essen- Of particular interest, however, is that they also found that
tial for normal tooth development. CTNNB1 mutations the odontogenic epithelium adjacent to the ghost cells
cause intracellular accumulation of β-­catenin that disrupts showed cytoplasmic and nuclear positivity for β-­catenin,
the pathway and interferes with cellular differentiation. and for LEF-­1, a downstream transcriptional factor that
Similar ectomesenchymal interactions occur in tooth, hair, interacts with β-­catenin in the WNT signalling pathway.
and pituitary development, and it is suggested that acti- Bilodeau et al. (2015) investigated LEF-­1 expression in sali-
vating mutations in the β-­catenin gene and disruption vary and odontogenic tumours and found that calcifying
of the WNT signalling pathway explain a common patho- odontogenic cysts (CCOT in their study) showed high
genic mechanism for development of calcifying odonto- expression in 64% (7 of 11) of cases. Furthermore, LEF-­1
genic cyst, pilomatrixoma, and craniopharyngioma (Sekine was always co-­expressed with nuclear β-­catenin, which
et al. 2003; Gomes et al. 2019). was seen in 82% (9 of 11) of cases.
More recently, de Sousa et al. (2016) and Yukimori et al. These data suggest that ghost cells in odontoma, a
(2017) interrogated calcifying odontogenic cysts for muta- hamartoma, may arise as a result of intracellular accumu-
tional hotspots in 50 cancer genes and both groups found lation of β-­catenin, in a similar mechanism as that seen
mutations only in CTNNB1, at a similar site to mutations in the ghost cell lesions. To date, however, CTNNB1 muta-
found in pilomatrixoma and craniopharyngioma. This sug- tions have not been identified in human odontomas,
gests that CTNNB1 mutations are a specific driver in the although studies have shown that activation of WNT sig-
pathogenesis of calcifying odontogenic cyst and other nalling by expression of mutated β-­catenin can generate
ghost cell lesions. Yukimori et  al. (2017) also showed odontoma formation in mice (Xavier et al. 2015).
nuclear staining for β-­catenin in cells surrounding ghost A number of studies have also shown accumulation
cells, but not in the ghost cells. However, ghost cells were of the enamel protein amelogenin in the ghost cells in cal-
positive for hair keratins. To test the association between cifying odontogenic cysts (Abiko et  al.  2001; Yoshida
CTNNB1 mutations and ghost cells, the authors transfected et  al.  2001). Takata et  al. (2000) found that ghost cells
embryonic kidney 293 cells with normal and mutant expressed amelogenin as well as enamelin, enamelysin
CTNNB1, and showed that the mutant cells were associ- (MMP-­20), and sheathlin (prism sheath protein). They also
ated with ectopic expression of hair keratins that initiate showed that the ghost cells of pilomatrixomas were nega-
the formation of ghost cells. tive. The significance of these findings is not clear, but they
This hypothesis is also supported by studies that have suggest that the aberrant differentiation, thought to be a
shown that ghost cells express cytokeratins (CKs) that are factor in the formation of ghost cells, may cause accumula-
associated with hair formation, including hard α-­keratins tion of enamel proteins as well as hair-­type keratins.
(Kusama et al. 2005; Kikuchi et al. 2012), CK6 (Kaminagakura Taken together, these data have been interpreted to sug-
et  al.  2013), AE1/AE3 (Rumayor et  al.  2015), and AE13 gest that most calcifying odontogenic cysts are neoplasms
(CK40; Yukimori et al. 2017). It is interesting that Yukimori caused by mutations in CTNNB1 (Yukimori et  al.  2017).
et al. (2017) found CTNNB1 mutations in two ameloblasto- The mutations are associated with intracellular accumula-
mas, but both were subsequently shown to contain ghost tion of β-­catenin and disruption of the WNT signalling
cells, which also expressed hair keratins. Although ghost pathway, causing aberrant epithelial differentiation and
cells express these ‘hard’ hair-­type keratins, they may not formation of ghost cells. These mechanisms provide a uni-
express many of the more common keratins such as CK8, fying hypothesis for the pathogenesis of ghost cell lesions,
CK13, CK18, or CK19 (Fregnani et al. 2003; Kaminagakura including calcifying odontogenic cyst, pilomatrixoma, and
et al. 2013; Rumayor et al. 2015). craniopharyngioma (Gomes et al. 2019).
These changes, however, are not specific for the ghost It should be noted, however, that β-­catenin gene muta-
cell lesions, since they have been identified in odontomas, tions are not specific to ghost cell lesions and have been
which is of particular interest given the association detected in a wide range of human tumours (reviewed by
between calcifying odontogenic cysts and odontomas. Kim and Jeong  2019). Also, only a single mutation has
Tanaka et al. (2007) examined 69 cases of odontomas using been recorded in calcifying odontogenic cyst and this
antibodies against human hair hard α-­keratins (Kusama may not be sufficient for the development of a neoplasm.
­Histopatholog 193

This single β-­catenin gene (CTNNB1) mutation may act as establish where this is located and its nature. The calcified
a common driver mutation or gatekeeper that initiates tissue may be calcified ghost cells, dentinoid material, or
altered cell differentiation at different anatomical sites, but tooth substance associated with an odontoma. In calcifying
further genetic events may be necessary for progression to odontogenic cyst, calcified ghost cells are usually located in
neoplasia. This lack of association with neoplasia is sup- the epithelial lining or the lumen, and dentinoid or odon-
ported by the fact that odontomas may be formed as a toma is located in the wall. If the lesion appears solid or the
result of aberrant WNT signalling induced by mutated lumen appears to be filled with dentinoid, then a dentino-
β- catenin (Xavier et al. 2015), and that ghost cells in odon- genic ghost cell tumour should be considered. It follows
tomas express hair keratins and are associated with nuclear from this that it is important to decalcify the hard tissue for
accumulation of β-­catenin in the adjacent epithelium, in a microscopic examination to establish its true nature.
pattern similar to that seen in calcifying odontogenic cyst The histological features of a classic calcifying odonto-
(Tanaka et al. 2007). genic cyst are characteristic and present few diagnostic
problems (Buchner 1991; Wright and Soluk ­Tekkeşin 2022;
Figures 11.8–11.10; Box 11.2). The typical cyst is lined by
­Histopathology epithelium, with a well-­defined basal layer of columnar
cells and an overlying layer of varying thickness that may
The majority of calcifying odontogenic cysts are enucleated resemble stellate reticulum. The characteristic feature is
with or without curettage, so the pathologist may receive a the presence of focal accumulations of ghost cells, which
collapsed cyst, or the specimen may be fragmented or sepa- may be in the epithelial lining or may form masses protrud-
rated into many parts. This is especially the case if the lesion ing into the lumen, or occasionally pushing into the fibrous
is associated with an odontoma (Figure 11.7). Gross exami- capsule. The ghost cells often become calcified. Calcifying
nation of the specimen and review of radiographs are espe- odontogenic cyst is almost always unicystic, but occasional
cially important in helping to diagnose this lesion. In a multicystic examples have been reported (Buchner 1991).
fragmented specimen it may be particularly difficult to These features are seen in all types of calcifying odonto-
appreciate the relationships between the components of the genic cyst, but there may be considerable variation in the
lesion and to determine its cystic nature. On gross examina- proportions of each component and a variety of features
tion it should be possible to identify a cyst wall, although may be seen in the lining or the connective tissue wall. For
the lumen may be filled, giving the impression of a solid the most part, the epithelial lining is thin and regular, as
tumour. It is helpful to open the cyst and search the wall can be seen in Figures  11.7–11.9, but with focal areas of
and lining for thickenings, which should be sampled. Many thickening, usually associated with accumulations of ghost
lesions will contain hard tissue and it is important to cells (Figures  11.8 and  11.10). The typical, and often

(a) (b)

Figure 11.7  A low-­power view of a specimen of a calcifying odontogenic cyst associated with an odontoma. The cyst (a) is collapsed
and fragmented and the hard tissue component (b) has been presented separately (see also Figure 11.13).
194 Calcifying Odontogenic Cyst

(a)

(b) (c)

Figure 11.8  Calcifying odontogenic cyst. (a) A typical simple cystic lesion. (b) The lining is thin and regular and, in this section, is
mostly ameloblastomatous. (c) Careful examination will show focal accumulations of ghost cells. Note a small island of odontogenic
epithelium in the wall (c, arrowhead).

diagnostic, lining resembles a unicystic ameloblastoma (Figures 11.9 and 11.10). Occasionally masses of ghost cells
and shows a columnar basal layer of ameloblast-­like cells, may form and may fill the lumen to form what appears to
with reversal of nuclear polarity and a superficial layer be a solid tumourous lesion. Prætorius et  al. (1981) and
resembling stellate reticulum (Figures  11.8b and  11.10c). Ledesma-­Montes et  al. (2008) classified a small number
Often however, areas of the lining are flattened and show a of  calcifying odontogenic cysts as a proliferative type,
simple stratified squamous epithelium (seen in with  ameloblastomatous proliferations that may extend
Figure 11.10b, e). into the lumen (Figure  11.9) or be seen in the cyst wall
The main diagnostic feature of the calcifying odonto- (Figure 11.11d). When this occurs the proliferation into the
genic cyst is the presence of ghost cells. The ghost cells are lumen usually resembles a plexiform ameloblastoma, but
large ballooned or elliptoid cells with amorphous and is associated with accumulations of coalesced ghost cells
eosinophilic cytoplasm, and with a pale clear space where (Figure  11.9), which often show areas of calcification.
the nucleus has been lost (Figure 11.10). Ghost cells may Similarly, when ameloblastoma-­like islands are seen in
arise as scattered single cells in the epithelial lining, but the  wall, they are usually associated with ghost cells
most often they form focal accumulates that produce epi- (Figure 11.11d). The ghost cells often calcify and individual
thelial thickenings or protrusions into the lumen or small numbers of calcified ghost cells can be seen in
­Histopatholog 195

(a)

(b) (c)

Figure 11.9  (a) Calcifying odontogenic cyst, showing luminal proliferations. These resemble plexiform ameloblastoma (b), but
contain sheets of ghost cells (c).

almost all lesions. Occasionally accumulations of coa- Dentinoid formation is also seen adjacent to the epithe-
lesced ghost cells may calcify to form calcified masses, but lial lining, and is seen in calcifying odontogenic cysts asso-
this is most prominent in calcifying odontogenic cyst asso- ciated with odontoma and in about 50% of simple cystic
ciated with odontoma (Figures 11.7b and 11.10f). lesions. It should be noted that dentinoid is not seen as fre-
Often, the ghost cells extrude from the epithelial lining quently in calcifying odontogenic cyst as it is in dentino-
through the basement membrane into the fibrous cyst wall genic ghost cell tumour, where it is a consistent finding.
(Figure  11.10d, e), where they may elicit a foreign-­body When present, it forms small masses or strips just below
giant cell reaction or be associated with dystrophic calcifi- the epithelial lining (Figure 11.12a, b). Large sheets of den-
cation (Figure 11.11). Other features that may be seen in tinoid are unusual except in those lesions associated with
the wall include islands of odontogenic epithelium that odontomas, where an admixture of dental hard tissues may
usually resemble quiescent rest cells of dental lamina be seen (Figure 11.7b and 11.13). Occasionally masses of
(Figure 11.11a), but may occasionally show ameloblast-­like dentinoid and ghost cells may fuse to form large amor-
differentiation with juxta-­epithelial formation of a dentine-­ phous sheets (Figures  11.12c and  11.13), but a simple
like material (Figure  11.11c). Takeda et  al. (1990) found van  Gieson stain will differentiate the two structures
islands of odontogenic epithelium or small satellite cysts in (Figure  11.12d). The ghost cells may also express some
9 out of 13 cysts examined. cytokeratins, as discussed in pathogenesis.
196 Calcifying Odontogenic Cyst

(a) (b)

(c) (d)

(e) (f)

Figure 11.10  Ghost cells are eosinophilic cells with a pale central area where the nucleus has been lost. They may form masses
replacing the epithelium (a, b) or may appear as small intraepithelial focal accumulations or even single cells (c). Often the ghost cells
protrude through the basement membrane (d) and form masses within the cyst wall (e). Calcification of ghost cells is common, either
as single cells (c, top left) or as coalesced masses (f). (b, e) Note that the adjacent cyst lining is thin stratified squamous epithelium.
­Histopatholog 197

Box 11.2  Calcifying Odontogenic Cyst: Diagnostic Criteria and Differential Diagnosis


●● Simple unicystic lesion ●● Lesions harbour a mutation in the β-­catenin gene
●● Lined predominantly by ameloblastomatous epithe- (CTNNB1)
lium, with columnar basal layer and superficial stel-
Caution: features that suggest an alternative diagnosis:
late reticulum-­like cells
●● All lesions contain ghost cells, which often show ●● Solid or multicystic lesion with prominent dentinoid
dystrophic calcification that is admixed with the epithelium: consider denti-
●● Ghost cells may be scattered single cells, but usually nogenic ghost cell tumour
form focal accumulations protruding into the lumen ●● Prominent calcified tissue with enamel and dentine,
●● Epithelium may proliferate into the lumen or into the but very little epithelium and no cystic areas: con-
wall, occasionally resembling plexiform ameloblastoma sider odontoma
●● About 50% contain dentinoid in the cyst wall adja- ●● Cyst lined by ameloblastomatous epithelium with or
cent to the epithelium without luminal proliferations, but with absence of
●● About 25% are associated with an odontoma ghost cells: consider unicystic ameloblastoma
●● Immunohistochemistry shows characteristic nuclear ●● Prominent epithelial proliferation with evidence of
and cytoplasmic staining for β-­catenin, but this is not cytological atypia, mitoses, or an infiltrative pattern:
specific, and may be seen in follicular ameloblastoma consider ghost cell odontogenic carcinoma

(a) (b)

(c) (d)

Figure 11.11  Features seen in the wall of calcifying odontogenic cyst. (a) Dystrophic calcifications associated with ghost cells
(arrow) and islands of odontogenic epithelium (arrowheads). (b) Ghost cells (arrow) elicit a foreign-­body giant cell response. (c) An
island of odontogenic epithelium that has induced a ring of dentinoid formation. (d) Islands resembling follicles of ameloblastoma,
but ghost cells are also present (arrow).
198 Calcifying Odontogenic Cyst

(a) (b)

(c) (d)

Figure 11.12  Features of dentinoid in calcifying odontogenic cyst. (a, b) Typically, small sheets of dentinoid can be seen in the
fibrous connective tissue in close proximity to the epithelial lining. (c) Occasionally amorphous masses of dentinoid and ghost cells
coalesce, but a simple van Gieson stain (d) can distinguish the red staining dentinoid (arrow) from yellow ghost cells. (b) Note that
cholesterol clefts can be seen (top right).

Figure 11.13  A high-­power view of part


of the lesion shown in Figure 11.7b. This
GC shows odontoma formation with
well-­formed enamel matrix (e), tubular
dentine (d) and dentinoid (de).
Accumulations of ghost cells are also
seen (gc), many of which are calcifying
(see also Figure 11.10f).

d
e
de
­Histopatholog 199

A rare feature, but one that is often remarked upon, is the varying between about 10% (Sedano and Pindborg  1975;
presence of melanin pigmentation in a small number of Soluk Tekkeşin et al. 2012b) and 67% (Tanaka et al. 2007).
cases (Buchner 1991). Han et al. (2007) reported a case and Odontomas are the most common of the odontogenic
reviewed 20 previous cases. Melanin pigment was found in tumours, and although they are see in about 25% of calcify-
variable amounts in the epithelial lining, including within ing odontogenic cysts, the converse is not true – less than
ghost cells. 1% of odontomas may contain a calcifying odontogenic
A small number of calcifying odontogenic cysts may be cyst. Care must be taken not to overdiagnose ghost cell
associated with other odontogenic lesions apart from lesions when ghost cells are seen in association with an
odontomas (Ledesma-­Montes et  al.  2008). These have odontoma. A calcifying odontogenic cyst should only be
been discussed in the section on pathogenesis. diagnosed if there is clear evidence of an associated
Buchner et  al. (1991) reviewed 45 cases of peripheral cystic lesion with the histological features described above
ghost cell lesions. At the time, all were classified as calcify- (Figure 11.7 and Box 11.2).
ing odontogenic cysts, but they found that 15 cases were A further diagnostic problem is to differentiate between
solid and would now be classified as peripheral dentino- a calcifying odontogenic cyst and a dentinogenic ghost cell
genic ghost cell tumours. Of the cystic lesions, they found tumour. As discussed at the beginning of this chapter, these
that the histological features were very similar to those two lesions have been regarded as variants of the same
described here for the intraosseous lesions. entity, originally as a ‘cystic’ and a ‘solid’ calcifying odonto-
Immunohistochemistry does not have an important role genic cyst, with overlapping features and no clear diagnos-
in diagnosis. Expression of β-­catenin, cytokeratins, and tic criteria. If the criteria for calcifying odontogenic cyst are
enamel proteins has been discussed in the section on followed, then the distinction should be clear. The calcify-
pathogenesis. Although these have provided some insight ing odontogenic cyst is primarily a unicystic lesion, and
into the histogenesis of the lesion, they have little role although occasional satellite cysts may arise in the wall,
in differential diagnosis. For example, although β-­catenin these are rare, few in number, and probably represent
shows unusual nuclear expression in calcifying odonto- microcystic degeneration in islands of odontogenic epithe-
genic cyst, this may also be seen in ameloblastomas, which lium. Dentinogenic ghost cell tumour, on the other hand, is
are the main lesions that may cause diagnostic confusion. a solid lesion that shows an admixture of ameloblastoma-­
A number of studies have also investigated proliferation like epithelium, ghost cells, and prominent dentinoid. Like
and apoptotic markers in calcifying odontogenic cyst and ameloblastoma, however, it may be multicystic and a
have shown evidence of greater expression of Bcl-­2, Ki-­67, biopsy of a cystic area may resemble calcifying odonto-
and proliferating cell nuclear antigen (PCNA) in prolifera- genic cyst. Errors can be avoided by properly considering
tive or ameloblastomatous lesions (Yoshida et  al.  2001; the clinical and radiological features and by careful exami-
Fregnani et al. 2003; Gomes da Silva et al. 2014), but these nation and sampling of the gross specimen. In particular,
findings are of little diagnostic value when examining a solid areas of apparently hard tissue should be decalcified
single case. and examined.

Histological Differential Diagnosis Ghost Cell Odontogenic Carcinoma


and Malignant Progression of Calcifying
In most cases the histological diagnosis is straightforward
Odontogenic Cyst
(Box 11.2), since the finding of a cyst lined by ameloblasto-
matous epithelium with ghost cells is diagnostic. However, Ghost cell odontogenic carcinomas are very rare, but many
in small or fragmented biopsies not all the features may be appear to arise in a precursor benign ghost cell lesion,
apparent, and it is important to properly sample the speci- either a calcifying odontogenic cyst or a dentinogenic ghost
men and examine multiple blocks. As Figure 11.8 shows, in cell tumour (van Heerden and Gomes  2022). De Arruda
any given section ghost cells may not be numerous and the et al. (2018a), in their systematic review, only identified 44
lining must be examined carefully. In this circumstance, if cases reported up to 2018, but found that 12 (27.3%) had
the ghost cells are missed, the main diagnostic error is to been preceded by a calcifying odontogenic cyst. In their
misdiagnose the lesion as a unicystic ameloblastoma. series of 122 ghost cell lesions, Ledesma-­Montes et  al.
Ghost cells are characteristic of calcifying odontogenic (2008) found only 3 carcinomas and showed that 2 arose
cyst, but they are not specific and the presence of ghost de novo and 1 had progressed from a simple type of calcify-
cells in an odontogenic lesion should not automatically ing odontogenic cyst. Lu et al. (1999) reported 4 cases and
lead to a diagnosis of a ghost cell lesion. In particular, ghost showed that 1  had arisen after recurrence of a typical
cells have been reported in odontomas with a frequency benign calcifying odontogenic cyst, and the other 3 showed
200 Calcifying Odontogenic Cyst

areas of typical calcifying odontogenic cyst associated with recurrent calcifying odontogenic cysts. Histological fea-
more solid areas showing cytological evidence of malig- tures that should arouse suspicion of malignant progres-
nancy. They also reviewed 12 previously reported cases and sion include proliferation of the lining with a cribriform
found 1 that had occurred after recurrence of a calcifying pattern, cytological atypia with increased mitoses, high
odontogenic cyst, and 2 associated with recurrence of expression of Ki-­67, and loss of ghost cells (Mokhtari
ameloblastomas. Of the total of 16  lesions, therefore, et al. 2013). Ghost cell odontogenic carcinoma is also twice
12 appeared to arise de novo, although most also showed as common in males as in females and in the maxilla as in
areas of typical calcifying odontogenic cyst on histology. the mandible.
Arashiyama et al. (2012) reported a single case that arose
18 years after removal of a typical calcifying odontogenic
cyst. In their literature review they identified 30 previously ­Treatment
reported cases of ghost cell odontogenic carcinoma, and
found that 11 (36.7%) had arisen secondary to a calcifying Calcifying odontogenic cyst is treated by surgical enu-
odontogenic cyst, and only 1 was secondary to a dentino- cleation unless it is associated with another odontogenic
genic ghost cell tumour. More recently, Nel et  al. (2020) tumour, in which case wider excision may be required. In
reported a calcifying odontogenic cyst in a 12-­year-­old the presence of an odontoma, conservative removal will
male that presented as a large, well-­demarcated radiolu- still be adequate. Although classic uncomplicated cases
cency filling the ramus of the mandible, with considera- of  calcifying odontogenic cyst may grow to a large size,
ble  bucco-­lingual expansion. Within three years the cyst reported recurrences are rare and are seen in less than 5%
expanded to also fill the body of the mandible, and histol- of cases (de Arruda et al. 2018a). Marsupialisation followed
ogy showed progression to a ghost cell odontogenic carci- by enucleation has occasionally been used for large lesions.
noma. Mokhtari et  al. (2013) also reported a case and Care must be taken with lesions showing unusual features
reviewed factors that may predict onset of malignancy or ameloblastomatous proliferations. It has been shown
in  calcifying odontogenic cysts. They concluded that that these may be more aggressive and should probably be
malignant transformation should be considered in all regarded as hybrid lesions (Nel et al. 2021).
201

12

Orthokeratinised Odontogenic Cyst

CHAPTER MENU
Clinical Features, 202
•• Frequency, 202
•• Age, 202
•• Sex, 202
•• Site, 202
•• Clinical Presentation,  203
Radiological Features, 204
Pathogenesis, 205
•• Relationship to the Dentigerous Cyst,  206
Histopathology, 207
•• Histological Differential Diagnosis,  211
•• Relationship to the Odontogenic Keratocyst,  211
•• Verrucous Odontogenic Cyst,  212
Treatment, 213

The orthokeratinised odontogenic cyst is a developmental Selvamani et  al.  2014). However, it still took almost four
odontogenic cyst lined by orthokeratinised stratified squa- decades for the lesion to be properly categorised as a dis-
mous epithelium (Speight and Neville  2022). Previously, tinct entity in the fourth (2017) edition of the WHO classifi-
most authors, including early World Health Organization cation (Speight et al. 2017b). The key distinguishing features
(WHO) classifications and previous editions of this book, of  the orthokeratinised odontogenic cyst are that it is
have referred to this cyst as a variant of odontogenic kerato- orthokeratinised and, in distinction to the odontogenic
cyst. It was Philipsen (1956) who first used the term odonto- keratocyst, it rarely recurs.
genic keratocyst, but in his original series of seven cases one Although we credit Philipsen (1956) and then Wright
was lined by orthokeratinised epithelium, and because of (1981) as the originators of our contemporary understand-
this subsequent authors and classifications stated that ing of keratinising jaw cysts, Ide et al. (2020) have pointed
odontogenic keratocysts are normally lined by  parakerati- out that they have been described since the mid-­nineteenth
nised epithelium, but may on occasions be orthokeratinised century. In this fascinating historical review, Ide and col-
(Pindborg and Kramer 1971; Brannon 1976, 1977; Kramer leagues trace the origin of keratocysts to 1855, when they
et al. 1992). Wright (1981) was the first to properly record were first referred to as ‘buttery cysts’ by Maisonneuve,
the distinctive features of the orthokeratinised variant of the term ‘buttery’ referring to the thick, keratinaceous con-
the odontogenic keratocyst and suggest that it should be tents that resemble butter. The authors record many exam-
recognised as a distinct entity. After Wright’s paper, Li et al. ples of references to keratinising cysts up to 1933, and
(1998) proposed the term orthokeratinised odontogenic cyst attribute the first record of an orthokeratinised jaw cyst to
and a number of authors subsequently recognised the Jeannel in 1886. In the early twentieth century Schultz
orthokeratinised odontogenic cyst as a distinct entity (Li (1927) described an orthokeratinised jaw cyst as a dermoid
et al. 1998; Dong et al. 2010; MacDonald-­Jankowski 2010; cyst, but other authors often referred to them as

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
202   Orthokeratinised Odontogenic Cyst

cholesteatoma. It was Philipsen (1956) who then described early papers that suggest that about 10% of odontogenic
‘cholesteatoma’ of the jaws as odontogenic keratocyst. keratocysts are orthokeratinised: 13% (Wright  1981);
9.7% (Brannon  1977); 12.2% (Crowley et  al.  1992); 8.0%
(Li et al. 1998); and 9.9% (MacDonald-­Jankowski 2011).
­Clinical Features
Age
Most descriptions of the orthokeratinised odontogenic cyst
have been included in large series of odontogenic kerato- Orthokeratinised odontogenic cyst presents in the third or
cysts. In his systematic review, MacDonald-­Jankowski fourth decades, with an average age of about 35 years
(2010) found 36 papers up to 2009 that, between them, (Table  12.1). In his systematic review, MacDonald-­
described 408 cases of orthokeratinised odontogenic cyst. Jankowski (2010) found 10 reports that recorded the age at
Since 2009, Dong et  al. (2010) and Oh et al. (2022) have presentation (n  =  181), with an average age of 34.8 years
reported series of 60 lesions, and a literature search shows and a peak in the third decade. Only 19 cases were under
a further 23 papers reporting findings in about 140 cysts, age 20 years and only 1 case was found in the first decade.
including three smaller series recording clinical features
(Aragaki et  al.  2010; Selvamani et  al.  2014; Uddin
Sex
et al. 2019). Table 12.1 summarises the clinical features of
orthokeratinised odontogenic cyst in selected series that There is a predilection for males, with studies showing that
have included 10 or more cases and from the systematic up to 75% of lesions have been encountered in males
review of MacDonald-­Jankowski (2010). (Table  12.1). MacDonald-­Jankowski (2010) found that 12
reports recorded sex (n = 192 cysts) and that 66.1% were
encountered in males (M : F 2 : 1).
Frequency
The orthokeratinised odontogenic cyst is rare, with a fre-
Site
quency of less than 2% of all odontogenic cysts. Most
studies recording the relative frequency of odontogenic All studies show that orthokeratinised odontogenic cyst is
cysts (see Table  1.3) have included orthokeratinised found most often in the mandible, with an overall fre-
odontogenic cyst in with odontogenic keratocysts, so quency of about 70% (Table 12.1), and with the single most
the frequency is uncertain. Only six of the reports in common site being the mandibular molar/ramus region.
Table 1.3 included specific reference to orthokeratinised The two largest series shown in Table 12.1 (Wright 1981;
odontogenic cyst, and showed frequencies of 0.3% Dong et  al.  2010) analysed the site distribution of their
(Grossmann et  al.  2007; Brazil), 0.3% (Tamiolakis cases and their combined data (n = 121) are illustrated in
et al. 2019; Greece), 1.5% (Ali 2011; Kuwait), 1.6% (Bhat Figure 12.1. Of these, 78.5% were located in the mandible
et al. 2019; India), 1.2% (Aquilanti et al. 2021; Italy), and and 62.8% of all cases were found in the posterior mandi-
1.7% (Kammer et al. 2020; Brazil). This accords well with ble. Crowley et al. (1992) found that 73.4% of their lesions

Table 12.1  Age, gender and site distribution from selected reports of orthokeratinised odontogenic cyst (where n ≥10), and from the
review of MacDonald-Jankowski (2010).

References Country n Mean age Peak decade Male (%) Mandible (%) Associated with impacted tooth (%)

Wright (1981) USA 60 35.4 3rd 75.0 63.3 72.9


Li et al. (1998) Japan/China 15 31.5 4th 66.6 93.3 46.7
Crowley et al. (1992) USA 55 33.8 NR 56.6 NR 75.7
Dong et al. (2010) China 61 38.9 4th 72.1 90.2 50.0
Aragaki et al. (2010) Japan 20 31.5 3rd 65.0 75.0 NR
Selvamani et al. (2014) India 11 29.1 3rd 72.7 81.8 NR
Uddin et al. (2019) Afghanistan 10 38.9 4th 70.0 70.0 30.0
Oh et al. (2022) S Korea 65 32.9 NR 72.3 80.0 70.8
MacDonald-­Jankowski (2010) SR 408a 34.8 3rd 66.1 70.8 69.4

NR, not reported; SR, systematic review.


a
 Total cases in review, but n for each parameter varies depending on number of reports.
­Clinical Feature  203

Figure 12.1  Site distribution of 121


orthokeratinised odontogenic cysts. Source: 8.3% 10.7%
2.5%
Data from Wright (1981) and Dong et al. (2010).
4.1%
11.6%

Percent of cases
Mandible
Maxilla
62.8%

Molar Premolar Anterior

were located in the posterior jaws, and that 20.8% were in et al. 2014; Oh et al. 2022). Joseph et al. (2021) reported a
the anterior regions. They also compared their 55 cases to patient with two cysts located on the upper and lower right
387 odontogenic keratocysts and specifically noted that third molars. Others have reported more than two lesions
16.3% (9 cases) of orthokeratinised odontogenic cyst were in the same patient. Cheng et al. (2015) described a patient
found in the midline, compared to only 22 (5.7%) odonto- with four cysts, associated with unerupted third molars in
genic keratocysts. One case has been reported in the condy- all four quadrants. Crane et al. (2020) reported two patients
lar head (Managutti et al. 2016), but this is very rare and with multiple cysts. The first had two cysts bilaterally in
may represent an unusual intraosseous epidermoid cyst. the mandible and a third cyst associated with an unerupted
There have been a number of reports of patients with maxillary third molar. The mandibular cases were found
multiple orthokeratinised odontogenic cysts. Most often, overlying the apices of the right first and second molars,
these have been two cysts located bilaterally in the mandi- and a larger lesion on the left side, in a dentigerous rela-
ble and usually associated with unerupted third molars tionship with an unerupted second molar and displacing
(Pereira et  al.  2012; Premalatha et  al.  2012; Pimpalkar the third molar (Figure  12.3). The second patient (Crane
et  al.  2020) had bilateral cysts associated with unerupted
maxillary third molars, one of which completely filled and
expanded the maxillary sinus.
Although these cases are striking, multiple orthokerati-
nised odontogenic cysts appear to be rare, with only seven
patients reported. In the series shown in Table 12.1, none
recorded multiple cysts. None of the patients with multiple
cysts showed any features of the naevoid basal cell carci-
noma syndrome (NBCCS).

Clinical Presentation
The majority of lesions present as painless swellings, and
often as an incidental finding. In MacDonald-­Jankowski’s
(2010) review, 47.6% were incidental findings, in 41.1% there
was evidence of swelling, and only 24.1% had a history of
pain. Wright (1981) found that 25 of his 60 cases (41.7%) were
completely asymptomatic, 13 (21.7%) presented with pain, 8
(13.3%) had swelling, and 5 (8.3%) had evidence of infection.
Figure 12.2  Orthokeratinised odontogenic cyst shows a
well-­demarcated unilocular radiolucency. Note that although the
In the other large series of 61 cases (Dong et  al. 2010), 46
lesion is associated with an impacted tooth, it is not in a (75.4%) showed swelling of the jaw and 13 (21.3%) also had
dentigerous relationship. pain. Two patients had evidence of infection (3.3%).
204   Orthokeratinised Odontogenic Cyst

Recurrence of the orthokeratinised odontogenic cyst is rare. ­Radiological Features


Indeed, it was the benign behaviour and lack of recurrence of
the orthokeratinised variant of the odontogenic keratocyst The typical orthokeratinised odontogenic cyst presents as a
that led to its recognition as a distinct entity (Wright 1981). In unilocular radiolucency with a well-­demarcated and often
the series shown in Table  12.1 only two reported recurrent corticated outline (Figures  12.2 and  12.3). Lesions are fre-
lesions (Wright  1981; Crowley et  al.  1992). These authors quently spherical and over 90% are unilocular (MacDonald-­
reported one recurrence each, a total of 2 out of 115 cysts Jankowski 2010). Wright (1981) found only one case that was
(1.7%). In his systematic review, MacDonald-­Jankowski multilocular (1.6%), although Dong et al. (2010) found seven
(2010), found only nine papers (n  =  165 cysts) that had cases in their series (13.0%). Overall, however, this is far fewer
reported recurrences, with an overall frequency of 4.2% (7 out than are found in odontogenic keratocyst, where about 30%
of 165 cases), compared to 28.1% of odontogenic keratocysts. of lesions are multilocular (Table 12.2).
There is some evidence that orthokeratinised odonto- In most reports, between about 50% and 75% of lesions
genic cyst may rarely be associated with NBCCS, but fur- (Table 12.1) have been associated with an impacted tooth.
ther research is needed. Although none of the series in This implies that they may be in a dentigerous relationship,
Table 12.1 reported a case associated with the syndrome, and that the lesion envelops the crown of an unerupted
MacDonald-­Jankowski (2010) found one paper that tooth. However, this may not always be the case, and most
recorded a syndrome-­associated case (Bolbaran reports do not provide sufficient information to determine
et al. 2000). Woolgar et al. (1987a) compared the histologi- exactly how many lesions may be truly dentigerous. Li et al.
cal features of solitary keratocysts with keratocysts associ- (1998) did make this distinction and noted that while seven
ated with NBCCS, and found that a similar number of cases were associated with an impacted tooth, only four of
cysts in each group contained orthokeratin (23% and 32%, them were radiographically in a dentigerous relationship.
respectively), but the text of the paper suggests that this As shown in Figure 12.2, a cyst may be associated with an
includes cysts with small focal areas of orthokeratin that impacted tooth but not dentigerous. In Figure  12.3, how-
would not meet the current definition of orthokeratinised ever, the lesion on the left side of the mandible does appear
odontogenic cyst (see Box  12.2). In a similar study, to truly envelop the crown of the impacted second molar.
Dominguez and Keszler (1988) compared 33 cases each of A review of the many case reports of orthokeratinised
syndromic and solitary keratocysts and found orthokerati- odontogenic cysts suggests that the majority of lesions
nisation in 9% (3 cases) and 24% (8 cases), respectively, have a well-­defined spherical outline (see Figures  12.2
suggesting that they had identified 3 cases of orthokerati- and  12.3), suggesting a ballooning or centripetal growth
nised odontogenic cyst associated with NBCCS. Of the 7 pattern with expansion of the cortical plates. MacDonald-­
patients reported in the literature with multiple Jankowski (2010) showed that 100% of the orthokerati-
orthokeratinised odontogenic cyst, there were no cases nised odontogenic cysts were well defined, corticated, and
associated with NBCCS (Pereira et  al.  2012; Premalatha had bucco-­lingual expansion, but that these features were
et al. 2012; Pimpalkar et al. 2014; Cheng et al. 2015; Crane less common in odontogenic keratocysts, where they were
et al. 2020; Joseph et al. 2021). seen in 64.5%, 60.0%, and 61.7% of cases, respectively.

Figure 12.3  A case of multiple orthokeratinised odontogenic cysts. Two cysts are noted in the mandible, both well demarcated and
corticated. On the left side the cyst is in a dentigerous relationship with the impacted second molar. A third cyst is associated with the
displaced and rotated left maxillary third molar. Source: Crane et al. (2020) / Springer Nature / CC BY 4.0.
­Pathogenesi  205

Table 12.2  A summary of the differences and similarities


between the orthokeratinised odontogenic cyst and odontogenic Box 12.1  Orthokeratinised Odontogenic Cyst: Clinical
keratocyst (see text for details). and Radiological Features
●● They are rare – less than 2% of odontogenic cysts
Orthokeratinised Odontogenic
●● Almost always in adults, mean age 35 years. Peak in
odontogenic cyst keratocyst
third and fourth decades
Clinical and radiological ●● About 70% are found in males (M : F 2 : 1)
features ●● About 75% arise in the mandible, and 63% in the
Age (Mean) 35 yr 38 yr mandibular molar/ramus region
Males 66.1% 60.0% ●● Well-­defined corticated radiolucency. Usually spheri-
Mandiblea 90.2% 74.6% cal and unilocular
a ●● Only about 10% may be multilocular
Posterior mandible 75.4% 59.9%
●● About 70% are associated with an impacted tooth
Swellingb 41.1% 58.3%
and may appear dentigerous
Incidental findingb 47.6% 21.0% ●● Recurrence is rare – less than 5%
Pain 24.1% 32.4%
Unilocular 93.0% 72.2%
Well defined/corticatedb 100% 60%
Associated with unerupted 69.4% 35.5% aetiology. The lesions share a similar site distribution and
toothb both are most commonly encountered in the posterior
Recurrencesb 4.2% 28.1% mandible, where residual dental lamina is abundant. Most
Histology authors therefore agree that the source of epithelium for
the orthokeratinised odontogenic cyst is the rest cells of the
Orthokeratinised 100% N
dental lamina.
Parakeratinised N 100%
Early studies have shown that dental lamina rests can
Corrugated surface N 100%
undergo cystic change and form microcysts lined by
Palisaded basal layer N Y orthokeratinised epithelium (Hodson  1962; Moskow and
Budding/rete pegs N Y Bloom 1983). These have been described as ‘microkerato-
Islands/satellite cysts 5% 20% cysts’ (Moreillon and Schroeder  1982) and have been
Ki-­67a 8%; basal 20%; described in Chapter  9 (see Figure  9.4). Most agree that
suprabasal this cystic degeneration in rest cells of dental lamina is the
Cytokeratins K1/K10 K4/3, K19 initiating event in the pathogenesis of a number of jaw
cysts, including lateral periodontal and botryoid odonto-
100%, present in all lesions; N, not present, rare, or focal;
Y, characteristic, often present. genic cysts, gingival cysts, and orthokeratinised odonto-
Bold: parameters that are significantly different (see text for details). genic cyst. There is however no explanation as to why each
a
 Dong et al. (2010). type of cyst has a histologically different type of epithe-
b
 MacDonald-­Jankowski (2010).
lial lining.
The mechanisms of cyst growth and enlargement have
not been described for orthokeratinised odontogenic cyst
Reports of the size of the cyst show the smallest to be
and are poorly understood. The cyst is not inflammatory in
10 mm in diameter (Wright  1981) and the largest to be
origin, so inflammatory mechanisms may not play a sig-
155 mm (Dong et al. 2010). However, this large size is unu-
nificant role. However, cyst formation may be promoted by
sual and both Wright (1981) and Li et al. (1998) found that
accumulation of cell debris within the microcysts, result-
their largest lesions were 70 mm, with an average of about
ing in an increased osmotic pressure that promotes cyst
25 mm. In their comparison, Dong et al. (2010) found that the
expansion and growth. The role of osmosis in cyst growth
average diameter of odontogenic keratocysts was 51 mm.
has been described in detail in Chapter 3. Cyst growth in
the odontogenic keratocyst is thought to be primarily by
­Pathogenesis cell proliferation in the epithelial lining, resulting in lateral
expansion of the cyst, but this does not seem to apply to the
Little has been written about the pathogenesis of the orthokeratinised odontogenic cyst. A number of studies
orthokeratinised odontogenic cyst, since it has been have shown that the cell proliferation rate and apoptotic
regarded as a variant of odontogenic keratocyst and most index in orthokeratinised odontogenic cyst are similar to
authors have assumed that the lesions have a similar dentigerous cyst, and lower than seen in the odontogenic
206   Orthokeratinised Odontogenic Cyst

keratocyst (Li et al. 1998; Thosaporn et al. 2004; Rangiani lesions. Gomes and Gomez (2011) proposed that PTCH1
and Motahhary 2009; Dong et al. 2010). Dong et al. (2010) may act as a gatekeeper gene for many types of odonto-
compared expression of Ki-­67 and p63  in 15 orthokerati- genic lesions, and that further genetic events drive the for-
nised odontogenic cysts and 15 odontogenic keratocysts, mation of different cysts or tumours.
and found significantly greater expression in the kerato- These studies suggest that aberrations in the PTCH gene
cysts. In particular, Ki-­67  was found mainly in the basal may be an early initiating event in a number of odontogenic
layers of orthokeratinised odontogenic cyst and in only 8% lesions, but that further genetic changes are needed to deter-
of cells, compared to suprabasal expression in 20% of cells mine the lesion type. Such a scenario could explain a role for
in keratocysts. Rangiani and Motahhary (2009) examined PTCH in orthokeratinised odontogenic cyst, but does not
expression of bcl-­2 and bax in 28  keratocysts and 9 exclude a further role for specific PTCH mutations in kerato-
orthokeratinised odontogenic cysts, and found that all cysts. A key event in keratocysts that supports a neoplastic
cysts showed strong expression of bax, but bcl-­2 was only origin is biallelic alterations in the PTCH gene, but there is
expressed in keratocysts, and all orthokeratinised odonto- no evidence for this in orthokeratinised odontogenic cyst or
genic cysts were negative. Since bcl-­2 is anti-­apoptotic, the other cyst types. This is discussed in detail in Chapter 7.
authors interpreted its expression in keratocysts as promot-
ing cyst growth. In a similar study, Vered et al. (2009) also
Relationship to the Dentigerous Cyst
showed that orthokeratinised odontogenic cysts, as well as
dentigerous and radicular cysts, were negative for bcl-­2, It has been noted above that the orthokeratinised odonto-
while positive expression was found in odontogenic kerato- genic cyst is often described as being in a dentigerous rela-
cysts and ameloblastomas. tionship with an unerupted tooth (Table  12.1). However,
These studies suggest that increased cell proliferation few papers accurately describe the relationship of the cyst
does not play an important role in driving growth of the to the tooth, and it is not known how many are actually in a
orthokeratinised odontogenic cyst, and that expansion may true dentigerous relationship. While Wright (1981) used the
be driven by increased intracystic osmotic pressure. This is term ‘dentigerous’, most other authors have described the
further supported by the observation that orthokeratinised cyst as associated with an ‘impacted’ or ‘unerupted’ tooth,
odontogenic cysts are often spherical with cortical expan- without defining or clarifying the relationship with the
sion, suggesting a centripetal growth pattern. crown (Crowley et  al.  1992; MacDonald-­Jankowski  2010;
The role of the PTCH gene in the pathogenesis and neo- Dong et al. 2010; Uddin et al. 2019). It is important to note
plastic potential of the odontogenic keratocyst has been that these terms are not synonymous, and that a cyst may be
discussed in Chapter 7. With regard to the orthokeratinised associated with an impacted tooth, but not be in a dentiger-
odontogenic cyst, there has been no suggestion that it is ous relationship (see Figure 12.2). In their series of 15 cases,
neoplastic. Indeed, when the odontogenic keratocyst was Li et al. (1998) did make this distinction and found that 7
renamed and designated as a neoplasm in the 2005 WHO cases were associated with an impacted tooth, but only 4 of
classification (Philipsen 2005), it was explicitly stated that them were radiographically in a dentigerous relationship.
the orthokeratinised odontogenic cyst did not form ‘part of Almost always, however, this description has been based on
the spectrum of keratocystic odontogenic tumour’. We clinical and radiological findings and there are few reports
have discussed the role of PTCH and the sonic hedgehog that have properly documented a case of an orthokerati-
signalling pathway in the dentigerous cyst (Chapter 5) and nised cyst in a true dentigerous relationship, where the cyst
the point was made that, although PTCH alterations are lining is attached to the tooth at the cementoenamel junc-
important in keratocysts, they do not seem to be specific tion (Shetty et al. 2016).
and that there is evidence that PTCH may be involved in Nevertheless, the frequency of a dentigerous relationship
other odontogenic lesions (Gomes and Gomez  2011), has led to speculation that some orthokeratinised odonto-
including dentigerous cyst, radicular cyst, and orthokerati- genic cysts may arise from reduced enamel epithelium, and
nised odontogenic cyst (Levanat et  al.  2000; Pavelić form a truly keratinised dentigerous cyst. Vuhahula et  al.
et al. 2001; Vered et al. 2009). (1993) were the first to suggest this. They identified 12
Diniz et  al. (2011) investigated loss of heterozygosity orthokeratinised jaw cysts and found that 9 (75%) were in a
(LOH) of the PTCH gene in seven each of odontogenic dentigerous relationship with an unerupted tooth on radio-
keratocyst and orthokeratinised odontogenic cyst, and logical examination, but did not demonstrate attachment at
found a similar pattern of LOH in both, with 5 of 7 and 4 of the cementoenamel junction. In the discussion they also
7, respectively, showing LOH in at least one locus of the noted that odontogenic keratocysts may envelop a tooth and
PTCH region of chromosome 9q. The authors concluded be in a ‘pseudo-­dentigerous’ relationship. Despite this they
that PTCH gene alterations may not be specific to kerato- still proposed that orthokeratinised cysts in this relationship
cysts, but may be shared by a number of odontogenic represent true dentigerous cysts with orthokeratinisation.
­Histopatholog  207

In his series of 312 odontogenic keratocysts, Brannon odontogenic keratocyst, and 1 case was an orthokeratinised
(1976, 1977) showed that 83 (26.6%) were found, on radio- odontogenic cyst.
graphs, to be associated with an impacted tooth, but he only These data suggest that focal keratinisation in dentigerous
reported 2 cases with histological evidence that the cyst was cysts is rare, but that complete keratinisation, resulting in a
attached at the cementoenamel junction. In other cases, the diagnosis of keratocyst or orthokeratinised odontogenic cyst,
cyst was separated from the tooth by a zone of fibrous con- is more common (7 of 119 cases: 5.9%). This was also noted
nective tissue. This latter relationship has been described as by Brannon (1977), who found that about 8.5% of cysts in a
extrafollicular, and suggests that the cyst is ‘pseudo-­ dentigerous relationship were keratinised and that about 30%
dentigerous’ and has arisen from dental lamina rests in the of these were orthokeratinised. There is no evidence that
tooth follicle, and has then grown and enveloped an dentigerous (or other) cysts may progressively keratinise and,
unerupted tooth. It is also possible that a tooth may actually if they did, then the frequency of focal keratinisation should
erupt into an overlying cyst with fusion of the reduced be much higher than complete keratinisation.
enamel epithelium and the cyst lining to produce a true den- Taken together, these observations cannot exclude the
tigerous relationship. In this case Browne (1971a,  1991b) possibility that an orthokeratinised odontogenic cyst (or an
suggested that the epithelial lining immediately adjacent to odontogenic keratocyst) may arise by keratinisation of a
the tooth is simple and non-­keratinised, resembling reduced dentigerous cyst, but overall it seems to be very unlikely.
enamel epithelium, while the remainder is keratinising. We suggest therefore that the source of epithelium is rest
A further possibility is that a typical dentigerous cyst cells of the dental lamina, and that those cysts that occur in
may undergo progressive keratinisation to produce a histo- a dentigerous relationship result from an adjacent
logical appearance of a keratocyst or orthokeratinised unerupted tooth becoming enveloped by a cyst that has
odontogenic cyst. Yeo et al. (2007) examined 119 cysts that, arisen in the overlying dental follicle. In rare cases, a tooth
on radiological examination, were dentigerous. Almost all may erupt into the cyst, with fusion of the lining with the
(108: 90.8%) were lined by non-­keratinised stratified squa- reduced enamel epithelium.
mous epithelium, and only 2 (1.7%) showed focal areas of
keratin formation (both parakeratin). These data support
an early study by Browne (1972) that showed focal keratin ­Histopathology
formation in only 2 of 81 dentigerous cysts (2.5%). Mucous
metaplasia or ciliated cells were much more common and Orthokeratinised odontogenic cysts are most often removed
were seen in 14 (11.8%) cases. Yeo et al. (2007) also found conservatively by enucleation, so the pathologist may
that 6 cases were fully parakeratinised and diagnosed as receive multiple fragments of cyst wall (Figure 12.4). The

Figure 12.4  Orthokeratinised odontogenic


cyst. The pathologist will typically receive
multiple fragments. The cyst is lined by thin
regular epithelium. Areas of folding can be seen
as well as accumulations of exfoliated keratin. A
microcyst and islands of epithelium are seen in
this cyst (square) and are shown in Figure 12.8.
208   Orthokeratinised Odontogenic Cyst

Box 12.2  Orthokeratinised Odontogenic Cyst: Histopathology and Diagnostic Criteria


●● Lined by a thin, regular stratified squamous epithelium
●● Orthokeratinised with a granular cell layer
●● Prominent keratin that forms lamellae filling the lumen (‘onion-­skin’ appearance)
●● Basal layer of flattened or cuboidal cells
●● Surface is not corrugated and basal layer is not palisaded
●● Only occasional epithelial islands or microcysts (less than 5% of cases)
Caution: features that suggest an alternative diagnosis:
●● If the lining shows areas of parakeratin, typical of a parakeratinised odontogenic keratocyst, it should be diag-
nosed as odontogenic keratocyst
●● If orthokeratin is only focal and the lining is mostly non-­keratinised, consider an odontogenic cyst with metaplasia,
especially a dentigerous cyst
●● If the lesion is outside of the tooth-­bearing areas or is peripheral, consider a non-­odontogenic cyst, especially an
epidermoid cyst
●● Focal or small areas of sebaceous cells are acceptable, but if these are extensive, or if other dermal appendages
are present, consider a diagnosis of intraosseous dermoid cyst

specimen may also include the cyst contents, which are orthokeratinised with a prominent granular cell layer and it
composed of keratinaceous material that is cream or yel- is not corrugated. Keratinisation is prominent and the kera-
low coloured, with a cheesy, fatty, or ‘buttery’ texture and a tin forms thick lamellated sheets, sometimes described as an
characteristically unpleasant odour. Similar contents may ‘onion-­skin’ pattern (Figures  12.5–12.7a). Keratin sloughs
be found in odontogenic keratocysts, but will also be famil- off from the surface and may fill the cyst lumen (Figure 12.6).
iar to many pathologists as a characteristic feature of cuta- The basal layer is composed of flattened or cuboidal epithe-
neous epidermal cysts. lial cells, but the palisading and reversal of nuclear polarity
In the context of intraosseous jaw lesions, the histological seen in keratocysts are not seen in orthokeratinised odonto-
features are virtually diagnostic (Box 12.2). The cyst lining is genic cyst. Separation of the epithelium from the fibrous
composed of thin, regular stratified squamous cyst wall is commonly seen (Figure 12.7a). Often the cyst lin-
epithelium, five to eight cells layers thick, lying on a flat ing is folded and may resemble the typical folds seen in
basement membrane (Figures  12.4–12.6). The surface is keratocysts (Figures 12.4 and 12.7b).

Figure 12.5  Typical lining of the orthokeratinised odontogenic cyst. There is prominent keratinisation with a well-­formed granular
cell layer. The basal layer is composed of flattened or cuboidal cells (inset). Lamellated sheets of keratin slough off into the lumen.
­Histopatholog  209

Figure 12.6  Orthokeratinised odontogenic cyst. The lining is thin and regular. Keratin sloughs off from the surface of the lining
and may fill the lumen.

(a) (b)

(c) (d)

Figure 12.7  Variable appearances of the lining of the orthokeratinised odontogenic cyst. (a) Typical lining, but with evidence of
separation from the underlying fibrous cyst wall. (b) The lining may appear folded. (c) The cyst wall is inflamed and here the lining
may be thickened and non-­keratinised. (d) In this area the lining is thin and non-­keratinised and overlies an area of inflammation and
loose granulation tissue.
210   Orthokeratinised Odontogenic Cyst

Focal areas of the cyst may be parakeratinised or non-­ that the keratin profiles appear to reflect the histological
keratinised, but this is usually in areas where the cyst wall differentiation of the lesions. Simple keratins (especially
has become inflamed (Figure  12.7c, d). Wright (1981) K4/13, K17, and K19) characterised keratocysts that are
found focal areas of parakeratin in only 4 of 60 cases parakeratinised, while high molecular weight keratins
(6.6%), but non-­keratinised areas in 51 (85.0%) cases, often (K1/10) and loricrin were seen in the orthokeratinised
associated with areas of inflammation. He also found that odontogenic cyst. A number of studies have shown simi-
27 cases had areas with rete pegs, but these were most lar keratin profiles (Li et al. 1998; da Silva et al. 2002; Tsuji
prominent in areas of inflammation (14 cases) and only et  al.  2014), but all have also shown wide variations
rarely seen where the cyst was orthokeratinised. between individual lesions, suggesting that, although the
The cyst wall is composed of fibrous connective tissue results confirm differences between the cyst types, the use
that is usually uninflamed, although focal areas of inflam- of keratin immunocytochemistry may not help in the
mation may be seen in about half of cases (Wright 1981) diagnosis of individual lesions. Other studies have inves-
and focal accumulation of cholesterol clefts may also be tigated expression of proliferation and apoptotic markers
seen. Occasional small microcysts or islands of epithelium (Thosaporn et  al.  2004; Rangiani and Motahhary  2009;
may be seen in the cyst wall (Figure 12.8). The islands may Dong et  al.  2010) and have been discussed earlier.
resemble rests of dental lamina (Figure 12.8b) or may be Although there is a difference between orthokeratinised
islands of squamous epithelium with evidence of central odontogenic cysts and keratocysts in the number and dis-
keratinisation (Figure  12.8a). Islands or microcysts are tribution of cells expressing bcl-­2 and p63 (Dong
only seen in about 5% of cases (Wright 1981; Li et al. 1998) et al. 2010), these have been shown in lesions with typical
and are not as common as in keratocysts, about 20% of histological features, so their value in diagnosing lesions
which show solid islands or satellite cysts (Woolgar from small biopsies, or where the lining is altered by
et al. 1987a; Cottom et al. 2012). inflammation, is not known.
Immunohistochemistry does not play a role in diagno- Occasional unusual orthokeratinised odontogenic cysts
sis, since the immunophenotype reflects the degree of have been reported where the histological features suggest
keratinisation of the lining epithelium, which is clearly divergent differentiation or hybrid lesions. De Fátima
visible on a routine haematoxylin and eosin (H&E)-­ Bernardes et al. (2008) described an orthokeratinised odon-
stained section. Aragaki et al. (2010) undertook a compre- togenic cyst associated with a calcifying odontogenic cyst.
hensive study of the keratin profiles of orthokeratinised Most of the cyst was lined by ameloblastomatous epithe-
odontogenic cyst and odontogenic keratocyst, and found lium with focal accumulation of ghost cells, but areas of

b
Figure 12.8  A portion of the lining of the cyst illustrated in Figure 12.4. The wall contains a small microcyst filled with keratin
and islands of epithelium. The epithelial islands may show focal keratinisation (inset a, arrows) or may resemble rest cells of
dental lamina (inset b).
­Histopatholog  211

orthokeratin were noted. The authors felt that the lesion non-­odontogenic cystic lesions, especially dentigerous
represented an example of diverse differentiation of the lin- cyst, odontogenic keratocyst, and ameloblastoma. Faced
ing, rather than a true hybrid lesion. Argyris and Koutlas with these appearances, it is very unlikely that a clinician
(2017) described a similar lesion in a patient with Gardner would consider an orthokeratinised odontogenic cyst as a
syndrome. In this case, the cyst was lined by epithelium likely diagnosis. In their series of 55 cases, Crowley et al.
showing areas of parakeratosis and orthokeratosis, as well (1992) found that the most common preliminary diagno-
as areas resembling ameloblastoma with ghost cells and sis was dentigerous cyst (22 cases; 40%), followed by
areas of calcification. They interpreted the lesion to be an odontogenic keratocyst (12 cases; 21.8%). Other diagno-
orthokeratinised odontogenic cyst in association with an ses included ameloblastoma (1) and lateral periodontal
odontogenic keratocyst with ghost cells. This lesion could cyst (1) and 19 diagnoses of ‘cyst of undetermined origin’.
be dismissed as an unusual oddity, although Ide et al. (2019) Li et al. (1998) found that the clinical diagnoses of their
have described another lesion with multiple features, 15 cases included dentigerous cyst (4 times), odontogenic
including areas of calcifying odontogenic cyst, orthokerati- keratocyst (7), ameloblastoma (4), radicular cyst (2), and
nised odontogenic cyst, and odontogenic keratocyst, but undetermined cyst (2). These diagnoses reflect the rela-
also with areas of mucous cells and duct-­like structures. It tive frequency of the lesions. Since about 70% of lesions
was a unilocular cyst in a true dentigerous relationship with are associated with an impacted tooth, it seems inevita-
an impacted maxillary canine, and contained luminal nod- ble that the most common presurgical diagnosis is denti-
ules typical of calcifying odontogenic cyst as well as the gerous cyst, and the site predilection in the posterior
other features. The authors regarded it as an example of the mandible will always arouse suspicion of keratocyst or
diverse histomorphological features that may be seen in ameloblastoma. Most times, therefore, a diagnosis of
odontogenic cysts as a result of the pluripotentiality of orthokeratinised odontogenic cyst will be first encoun-
odontogenic epithelium. These cases are difficult to inter- tered on pathological examination and will be unex-
pret, but overall they probably represent examples of mor- pected by the surgeon.
phologically diverse cysts with areas of orthokeratin, rather It should be noted that the histology of the orthokerati-
than lesions that can be regarded as orthokeratinised odon- nised odontogenic cyst is almost identical to the epidermal
togenic cysts with unusual features. (‘sebaceous’) cyst of the skin. Occasional epidermoid cysts
Chi et  al. (2007) reported 5 cases of orthokeratinised have been reported in the jaws, but these are unusual (see
odontogenic cyst with focal areas of sebaceous glands. This Chapter 18). If an orthokeratinising cyst arises within the
is interesting, because usually a keratinising cyst with evi- tooth-­bearing areas, then a diagnosis of orthokeratinised
dence of dermal appendages (including sebaceous glands) odontogenic cyst can be given, but if it is outside the tooth-­
would be regarded as a dermoid cyst (see Chapter  18). bearing areas, or within the soft tissues, then an epider-
However, the 5 cases reported by Chi et  al. (2007) all moid cyst should be considered. Similarly, if dermal
showed clinical and radiological features typical of an appendages are found, even if the lesion is within the
odontogenic cyst, and the authors strongly argued that tooth-­bearing areas, then a diagnosis of dermoid cyst
they should be regarded as orthokeratinised odontogenic should be considered (Vuhahula et al. 1993; Box 12.2).
cysts with sebaceous differentiation. Vuhahula et al. (1993)
examined 12 orthokeratinised jaw cysts and found 2 that
Relationship to the Odontogenic Keratocyst
contained sebaceous cells. In one the sebaceous cells were
focal and few in number and the authors interpreted the As discussed above, faced with an individual lesion it is
case as an odontogenic cyst with sebaceous metaplasia. almost impossible to differentiate an orthokeratinised
The second cyst was unusually large and had prominent odontogenic cyst from an odontogenic keratocyst based
sebaceous glands, as well as other dermal appendages on the clinical and radiological features. Some of the
including hair follicles and sweat glands. This was diag- differences have been discussed previously and the differ-
nosed as an intraosseous dermoid cyst. More recently, Oh et ences and similarities between the two lesions are sum-
al. (2022) examined 65 orthokeratinised odontogenic cysts marised in Table 12.2. The orthokeratinised odontogenic
and found focal areas of sebaceous differentiation in 2 cases. cyst is slightly more common in the posterior mandible, is
more often in a dentigerous relationship, and is more
likely to be spherical, unilocular, and corticated. It is also
Histological Differential Diagnosis
less likely to present as a swelling and more likely to be an
The clinical and radiological features of the orthokerati- incidental finding than an odontogenic keratocyst
nised odontogenic cyst (Box 12.1) are not specific, since (MacDonald-­Jankowski 2010). This is in keeping with the
they are shared with many other odontogenic and observation that orthokeratinised odontogenic cysts are
212   Orthokeratinised Odontogenic Cyst

usually smaller at presentation (Dong et al. 2010). These may be seen (e.g. see Figure 12.7c, d), but in these cases
differences, however, are unlikely to be helpful in making immunocytochemistry does not help and further sections
a distinction between the two lesions when faced with a or blocks must be examined.
single patient.
Once a biopsy specimen has been examined, a diagnosis
Verrucous Odontogenic Cyst
of orthokeratinised odontogenic cyst is usually straightfor-
ward, since a finding of an orthokeratinised lining is virtu- There are a small number of case reports of a keratinising
ally diagnostic. However, orthokeratinised odontogenic odontogenic cyst with a verrucous or verruciform prolifer-
cysts may occasionally show focal areas of parakeratin and ation of the cyst lining. These are summarised in Table 12.3.
odontogenic keratocysts may show focal areas of orthoker- Few have been specifically diagnosed as orthokeratinised
atin. In Brannon’s (1977) early series of 312 odontogenic odontogenic cyst, but in most cases the lining has been at
keratocysts, he found that 9.7% would meet the criteria for least partially composed of orthokeratin.
an orthokeratinised odontogenic cyst, but that 22 cases Aldred et al. (2002) reported the first case in a 13-­year-­old
(7.1%) showed both parakeratin and orthokeratin. Because girl, in which the lining comprised a thin stratified squa-
the orthokeratin was focal, he regarded these lesions as mous epithelium with an area of markedly hyperplastic
odontogenic keratocysts. Crowley et al. (1992) also found a stratified squamous epithelium thrown up into a series of
small number of lesions with both para-­ and orthokeratin verrucous projections shedding keratin into the lumen
(7 of 449 cases: 1.6%) and also considered these to be (Figure 12.9). The cases of Ueeck et al. (2007) and Argyris
keratocysts. In his series of 60 orthokeratinised odonto- et  al. (2015a) were similar, with linings of ortho-­ or par-
genic cysts, Wright (1981) found 4 (6.6%) with focal areas akeratinised epithelium and focal areas of verrucous
of parakeratin. hyperplasia. The verrucous areas were exophytic and pro-
It appears therefore that there is no clear cut-­off for truded into the lumen, with no evidence of pushing or
how much orthokeratin must be present to determine a invasion into the cyst wall that may suggest verrucous car-
diagnosis of orthokeratinised odontogenic cyst. The WHO cinoma. All the lesions have been negative for human pap-
definition states that the lesion is lined by orthokerati- illomavirus (HPV), although the case of Argyris et  al.
nised odontogenic epithelium, but may show small areas (2015a) stained for p16. The case of Crane et  al. (2020)
of para-­or non-­keratinised epithelium, usually associated showed only small focal areas of verrucous hyperplasia in
with inflammation (Speight and Neville 2022). We would one cyst of a patient with three orthokeratinised odonto-
interpret this to mean that most of the cyst lining is com- genic cysts. The case is illustrated in Figure  12.3 and the
posed of orthokeratin. If the lining shows prominent verrucous lesion was associated with the left mandibular
areas of a typical parakeratinised odontogenic keratocyst second molar.
(see Chapter 7), then a diagnosis of odontogenic kerato- Lalla et al. (2016) examined the histology of 29 keratinis-
cyst should be given. As discussed above, immunocyto- ing jaw cysts and found 13 that met the criteria of orthokerati-
chemistry is of little help, because the cytokeratin profiles nised odontogenic cyst, while the remainder showed a
only reflect the pattern of keratinisation, which can be mixed keratin pattern. They also identified 5  in which at
clearly seen on a routine H&E-­stained section. Doubt least one-­third of the lining showed verruciform hyperpla-
may arise in a small biopsy or if the lining is inflamed, sia. This verruciform architecture was found in 3 of the 13
when only non-­keratinised or proliferative epithelium (23.1%) orthokeratinised odontogenic cysts, while the other

Table 12.3  Cases reported as keratinising odontogenic cysts with verrucous features.

References Age/Sex Site Histology Radiology

Aldred et al. (2002) 13/F Maxilla Mostly non-­keratinised Unilocular


Canine/premolar area Partially verrucous with parakeratin
Ueeck et al. (2007) 46/M Mandible Ortho-­and parakeratin Multilocular, corticated
Molar/ramus Mostly verrucous
Argyris et al. (2015a) 32/M Mandible Orthokeratin Multilocular, corticated
Molar/ramus Partially verrucoid
Crane et al. (2020) 23/M Mandible Orthokeratin Unilocular, corticated
Third molar region Small focal verrucous area
­Treatmen  213

Figure 12.9  An odontogenic cyst showing transition of


the epithelial lining to a verrucous hyperplasia. Source:
From the case reported by Aldred et al. (2002).

2 were in cysts with a mixed keratin pattern. All the cysts surface, it also showed bulbous ‘pushing’ rete pegs and the
were negative for HPV. This study suggests that verrucous lesion subsequently recurred and metastasised, suggesting
hyperplasia may be more common than previously reported, that it represented an unusual malignancy, possibly a ver-
but may have been overlooked as unimportant by reporting rucous carcinoma arising in an odontogenic cyst.
pathologists. In the paper by Lalla et al. (2016) only a portion
of one case is illustrated, and this shows a small area with
columns of parakeratin, described as a ‘verrucous-­like pat- ­Treatment
tern’. Such a feature may be regarded as focal hyperplasia
rather than true verrucous change by some pathologists. In reports where the treatment of the orthokeratinised
A further case was reported by Manaktala et al. (2017) in odontogenic cyst has been described, lesions have been
a 13-­year-­old male. This appeared to be similar to the previ- removed conservatively by enucleation. Recurrences are
ously reported lesions, but as well as an unusual verrucous rare and have been reported in less than 5% of cases.
214

13

Nasopalatine Duct Cyst

CHAPTER MENU
Terminology and the Anatomy of the Incisive Canal, 214
Clinical Features, 218
●● Frequency, 218

●● Age, 219

●● Sex, 219

●● Site, 219

●● Clinical Presentation,  220

Radiological Features, 221
●● Radiological Differential Diagnosis,  223

●● Median Palatal Cyst,  223

Pathogenesis, 225
Histopathology, 226
Treatment, 229

The nasopalatine duct cyst is a non-­odontogenic cyst that ­ erminology and the Anatomy of the
T
arises in the midline of the anterior maxilla, just posterior Incisive Canal
to the central incisors. The cyst is thought to arise from epi-
thelial remnants of the nasopalatine duct that are found in The terms incisive canal and nasopalatine canal are used
the incisive canal (Rich and Neville 2022). The first descrip- interchangeably to describe the bony channel that links the
tion of the nasopalatine duct cyst is attributed to Meyer, anterior oral cavity with the floor of the nose. Either term
who described it as a supernumerary midline paranasal can be used, but here we will refer to the canal as the
sinus (Meyer 1914) and subsequently as a median anterior ­incisive canal, since this term is more commonly used in
maxillary cyst (Meyer 1931). This latter term remained in the dental literature. For a more detailed account of the
the literature for many years and is still used in some text- anatomy and embryology of the incisive canal and related
books as a synonym. However, the association with the structures, readers should consult current textbooks of oral
nasopalatine duct led the World Health Organization anatomy and embryology. Early studies of the anatomy of
(WHO) classifications to adopt the term nasopalatine duct the region involved examination of dried skulls (Roper-­
cyst (Pindborg and Kramer  1971; Kramer et  al.  1992; Hall  1938; Chamda and Shear  1980) or fetal material
­El-­Naggar et  al.  2017; WHO  2022a). The cyst is found in (Abrams et al. 1963), but these studies yielded little detailed
association with the incisive canal and the alternative term information on the structure. More recently, the advent of
of incisive canal cyst is sometimes used. To fully understand cone beam computed tomography (CBCT) and interest in
the pathogenesis and clinical features of the nasopalatine the microanatomy of alveolar bone in the context of
duct cyst, it is helpful to have an appreciation of the termi- implant placement has resulted in more detailed studies of
nology, embryology, and anatomy of the incisive canal and the anterior maxillary region (Jacob et  al.  2000; Song
related structures. et al. 2009; Bornstein et al. 2011; Falci et al. 2013; Al-­Amery

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
­Terminology and the Anatomy of the Incisive Cana 215

Nasal septum

Nasopalatine foramen

Incisive canal

Incisive foramen Incisive foramen


(a) (b)

Figure 13.1  The anatomy of the incisive canal. The canal passes from the incisive foramen in the oral cavity to the nasopalatine
foramen in the floor of the nose. (a) Coronal view. The canal forms a Y shape with two foramina in the nasal floor separated by the
nasal septum. (b) Sagittal view. The canal may be vertical (solid lines) entering the nose perpendicular to the nasal floor, or may be
slanted (dotted lines) and enter the nasal floor more posteriorly.

et al. 2015; Friedrich et al. 2015; Lake et al. 2018). The ter- incisive canal is a Y-­shaped structure with a single opening
minology and key features of the region are summarised in on the oral side (the incisive foramen) and two openings on
Box  13.1, and Figure  13.1 presents a simple diagram to the nasal side (the nasopalatine foramina; Song et al. 2009;
illustrate the main anatomical features. Figures 13.1a and 13.2). However, between the single infe-
The incisive canal runs from the oral cavity to the floor of rior opening and the branching of the Y, the canal is often
the nose (Figure  13.1). In the oral cavity, the foramen or divided into two or more smaller channels or ‘canaliculi’
opening is in the midline of the anterior palate, posterior to (Bornstein et  al. 2011). Bornstein et  al. (2011) and
the incisor teeth and just deep to the incisive papilla. The ­Al-­Amery et  al. (2015) studied 100 and 96 subjects,
foramen is found in a shallow funnel-­shaped indentation respectively, by CBCT and found a single channel in 45%
of bone called the incisive fossae. The shape and size of the and 65% and two channels in 15% and 34%, respectively.
fossae and the foramen show considerable variations In  Bornstein’s study 40% of cases had multiple channels,
(reviewed by Abrams et  al.  1963), but in general it is the but Al-­Amery et al. (2015) found only one case with three
incisive foramen that is seen on anterior occlusal radio- channels. In a more a detailed micro-­computed tomogra-
graphs as a radiolucency in the anterior palate. The incisive phy (CT) analysis of dissected cadavers, Song et al. (2009)
canal passes superiorly and opens into a fossa in the floor found that 43% of incisive canals had one channel, 23% had
of the nose that is divided by the nasal septum into two two, 25% had three, and 9% had four. Occasionally, multi-
foramina, each referred to as a nasopalatine foramen or ple channels may be a serendipitous finding on CT exami-
foramen of Stensen.1 The foramina in the floor of the nose nation of the anterior maxilla (Figure 13.2). This complex
lie either side of the nasal septum and are found approxi- anatomy, and the presence of more than one channel
mately 20 mm inside the nasal cavity, at the junction of the within the bony canal, may explain why, on occasion, a
septum and the nasal floor (Jacob et  al.  2000). Thus the nasopalatine duct cyst is located off-­centre or lateral to the
midline.
A number of workers have determined the length and
1
A historical note: Niels Stensen (also known as Nicolas Steno) was a direction of the incisive canal. On average, the distance
Danish anatomist and geologist (1638–1686) who described a between the oral and nasal foramina is about 10 mm (Lake
number of anatomical and geological features. He is also known for et  al.  2018), but there is wide variation, with maximum
describing the duct of the parotid gland – called Stensen’s duct.
Occasionally both the duct and the foramen are incorrectly attributed lengths of 15.8 mm (Song et  al.  2009; South Korea),
to ‘Stenson’. 17.8 mm (Bornstein et  al.  2011; Switzerland), 19.8 mm
216 Nasopalatine Duct Cyst

(a) (Friedrich et  al.  2015; Germany), 21.3 mm (Etoz and


Sisman 2014; Turkey), and 30.2 mm (Al-­Amery et al. 2015;
Malaysia) (Table  13.1). Sekerci et  al. (2014) recorded a
mean length of 10.83 mm in a paediatric population
(<16 years), with a range of 2.4–16.1 mm. There is evi-
dence that the size and shape of the canal may vary
between different ethnicities (Table  13.1). The smallest
dimensions have been recorded in South Korea (Song
et al. 2009) and Switzerland (Bornstein et al. 2011) and the
largest in Malaysia (Al-­Amery et al. 2015). However, the
(b)
length of the canal is not a simple measure of the thick-
ness of the anterior maxillary bone, because the canal also
varies in direction and shape. Song et al. (2009) classified
the canals as vertical or slanted and each as curved or
straight. Vertical canals are the shortest and run vertically
upwards and perpendicular to the floor of the nose, while
slanted canals mostly run in an anterior to posterior direc-
tion, with an angle of up to 35° (Figure 13.1b). Song et al.
(2009) found that 60.7% of canals were vertical, of which
(c) about one-­third were curved. The remainder (39.3%) were
slanted and about one-­quarter of these were also curved.
Al-­Amery et  al. (2015), however, found that 96.6% of
canals were slanted and half of these were curved. The
slanted morphology means that many canals are longer
and may be orientated posteriorly towards the centre of
the palate. This explains why some nasopalatine duct cysts
may appear posterior to the incisive papilla and be diag-
nosed as ‘median palatal cyst’.
Figure 13.2  The anatomy of the incisive canal is illustrated in The incisive canal contains fibrous tissue and neurovas-
this series of axial computed tomography (CT) images from the cular bundles that provide nerve (the nasopalatine nerve)
same individual. (a) In the oral cavity, at the level of the incisive and blood supply (palatine arteries) to the anterior palate. It
foramen, there is a single channel. (b) In the midline of the canal
may also contain mucous glands, especially in the region of
three small channels can be seen, each with a corticated outline.
(c) In the floor of the nose there are two openings, the the incisive papilla and nasopalatine fossae in the nose. In
nasopalatine foramina, one on each side of the nasal septum. animals (in particular cats, cattle, horses, and some rodents),

Table 13.1  Dimensions of the incisive canal and related foramina.

Diameter of the incisive canal

Centre of Nasopalatine Incisive


Reference Country n Incisive foramen the canal foramen canal length

Bornstein et al. (2011) Switzerland 100 4.45 (1.5–9.7) NR 3.49 (0.9–8.8) 10.99 (5.9–17.8)
Etoz and Sisman (2014) Turkey 500 5.26 (1.8–13.7) NR 3.25 (0.4–6.7) 13.50 (3.8–21.3)
a
Sekerci et al. (2014) Turkey 368 4.13 (1.5–9.0) NR 2.53 (0.3–5.4) 10.83 (2.4–16.1)
Al-­Amery et al. (2015) Malaysia 90 3.49 (1.5–6.6) 3.85 (1.5–7.2) 6.06 (1.3–16.0) 16.33 (4.5–30.2)
Friedrich et al. (2015) Germany 200 4.49 (0.7–11.4) 2.48 (0.3–9.1) 3.43 (0.7–7.7) 11.15 (2.1–19.8)
Suter et al. (2016) Belgium 100 3.27 [1.20] 2.26 [1.08] 2.86 [1.10] 8.81 [1.89]

Data are maximum diameters or length: mean (range) or standard deviation [SD].
NR, not recorded.
a
 Sekerci et al. reported a paediatric population only (<16 years).
­Terminology and the Anatomy of the Incisive Cana 217

Box 13.1  The Incisive Canal: Key Features, Definitions, and Synonyms


Incisive canal: a bony channel in the anterior maxilla, passing from the oral cavity to the floor of the nose. May be
referred to as the nasopalatine canal. The canal is Y shaped – divided into two parts superiorly by the nasal septum.
About 50% of people may have two canals and rarely an individual may have three or four.
Incisive foramen: also called the incisive fossae. The opening of the canal in the oral cavity.
Incisive papilla: a soft-­tissue papilla, in the midline of the anterior hard palate just posterior to the central incisors. The
papilla overlies the incisive foramen.
Nasopalatine foramen: also called the nasopalatine fossae. The opening of the incisive canal in the floor of the nose.
There are actually two nasopalatine foramina, one on each side of the nasal septum. The foramina are also called the
foramen of Stensen.
Nasopalatine duct: an epithelial-­lined duct that lies in the incisive canal and joins the oral cavity to the nose. It is pat-
ent in many mammals, but in humans it usually disintegrates at about 10 weeks of fetal life and remains as epithelial
remnants. Very rarely an individual may have a patent duct.
Vomeronasal organ or Jacobson’s organ: an accessory olfactory organ found in mammals in the floor of the nose just
anterior to the nasopalatine foramen. It is vestigial in humans, but may be seen as a blind pouch in the floor of the nose.

the canal also contains an epithelial-­lined ­nasopalatine (Abrams et  al.  1963; Falci et  al.  2013; Lake et  al.  2018).
duct, which provides a patent conduit between the oral and These epithelial remnants may persist into adult life and
nasal cavities. On the nasal side the duct opens close to an give rise to the nasopalatine duct cyst (see ‘Pathogenesis’).
accessory olfactory organ called the vomeronasal organ Bodin et  al. (1986) examined histologically tissue extir-
(also called Jacobson’s organ) that is found bilaterally on pated from clinically and radiologically normal incisive
either side of the nasal septum. Together the nasopalatine foramina from 20 patients who had had a palatal flap raised
duct and vomeronasal organ provide an enhanced sense of for other reasons. Of the 20 cases, 10 (50%) contained epi-
smell for identifying prey or predators and for detecting thelial remnants, including oral and respiratory types.
pheromones for sex recognition and courtship. These authors also found that 6 (30%) of the normal speci-
In humans the nasopalatine duct and vomeronasal organ mens contained a small nasopalatine duct cyst. In 1 case
are usually lost during development, but vestigial structures, they also found a small mucous extravasation cyst.
including evidence of a nasal aperture and a blind pouch Many studies have been undertaken to determine the
representing the vomeronasal organ, may be found in the dimensions of the bony incisive canal. This is particularly
nasal mucosa in over 90% of individuals (Jacob et al. 2000). relevant to radiological examination and the criteria for
In addition, a small number of individuals have a patent diagnosis of a cyst. Early studies measured the dimensions
nasopalatine duct that traverses the canal and results in a on dried skulls (Roper-­Hall 1938; Chamda and Shear 1980)
direct communication between the oral cavity and nasal and showed that the incisive foramen is oval shaped, ori-
floor. The prevalence of this occurrence is not known, but entated with the greatest dimension in the antero-­posterior
von Arx et al. (2018) documented 4 cases and reviewed 37 plane. The average dimensions were approximately 10 mm
publications reporting 57 cases between 1881 and 2016. antero-­posteriorly and 5 mm wide, although the greatest
Patent ducts have been found in all age groups and are often dimension was 17.5 mm (Chamda and Shear 1980).
symptomless, although patients may be aware of the passage More recent studies have been highly detailed and have
of air or secretions, or may be able to elicit a squeaky or been undertaken using CBCT (Song et  al.  2009; Bornstein
whistling noise. In 60% of the reports there were two bilat- et al. 2011; Al-­Amery et al. 2015; Friedrich et al. 2015; Suter
eral openings of the duct in the oral mucosa on either side of et al. 2016). These studies have measured the dimensions of
the incisive papilla. Treatment was rarely needed. the canal and also of the associated foramina. The dimen-
In most humans, however, the nasopalatine duct is only sions on imaging have been shown to be lower than actual
found during fetal life and is lined by simple stratified measurements on dried specimens (Chamda and
squamous (oral-­type) epithelium or by pseudostratified Shear 1980) and therefore the imaging data is of more value
columnar ciliated (respiratory-­type) epithelium. After to the diagnostic radiologist. The dimensions are summa-
about nine weeks of intrauterine life the duct disappears, rised in Table 13.1. The incisive foramen has a mean diame-
but epithelial remnants and mucous glands remain ter of between 3.5 and 5.3 mm and a maximum dimension of
218 Nasopalatine Duct Cyst

about 13.5 mm (Etoz and Sisman 2014). All the studies have have been taken from a number of selected larger case
shown that the dimensions of the canal, including diameter series that are summarised in Tables 13.2 and 13.3.
and length, are greater in males than in females, but the dif-
ference rarely exceeds 1 mm. The diagnostic ­relevance of the
Frequency
dimensions of the incisive foramen is discussed later in this
chapter (see ‘Radiological Features’). The nasopalatine duct cyst represents about 4% of cysts of
the jaws, but is by far the most common of the non-­
odontogenic cysts. Shear’s series from the University of the
­Clinical Features Witwatersrand shows the highest frequency, where it con-
stituted 404 (11.6%) of the 3498 jaw cysts registered over a
Most descriptions of the nasopalatine duct cyst are to be 46-­year period and was slightly more common than the
found in short case reports that often record unusual or odontogenic keratocyst (see Table  1.1). In other series,
interesting findings that may not be typical. There have however, the frequency has ranged from about 1% to 5.5%.
also been a number of case series that record the overall In two large studies from Sheffield, Jones and Franklin
features of this lesion. Descriptions of the clinical features (2006a,b) reviewed almost 50 000 oral and maxillofacial

Table 13.2  Nasopalatine duct cyst. Age, sex, and diameter, from selected case series.

Age Diametera

Reference Country n Mean Range Males (%) Mean (mm) Range

Allard et al. (1981a) Netherlands 22 44.1 11–71 59.1 12.8 5–22


Bodin et al. (1986) Sweden 70 45.0 8–77 67.1 NR 4–40
Anneroth et al. (1986) Sweden 32 54.0 23–88 65.6 15.0 8–25
Nortjé and Wood (1988) S Africa 46 31.0 7–61 75.0 32.0 9–52
Swanson et al. (1991) USA 334 42.5 9–84 54.2 17.1 6–60
Vasconcelos et al. (1999a) Brazil 31 37.4 20–69 77.4 NR NR
Escoda Francolí et al. (2008) Italy 22 46.0 16–73 54.5 14.0 NR
Suter et al. (2015) Switzerland 40 51.3 7–80 62.5 15.0 7–47
Barros et al. (2018) Brazil 30 42.7 17–90 50.0 23.7 NR
a
 In all studies the size is recorded as the maximum diameter of the lesion.

Table 13.3  The frequency of clinical signs and symptoms (%) reported in selected case series of nasopalatine duct cysts.

Clinical signs or symptoms

References n No symptomsa Swelling Pain Infectionb

Allard et al. (1981a) 22 32 55 23 NR


Bodin et al. (1986) 70 20 49 51 37
Anneroth et al. (1986) 32 69 38 31 22
Swanson et al. (1991) 334 29 52 20 25
Vasconcelos et al. (1999a) 31 87 3 1 10
Escoda Francolí et al. (2008) 22 64 18 18 18
Suter et al. (2015) 40 35 25 20 10
Average 52 34 23 20
a
 Usually reported as an incidental finding on clinical examination or radiology.
b
 Signs of infection include fistula, discharge, or foul taste. Totals are over 100% because more than one symptom may be present.
­Clinical Feature 219

biopsy specimens and found a total of 254  nasopalatine from 7 to 90 years, with a mean age of 30–55 years. The age
duct cysts. These represented 3.5% of all jaw cysts. The fre- distribution of 227 of the University of the Witwatersrand
quency in children (2.9% of all jaw cysts; n  =  586; Jones cases is given in Figure 13.3 and shows a peak in the fourth
and Franklin 2006b) was slightly less than in adults (3.6% decade. Cases in children and adolescents are rare.
of all jaw cysts; n  =  6634; Jones and Franklin  2006a).
In  other series, nasopalatine duct cysts have constituted
Sex
from 0.74% (Italy; Aquilanti et  al.  2021) to 5.6% (Kuwait;
Ali 2011) of total jaw cysts. Other frequencies include 1.3% Almost without exception, studies have shown a predilec-
(Soluk Tekkeşin et  al.  2012b; Turkey), 2.2% (Grossmann tion for males, with overall about 65% of lesions being
et al. 2007; Brazil), 2.9% (Tamiolakis et al. 2019; Greece), found in males (Table 13.2). In our material (Figure 13.3)
and 4.0% (Daley et al. 1994; Canada). there were 156  males (68.5%), and other studies have
As a proportion of non-­odontogenic cysts, the nasopala- shown from 54% to 77% in males (Table 13.2).
tine duct cyst represents between about 30% and 70%.
Daley et  al. (1994) found 403  non-­odontogenic cysts in a
Site
review of 40 000 oral biopsies, of which the nasopalatine
duct cyst was the most frequently diagnosed, comprising The nasopalatine duct cyst is always located in the mid-
295 (73.4%). In other studies, nasopalatine duct cyst has line of the anterior palate. Clinically the cyst presents as a
constituted 39.1% (Jones and Franklin 2006a,b) and 32.8% swelling on, or just posterior to, the incisive papilla.
(Nonaka et al. 2011) of non-­odontogenic cysts. Almost without exception, radiology shows that the cyst
The reason for the variability in reports of frequency is is located within the incisive canal. In rare cases, how-
probably due to the inclusion criteria for the total number ever, the cyst may arise in the superior/posterior aspect of
of cysts used in the denominator. Some studies only include the canal, close to the nasal floor and towards the centre
cysts in the bone of the jaws, while others have incorpo- of the palate (see Figure  13.1b). In this case a plane
rated a wider range of maxillofacial cysts (e.g. lymphoepi- occlusal radiograph may show the cyst located posterior
thelial cysts or epidermoid cysts). to the shadow of the normal incisive foramen. A sagittal
CT, however, will show that the cyst is continuous with or
located within the superior aspect of the canal. The cyst is
Age
almost always located in the midline of the palate, but
Nasopalatine duct cysts are found in adults, but the age occasionally it may be located laterally if it has arisen in a
range is wide (Table 13.2). The age of presentation ranges secondary canal.

70

61
60

19
50
43 43
No. of cases

40
40
12 Females
14
13 Males
30

42 19
20
16
31 6
5 29
27
10
11 13
3
1 1
0
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89
Age

Figure 13.3  Age and sex distribution of 227 patients with nasopalatine duct cysts.
220 Nasopalatine Duct Cyst

Clinical Presentation sometimes describe a salty taste, or it may be purulent and


patients complain of a foul taste. Although swelling or ten-
Clinical signs and symptoms vary, but in a significant pro-
derness is often found on clinical examination, it may not
portion of patients the lesion is symptomless and may be
be appreciated by the patient. Anneroth et al. (1986) found
found as an incidental finding during a clinical or radio-
that although clinical signs were found in 75% of cases (24
logical examination for another purpose (Table 13.3). The
of 32), 22 (69%) cysts were symptomless and were an inci-
most common sign or symptom is swelling in the anterior
dental finding on clinical or radiological examination.
region of the midline of the palate (Figures 13.4 and 13.5).
Displacement or splaying of the incisor teeth is observed
Occasionally the swelling may extend to the midline on the
fairly often.
labial aspect of the alveolar ridge (Figure  13.6). In some
Occasionally cysts are superficial and appear to affect
cases, ‘through and through’ fluctuation may be elicited
only the palatine papilla (incisive papilla cyst). In these
between the labial and palatal swellings. The cyst may also
cases, there may be a history of recurrent swellings that
produce bulging of the floor of the nose. Pain or tenderness
periodically discharge and then ‘go down’ and recur. Some
is less often encountered, but when present is usually
of these superficial lesions may not be true nasopalatine
accompanied by swelling and evidence of infection.
duct cysts, but may represent a superficial mucocele (see
Various combinations of swelling, pain, and discharge may
‘Pathogenesis’).
occur. The discharge may be mucoid, in which case patients
The swellings are usually located in the anterior maxilla,
just behind the incisor teeth, and rarely exceed 20 mm in
diameter (Figure 13.4), although radiological diameters of
up to 60 mm have been recorded (Swanson et  al.  1991;
Table 13.2). Occasionally the lesion may be large and may
extend towards the middle of the palate (Figure  13.5).
Midline swellings of the palate that occur further posteri-
orly have sometimes been diagnosed as ‘median palatal
cysts’ and have been thought to be a specific type of mid-
line fissural cyst. However, they are probably posteriorly
located nasopalatine duct cysts that have arisen towards
the postero-­superior aspect of a slanting incisive canal (see
Figure 13.1b). The median palatal cyst is discussed later in
this chapter.
In general, symptoms are not severe and patients often
disregard them for many years. The cyst may also be com-
Figure 13.4  Nasopalatine duct cyst. A typical lesion presenting pletely symptomless and be discovered fortuitously by the
as a soft ovoid swelling in the midline of the anterior maxilla,
posterior to the central incisor teeth. Source: Courtesy of the late dentist during routine radiological examination, and occa-
Prof. J.J. Pindborg. sionally the presence of a cyst may become apparent only

Figure 13.5  Large nasopalatine duct cyst extending posteriorly Figure 13.6  Nasopalatine duct cyst producing a midline
to involve much of the anterior third of the hard palate. swelling of the labial aspect of the alveolar ridge.
­Radiological Feature 221

after dentures are placed. Sometimes the cyst may become roots and the lamina dura are intact (Figure  13.7a, b).
apparent due to trauma from the lower anterior teeth, and Larger cysts cause the incisor tooth roots to diverge and
this has also been thought to be an aetiological factor. become splayed (Figures  13.7a and  13.8; Anneroth et  al.
However, a history of trauma is not often reported and 1986; Bodin et al. 1986; Escoda Francolí et al. 2008).
when it has been seen, it is only in up to 16% of patients Cysts that arise higher in the incisive canal may appear
(Bodin et al. 1986; Anneroth et al. 1986). above the apices of the incisor teeth and be located towards
the floor of the nose (Figure 13.7b). Escoda Francolí et al.
(2008) reported the features of 22 cysts and found that 15
­Radiological Features (68.2%) were palatal to the incisors and that 7 (31.8%) were
located above the apices towards the nasal regions. Very
The nasopalatine duct cyst is always located in the midline large cysts may extend posteriorly and superiorly and may
of the anterior palate and presents most often as a well-­ be associated with labial swelling (Figures 13.6 and 13.8),
demarcated, round or oval radiolucency (Figure  13.7). or even swelling in the floor of the nose.
Between 75% and 85% of lesions are round or oval The margins of nasopalatine duct cysts are well demar-
(Anneroth et al. 1986; Bodin et al. 1986), with a slight pre- cated, but exhibit varying degrees of cortication (Bodin
dilection for an oval or slight ‘pear shape’ (Figure 13.7a). Of et al. 1986; Nortjé and Wood 1988). Bodin et al. (1986) found
the remainder, up to 20% (Bodin et  al.  1986) may appear that 80% of their 70 cysts had a well-­demarcated border that
heart shaped, either because they become notched by the was corticated to varying degrees. Most cases (60%) were
nasal septum during their expansion or because the ante- completely surrounded by a clearly perceptible corticated
rior nasal spine is superimposed on the radiolucent area margin (Figures 13.7b and 13.8b), whereas in others the infe-
(Figure 13.7c). rior border may be less distinct or not visible (Figures 13.7a
Most cysts arise within the inferior (oral) aspect of the and  13.8a). In 2 of their 70 cases, the cysts were located
incisive canal, palatal to the central incisors, and on a plain entirely within the soft tissues of the palatine papillae and
radiograph the radiolucency may be superimposed over were not visible on radiographs (Bodin et al. 1986).
the tooth roots. A key feature of the nasopalatine duct cyst, The nasopalatine duct cyst occurs in the incisive canal
however, is that the periodontal ligament of the involved and on radiological examination it may be difficult to

(a) (b) (c)

Figure 13.7  Variable radiological features of the nasopalatine duct cyst. (a) The cyst is oval or slightly pear shaped. Note that the
inferior border is less corticated than the superior margin. The roots of the maxillary incisor teeth are displaced laterally. (b) A rounded
radiolucency located above the apices of the teeth. (c) The cyst is heart shaped. A key feature is that the periodontal space and lamina
dura remain intact (arrows) even when the tooth apex appears to be in the cyst (a).
222 Nasopalatine Duct Cyst

(a) (b)

Figure 13.8  Radiographs of large nasopalatine duct cysts. (a) Large cysts may cause splaying or separation of the incisor roots. This
cyst is corticated at the superior margin, but is indistinct inferiorly. (b) A rare, very large cyst almost filling the hard palate and abutting
the nasolacrimal canals.

decide whether a radiolucency in the anterior maxilla is a The mean maximum diameter is recorded as between
small cyst or a normal incisive foramen. As shown in about 13 mm (Allard et al. 1981a) and 32 mm (Nortjé and
Table 13.1, many studies have recorded the dimensions of Wood 1988), with a range from 4 to 60 mm. Most studies,
the incisive canal, but it is the incisive foramen that is seen however, agree that the majority of lesions are small and
on occlusal radiographs. Overall, the studies summarised rarely exceed 20 mm.
in Table 13.1 show a mean diameter of the incisive foramen Anneroth et  al. (1986) found that 24 of their 32 cysts
of between 3.5 and 5.3 mm and a maximum diameter of (75%) were 15 mm or less in diameter, and only 7 (21.9%)
13.7 mm. Most studies, however, suggest that a normal were greater then 15 mm. Only one cyst was found to be
foramen over 6 mm in diameter is unusual. Roper-­Hall less that 10 mm. Swanson et al. (1991) recorded the size dis-
(1938) examined 2162 dried skulls and found that only 7 tribution of 116 of their cysts and found that 75% (87 cases)
(0.3%) were larger than 6 mm. All 7 were oval, with maxi- were 20 mm or less and 50% were between 11 and 20 mm.
mum antero-­posterior dimensions between 8 and 15 mm. Only 8 cysts were 6 mm or less in diameter. Of these, 7 were
He concluded that any radiograph that showed a width of diagnosed because the patient had symptoms. Bodin et al.
6 mm or less may be considered to be within normal limits, (1986) studied 70  nasopalatine duct cysts and found that
provided the patients have no other symptoms. Chamda the maximum dimension was 40 mm, but that 79% (55)
and Shear (1980) agreed with this finding and showed that were 12 mm or less. Only 2 lesions (3%) were found to be
incisive foramen widths of greater than 6 mm were only less than 6 mm in diameter and both were found within the
found in 148 (15.3%) of 970 specimens, but that antero-­ incisive papillae.
posterior dimensions of greater than 7 mm were found in The use of CT or CBCT may be valuable in the investiga-
799 (82.4%). tion of nasopalatine duct cysts, especially to appreciate the
These data show that the normal incisive foramen is usu- relationship of larger lesions to the teeth and to the floor of
ally oval in shape and may have a width up to 6 mm, but the nose and other vital structures. Suter et al. (2011, 2015)
antero-­posterior dimensions of as much as 10 mm or more have undertaken detailed CBCT analyses of nasopalatine
are also found. Bodin et  al. (1986) proposed that if the duct cysts. They found that CBCT was particularly useful
width of the foramen was less than 6 mm, then a cyst was to determine the relationship of the cyst to the apices of the
very unlikely. If a radiolucency was between 6 and 8 mm, teeth, and enabled informed decisions regarding the need
then further observation may be indicated, and if greater for preoperative endodontic treatment (Suter et al. 2015).
than 8 mm and with a thin corticated border, exploratory They found that 15% of patients required endodontic treat-
surgery should be considered. Radiolucencies exceeding ment because of the involvement of tooth apices in the
14 mm in diameter (width) were always cysts. lumen of larger cysts. Overall, however, they found no cor-
A number of studies have measured the size of nasopala- relations between the diameters or volume of cysts and
tine duct cysts, and these are summarised in Table  13.2. clinical symptoms, although larger cysts were more likely
­Radiological Feature 223

to have postoperative complications (Suter et  al.  2011). are of inflammatory origin and are actually radicular cysts in
These data suggest that although CBCT is able to provide the median maxilla (Tsuneki et al. 2013; see ‘Pathogenesis’
more information, it has few advantages over conventional for further discussion). Nevertheless, if the diagnostic crite-
radiology for treatment planning for the majority of lesions. ria are followed (Box 13.2), a correct diagnosis of nasopala-
CBCT may, however, be of value for careful analysis of tine duct cyst should be possible in the vast majority of cases.
large lesions, where knowledge of the relationships to adja- Occasionally, however, it may not be possible to distinguish
cent structures is helpful for surgical planning. between a nasopalatine duct cyst and a radicular cyst. This
difficulty is most likely to arise if a nasopalatine duct cyst is
laterally placed, since a midline radicular cyst is rarely
Radiological Differential Diagnosis
encountered.
The nasopalatine duct cyst shows characteristic clinical, Although it is possible that other jaw lesions may arise in
radiological, and histological features (Box 13.2) that, when the anterior palate, it is very unusual. Of the odontogenic
considered together, enable an accurate diagnosis in almost cysts, the keratocyst appears to be the most likely to arise at
all cases. Although the radiological features are typical, a this location. Woo et al. (1987) reported a single case of a
number of other lesions may arise in, or close to, the mid- keratocyst that occurred in the anterior midline of the
line of the anterior palate and be mistaken for a nasopala- maxilla, and Neville et al. (1997) reported 18 cases at this
tine duct cyst. site. Many of these lesions had been diagnosed clinically or
The most common alternative diagnosis that must be radiologically as nasopalatine duct cyst and images show
considered is radicular cyst. In establishing a diagnosis of radiological features indistinguishable from those illus-
nasopalatine duct cyst, it is important to attempt to exclude trated in Figures 13.7 and 13.8. Faced with these features, it
the possibility of a periapical lesion. Clinically and radio- would not be possible to distinguish these keratocysts from
logically, nasopalatine duct cysts may occasionally be a nasopalatine duct cyst. The only distinguishing feature
found lateral to the midline, or may extend over the apical was that 90% of Neville et al.’s cases occurred over the age
regions of the incisor teeth and be indistinguishable from a of 60 years, with a mean age of 69.9 years, which is one or
radicular cyst. The key diagnostic features are that in a two decades older than seen in nasopalatine duct cyst
nasopalatine duct cyst the incisor teeth should be vital on (Table  13.2). Of relevance to the above discussion, it is
pulp vitality tests and the lamina dura and periodontal interesting to note that Neville et al. (1997) included repre-
space should be intact on radiographic examination. sentative radiographs from four cases and all four showed
However, the lamina dura is not always clearly visible (e.g. that an associated tooth had been root treated.
see Figures 13.7c and 13.8b), and a review of the images in
case reports will show that it is not uncommon to see that
Median Palatal Cyst
one or more of the associated teeth have been root filled.
In their series of 22 cases, Escoda Francolí et al. (2008) A further consideration in the differential diagnosis is the
showed that in 8 (36.4%) cases an associated tooth had a median palatal cyst (often also call the median palatine
necrotic pulp or had been root canal treated. In a similar cyst). The very existence of this cyst is controversial. It is
study, Suter et al. (2015) investigated 40 cases by CBCT and thought to be a fissural cyst, arising from epithelial rem-
found that 15 (37.5%) patients had a root filling in an asso- nants entrapped in the midline of the developing palate.
ciated tooth. In a study of 41 lesions, Tsuneki et al. (2013) The hard palate forms in the seventh to eighth week of
showed that a tooth root was close to the lesion in 18 intrauterine life by the fusion of the palatal shelves or pro-
(43.9%) cases and that in 9 of these the tooth was non-­vital cesses. The shelves come into contact above the tongue
by electric pulp tests, although some of these cases (num- along the midline, and to fuse fully the epithelial covering
ber not given) had been root canal treated. of the shelves must disintegrate to allow merging of the
It is difficult to know how to interpret these data. Clearly, mesenchyme to form the secondary palate. At first the epi-
in most cases the cyst had not been removed and a tooth in thelium merges, but then breaks up and fragments into
close proximity to the cyst lumen had been root treated. This multiple small epithelial remnants. It is the existence and
suggests that a (mis)diagnosis of radicular cyst had been location of these remnants that are controversial and fuel
made on finding a radiolucency close to the tooth apex. It is the debate on the existence of cysts in the palatal midline.
also possible that a tooth may be rendered non-­vital by the It is thought that the remnants ‘dissolve’ or disintegrate
presence of a cyst that impinges onto the apex, especially if and disappear at or shortly after birth. The actual process
the cyst itself were to be secondarily infected. Others, how- involves an epithelial–mesenchymal transformation,
ever, have proposed that the close association of the cyst to whereby the epithelial cells change into mesenchymal cells
non-­vital teeth suggests that some nasopalatine duct cysts and disappear. However, there is good evidence that
224 Nasopalatine Duct Cyst

epithelial rests may persist in soft tissues of the midpalate one-­third or more of nasopalatine duct cysts (see
in the first year of life, and these remnants give rise to the ‘Histopathology’ and Table 13.4).
midpalatal raphe cyst or Epstein pearls. The formation of Most authors therefore do not accept the concept of
the palate and these cysts are discussed in detail in intraosseous midpalatal fissural cysts and regard all
Chapter 9. Although it is accepted that Epstein pearls arise ‘median palatal cysts’ as variants of nasopalatine duct cyst.
from epithelial remnants following palate formation, these Nevertheless, a number of standard textbooks on oral and
cysts are found only in neonates and there is very little evi- maxillofacial pathology still include the median palatal
dence that remnants remain or can give rise to cysts in cyst as a rare, but controversial, entity (Neville et al. 2016;
adults. Because of doubts about the fissural pathogenesis, Woo 2016), while others either do not mention it, or regard
the concept of the median palatal cyst fell out of favour and it as a posterior extension of the nasopalatine duct cyst
there have been few reports after 1985. In the 1992 WHO (Regezi et al. 2017).
classification it was clearly stated that the existence of the Some authors, however, have shown a renewed enthusi-
cyst was in doubt and the ‘fissural cysts’ were not included asm for this cyst (Hadi et  al.  2001; Manzon et  al.  2009;
(Kramer et al. 1992). Allmendinger et  al.  2009; Bacci et  al.  2011; Rangaswamy
Gingell et al. (1985) reviewed 17 cases of median palatal et al. 2018). In part, this is because CT or CBCT imaging in
cyst reported between 1930 and 1985. The cysts were the sagittal plane can more accurately determine the
described as fissural cysts that presented in the midline of antero-­posterior relationship of the cyst and the incisive
the palate posterior to the incisive papilla. These authors canal. Sadly, although many of these authors cite Gingell’s
suggested five criteria that must be met for a diagnosis of a five criteria, they do not meet them, and imaging often
true median palatal cyst: shows a large lesion in the anterior maxilla obliterating the
incisive foramen. Bacci et al. (2011) report a case with CT
1) The cyst must arise in the midline of the palate.
imaging that they describe as separate from the incisive
2) It should be located posterior to the palatine (incisive)
canal, but their illustrations suggest a direct communica-
papilla.
tion with the superior/posterior aspect of the canal and
3) It is an ovoid or circular radiolucency.
also with the nasopalatine foramen in the floor of the nose.
4) It is not associated with a non-­vital tooth or with the
Hadi et al. (2001) show a case that fills the anterior maxilla
incisive canal.
and histologically resembles an orthokeratinised odonto-
5) There should be no histological evidence of nerve
genic cyst. Both these authors and others (Rangaswamy
trunks, large vessels, cartilage, or mucous glands in the
et al. 2018) have cited a lack of large nerves or vessels as
cyst wall.
supportive of their diagnosis of median palatal cyst, but
It can be seen that these criteria are not specific and all this finding does not exclude a nasopalatine duct cyst
except number four would be consistent with a nasopala- (Table 13.4).
tine duct cyst. If the median palatal cyst truly exists, it must Allmendinger et  al. (2009) have reported a well-­
be posterior to the incisive papilla, and on radiology it must demarcated intrabony cyst in the centre of the hard palate.
be shown to not be associated with the incisive canal. Most They illustrated a sagittal CT that shows the cyst located in
case reports of median palatal cyst illustrate a clinical the hard palate with no communication with the incisive
swelling posterior to the incisive papilla, but almost all canal, which is clearly visible approximately 10 mm ante-
show radiographs of a lesion that impinges on, or is super- rior to the lesion. This report does suggest that a true
imposed over, the incisive canal and is indistinguishable median palatal cyst may exist, and that the defining crite-
from a nasopalatine duct cyst. Although most nasopalatine rion to distinguish it from a nasopalatine duct cyst should
duct cysts arise towards the inferior (oral) aspect of the be a CT to show that it does not involve the incisive canal.
incisive canal, close to the incisive papilla, some may arise Note, however, that the lesion was not removed and the
towards the posterior/superior aspect of the canal (Escoda authors did not illustrate any histology.
Francolí et al. 2008; Figure 13.1b) and in these cases, the Taken together, the literature rarely offers strong evi-
clinical findings may suggest a cyst posterior to the papillae dence for the existence of a median palatal cyst as a sepa-
and a plain occlusal radiograph may show a cyst posterior rate and independent entity. We would support the views
to the incisive foramen. of Rapidis and Langdon (1982) four decades ago that most
Many authors also cited an absence of large nerves of these cysts represent large or posteriorly placed naso-
and other structures (criterion 5) as evidence that a palatine duct cysts, while others represent the random
median palatal cyst has not arisen in the incisive canal localisation of other cyst types. However, there are few
and must be a separate entity to nasopalatine duct cyst. immutable rules in biology and it remains possible that
However, these structures may be absent in up to such a cyst may exist. The case of Allmendinger et  al.
­Pathogenesi 225

(2009) is one of the most convincing, and further reports, non-­vital teeth: first, that a nasopalatine duct cyst may
with high-­resolution imaging and histology, may shed fur- expand to resorb the lamina dura and involve the apices of
ther light on this conundrum. the tooth, which is then rendered non-­vital; secondly, that
a radicular cyst associated with a central incisor expands
into the incisive canal and is indistinguishable from a naso-
­Pathogenesis palatine duct cyst; and thirdly, that chronic periapical
infection at the apices of an incisor involves the incisive
Nasopalatine duct cysts arise from epithelial remnants of canal and initiates cyst formation by inflammatory stimu-
the nasopalatine duct in the incisive canal. The anatomy of lation of the epithelial remnants of the nasopalatine duct.
the incisive canal and nasopalatine duct have been dis- These possibilities are speculative, but cannot be dismissed,
cussed earlier in this chapter (Figures 13.1 and 13.2) and it and it has to be accepted that occasionally it may not be
was shown that there is good evidence that epithelial rem- possible to distinguish between a nasopalatine duct cyst
nants may persist into adult life. Bodin et al. (1986) demon- and a radicular cyst.
strated that 50% of normal canals contained epithelial The fact that mucous glands develop in association with
remnants, and we and others (Abrams et  al.  1963) have nasopalatine ducts and are sometimes seen in the walls of
noted epithelial cell rests in the wall of up to about one-­ the cysts has led to the suggestion that some nasopalatine
third of nasopalatine duct cysts. duct cysts are in fact a retention or extravasation type of
Although it is widely accepted that the nasopalatine duct mucocele. A number of authors have considered this
cyst arises from epithelial remnants in the incisive canal, the mechanism, but have dismissed it on the basis that mucus
factors associated with cyst formation are poorly understood is rarely found in the lesion, there is rarely an association
and the pathogenesis is largely speculative. It has been sug- between the cyst lumen and glandular ducts, and histology
gested that trauma or bacterial infection could stimulate the does not show mucous extravasation nor a lining of duct
nasopalatine duct remnants to proliferate. There is, how- epithelium (Abrams et  al.  1963; Allard et  al.  1981a;
ever, very little evidence to support such hypotheses, and a Anneroth et al. 1986; Bodin et al. 1986). These authors also
number of observations tend to preclude these mechanisms. felt that the secretory pressure that would exist in a
Very few reports record trauma as a significant clinical find- mucocele is unlikely to be sufficient to produce bone
ing and when it has been noted it has only been reported in resorption and form an intraosseous cyst. As discussed pre-
up to 16% of cases (Bodin et al. 1986; Anneroth et al. 1986). viously, it has also been noted that mucoceles in the ante-
If trauma to the area during mastication were the cause, it rior palate must be extremely rare, since large series have
would not explain why the cysts are found so infrequently not recorded a single case (Hayashida et  al.  2010; Chi
when such trauma is common, and it cannot explain the et al. 2011).
male predilection. It is also noted that although many cysts More recently, Tsuneki et al. (2013) found small mucous
show inflammation in the wall, it is not consistent and when glands in about half of their cysts and showed that the
present is often focal and mild. Occasionally this can be immunohistochemical profiles of the glandular ducts and
shown to be associated with secondary infection and may the cyst lining were similar. They also found ‘daughter cysts’
also be a result of trauma to the developed cyst, but it would with similar features, but on review of the text and images,
not appear to be an aetiological factor. these appear to be dilated ducts or cross-­cut ­sections of the
These observations do not of course definitely exclude cyst walls. Nevertheless, Tsuneki et al. (2013) suggested that
the possibility of an inflammatory origin, as the inflamma- the similar immunohistochemical profiles were evidence
tory process could have subsided before the cyst was that up to 50% of nasopalatine duct cysts are in fact a type of
removed. As discussed previously (see ‘Radiological mucous retention cyst. Based on these findings and on their
Differential Diagnosis’), on occasion it may not be possible finding of associations with non-­vital teeth, these authors
to distinguish between a nasopalatine duct cyst and a have dismissed the concept of the nasopalatine duct cyst
radicular cyst, and a number of studies have demonstrated and propose that cysts in the midline of the anterior palate
an association with non-­vital teeth. In particular, Tsuneki are all either radicular cysts (with evidence of metaplastic
et al. (2013) showed that a tooth root was close to the cyst change) or mucous retention cysts. The evidence to support
in about 45% of cases and that in half of these the tooth was their proposal is very circumstantial and does not appear to
found to be non-­vital. They suggested that the close asso- be well supported in the literature. Further studies may
ciation to non-­vital teeth was evidence that some nasopala- shed light on their suggestion.
tine duct cysts are of inflammatory origin and are actually In the absence of evidence to the contrary, most authors
radicular cysts in the midline of the maxilla. There are accept that the nasopalatine duct cyst is a developmental
three possible explanations for the association with cyst that arises from cystic degeneration of epithelial
226 Nasopalatine Duct Cyst

Box 13.2  Nasopalatine Duct Cyst: Key Features and Diagnostic Criteria

Clinical Features
●● Comprise only about 4% of all jaw cysts, but are the most common non-­ odontogenic cyst
●● Found in adults with a mean age of about 45 years and peak in the fourth decade

●● There is a wide age range (7–90 years), but lesions in children are rare

●● About 65% arise in males

●● Always located in the midline of the anterior hard palate

●● Most lesions are 10–15 mm in diameter

●● Most common presentation is a swelling, but 50% or more may be symptomless and an incidental finding

Radiological Features
●● Well-­
demarcated and corticated radiolucency
●● Always in the midline of the anterior palate. Arise in the incisive canal

●● Most are round or oval, but a ‘heart shape’ may be seen

●● 15 mm to 20 mm in diameter. Rare lesions may be up to 60 mm and fill the palate

●● Often involve the apices of the incisors

●● The lamina dura and periodontal space are intact

Histological Features
●● Lined by stratified squamous and/or pseudostratified columnar epithelium

●● Most lesions show both types of epithelium

●● Rarely have three or more types, with areas of columnar or cuboidal epithelium

●● Large nerves and blood vessels are typically seen in the wall

●● Often inflamed, but usually focal and mild

●● Mucous glands are seen in up to one-­ third of cases

remnants of the nasopalatine duct. Although there is no are usually enucleated, and because most are between 10
proof for this, the concept is supported by a number of and 20 mm in diameter, the pathologist may often receive
observations. First, there is the observation that small the lesion in one piece, although it may be ruptured
cystic dilatations of portions of the nasopalatine ducts are (Figure 13.9a). Otherwise multiple fragments of cyst wall
occasionally seen in fetal material (Abrams et  al.  1963), are received. The wall may be thick and fibrous, but is usu-
and secondly, remnants of the duct and small cysts may be ally of even thickness. If present, the luminal contents are
found in normal incisive canals and in the wall of cysts usually brown and watery. Occasional cysts may contain
(Abrams et  al.  1963; Bodin et  al.  1986; Falci et  al.  2013; mucus or, if infected, pus.
Lake et  al.  2018). It would also explain the absence of On histological examination, the nasopalatine duct cyst
inflammatory cell infiltrates in so many cases, as well as is characterised by having a variable microscopic appear-
the relative infrequency of an association with trauma in ance, so the finding of more than one type of epithelium,
the nasopalatine area. and of nerves, vessels, and mucous glands in the wall, are
Like other developmental cysts, although the stimulus in themselves almost diagnostic. Table  13.4 summarises
for proliferation is not known, the mechanism for growth the variable histological features reported in a number of
and enlargement appears to be through a process of larger cases series and in 86 of our own cases.
osmotic pressure, since the cysts are almost always sym- Although most cyst linings include a typical stratified
metrically round or oval, suggesting even centripetal squamous epithelium, only up to 50% are lined in their
expansion. entirety by one type of epithelium. Most of the remainder
have two types of epithelium, but up to four types have
been reported in a single cyst. The four types of epithelium
­Histopathology that may be seen are stratified squamous, pseudostratified
columnar, cuboidal, and columnar. These are seen individ-
The nasopalatine duct cyst does not have any distinctive ually or in combination, and often there is a transition
features on gross or macroscopic examination. The cysts between them.
­Histopatholog 227

Table 13.4  Histological features of nasopalatine duct cysts (proportion of cysts showing each feature (%). Totals may be greater than
100% because multiple features are seen).

Abrams et al. Allard Anneroth et al. Bodin et al. Vasconcelos et al. Barros et al.
Shear (1963) (1981a) (1986) (1986) (1999a) (2018)

n = 86 n = 61 n = 22 n = 32 n = 50 n = 31 n = 30

Epithelial lining
Stratified squamous 77.9 82.0 72.7 50.0 42.0 93.5 73.3
Pseudostratified columnar 45.3 44.3 27.3 23.3 42.0 77.4 53.3
Simple columnar 4.7 14.8 31.8 73.3 34.0 3.2 30.0
Simple cuboidal 21.0 31.1 27.3 29.0 40.0
One type of epithelium 46.5 36.1 54.5 53.3 46.0 35.5 40.0
Two types of epithelium 51.2 54.1 31.8 40.0 24.0 38.7 30.0
Three or more types 2.3 9.8 13.6 6.7 30.0 25.8 30.0
Mucous (goblet) cells NR 8.2 NR 6.7 14.0 16.1 50.0
Fibrous cyst wall
Neurovascular bundle 45.3 88.5 77.3 78.1 72.0 48.4 50.0
Mucous glands 7.0 31.1 36.7 25.0 18.0 12.9 23.3
Inflammatory infiltrate 75.6 92.8 77.3 71.9 62.0 80.6 83.3

(a) (b)

(c)

(c)

(b)

Figure 13.9  Nasopalatine duct cyst. (a) Low power shows a typical cyst with a fibrous wall of regular thickness lined by a number of
types of epithelium. There are focal accumulations of inflammatory cells (arrows). (b) Inset shows a pseudostratified ciliated columnar
(respiratory) epithelium. (c) Inset shows a proliferative stratified squamous epithelium, associated with inflammation.
228 Nasopalatine Duct Cyst

Stratified squamous epithelium is always non-­keratinised epithelium of this type and only 6.6% of cysts were lined
and may be thin and regular, but is often thickened and entirely by pseudostratified columnar epithelium. Bodin
acanthotic or hyperplastic, especially when the cyst is et al. (1986) found 16.0% and 14.0% lined entirely by strati-
inflamed (Figures  13.9c and  13.10). Overall, stratified fied squamous epithelium or pseudostratified columnar
­squamous epithelium is the most frequently encountered epithelium, respectively, while the equivalent numbers
type of epithelium and is found in between 50% and about reported by Barros et al. (2018) were 30% and 10%.
94% of lesions (Table  13.4), but is most often seen in Pseudostratified columnar epithelium is usually ciliated
­combination with pseudostratified columnar epithelium and is of typical respiratory type. It often contains mucous
(Figure 13.9). In our series of 86 cases, 67 (77.9%) showed or goblet cells (Figure  13.9b). Overall, up to 50% of cysts
stratified squamous epithelium, but only 33 (38.4%) were may show prominent goblet cells. These may be found in
entirely lined by this epithelium. Furthermore, 39 cysts the respiratory-­type epithelium, or at the surface of strati-
(45.3%) contained pseudostratified ciliated columnar epi- fied squamous epithelium, or occasionally in association
thelium in part of their linings, but only 7 (8.1%) were lined with columnar or cuboidal epithelium (Figure 13.10c).
entirely in this way. Only 4 cysts showed areas of simple A simple cuboidal or columnar epithelium is frequently
columnar epithelium, although 18 (20.9%) had areas of seen and has been reported in up to 73% of cysts, but almost
cuboidal epithelium. One cyst was lined entirely by cuboi- always in combination with stratified squamous or pseu-
dal epithelium. dostratified epithelium (Anneroth et al. 1986; Table 13.4).
These frequencies are very similar to those reported by This epithelium is difficult to define, but usually appears as
others (Table 13.4). For example, Abrams et al. (1963) found a simple epithelium two to five cell layers thick
that 82% of their cysts had stratified squamous epithelium (Figures 13.10b and 13.11). Most often, however, a transi-
in the lining, but only 26.2% (16 of 61) were entirely lined by tion is seen between epithelial types, with cuboidal or low

(a) (b)

(c) (d)

Figure 13.10  Nasopalatine duct cysts, showing different patterns of epithelium. (a) Stratified squamous epithelium of variable
thickness. (b) A thin, regular epithelium composed predominantly of cuboidal cells. (c) Stratified squamous epithelium, with prominent
mucous (goblet) cells in the superficial layer. (d) This cyst wall contains mucous glands. A duct enters the cyst lumen in the lower left
corner of the field.
­Treatmen 229

Small foci of mucous glands (Figure 13.10d) have been


found in the fibrous capsules of up to one-­third of cases
(Abrams et al. 1963; Allard et al. 1981a; Table 13.4). As the
foci are small, it is probable that their frequency would be
higher if biopsy material were more extensively sampled.
Abrams et al. (1963) found evidence of fibrosed glands in
association with areas of inflammation and suggested that
glands may have degenerated in some lesions. Mucous
glands are a consistent finding within the incisive canal,
but in fetal tissues they were found only towards the nasal
aspect. Since nasopalatine duct cysts most often arise in the
inferior portions of the canal, this may explain the paucity
of glandular tissue.
Nasopalatine duct cysts are often inflamed, but this is not a
Figure 13.11  Large nerves in the wall of a nasopalatine duct consistent finding. In our series of 86 cases, 65 (75.6%) showed
cyst. In this field the cyst is lined by simple cuboidal epithelium. evidence of inflammation, but this was only prominent or
Courtesy of Dr Daniel Brierley. severe in 9 (10.5%). Bodin et al. (1986) and Barros et al. (2018)
also found that prominent inflammation was unusual (12%
columnar cells admixed with squamous cells. Often, a pre- and 10% of cysts, respectively), but others found a heavy
dominantly stratified squamous epithelium may be seen inflammatory infiltrate in about half of cases (Abrams
but with superficial columnar, goblet or ciliated cells et al. 1963; Allard et al. 1981a). When present, the inflamma-
(Figure 13.10c, d). tion is usually focal and is associated with a hyperplastic or
It has frequently been suggested that cysts lined by res- proliferative squamous epithelial lining (Figure 13.9c).
piratory epithelium probably originate within the superior Occasionally, epithelial cell rests may be seen in the cyst
aspect of the incisive canal adjacent to the nasal cavity, wall. Some of these may be cross-­sectioned fragments of
whereas those lined by stratified squamous epithelium mucous glands or small ducts, but others appear to be
develop in the lower portion of the canal. However, this is squamous and probably represent remnants of the naso-
not a consistent finding and no studies have been able to palatine duct. These were found in 19 (22.1%) of our 86
establish a relationship between the nature of the lining cases and Abrams et al. (1963) found epithelial cell rests in
and the site of the cyst. Abrams et al. (1963), for example, 22 (36.1%) cysts, of which 19 were squamous and 3 showed
found two cysts in the incisive papilla lined by pseudostrat- small proliferations of cuboidal cells.
ified ciliated columnar epithelium. The fact that the major- Other features include small islands of hyaline cartilage
ity of cyst linings have a combination of types of epithelium that may very rarely be seen in the cyst walls. They were
is suggestive of an origin from pluripotential epithelium, present in 6 of our cases (7%) and Abrams et  al. (1963)
but the possibility that metaplasia occurs must also be found cartilage in 5 (8.2%) of their 61 cysts. However, 4 of
considered. these were cysts in the palatine papilla (‘incisive papilla
The cyst wall is composed of collagenous fibrous connec- cysts’) and cartilage was only seen in 1 of 57 cysts that they
tive tissue, which contains a number of characteristic struc- felt had actually arisen within the canal. It is not unusual to
tures. A valuable diagnostic feature of nasopalatine duct find small amounts of cartilage in the region of the normal
cyst is the presence of prominent neurovascular bundles palatine papilla, so its presence in the cyst wall should not
with obvious moderate to large nerves in the fibrous cap- be regarded as a pathological process.
sule (Figure 13.11). These have been reported in up to about
90% of cases (Table 13.4). Abrams et al. (1963) noted nerves
in all their cases, but found they were moderate to large in ­Treatment
88.5%. In 87% of their cases, muscular arteries and numer-
ous small veins were also present. Our own observations Nasopalatine duct cysts are treated by surgical enucleation.
support the frequency with which these features are found. Most cysts are less than 20 mm in diameter and do not
In our series of 86 cases, prominent neurovascular bundles involve the teeth, which can be preserved. Occasionally,
were found in 45% and large muscular arteries in 72%. The however, the apices of the incisor teeth may be involved in
explanation for this is that the long sphenopalatine (naso- the cyst lumen and some authors advocate presurgical
palatine) nerve and vessels that pass through the incisive endodontic treatment (Suter et al. 2015). Nasopalatine duct
canal are either included in the cyst wall or are removed cysts rarely recur. In their series of 334 cases, Swanson
with the cyst in the course of surgical enucleation. et al. (1991) found recurrences in only 2% of cases.
230

14

Nasolabial Cyst

CHAPTER MENU
Clinical Features, 230
●● Frequency, 230
●● Age, 230
●● Sex, 231
●● Clinical Presentation,  231
Radiological Features, 233
Pathogenesis, 234
●● Nasolacrimal Cyst,  234
Histopathology, 234
Treatment, 236

The nasolabial cyst occurs outside the bone in the soft 79 publications reporting 311 cases up to 2016. Tables 14.1
­tissues of the upper lip below the alae of the nose. Because and  14.2 summarise data from selected case series and
it arises in the maxillofacial region, in close proximity from the reviews of Roed-­Petersen (1969) and Sheikh
to  the maxillary alveolus, it is often classified among et al. (2016).
the jaw cysts. The nasolabial cyst has also been called the
­nasoalveolar cyst, but as the alveolus is not involved,
Frequency
the term nasolabial is preferred. Occasionally the literature
still includes the term Klestadt cyst after Klestadt, who Nasolabial cysts are rare lesions and probably represent
described the cyst in a number of papers in the early 1900s only about 0.5% of all cysts of the maxillofacial region.
(reviewed by Klestadt 1953; Roed-­Petersen 1969). Shear found only 21 examples in the archives of the
University of the Witwatersrand over a period of 46 years,
representing 0.6% of jaw cysts (see Table  1.1). Jones and
­Clinical Features Franklin (2006a), in a review of almost 45 000 biopsy speci-
mens in Sheffield, found only 20  nasolabial cysts, repre-
Nasolabial cysts are rare and most reports in the literature senting 3.4% of non-­odontogenic cysts and only 0.3% of all
are single case reports or small case series. Roed-­Petersen jaw cysts. Data from other large series have shown a fre-
(1969) undertook a review of the literature between 1882 quency among jaw cysts of 0.04% (Soluk Tekkeşin
and 1969 and found 155 patients reported with nasolabial et al. 2012b; Turkey), 0.1% (Daley et al. 1994; Canada) and
cysts. He was able to carry out an analysis of the combined 1.0% (Grossmann et al. 2007; Brazil).
data of 111 of these patients plus 5 of his own cases. Van
Bruggen et  al. (1985) identified a further 45 cases up to
Age
1985 and added 10 cases of their own. Subsequently there
have been many case reports and small series, but overall The nasolabial cyst occurs in adults, with a mean age of
fewer than 350 cases have been reported. Sheikh et  al. about 42 years (Table  14.1). The age distribution of 167
(2016) undertook a systematic review and identified patients is shown in Figure 14.1. These data are taken from

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
­Clinical Feature 231

Table 14.1  Age, sex, and clinical findings from selected case series of nasolabial cysts and from the large reviews of Roed-­Petersen
(1969) and Sheikh et al. (2016).

Age Sizea

Reference Country n Mean Range Female (%) Mean Range Bilateral (%)

Chinellato and Damante (1984) Brazil 8 26.6 24–35 87.5 NR 10–30 0


van Bruggen et al. (1985) S Africa 10 43.3 30–72 100 23.3 20–25 0
Vasconcelos et al. (1999b) Brazil 15 42.0 20–69 86.7 NR NR 6.6
Choi et al. (2002) S Korea 18 42.8 17–67 72.2 NR 10–50 0
Yuen et al. (2007) Singapore 17 41.0 31–75 64.7 NR NR 5.8
Tiago et al. (2008) Brazil 8 47.6 20–67 100 21.8 13–20 12.5
Lopes-­Rocha et al. (2011) Brazil 7 40.3 27–62 100 14.7 10–30 0
Roed-­Petersen (1969) Review 116 42.4 12–75 77.6 NR NR 11.2
Sheikh et al. (2016) SR 311 41.8 4 m-­78 78.2 NR NR 9.6

NR, not reported; SR, systematic review.


a
 Size (mm): in all studies the size appears to be recorded as an estimate of the diameter of the lesion on clinical examination.

Table 14.2  Clinical signs and symptoms (%) reported in selected case series of nasolabial cysts and in the large reviews of Roed-­
Petersen (1969) and Sheikh et al. (2016).

Clinical findings (%)

Reference n Swelling Nasal obstruction Paina Infectionb

Chinellato and Damante (1984) 8 100 NR 25.0 12.5


Vasconcelos et al. (1999b) 15 100 13.3 0 0
Choi et al. (2002) 18 55.6 27.8 16.7 NR
Yuen et al. (2007) 17 41.2 23.5 11.8 29.4
Tiago et al. (2008) 8 50.0 62.5 25.0 0
Roed-­Petersen (1969) 116 83.6 31.0 31.9 NR
Sheikh et al. (2016) 311 70.9 17.3 41.9 3.8

NR, not reported.


a
 Includes complaints of pain and tenderness on clinical examination.
b
 Signs of infection include fistula, discharge, foul taste. Totals are over 100% because more than one symptom may be present.

10 cases of Shear’s from the University of the Witwatersrand Sex


and from a number of reviews or case series where the age
Of all the cysts of the maxillofacial region, the nasolabial
distribution was known: Roed-­Petersen (1969; n  =  106);
cyst is the only one that shows a predilection for females.
van Bruggen et al. (1985; n = 25); Choi et al. (2002; n = 18);
Table 14.1 shows that in some series 100% of patients have
Chinellato and Damante (1984; n = 8). There is a wide age
been female, but the largest review found that 78.2% of
range, with a peak frequency in the fourth and fifth dec-
cases occurred in females (Sheikh et al. 2016). Of the 167
ades. Very few series have reported cases in children,
cases shown in Figure  14.1, 78.4% (131 cases) arose in
although a few were found in the second decade
females.
(Figure 14.1). In a review of over 4400 maxillofacial biop-
sies from children, Jones and Franklin (2006b) found only
one case of nasolabial cyst, in a 12-­year-­old female. In their
Clinical Presentation
systematic review, Sheikh et  al. (2016) found a wide age
range and recorded one case in a 4-­month-­old child, but The nasolabial cyst presents as a swelling of the upper lip,
details were not provided. which is often visible on extraoral examination. The cyst
232 Nasolabial Cyst

50
46
45
40
40

35

30
No. of cases

26 36
Females
25 31 23
Males
20
16
15 21
20
10 8 12 8

5
5 6 10 9
0 5 4 0
2 3 3
0
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89
Age

Figure 14.1  Age and sex distribution of 167 patients with nasolabial cysts.

found swelling as a symptom in only about 50% of cases


(Choi et  al.  2002; Tiago et  al.  2008). In their systematic
review, Sheikh et  al. (2016) found that 70.9% of patients
complained of swelling, the majority of which involved the
nasolabial fold and the alar region. Only 16 cases (6.8%)
showed lip swelling only. They recorded only 2 cases that
were found incidentally. Physical examination of the
lesions was recorded in 93 cases: 40 (43.0%) were described
as fluctuant, 14 (15.0%) as mobile, and 39 (41.9%) were ten-
der. Fluctuance may be elicited between the swellings in
the floor of the nose and in the labial sulcus.
Patients may complain of nasal obstruction or difficulty
in nasal breathing, but the frequency of this is variable
(Table  14.2). This may depend on where the cyst was
managed. For example, Tiago et  al. (2008) reported that
Figure 14.2  Nasolabial cyst producing a swelling of the right 62.5% of their patients had nasal obstruction, but these
upper lip, forming a bulge high in the labial sulcus (arrows).
patients were seen in a department of otorhinolaryngology
(Ear, Nose, and Throat), whereas lower frequencies were
fills out the nasolabial fold, often lifts the ala, and may dis- seen in a dental clinic environment (13.3%; Vasconcelos
tort the nostril. It may produce a swelling of the floor of the et al. 1999b). Pain, or tenderness on palpation, is seen in up
nose and can displace the inferior turbinate. Intraorally it to about 40% of patients and occasionally there may be
forms a sessile bulge high in the labial sulcus (Figure 14.2). clear signs of infection (Table 14.2). Infected cysts may dis-
Signs and symptoms are summarised in Tables  14.1 charge into the labial sulcus, or into the nose. In some
and  14.2. The most frequent finding is of a swelling and cases, difficulty with an upper denture has drawn attention
very often this was the only complaint. However, some- to the problem.
times if the swelling is small the cyst may be an incidental A number of series have recorded the size of the cyst and
finding, diagnosed fortuitously during routine examina- have shown mean diameters up to 23.3 mm (van Bruggen
tion. Roed-­Petersen (1969) found that only 97 (83.6%) of et al. 1985; Table 14.1). The cysts ranged in size from less
116 patients complained of swelling, while others have than 10 mm to as large as 50 mm (Choi et  al.  2002).
­Radiological Feature 233

Occasional ‘giant’ or massive nasolabial cysts have been If a radiopaque contrast medium is introduced, the cyst
recorded. Cohen and Hertzanu (1985) reported a case that appears as a spherical or bean-­shaped lesion lying against the
reached 70 mm in diameter, causing severe facial deformity. inferior and lateral borders of the anterior bony aperture of
In most cases the cysts are unilateral, but about 10% of the nose, extending from the midline to the canine fossa
patients have been found to have bilateral lesions. In the (Figure 14.4; Chinellato and Damante 1984). Computed tomog-
two large reviews, the frequency of bilateral cases was raphy (CT) and magnetic resonance imaging (MRI) show these
11.2% and 9.6% (Table 14.1). features with more clarity. Choi et al. (2002) reported CT find-
ings on 11 patients, and found that the cysts presented as well-­
demarcated, low-­density lesions lying just lateral and inferior to
­Radiological Features the nasal opening (the pyriform aperture). They observed no
invasion of bone in any of their patients.
Radiological examination can assist in the diagnosis of MRI shows a well-­demarcated, hypointense cystic lesion,
nasolabial cyst, in the first instance by excluding a lesion of but T2  weighting highlights the lesions as hyperintense
intraosseous origin, but the cyst also has some characteristic (white). In axial views, the cyst lies just below the alae of
radiological features. The lesion overlies the maxilla and may the nose and displaces the nasal cavity and nasolabial fold
cause pressure resorption of the cortical plate that appears as (Iida et al. 2006b; Sumer et al. 2010). In coronal and sagittal
a slight radiolucency of the alveolar process above the apices views, the cyst often has a characteristic bean shape with
of the incisor teeth (Figure  14.3). A more characteristic the concave margin orientated towards the medial or pos-
feature, however, is seen on an anterior occlusal plane terior aspect, where the cyst impinges on the lateral or
radiograph. In this view, the normal nasal septum and lateral anterior nasal margins of the maxilla. Occasionally, a fluid
and anterior margins of the nasal aperture form an ‘anchor’ level may be seen that shows as slight hyperintensity on T1
shape that is symmetrical about the midline. The nasolabial and hypointensity on T2  weighting (Iida et  al.  2006b).
cyst may distort or displace the radiopaque line of the inferior Although there may be displacement of bone or slight sau-
margins of the nose to form a convexity towards the posterior cerisation of the cortical plates, there is no evidence of
aspect (Figure 14.3, arrows). This “distorted anchor” feature bone destruction or perforation.
was first described by Seward (1962) and was seen in 6 of the A number of workers have used ultrasound to examine
8 cases reported by Chinellato and Damante (1984). the nasolabial cyst. This shows a well-­defined anechoic
cystic lesion (Ocak et al. 2017). An advantage of ultrasound
is that it does not involve ionising radiation and it is quick
and cheap. It is also valuable in easily distinguishing
between a cystic lesion and a solid lesion that may present

Figure 14.3  Anterior occlusal radiograph of a patient with a


nasolabial cyst. There is a posterior convexity of the left half of
the radiopaque line that forms the bony border of the nasal
aperture (arrows). ⃝ is located within the cyst and shows an Figure 14.4  Nasolabial cyst. The extraosseous position of the
area of very slight radiolucency. Source: Courtesy of cyst is demonstrated by injection of a radiopaque fluid. Source:
Drs L.E.M. Chinellato and J.H. Damante. Courtesy of Drs L.E.M. Chinellato and J.H. Damante.
234 Nasolabial Cyst

at this site. In particular, benign salivary gland tumours are The nasolacrimal cyst is an acquired retention cyst arising
not uncommon in the upper lip. due to fluid retention within the nasolacrimal duct. The
developing nasolacrimal canal opens into the lateral wall
of the nose in the inferior meatus, but is covered by a mem-
­Pathogenesis brane (the membrane of Hasner) and does not become pat-
ent until birth or shortly thereafter. In a small number of
There is general agreement that nasolabial cysts are of devel- neonates, the duct does not become patent and the block-
opmental origin. Early workers regarded the lesion as a fis- age results in retention of tears and cyst formation in the
sural cyst that arose from epithelial remnants entrapped after duct or lacrimal sac, called a dacryocystocele. These may be
fusion of the lateral nasal and maxillary processes. A fissural congenital or arise in neonates, but are very rare after a few
origin is now regarded as not tenable, since the embryologi- months of age. However, occasionally a dacryocystocele
cal basis for this has been disputed and discredited. may arise in adults if the nasolacrimal duct is blocked due
It is now thought that the cyst arises from the lower to another reason, such as infection or trauma. Nevertheless,
­anterior part of the nasolacrimal duct, a proposal that was these lesions arise in the lateral wall of the nose adjacent to
first suggested in 1920 by Brüggemann (reviewed by the inferior turbinate, and differentiation from nasolabial
Allard  1982). The nasolacrimal duct (also called the tear cysts should be straightforward (Eloy et al. 2012; Box 14.1).
duct) runs from the lacrimal sac in the medial aspect of the
orbit and drains tears into the nose. It runs in a bony canal
or groove (the nasolacrimal canal) that lies between the ­Histopathology
maxilla and the lacrimal bone superiorly and through the
inferior turbinate bone inferiorly to open in the lateral wall Nasolabial cysts are soft-­tissue lesions and may be removed
of the nose into the inferior meatus below the inferior tur- by a conservative excision, so that the pathologist receives
binate. The nasolacrimal canal can sometimes be seen on an intact cyst with a fibrous wall. On dissection, the lesion
occlusal radiographs, superimposed over the posterior part may reveal watery or mucous fluid. Larger cysts may be
of the hard palate (for example, see Figure 13.8). enucleated and in some cases may be marsupialised, often
The nasolacrimal duct begins to form at about six weeks by a transnasal route. In these cases, the specimen may be
of intrauterine life, at the junction of the maxillary process composed of cystic fragments.
and the lateral nasal process. As the processes fuse, a deep The nasolabial cyst is usually lined by a non-­ciliated
groove is formed and the epithelium at the base of the pseudostratified columnar epithelium (Figure  14.5),
groove thickens to form a solid cord, the nasolacrimal although other epithelial types may be seen, including
ridge, which eventually canalises and separates from the stratified squamous, cuboidal, or respiratory epithelium. In
surface to form a duct within the mesenchyme. The supe- Roed-­Petersen’s (1969) review, 64 cases that had been eval-
rior (cranial) end of the duct connects with the developing uated histologically were summarised. Pseudostratified
lacrimal sac, while the inferior (caudal) end proliferates to columnar epithelium was seen in 53 (82.8%) cysts and in 26
connect with the lower part of the lateral nasal wall. The (40.6%) was the only type of lining present. In 9 cases,
location of the nasolabial cyst is such that it most likely pseudostratified columnar epithelium was present in asso-
arises from epithelial remnants at the inferior end of the ciation with stratified squamous epithelium, and in 15 with
embryonic nasolacrimal rod or duct, or possibly from the cuboidal epithelium. Only 4 cases (6.3%) were lined solely
lower anterior portion of the mature duct. The mature by stratified squamous epithelium and 4 solely by cuboidal
nasolacrimal duct is lined by pseudostratified columnar epithelium. Overall, 53.1% (n = 34) were lined by one epi-
epithelium and this is the type of epithelium most often thelial type, 42.2% (n  =  27) by two epithelia, and 4.7%
found lining nasolabial cysts. (n = 3) by all three types. The most frequent combinations
A developmental origin for the nasolabial cyst is further of epithelial types were pseudostratified columnar with
reinforced by the finding that about 10.0% are bilateral. cuboidal epithelium (15 cases: 23.4%) or with stratified
Another remarkable feature is the high frequency in squamous epithelium (9 cases: 14.1%). Mucous or goblet
females, the only cyst of the oral regions that shows a cells were present in only 51.5% (n = 33) of cysts and only
female preponderance. The reason for this is unknown. 34.3% (n = 22) contained ciliated cells.
Most workers have found a similar type of epithelial lin-
ing. Chinellato and Damante (1984) found that all 8 of
Nasolacrimal Cyst
their cysts were lined by pseudostratified columnar epithe-
Another cyst that may arise in this region, and must not be lium and that 4 also had areas of stratified squamous epi-
confused with the nasolabial cyst, is the nasolacrimal cyst. thelium; 7 cases also contained goblet cells. Yuen et  al.
­Histopatholog 235

Box 14.1  Nasolabial Cyst: Key Features

Clinical Features
●● Rare cyst, comprising about 0.5% of jaw cysts

●● Affects adults with a mean age of 42 years and peak in the fourth and fifth decades

●● About 80% arise in females

●● Presents as a swelling in the upper lip

●● May displace the nasolabial fold and the floor of the nose

●● Most are about 20 mm in diameter

●● 10% may be bilateral

Radiological Features
●● They are extraosseous, but often resorb the alveolar bone

●● May see a slight radiolucency above the incisor roots

●● On an occlusal radiograph, they may displace the inferior margins of the nasal aperture to form a characteristic

­convexity towards the inside of the nose


●● Magnetic resonance imaging (MRI) shows a hyperintense T2-­ weighted image, well demarcated and ‘bean’ shaped
on coronal and sagittal views
Histological Features
●● Lined predominantly by pseudostratified columnar epithelium

●● About 40% lined by two types of epithelium, usually pseudostratified with stratified squamous or columnar

●● Often show subepithelial hyalinisation of the collagen

●● May see goblet cells, or mucous glands in the wall

Figure 14.5  Nasolabial cyst lined predominantly by a pseudostratified columnar epithelium containing many goblet cells. A small
area of lining is composed of simple cuboidal epithelium (arrow). The cyst wall shows prominent eosinophilic hyalinisation below the
epithelium. Source: Courtesy of Dr Daniel Brierley.

(2007) reviewed 17 cases and found that 9 (52.9%) were epithelium and that 47.9% contained goblet cells or mucous
lined by pseudostratified columnar epithelium, 4 (23.5%) glands. Only 33 (10.6%) were reported to be inflamed.
by stratified squamous epithelium, and 3 (17.6%) by both The cyst wall is composed of relatively acellular fibrous
types. One case was lined by cuboidal epithelium alone. connective tissue, which may contain small numbers of
In their systematic review of 311 cysts, Sheikh et  al. mucous glands. Focal areas of inflammation may also be
(2016) found that 71.1% (n = 221) were lined by a mixed seen. Roed-­Petersen (1969) noted that in 4 of their own 5
236 Nasolabial Cyst

cases there was a prominent subepithelial zone of eosino- outcomes of transnasal marsupialisation (n = 34) with sub-
philic hyalinised connective tissue. Although this feature is labial excision (n = 23) and found that both were equally
rarely mentioned in publications, we have noted it in our effective, but that transnasal marsupialisation was cheaper
own cases (Figure 14.5). and involved fewer days of hospitalisation. Marsupialisation
was used for larger lesions with a mean diameter of
21.2 mm. In their systematic review, Sheik et  al. (2016)
­Treatment found information about treatment in 263 cases. Most
(70.0%; n = 184) were treated by sublabial excision or enu-
Although nasolabial cysts are extraosseous, careful surgi- cleation and 79 (30.0%) were treated by transnasal marsu-
cal excision or enucleation via a transoral sublabial pialisation. They found that complications following
approach is the most common treatment. Cysts lying close surgery were seen in 27.2% of the patients treated by the
to the floor of the nose can be treated by a transnasal endo- sublabial approach, compared with only 13.9% in the trans-
scopic marsupialisation technique. This was developed by nasal group. The most common complication was facial
Su et al. (1999), who treated 16 patients and found that the swelling, seen in 19 sublabial patients, but only 5 treated by
cyst became replaced by an air-­containing sinus with a per- marsupialisation. Other complications of the sublabial
sistent opening at the anterior or anterolateral nasal floor. approach included perforation of the nasal mucosa, pain or
More recently, Chao et al. (2009) compared the costs and paraesthesia, and toothache.
237

15

Cysts of the Salivary and Minor Mucous Glands

CHAPTER MENU
­Mucoceles, 237 ­Retention Cyst and Pseudocyst of the Maxillary Sinus,  253
•• Classification and Terminology,  238 •• Clinical Features,  253
•• Clinical Features,  238 –– Frequency,  254
–– Frequency,  238 –– Age,  254
–– Age,  239 –– Sex,  254
–– Sex,  240 –– Site,  254
–– Site,  240 –– Clinical Presentation,  254
–– Clinical Presentation,  241 •• Radiological Features,  254
–– Superficial Mucoceles,  241 •• Pathogenesis,  254
–– Mucocele of the Glands of Blandin–Nuhn,  242 •• Histopathology,  255
•• Ranula,  242 •• Treatment,  255
•• Pathogenesis,  244
­Lymphoepithelial Cysts,  255
•• Histopathology,  244
–– Mucous Extravasation Cysts,  244 ­Intraoral Lymphoepithelial Cysts,  256
–– Superficial Mucoceles,  247 •• Clinical Features,  256
–– Mucous Retention Cysts,  248 •• Frequency,  256
–– Histological Differential Diagnosis,  249 •• Age,  256
•• Treatment,  250 •• Sex,  256
­Salivary Duct Cysts,  251 •• Site,  256
­Cysts of the Maxillary Sinus,  252 –– Clinical Presentation,  256
­Mucocele of the Maxillary Sinus,  252 •• Pathogenesis,  256
•• Histopathology,  257
•• Clinical Features,  252
–– Frequency,  252 •• Treatment,  258
–– Age and Sex,  252 ­Lymphoepithelial Cysts of the Parotid Gland,  258
–– Clinical Presentation,  253 –– HIV-­Associated Salivary Gland Disease,  259
•• Radiological Features,  253
•• Pathogenesis,  253 ­Polycystic (Dysgenetic) Disease of the Parotid Glands,  259
•• Histopathology,  253 ­Sclerosing Polycystic Adenoma,  260
­Keratocystoma,  260

Cysts of the salivary and minor mucous glands of the the oral cavity, the major salivary glands, and the maxil-
head and neck are common and fall into three broad cat- lary sinus.
egories: developmental, reactive, and neoplastic. Cysts
may arise in the major salivary glands or in the minor
glands that are found in the submucosa of the oral cavity. ­Mucoceles
Similar lesions arise in the minor mucous and seromu-
cous glands that are dispersed throughout the upper aer- Mucoceles are the most common of all lesions of the sali-
odigestive tract and the paranasal sinuses. This chapter vary glands and are mucus-­filled cystic cavities that arise in
will discuss the more common cystic lesions that affect the mucosa of the upper aerodigestive tract. Although

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
238 Cysts of the Salivary and Minor Mucous Glands

mucocele is a well-­recognised and widely used term, there The pooling of salivary mucus within a cystic cavity
is still much debate and disagreement about how it should resulting from blockage or rupture of a salivary duct.
be defined and its precise usage.
This definition describes the lesion as a cystic cavity regard-
less of lining and includes both the retention type (caused by
Classification and Terminology
blockage) and the extravasation type (caused by rupture).
The term mucocele is used in different ways by different In this chapter we use this definition and will refer to the
authors and in different countries, leading to confusion extravasation type of mucocele as mucous extravasation cyst
and lack of consistency when considering case series and and the retention type as mucous retention cyst. Furthermore,
case reports. We have been unable to find a consensus on we regard salivary mucoceles as intraoral lesions and use
the correct terminology or even spelling of what consti- the term salivary duct cyst for retention cysts of the major
tutes a mucocele.1 Most clinicians and especially paediat- salivary glands. Although the clinical appearance of mucous
ric dentists recognise the mucocele as a cystic lesion extravasation cysts and mucous retention cysts may be
arising on the oral mucosa, and use the term regardless of indistinguishable, they do have some distinctive clinico-
the histological findings. It is recognised, however, that pathological features (Table 15.1; Box 15.1).
there are two types of mucocele. One arises as a result of
mucus extravasation and the other as a result of mucus
Clinical Features
retention within a dilated duct. Many pathologists never-
theless choose to make a clear distinction between these Frequency
two types and some decline to use the term ‘cyst’ for the Mucoceles of the mouth are very common. Their true inci-
extravasation type because it does not have an epithelial dence is difficult to determine as many lesions may rupture
lining. Thus, many authors in the USA prefer to use the and resolve, patients do not always seek treatment, and of
term ‘extravasation phenomenon’ or ‘mucus escape reac- those diagnosed a large number are not surgically excised
tion’ for the extravasation type, and often use ‘mucocele’ and do not reach a pathology department. In series of oral
as synonymous with this type of lesion. It is ironic that biopsies, however, mucoceles are one of the most com-
these authors still use the terms ‘solitary bone cyst’ and monly encountered diagnoses. Jones and Franklin
‘aneurysmal bone cyst’ for other cystic lesions that are also (2006a,b) reviewed more than 48 000 biopsies received
not lined by epithelium. from adults and children over a 30-­year period and found a
The retention type is usually called a ‘mucous retention total of 2472 mucoceles, representing 5.1% of all biopsies
cyst’, but others use the term ‘salivary duct cyst’ and do not and 13.8% of biopsies of the oral mucosa. As a comparison,
regard this as a type of mucocele. The term sialocyst is also radicular cysts and dentigerous cysts represented 7.1% and
commonly used, but it is ill defined and is used by different 2.5%, respectively, of all biopsies. Other studies have shown
authors as a name for a variety of lesions. a similar proportion of all biopsies, with reported frequen-
Clinicians will appreciate that extravasation and reten- cies of 4.6% (Re Cecconi et al. 2010), 5.3% (Chi et al. 2011),
tion mucoceles are indistinguishable on clinical examina- 5.8% (de Brito Monteiro et  al.  2016), and 5.8% (Bezerra
tion and that both present as a cystic lesion. They are often et  al.  2016). Note that some of these studies reported
therefore referred to as mucous extravasation cyst and extravasation cysts only.
mucous retention cyst. This terminology is clear and pre- Mucoceles are more common in children than in adults,
cise, is in common usage, and recognises that the lesions with the single most common occurrence being lesions on
are clinically cystic. the lip in children. In their reviews, Jones and Franklin
With regard to a definition for mucocele, this is hard to found that the frequency in children was 17.1% of all biop-
come by, but the best we have found is that of Richardson sies (n = 4406; 752 mucoceles; Jones and Franklin 2006b),
(2019) used in a contemporary pathology textbook: compared with 3.9% in adults (n = 44 007; 1720 mucoceles;
Jones and Franklin 2006a).
With regard to the different types of mucocele, the
1  The alternative spelling is mucocoele. Both –cele and –coele are
mucous extravasation cyst is the most common. Jones and
suffixes derived from Greek, but have similar meanings. Cele is
derived from kēlē, meaning tumour, swelling, or hernia, while coele is Franklin (2006a,b) found that of all mucoceles (n = 2427),
derived from koîlos, meaning a cavity or hollow. The two are used 90.3% were extravasation cysts and only 9.7% (n = 235) were
interchangeably and synonymously in medicine as a suffix to refer to mucous retention cysts, but they also found a difference
swellings, cystic or not, involving a particular organ or tissue. In the
between children and adults. Mucous retention cysts were
literature, mucocele is used far more commonly (by a factor of more
than 10) than mucocoele. For these reasons, we have chosen to use relatively more common in adults, constituting 12.7% of all
the term mucocele. mucoceles, while in children they only represented 2.3%.
­Mucocele 239

Table 15.1  Mucoceles: age, sex, and site distribution from selected reports that have distinguished extravasation and retention cysts.

Type of mucocele

Extravasation Retention

References N Male (%) n Age mean (range)a Lower lip (%) n Age mean (range)a Lower lip (%)

Harrison (1975)b 400 58.4 360 2nd 71.7 40 7th 2.5


Southam (1974) 236 NR 224 2nd 68.3 12 7th 0
Eversole (1987) 121 43.0 – – – 121 51.7 (10–80) 14.8
Stojanov et al. (2017) 177 41.8 – – – 177 56.0 (2–95) 15.8
Chi et al. (2011) 1715 50.7 1715 24.9 (1–87) 81.9 – – –
Conceição et al. (2014) 100 60.0 100 23.2 (1–57) 92.0 – – –
Bezerra et al. (2016) 719 45.3 719 20.8 (1–82) 67.3 – – –
c c
Mínguez-­Martinez 89 42.7 89 6.1 (0–14) 79.8 – – –
et al. (2010)
c c
Bodner et al. (2015) 56 43.0 56 11.2 (1–16) 67.9 – – –

N = total number of patients; n = number of patients with each type; NR, not reported.
a
 In some studies no means are given. Peak decades are quoted.
b
 Harrison reported 45 cases, but reviewed 400.
c
 Paediatric cases only.

Box 15.1  Key Distinctive Features of the Two Types of Mucocele

Mucous extravasation cyst Mucous retention cyst

Most common site Lower lip Buccal mucosa, floor of mouth


% of all mucoceles 90% 10%
Frequencya 5.0% (17% in children) <0.5%
Proportion in males 50%. Equal sex distribution 45%. Slightly more common in females
Age 80% below age 40. Mean age 80% above age 40. Mean age about 55. Peak in
about 22. Peak in second seventh decade
decade
Clinical features Sessile swelling, usually on Sessile swelling, buccal mucosa, floor of
the lower lip. Pale blue/grey mouth, occasionally palate. Pale blue/grey or
or normal colour. Contains normal colour. Contains fluid/fluctuant.
fluid/fluctuant. May be 10 mm diameter
superficial and resemble a
blister. 10 mm diameter
Histology Pools of mucus surrounded Cystic cavity filled with mucus. Lined by
by inflamed fibrous or simple cuboidal or columnar bilayered
granulation tissue, with duct-­like epithelium. Minimal inflammation
prominent mucus-­filled
histiocytes
a
 Frequency as a proportion of all oral biopsies.

These proportions of retention cysts are similar to those Age


reported in other series: 4.0% (Cataldo and Mosadomi, 1970; The age distribution of mucoceles is illustrated in
n = 594), 5.1% (Southam 1974; n = 236), 8.8% (Eversole 1987; Table 15.1. In many clinical studies a bimodal distribution
n  =  1380), 7.5% (Oliveira et  al.  1993; n  =  112), and 7.8% has been noted and, as can be seen in Table  15.1, this is
(Stojanov et al., 2017; n = 2275). due  to different ages of occurrence of extravasation and
240 Cysts of the Salivary and Minor Mucous Glands

retention cysts. However, because extravasation cysts are Site


more common, the vast majority of mucoceles are encoun- Overall the great majority of mucoceles, up to about 80%,
tered in children and young adults, below the age of are found in the lower lip and they are rare in the upper lip.
30 years. They are located on the mucosal side of the lower lip
Mucous extravasation cysts occur at an average age of towards the commissure (Figure  15.1). This predilection
about 20–25 years and all studies show a peak incidence in for the lower lip can be explained by the susceptibility of
the second decade. In the largest series (Chi et  al.  2011), this site to trauma from biting, lip chewing, or acciden-
507 (29.6%) of the 1715 cases arose in the second decade tal trauma.
and 65.9% of all cases were found below the age of 30 years. There is a difference, however, in the site distribution of
In his review, Harrison (1975) showed the age distribu- mucous extravasation cysts and mucous retention cysts
tion  of 356 extravasation cysts and found that 34.6% (Tables 15.1 and 15.2). Table 15.2 summarises the locations
occurred in the second decade, and 70.8% were below the of extravasation and retention cysts. These data are esti-
age of 30 years. Only 11.2% arose in patients over the age mated from a number of publications that have recorded
of  40 years. More recently, Bezerra et  al. (2016) reported the site distribution (Southam  1974; Harrison  1975;
719  mucous extravasation cysts and found that 36.7% Eversole 1987; Chi et al. 2011; Bezerra et al. 2016; Stojanov
occurred in the second decade and 78.7% below the age of et al. 2017). The proportions are rounded to the nearest 1%
30 years. or 5% for illustrative purposes and to aid recall.
In contrast, mucous retention cysts are found in older age In the largest study, Chi et al. (2011) found that 81.9% of
groups, with an average age of about 55 years and a peak extravasation cysts were located in the lower lip, and about
incidence in the sixth and seventh decades (Table  15.1). 5% each in the floor of the mouth and buccal mucosa.
Harrison (1975) reported that 42.5% of cases occurred in
the seventh decade and that 85% were found in patients
over 40 years of age. Only one case was found in the sec-
ond decade.
Mucoceles are frequently found in the first decade, and
are often diagnosed in neonates and even newborns. All
mucoceles in children are of the extravasation type. In a
large series of 594 cases reported by Cataldo and Mosadomi
(1970), 16 (2.7%) were in infants less than 1 year old.
Mínguez-­Martinez et al. (2010) described 89 cases in chil-
dren ( 14 years) and found 11 cases in children under
1 year and 4 cases that were congenital; 3 occurred on the
lower lip and 1 was found on the uvula. Shapira and Akrish
(2014) reviewed the literature up to 2014 and found 60
cases of mucoceles in infants ( 1 year), of which 30 were
Figure 15.1  Mucocele of the lower lip. This is the most
found in neonates ( 1 month). Of these 30 cases, 19 (63.3%) common site and clinical presentation of this lesion.
arose in the floor of the mouth and were designated
as ranulas. Table 15.2  Site distribution of mucoceles (%).

Sex Type of mucocele


Most studies show that the frequency of mucoceles in
males and females is almost equal, but with an overall Site Extravasation Retention
slight predilection for females (Table 15.1). This predilec-
tion may arise because mucous retention cysts may be Lower lip 75 10
found somewhat more frequently in females than in males. Floor of mouth 10 25
Chi et al. (2011) reported only extravasation cysts (n = 1715) Buccal mucosa 5 30
and found an equal sex distribution (50.7% males), whereas Ventral tongue 10 5
Stojanov et al. (2017) reported only retention cysts (n = 177)
Palate 1 20
and found that 58.2% arose in females. In Harrison’s (1975)
Upper lip <1 5
review, he found that 54.8% of extravasation cysts (n = 356)
arose in males, but that 27 of 40 retention cysts (67.5%) Data are estimates from a number of publications that have recorded
were found in females. site (see text for details).
­Mucocele 241

Conceição et  al. (2014) also found a very high frequency a history of repeated rupture (Chi et  al.  2011; Bezerra
(92%) in the lower lip, but others have found lower propor- et al. 2016).
tions, ranging from 67.3% (Bezerra et  al.  2016) to 79.8% Most mucoceles arise in the lower lip and give few
(Mínguez-­Martinez et  al.  2010), and most studies record ­diagnostic problems, with most studies showing that 85%
about 10% each in the floor of the mouth and buccal are correctly diagnosed on clinical examination (Chi
mucosa (Table  15.2). Extravasation cysts in the upper lip et al. 2011; Bezerra et al. 2016). Deeper, firmer lesions may
are very rare. Chi et al. (2011) and Conceição et al. (2014) be confused with a fibroepithelial polyp or a small salivary
reported nearly 2000 cases between them and did not find gland tumour. However, when considering the differential
a single cyst in the upper lip. Bezerra et  al. (2016) found diagnosis, it should be noted that mucoceles affect the
only 2 cases (0.3%) in the upper lip. lower lip while salivary gland tumours almost always affect
In contrast, retention cysts are most commonly found in the upper lip. Some mucoceles have been diagnosed as
the floor of the mouth and buccal mucosa, accounting for lipomas or, if they become blood filled, may resemble
about 55% of the total, but are relatively uncommon in the haemangioma.
lower lip. About 5% are found on the upper lip. The palate is At some sites, oral mucoceles have a characteristic clini-
rarely affected by mucoceles, but when they do occur they cal presentation and may pose particular management or
are almost always retention cysts, and are usually found in diagnostic problems. These clinical variants include super-
the soft palate or at the junction of hard and soft palate. ficial mucoceles, mucoceles of the anterior ventral tongue
A distinctive type of extravasation mucocele has been affecting the glands of Blandin–Nuhn, and the ranula.
reported in the anterior aspect of the ventral surface of the
tongue associated with the glands of Blandin and Nuhn Superficial Mucoceles
(Sugerman et al. 2000). A number of series have been pub- Occasionally, mucoceles may be very superficial and lie
lished that suggest that these cysts may represent up to 15% just below the epithelium and resemble a subepithelial
of extravasation mucoceles (Jinbu et al. 2003; de Camargo blister or vesicle (Figure 15.2). These were first described
Moraes et al. 2009). The clinical variants are discussed in by Eveson (1988), who reported eight cases. All presented
more detail in the next section. as  small superficial lesions that resembled blisters, and
all  had been given a preliminary clinical diagnosis of a
Clinical Presentation vesiculo-­bullous disorder, either bullous lichen planus or
Mucoceles present as painless swellings that are round or mucous membrane pemphigoid. All the lesions occurred
oval with a smooth surface (Figure 15.1). The swelling is in females and six were associated with lichen planus and
often firm and the colour of normal mucosa, but about 30% one with pemphigoid. The soft palate (six cases) was the
of lesions are quite superficial and may be blue or grey in most common site. Superficial mucoceles often rupture
colour and fluctuant (Chi et al. 2011). Occasional lesions and may present as small ulcers.
are clear or blister-­like or may be red due to intralesional
haemorrhage. The colour and texture of the lesion depend
on its size and depth, with more superficial lesions being
blueish or clear and fluctuant. Mucoceles are almost always
sessile, but occasionally a superficial pedunculated and
‘ballooning’ lesion may be seen.
Sometimes the swelling may develop suddenly at meal-
times and then resolve. Patients often report that the cyst
has recurred or that it periodically bursts and drains. They
may have been present for only a few days, but some
patients tolerate them for months or even years before
seeking treatment. The mucocele may be only 1–2 mm in
diameter, but are usually larger: the majority are between 5
and 10 mm, but they may reach up to 40 mm (Chi
et al. 2011).
Mucoceles are usually symptomless and complaints of
pain or discomfort are rare. Although they are caused by
trauma and patients are often aware of biting them, only
Figure 15.2  A superficial mucocele in the retromolar region.
about one-­quarter of patients are able to relate the develop- The lesion is raised and slightly polypoid, and resembles a
ment of the cyst to trauma and a similar proportion report blister or vesicle. Source: Courtesy of Prof. John Eveson.
242 Cysts of the Salivary and Minor Mucous Glands

Subsequently there have been a number of reports that there have been many case reports and two series (Jinbu
have confirmed a predilection for females and an associa- et  al.  2003; de Camargo Moraes et  al.  2009) that are
tion with lichen planus (Bermejo et al. 1999; Lv et al. 2019). ­summarised in Table 15.3.
Lv et  al. (2019) reported nine patients with oral lichen These mucoceles occur in a wide age range, but with a
planus who presented with multiple small superficial mean of about 15–17 years and a peak in the second dec-
mucoceles. All cases involved the soft palate and some also ade. The cysts are usually less than 10 mm in diameter, and
had lesions on the buccal mucosa and lower lip. The appear as polypoid or pedunculated swellings. Most lesions
authors speculated that the lesions may be caused by (65–70%; Table 15.3) arise in the midline towards the tip of
­damage or blockage of small salivary ducts secondary to the ventral surface of the tongue. The remainder are found
lymphocytic infiltration and fibrosis associated with the in the midline towards the root of the tongue, although
lichenoid reaction. very occasional lesions may be lateral to the midline. They
Superficial mucoceles have also been associated with are always extravasation in type, and are characterised by a
graft-­versus-­host disease (Campana et al. 2006) and, more blueish colour and fluctuance. Cysts at this site are proba-
recently, 10 cases have been reported associated with bly associated with biting or tongue thrusting and there is
mucositis following radiotherapy for head and neck cancer often a history of trauma. Occasionally, cysts of Blandin–
(Prado-­Ribeiro et al. 2018). These cases were seen in 1.2% Nuhn may reach a large size and cases of 30 mm or greater
of patients who had received radiotherapy, and affected have been reported (Andiran et al. 2001). One case that was
mostly the floor of the mouth or buccal mucosa. 21 mm in diameter has been reported in a neonate (Ochiai
Overall, superficial mucoceles are uncommon. In their and Nakayama  2015). At this site, and in infants, large
analysis of 1715  mucous extravasation cysts, Chi et  al. lesions may be alarming and may interfere with normal
(2011) found only 3 (0.2%) that met the clinical and histo- feeding or may compromise the airway.
logical criteria for a superficial mucocele, although a fur-
ther 4.1% showed either clinical or histological features.
Ranula
Similarly, in a histological review of 667  mucoceles, de
Brito Monteiro et al. (2016) found only 20 (2.9%) cases. The term ranula2 is used to describe a characteristic type of
mucocele that occurs in the floor of the mouth. It is worthy
Mucocele of the Glands of Blandin–Nuhn of special consideration because on occasion, and espe-
Mucoceles are relatively rare on the tongue and when cially in children, a ranula may be so extensive as to cause
they  do arise, a distinctive clinical presentation is of a gross swelling in the neck and may compromise the airway.
mucocele arising towards the anterior aspect of the ventral The clinical presentation, treatment, and pathogenesis of
tongue, affecting the seromucous glands of Blandin–Nuhn ranula have been well reviewed by Baurmash (2003) and
(Figure  15.3). They were first described in detail by Harrison (2010).
Sugerman et al. (2000), who reported five cases. Subsequently Ranulas appear as soft, fluctuant swellings in the floor of
the mouth and are always to one side of the midline
(Figure 15.4), although rare examples have been reported
that are bilateral. The majority arise from the sublingual
glands, although some may arise from other small glands
in the anterior floor of the mouth. The sublingual gland is
actually a collection of multiple, small unencapsulated
mucous glands that lie between the floor of the mouth and
the mylohyoid muscle. There is often a large duct (Bartholin
duct) that opens into the submandibular duct or directly
into the mouth at the sublingual papillae (carunculae sub-
lingualis), but there are also between 10 and 30 small ducts
(the ducts of Rivinus) that open along the sublingual folds,
each one from a small gland. The posterior part of the

2  Ranula is a slightly unfortunate term, since it is derived from the


Latin word for frog and refers to the similarity of the lesion to the
underbelly or to the air sac of a small frog. The derivation is from the
Figure 15.3  A mucocele on the ventral aspect of the tongue, Latin rāna (frog) and the suffix -­ula (small). Harrison (2010)
associated with the glands of Blandin–Nuhn. Source: Courtesy of attributes its first usage to John Bannister in A Compendious
Prof. John Eveson. Chyrurgerie, 1585.
­Mucocele 243

Table 15.3  Mucocele of the glands of Blandin–Nuhn.

  Size (mm)
References n Male (%) Age mean (range) Mean (range) polypoid (%) Tip, midlinea (%) Frequencyb (%)

Jinbu et al. (2003) 26 19.2 17.0 (5–36) 6 (3–12) 73.1 65.4 9.9
De Camargo Moraes et al. (2009) 48 58.3 15.3 (5–49) NR 85.7 70.8 15.4

Summary of the key features from two large series.


n, number of patients.
a 
Refers to lesions towards the tip of the tongue in the midline. Other lesions arose towards the root of the tongue.
b 
Frequency as a proportion of all mucous extravasation cysts.

submandibular space and tissues of the neck. Examination


of human cadavers has shown that mylohyoid perforations
and herniation of the sublingual gland may be found in up
to 43% of individuals (Nathan and Luchanski 1985; Harrison
et al. 2013). Lee et al. (2016) reviewed computed tomogra-
phy (CT) scans of 41 patients with plunging ranulas and
found that in 36 (87.8%) cases the mucus passed directly
through a defect in the mylohyoid muscle, and only 5 (12.2%)
showed spillage of mucus over the posterior margin of the
muscle. They also demonstrated that in 31 (75.6%) patients
there was herniation of the sublingual gland through the
mylohyoid defect into the submandibular space.
In the largest series of ranulas, Zhao et al. (2004) reviewed
Figure 15.4  Ranula. The cyst arises to one side of the midline 580 cases and found that the majority (67.9%; n  =  394)
in the floor of the mouth. The lesion is superficial and appears were simple oral mucoceles presenting as a swelling in the
as a blueish fluctuant swelling.
floor of the mouth. The remainder (32.1%; n = 186) were
plunging ranulas.
sublingual gland often merges with a superior extension of Of the plunging ranulas, the majority (64.0%; 119 cases)
the submandibular gland (the uncinate process) that presented in the submandibular or submental region only
curves upwards and extends over the posterior aspect of without an oral swelling, and 36.0% (67 cases) presented as
the mylohyoid muscle. Occasionally a small oral ranula a mixed intraoral and extraoral swelling. The peak age of
may be of the retention type, arising from a blocked duct of presentation was in the second decade (range 3 months to
Rivinus, but the vast majority are extravasation cysts 80 years) and there was a slight predilection for females
caused by spillage of mucus from a ruptured duct or acini (54.5%). Most lesions, whether plunging or not, were 2–3 cm
(Harrison 2010; McGurk et al. 2008) in diameter. Zhao et al. found 9 cases (1.6%) that were bilat-
Ranulas may present in three different ways. Most com- eral. All 580 lesions were mucous extravasation cysts.
monly they are a simple oral or superficial ranula and pre- Occasionally a ranula may be associated with a swelling
sent as an intraoral swelling only. They may however extend of the submandibular gland that increases and decreases in
into the submandibular space as a plunging ranula. Plunging size with gustatory stimulation. This was found in 13 (2.3%)
ranulas may present as a submandibular or submental swell- cases in Zhao’s series. Although mucoceles may arise from
ing only, or as a mixed intraoral and extraoral swelling. the submandibular gland (Anastassov et  al.  2000), this is
Plunging ranula are mucous extravasation cysts of sublin- thought to be extremely rare (Harrison 2010) and the sub-
gual gland origin that ramify diffusely into the subman- mandibular swelling associated with eating is thought to
dibular space and may extend into the neck. The extravasated be due to partial blockage of the submandibular duct by
mucus enters the submandibular space by either ‘spilling’ fibrosis or by pressure (Zhao et al. 2004; Harrison 2010).
over the posterior aspect of the mylohyoid muscle, or In most cases of plunging ranula the swelling is limited
through a perforation in the mylohyoid muscle. A deficiency to the submandibular or submental region, but occasional
or hiatus between the deep and superficial aspects of the lesions may be extensive, with evidence of mucus extrava-
mylohyoid muscle has been reported to be fairly common sation into the parapharyngeal space, or extending deep
and herniated projections of the sublingual gland through into the cervical tissues and occasionally into the thorax
these perforations may permit mucus extravasation into the (Zhao et al. 2004; Pang et al. 2005). Extensive ranulas in the
244 Cysts of the Salivary and Minor Mucous Glands

neck, especially in children, may be mistaken for a cystic with ductal dilatation, which they interpreted as indicative
hygroma (Jain et al. 2012). of an obstructive aetiology. Eversole (1987) had also noted
sialadenitis and ductal ectasia and suggested an obstruc-
tive aetiology associated with mucous plugs, or with duct
Pathogenesis
constriction secondary to trauma and fibrosis.
Most mucoceles, whether of the extravasation or retention Lesions similar to mucoceles are occasionally associated
type, are thought to arise as a result of trauma that causes with persistent sialadenitis of the minor glands of the lips or
rupture or blockage of a salivary duct or damage to acini. buccal mucosa. Such lesions have been termed cheilitis
The evidence for a traumatic aetiology is most convincing glandularis and stomatitis glandularis, respectively (Tal
for mucous extravasation cysts, the vast majority of which et al. 1984; Williams and Williams 1989; Cannell et al. 1997;
arise in young people and at sites that are most likely to be Musa et  al.  2005). Affected patients present with multiple
exposed to trauma, including the lower lip and ventral cysts, often with suppuration, that histologically showed
tongue. Acute trauma causes rupture of the salivary ducts so mucous extravasation, as well as cystic spaces lined by ductal
that the continually secreted saliva spills into the adjacent epithelium that had often undergone oncocytic metaplasia.
tissues, resulting in a mucus-­filled cavity. Overall, only about
25% of patients are able to associate their lesions with trauma
Histopathology
(Chi et  al.  2011), but a number of early animal studies,
which are unlikely to be repeated, have convincingly shown The optimal treatment for oral mucoceles is excision of the
that damage or rupture of salivary ducts causes pooling of lesion along with the underlying glands. On gross examina-
mucus in the connective tissues and formation of mucous tion the specimen is often a thin-­walled cyst with associated
extravasation cysts (Bhaskar et  al.  1956; Standish and salivary gland, and frequently a portion of oral mucosa is pre-
Shafer  1957,  1959; Chaudhry et  al.  1960). In other experi- sent on the superficial surface (Figure 15.5). When the speci-
ments on cats, it has been shown that ligation of the sublin- men is cut, the cyst may be discrete, bound by a thin lining,
gual ducts, without impairment of the parasympathetic and filled with gelatinous mucoid material. For extravasation
nerve supply, resulted in rupture of acini and extravasation mucoceles the lumen may not always be obvious and the
of mucus, without dilatation or rupture of ducts (Harrison specimen may resemble loose connective tissue with a mucoid
and Garrett  1972,  1975). The authors suggested that the texture. If the lesion has been marsupialised or removed
minor salivary glands of humans, like the cat sublingual piecemeal, then only fragments of t­ issue may be received.
gland, secreted spontaneously, and that duct blockage with Microscopically, mucous extravasation and retention
subsequent rupture of the acini may also be a factor in the cysts show distinctive features. The mucous extravasation
pathogenesis of mucoceles of the oral mucosa. Ranulas are cyst is not lined by epithelium and should properly be
mucous extravasation cysts and have a similar pathogenesis, regarded as a pseudocyst, whereas the retention cyst is a
although the distinctive anatomy of the floor of the mouth true cyst lined by epithelium that is derived from the duct
determines the clinicopathological features of the plunging of the involved gland.
ranula. This has been discussed in the previous section.
The pathogenesis of the mucous retention cyst is less Mucous Extravasation Cyst
clearly established, although it is thought that complete or Extravasation cysts most often show a well-­defined cystic
partial obstruction may lead to ductal dilatation and forma- space lined by a wall of inflamed fibrous or granulation tis-
tion of an epithelial-­lined cystic space. In most cases, it is sue (Figures 15.5a and 15.6a). The granulation tissue may
likely that chronic persistent trauma may lead to hyper- be of variable thickness and is often surrounded by a zone
keratosis or periductal fibrosis that constricts the ducts, of denser, collagenous fibrous tissue with variable degrees
resulting in complete blockage or slowing of salivary flow of inflammation. Macrophages are almost always promi-
and formation of mucous plugs. nent and are seen within the granulation tissue wall as well
Stojanov et  al. (2017) examined 177  mucous retention as within the lumen of the cyst (Figures  15.6a and 15.8),
cysts (they used the term ‘intraoral salivary duct cyst’) and where they may be few in number and scattered, or occa-
found that 41.8% showed histological evidence of ductal sionally may fill the lumen. The macrophages are often
obstruction in the form of mucus stasis, which varied from mucus filled and foamy and will stain positive with mucin
intraluminal accumulations of mucus to well-­formed stains, for example periodic acid–Schiff (PAS) or mucicar-
mucous plugs that obstructed the lumen. Occasional cysts mine. Often macrophages are prominent on the luminal
(4.5%) contained calcifications, suggesting that sialoliths surface of the granulation tissue and occasionally they may
may sometimes be a factor. Stojanov et al. also found that be epithelioid in morphology and form a palisade that
90% of the cysts were associated with chronic sialadenitis shows a close resemblance to a simple epithelial lining
­Mucocele 245

(a) (b)

*
Figure 15.5  A mucous extravasation cyst (a) and a mucous retention cyst (b). The cysts have been received intact and show oral
mucosa on one aspect (top of both images) and lobules of salivary tissue at the base (*). Both cysts contain mucus, and in b there are
focal accumulations of inflammatory cells. (Insets a and b: see Figure 15.6.)

(a) (b)

Figure 15.6  The typical linings of a mucous extravasation cyst (a) and a mucous retention cyst (b). (a) The lumen of the
extravasation cyst is filled with mucus containing scattered macrophages. (b) The retention cyst is lined by cuboidal or columnar
epithelium showing oncocytic change. Images are taken from the cysts shown in Figure 15.5, insets a and b.
246 Cysts of the Salivary and Minor Mucous Glands

Figure 15.7  A mucous extravasation cyst. The


wall is folded into papillary projections and is lined
by an amorphous eosinophilic ‘membrane’
overlying a layer of prominent macrophages with
evidence of palisading.

(Figure 15.7). Adjacent mucous glands are usually inflamed synovial metaplasia that have been described in association
and show dilatation of the ducts. Sometimes a ruptured with implants and prostheses in other tissues including
duct may be seen feeding into the cyst lumen. breast and hips. They have called this feature ‘papillary
Chi et  al. (2010) described two mucous extravasation synovial-­like change’ and suggest it is a variant of mucocele
cysts where the granulation tissue wall was folded into pap- that must be distinguished from other salivary lesions that
illary projections, with an amorphous eosinophilic surface may exhibit a papillary growth pattern. In their large series
layer resembling a membrane. Below this was a layer of of lesions, the same authors found only two extravasation
rounded to spindled cells with occasional palisading. These cysts (0.1%) with these features (Chi et al. 2011).
cells were shown to be macrophages. The authors were We have seen similar changes (Figure 15.7) and feel that
struck by the resemblance of these features to synovial they are rather more common than Chi et al. (2010, 2011)
membrane, and in particular to examples of papillary suggested, since on occasion they may only involve a por-
tion of the cyst wall. A number of authors have drawn
attention to, or have illustrated, similar appearances
(Harrison  1975; Conceição et  al.  2014; de Brito Monteiro
et al. 2016). Conceição et al. (2014) found palisaded mac-
rophages in 4% of cases and papillary projections in 3%,
while de Brito Monteiro et  al. (2016) found synovial-­like
changes in 54 (8.1%) of their 667 cysts, and papillary pro-
jections in a total of 86 (12.9%) cases. Often, however, illus-
trations of the papillary processes show folding of the
lining rather than true papillary outgrowths.
Li et al. (1997) reported a single case of a mucous extrava-
sation cyst that was lined by typical granulation tissue, but
the lumen was filled with round or polygonal, slightly lam-
inated eosinophilic globules. Many of the globules were
surrounded by spindled fibroblast-­like cells and in places
similar globular protrusions were in continuity with the
granulation tissue of the wall. The authors noted that the
globules were similar to those reported in mucoceles of
the  appendix, which have been called ‘myxoglobulosis’
of the appendix. Li et al. (1997) therefore were the first to
Figure 15.8  Mucous extravasation cyst. The lumen is filled with
macrophages, most of which are foamy and filled with mucus. In
introduce this term in relation to oral extravasation cysts.
the centre is an eosinophilic, slightly laminated or fibrillar Subsequently there have been a number of reports of simi-
globular structure. lar globular structures and authors have continued to use
­Mucocele 247

the term myxoglobulosis (Ide and Kusama  2002a; Shah Shah’s views were supported by Ide et  al. (2010), who
2003; Paremala et  al.  2011; Schulman and Jordan  2014). also illustrated an extravasation cyst that showed transi-
Most reports have shown a striking accumulation of glob- tions between papillary processes and collagenous globules
ules that fill the cyst lumen and have noted that this is a in the lumen. They suggested that the intraluminal globu-
rare occurrence. In Li et al.’s original report, they only saw lar structures could be isolated spherules, or be a result of
this feature in 1 of 657 cysts examined (0.15%). Chi et al. cross-­cutting of papillary structures protruding into the
(2011) defined oral myxoglobulosis as globules occupying lumen (e.g. Figure 15.7). Furthermore, Ide et al. (2010) also
30% or more of the lesion and found a frequency of only showed that the papillary processes that gave rise to the
6 cases in 1715 cysts examined (0.4%). Others have found globules had a hyalinised or eosinophilic surface similar to
them to be more common and similar globules have been that described by Chi et al. (2010) as synovial-­like (Figure
reported in 2.9% (de Brito Monteiro et  al.  2016), 9% 15.8). They suggest that the two features are associated and
(Conceição et al. 2014), and 31% (Shah 2003) of cases. represent an evolving continuum in the formation of the
We have also found eosinophilic, slightly laminated, or cyst, associated with the ongoing repair process and
fibrillar globules to be quite common (Figure 15.8), and it is walling-­off of mucus by granulation tissue.
apparent that the reported frequencies may depend on how We would support this concept and would not regard
much importance each author places on the findings and on the globules or synovial-­like changes as representing vari-
different definitions of ‘myxoglobulosis’. Shah (2003) found ants of mucoceles, but rather regard them as random inci-
globular structures in 31% of 71 mucous extravasation cysts dental findings that have probably been noted but ignored
examined. He described a spectrum of change, from mucous by pathologists when making a straightforward diagnosis of
extravasation without cyst formation, to fully formed cystic common lesions. The term myxoglobulosis should be used
lesions lined by granulation tissue. In lesions with early only as a descriptive term for those cases where the lumen
mucous extravasation, he noted separation of collagen bun- is filled with globules, but even then it is not associated with
dles that in places rounded up to form globular structures any distinctive clinical features or management problems.
that continued capillaries and were associated with mac- In the majority of cases mucous extravasation cysts will
rophages. Similar globular structures were found in 11 of 38 show obvious cystic cavities filled with mucus, but over time
(28.9%) fully formed cysts, but only between 1 and 5 glob- the lesion may become more solid as the tissue organises to fill
ules were seen in most lesions (e.g. Figure 15.8). He found the lumen and the cyst is replaced by loose, inflamed granula-
that the globules had the staining characteristics of collagen tion tissue containing pools of mucus. Mucin stains may be
and proposed that they arose as a result of walling-­off of helpful to demonstrate residual pools of mucus and mucus-­
separated fragments of collagen that then become incorpo- filled macrophages. Many cysts are also ruptured or collapsed
rated into the cyst lumen. Shah (2003) did not support the and the lumen may appear only as ramifying slit-­like spaces.
use of the term ‘myxoglobulosis’ for oral lesions. He and
others (Henry et al. 2008) have pointed out that the similar Superficial Mucoceles
structures seen in appendix lesions are composed primarily Superficial mucoceles are extravasation cysts where the
of mucins, whereas those in oral mucoceles appear to be spilled mucus accumulates at the epithelial–connective
collagenous, and are probably associated with fragmenta- ­tissue interface and raises the epithelium to form a blister
tion of collagen during the evolution of the lesion. or vesicle (Figure  15.9). At the base the cyst is lined by

Figure 15.9  A superficial mucocele. The


base of the lesion is composed of
inflamed granulation tissue and the roof
of atrophic oral epithelium. Note the
inflamed salivary tissue and dilated
ducts beneath the lesion.
248 Cysts of the Salivary and Minor Mucous Glands

inflamed granulation tissue and the roof is composed of a The epithelial lining often shows additional features due to
thin atrophic epithelium. Often, as part of the healing pro- reactive or metaplastic changes. The histopathology of mucous
cess, the adjacent epithelium may migrate across the floor retention cysts and the variable pattern of the epithelium have
of the mucocele, and in a certain plane of section the lesion been well described by a number of authors (Southam 1974;
may appear as wholly intraepithelial. Care must be taken Harrison  1975; Eversole  1987), but more recently Stojanov
not to misdiagnose these lesions as a vesiculobullous disor- et al. (2017) have undertaken a detailed quantitative analysis
der. Mucin stains are helpful, but the presence of foamy of the histopathological features of 177 cysts. Their findings
macrophages and inflamed salivary tissue at the base help are representative of the findings of the previous papers.
to make the correct diagnosis (Figure 15.9). Stojanov et al. (2017) found that 85.3% of cysts were lined
by a layer of flattened cuboidal or columnar epithelium,
Mucous Retention Cysts one to two cell layers thick (Figure 15.10a), but that 68.4%
Retention cysts are true cystic lesions lined by epithelium. of cases showed additional reactive or metaplastic changes
Most are lined by a simple cuboidal or columnar epithe- that involved all or part of the lining. The most common
lium, which may resemble a normal salivary duct feature was oncocytic metaplasia, which was seen in 50.8%
(Figures 15.5b, 15.6b, 15.10). Most retention cysts are uni- of cysts. Oncocytes were seen as deeply eosinophilic and
cystic, but multicystic lesions have been described in up to sometimes granular cells that often occupied the whole of
18% of cases (Eversole 1987; Stojanov et al. 2017). the cyst lining (Figures 15.6a and 15.10d).

(a) (b)

(c) (d)

Figure 15.10  Mucous retention cysts. Variable features of the epithelial lining. (a) The typical lining is of flattened cuboidal or
columnar cells. (b) Pseudostratified ciliated epithelium. (c) A dilated duct is continuous with the cyst lumen. (d) Folding of the cyst wall
produces infoldings or undulations that resemble intraluminal papillae. This lining also shows oncocytic change.
­Mucocele 249

Other features that were seen in the lining epithelium that sialoliths are an important factor in the pathogenesis
included mucous (goblet) cells (36.2%), squamous cells of retention cysts. In their examinations of more than
(23.7%), pseudostratified ciliated epithelium (16.4%; 300 lesions, none of the authors has been able to identify
Figure  15.10b), and apocrine changes (14.1%; Stojanov any obvious cause of ductal obstruction that may be associ-
et al. 2017). These authors also found that 40.1% of cases ated with mucous retention (Southam 1974; Harrison 1975;
showed more than one type of metaplasia and that 7.3% of Eversole 1987; Stojanov et al. 2017).
lesions were multicystic (Figure 15.11). More than 70% of cysts are inflamed and in over 90%
Many cysts also show undulation or folding of the cyst of cases the associated salivary glands show inflammation
wall to form variably sized papillary-­like processes that pro- and ductal ectasia consistent with chronic obstructive
trude into the lumen (Figures 15.5b and 15.10d). Stojanov sialadenitis (Stojanov et al. 2017). Occasionally a fortuitous
et al. (2017) found an undulating architecture in 41.2% of plane of section may show a dilated duct entering the
cases, and interpreted this as folding due to collapse of the lumen of the cyst (Figure 15.10c).
cyst wall, rather than true papillary processes with a fibro-
vascular core. Nevertheless, their photomicrographs show Histological Differential Diagnosis
papillary-­like structures and 39 (22.0%) of their cases were A diagnosis of mucous extravasation cyst is usually
lined by oncocytic epithelium with infoldings, closely straightforward and should not present a problem. The
resembling oncocytic papillary cystadenomas. main area of confusion arises in the diagnosis of a superfi-
Eversole (1987) described similar features. He examined cial mucocele, which may clinically and histologically
121 epithelial-­lined mucoceles (which he preferred to call resemble a vesiculobullous lesion. In Eveson’s (1988) origi-
sialocysts) and found that 41 (33.8%) were lined by onco- nal report, all eight cases were misdiagnosed clinically as
cytic epithelium and that 27% of these had small papillary vesiculobullous disorders, and three were misdiagnosed
projections. A further 10 (8.3%) cases were described as histologically as mucous membrane pemphigoid. On a sin-
‘mucopapillary cysts’, which were lined by a cuboidal and gle haematoxylin and eosin (H&E)-­stained section, distinc-
columnar epithelium with prominent mucous cells, and tion between a mucocele and pemphigoid may be difficult.
papillary projections that protruded into the lumen. There Direct immunofluorescence can be used to confirm a diag-
is no doubt that some of these lesions resemble papillary nosis of pemphigoid, but false negative staining is com-
cystadenoma and Eversole (1987) suggested that in some mon. More useful is a mucin stain that will confirm mucus
cases this may be the correct diagnosis. in a mucocele. Other helpful features include the presence
Most cysts contain mucus in the lumen, which in places of foamy macrophages and of inflamed salivary tissue with
may form amorphous eosinophilic ‘plugs’, and about 5% dilated and sometimes ruptured ducts at the deep aspect.
may show focal areas of calcification resembling a salivary A histological diagnosis of retention cysts may be more
sialolith (Figure  15.11). However, this is probably associ- problematic, because a number of salivary neoplasms may
ated with stasis within the cyst and there is no evidence present as cystic lesions. The two most commonly

Figure 15.11  Mucous retention cyst. This lesion is


multicystic, but also shows a number of other
features. There is folding of the wall with a
papillary pattern and the lumina contain mucus
with amorphous eosinophilic ‘plugs’. Associated
glands show features consistent with obstructive
sialadenitis, with prominent dilated ducts. One of
these (arrow) contains a sialolith.
250 Cysts of the Salivary and Minor Mucous Glands

misdiagnosed lesions are cystadenomas and low-­grade children with mucous extravasation cysts, Mínguez-­Martinez
mucoepidermoid carcinoma. et al. (2010) found that 39 (43.8%) cysts resolved spontane-
Mucous retention cysts are lined by a simple cuboidal ously after an average time of three months. Most lesions,
to columnar epithelium, which often shows oncocytic change. however, are removed surgically and the basic principle is
These features are also seen in cystadenomas of both the that the offending gland should be removed with the cyst, to
minor and major glands. On occasion it may be difficult to avoid recurrence. Marsupialisation of extravasation cysts is
make a distinction and a definitive diagnosis may not always sometimes carried out and can be successful if the underly-
be possible. A particular problem may arise when faced with ing gland is also removed (Baurmash 2003). Marsupialisation
a retention cyst that shows infoldings of the lining and onco- alone has a recurrence rate of up to 50% or more.
cytic change. Cystadenomas, however, are more often multi- Treatment of ranulas is particularly problematic because
cystic with variably sized cystic spaces, and more often contain lesions may be large and surgery in the floor of the mouth
intraluminal papillary projections. These are true epithelial may be extremely difficult and hazardous. Management of
papillae that may proliferate and occupy much of the lumen. ranulas is the subject of an extensive literature that is well
Retention cysts are more likely to be associated with inflamed covered in more appropriate surgical texts. Overall, most
salivary tissue that shows features typical of chronic obstruc- agree that surgical removal of the sublingual gland through
tive sialadenitis (Stojanov et al. 2017). Occasionally retention the mouth without any cervical approach is the favoured
cysts are associated with dense lymphocytic infiltrates and form of treatment. This removes the secreting source,
may resemble a Warthin tumour. Stojanov et al. (2017) found thereby preventing recurrence, and also avoids the problem
7 (4.0%) cases with a prominent lymphoplasmacytic infiltrate of a difficult neck dissection. In the large series of Zhao
that resembled Warthin tumour. However, the cysts did not et al. (2004), only 1.2% of lesions recurred if the ­sublingual
have the typical prominent papillae seen in Warthin tumour, gland was removed, compared with 57.7% if the ranula was
and their photomicrographs show diffuse lymphocytic infil- excised alone and 66.7% following marsupialisation only. A
trates without the germinal centres or follicles that are typi- more recent meta-­analysis of 39 reports supported this and
cally seen in the lymphoid infiltrate of Warthin tumour. Some showed that intraoral removal of the sublingual gland had a
of these cysts resemble intraoral lymphoepithelial cysts, some cure rate of 98.8% (Chung et al. 2019).
of which probably arise from salivary ducts and may be Harrison (2010) undertook a detailed review of the litera-
regarded as a mucous retention cyst. ture and found reports of 13 different treatments for oral
A more important diagnostic trap is to miss a diagnosis ranula, and 12 for plunging ranula. Treatments that
of low-­grade mucoepidermoid carcinoma when faced with included removal of the sublingual gland were always the
a small incisional biopsy of an epithelial-­lined cyst. On most successful, with recurrence rates as low as 0%.
examination of a whole specimen, the distinction should Marsupialisation alone had recurrence rates of 50% or
be easy since a mucoepidermoid carcinoma is usually mul- more, but if marsupialisation was followed by packing the
ticystic, but also shows evidence of infiltration and areas success rate rose to close to 100%. Harrison (2010) suggested
with an admixture of squamous, intermediate, and mucous that packing encouraged fibrosis that may seal the source of
cell. However, in places a cystic space may be lined by sim- mucus extravasation. Since the lumen of the ranula itself is
ple ducal epithelium and the fact is that on a small inci- lined by granulation tissue, the cyst wall does not have to be
sional biopsy, it may be impossible to differentiate between removed, as it will eventually resolve. These observations
a simple retention cyst and a mucoepidermoid carcinoma. have led to a more conservative approach for the manage-
It must be noted that mucoepidermoid carcinomas are one ment of the ranula, which reduces surgical intervention in
of the most common malignant salivary gland tumours in an anatomically compromised area. McGurk and colleagues
children, while retention cysts are uncommon in children. (McGurk et al. 2008; Harrison 2010; Hills et al. 2016) have
The best advice is that a pathologist must always be cau- described conservative treatments that involved decom-
tious when faced with a small biopsy of a simple cystic pression of the cyst followed by local removal of only the
lesion lined by ductal or oncocytic epithelium. Such lesions attached part of the sublingual gland, or by ligation of the
in children, especially at unusual sites such as the palate or leaking part of the gland with a mattress suture.
retromolar area, should always be treated with caution. Ranulas have also been treated by injection of the scle-
Never hesitate to defer a diagnosis and to seek imaging or a rosing agent OK-­432 (Rho et al. 2006). This substance only
second, larger or excisional biopsy. appears to have been used in South Korea and Japan and is
a sclerosing and immune system–stimulating agent derived
from lyophilised Streptococcus pyogenes. Rho et al. (2006)
Treatment
treated 21 patients with plunging ranulas and found total
Small mucoceles may require no surgical treatment provided resolution in 7 (33.3%) cases and near-­total or marked
that the patient finds them no hindrance. In a review of 89 shrinkage in most of the remainder. They propose that
­Salivary Duct Cyst 251

sclerotherapy using OK-­432 is a safe and potentially cura- Yamamoto  2001; Vinayachandran and Sankarapandian
tive substitute for surgery. Overall, however, success rates 2013; Belli et al. 2004), although occasional cases have been
are variable and sclerotherapy does not appear to be as reported in children (Munteanu et al. 2019). Most lesions
effective as surgery. In their meta-­analysis, Chung et  al. have affected only one gland, but occasional cases of bilat-
(2019) found 6 studies that had used OK-­432 and reported eral parotid cysts have been reported (Belli et al. 2004).
overall cure rates of only 76.4% for oral ranulas, and 65.6% Salivary duct cysts are thought to be retention cysts,
for plunging ranulas. with a similar pathogenesis and histopathological features
to retention cysts of minor salivary glands. However, some
authors suggest that at least some salivary duct cysts may
­Salivary Duct Cysts be developmental in origin, although it is difficult, if not
impossible, to establish the true cause of these lesions. A
The term salivary duct cyst is often used to refer to a cystic review of the few reports and published images of cases
lesion of the salivary glands caused by dilatation of the often shows inflamed salivary tissue and dilated ducts
ducts secondary to ductal obstruction. In this context it consistent with obstructive sialadenitis. Obstructive
would include mucous retention cysts, as described in the lesions in the parotid gland are frequently assumed to be
previous sections. Thus, in some current textbooks and in caused by mucus plugs, but such plugs or other causes are
the literature, intraoral retention mucoceles are sometimes rarely documented. Trauma, and stenosis to the parotid
called salivary duct cyst (Stojanov et al. 2017). However, it duct, has been reported as a possible cause (Belli
is more common that authors make a distinction between et al. 2004). It is ironic that so few cases are reported in the
intraoral and major gland lesions by using the term sali- submandibular gland, which is commonly obstructed by a
vary duct cyst to describe mucous retention cysts arising in sialolith. In the recent past, it was commonplace for sub-
the major salivary glands, most often the parotid (Batsakis mandibular glands to be removed if there was evidence of
and Raymond 1989). ductal obstruction due to a sialolith, and in histological
Major gland salivary duct cysts appear to be rare and examination of such glands we have noted that cystic dila-
there are few reports in the literature and no large case tation of ducts was common. However, this was often
series. In a review of 586  non-­neoplastic salivary gland regarded as part of the spectrum of changes associated
cysts, Takeda and Yamamoto (2001) found that 580 (99.0%) with obstructive sialadenitis and a diagnosis of duct cyst
were extravasation cysts, of which 113 (19.3% of the total) was not made. In contrast, such lesions are unusual in the
were ranulas. There were only 3  intraoral mucous reten- parotid gland and obstruction is not evident clinically,
tion cysts and 3 (0.5%) salivary duct cysts. More than 90% making it more likely that cystic change would be noted. It
arise in the parotid gland, and about 8.0% are seen in the is possible therefore that salivary duct cysts are equally
submandibular gland (Ellis and Auclair 2008). common in the submandibular gland as in the parotid
Most lesions are slowly enlarging, painless swellings up gland, but are not reported.
to 20 mm in diameter. They are usually mobile and may be Histologically, salivary duct cysts are usually unilocular
fluctuant. Salivary duct cysts are found in adults, most cysts lined by a simple cuboidal or columnar ductal epithe-
often in the sixth and seventh decades (Takeda and lium (Figure 15.12). Mucous cells and oncocytic metaplasia

Figure 15.12  A salivary duct cyst of the parotid


gland. An irregular cyst wall lined by ductal
epithelium.
252 Cysts of the Salivary and Minor Mucous Glands

similar to that seen in mucous retention cysts may be


Box 15.2  Key Features of Mucoceles and Cysts of the
found. Occasional lesions may be multilocular or papillary
Maxillary Sinus
and must be distinguished from cystic neoplasms. The
cysts are usually simple with a uniform lining, whereas
Mucocele
neoplasms show a more complex morphology, with a range
●● Occupies the entire sinus
of cell types. Cysts with an associated lymphocytic infil-
●● Caused by blockage of the ostium, often secondary
trate are discussed later in this chapter (‘Lymphoepithelial
to chronic sinusitis
Cysts’), but it is quite possible that some of these are sim-
●● Cystic structure filled with mucus and lined by
ple retention cysts that have arisen from ducts associated
sinus epithelium
with an intraglandular lymph node or duct-­associated
●● Expands the sinus and may destroy and perforate
­lymphoid tissue.
adjacent bone
Retention Cyst
­Cysts of the Maxillary Sinus ●● Epithelial-­
lined cyst
●● Caused by mucus retention as a result of blockage

Although not of salivary gland origin, these lesions are of a duct of a minor gland
associated with mucosal seromucous glands and are suffi- ●● Often small and clinically silent and found associ-

ciently similar to salivary mucous cysts that they can be ated with sinusitis or polyps
included in this chapter. ●● Dome-­ shaped radiopacity of antral wall. No expan-
When applied to the paranasal sinuses, the term sion and not destructive
mucocele has often been used loosely to refer to any type Pseudocyst
of cystic lesion associated with extravasation or accumula- ●● Accumulation of mucus and inflammatory exudates
tion of mucus. However, there are a number of distinctive that raise the mucosa from the bone of the sinus floor
entities that may affect the sinuses, including what is ●● Inflammatory in origin
regarded as a true sinus mucocele, retention cysts, and ●● Often secondary to odontogenic infection or sinusitis
mucosal pseudocysts. The distinction is important, because ●● Dome-­ shaped radiopacity on the floor of the sinus
these lesions have different clinicopathological features
and behaviours. The distinctive features and criteria for
diagnosis of this group of lesions were reviewed by Meer behaviour and the importance of early diagnosis and man-
and Altini (2006) and a simple classification with key fea- agement, warrants its separate consideration
tures is shown in Box 15.2. A further cyst that may arise in
the maxilla is the surgical ciliated cyst, which is discussed Clinical Features
in Chapter 16.
Frequency
Mucoceles are uncommon cystic lesions affecting the para-
­Mucocele of the Maxillary Sinus nasal sinuses, but the frontal and ethmoid sinuses are most
often affected, with only about 10% arising in the maxillary
A true sinus mucocele completely fills the sinus and is sinus (Natvig and Larsen  1978). Overall, therefore, true
caused by blockage of the outlet or ostium, usually second- maxillary sinus mucoceles appear to be rare. In a study of
ary to inflammatory changes associated with chronic rhi- symptomless young men, Savolainen et  al. (1997) exam-
nosinusitis. It most often affects the frontal and ethmoidal ined radiographs of 404 subjects and recorded ‘cysts or pol-
sinuses, but about 10% occur in the maxillary sinus. The yps’ in 7.2%. However, only 3.3% had a completely opaque
lesion is a true cyst filled with mucus and lined by the sinus, suggesting that this would be the highest possible
mucoperiosteum of the involved sinus. Sinus mucoceles prevalence of true mucoceles. Pérez-­Sayáns et  al. (2020)
are characterised by ballooning expansion, with destruc- reviewed the features of 214 patients with maxillary sinus
tion and perforation of the surrounding bone and dis- pathologies and found 8 (3.7%) retention cysts, but only 5
placement of adjacent structures. This may result in nasal (2.3%) mucoceles. The most common lesions were sinusitis
blockage, proptosis, and a range of other clinical symptoms (54.2%) and polyps (18.2%).
that are often mistaken for more aggressive neoplastic
lesions. The lesion has characteristic clinicopathological Age and Sex
features that serve to differentiate it from other cystic Mucoceles affect all age groups, with one of the largest
lesions of the sinus. This, along with its locally destructive series reporting a range from 13 to 71 years with a mean age
­Retention Cyst and Pseudocyst of the Maxillary Sinu 253

of 49.9 years (Busaba and Salman 1999). There appears to Pathogenesis


be no sex predilection for this lesion.
Sinus mucoceles are caused by obstruction of the ostium
with accumulation of mucus within the sinus that causes
Clinical Presentation
increased pressure and ballooning expansion. Often the
The characteristic clinical presentation is of a gradually
cause of obstruction is not apparent, but most patients
enlarging swelling of the cheek and lateral nasal region,
record a history or show symptoms of chronic sinusitis.
with obliteration of the nasolabial fold or of the buccal sul-
Occasionally blockage may follow nasal surgery, or may be
cus intraorally (Meer and Altini 2006). The lesion is expan-
due to a neoplasm or other pathology.
sile and in over 75% of cases there is bulging of the medial
wall of the sinus, causing partial obliteration of the nasal
cavity (Busaba and Salman  1999). Other features include Histopathology
pain or tenderness of the cheek or teeth, nasal drainage,
The histological features are not specific, but will con-
headache, and occasionally the orbit is involved, causing
firm the diagnosis and, more importantly, exclude another
proptosis. Patients may report a long history of symptoms
cause of expansion and bone destruction. Pathologists
of nasal congestion and sinusitis.
should note that occasionally the blockage is caused by a
neoplastic process or other pathology and one should be
Radiological Features alert to this possibility. Histologically, the appearance is
of a cyst filled with mucus or mucoid material, and lined
Radiology is important and usually the diagnosis is made
by  essentially normal sinus mucosa, covered by ciliated
on imaging and then confirmed by histology. The lesions
respiratory-­type epithelium or flattened simple cuboidal or
are better visualised using CT or magnetic resonance imag-
squamous epithelium. It is unusual to receive an intact
ing (MRI; Han et al. 1995; Busaba and Salman 1999). The
cyst, so most often the pathologist will examine multiple
defining criteria are complete opacification of the maxil-
fragments of mucoid material lined in places by epithe-
lary sinus on CT associated with evidence of bony expan-
lium. Portions of reactive bone may be seen at the margins.
sion (Figure 15.13). Early lesions may show only an opacity
There may be chronic inflammation in the wall and if there
and the overall morphology and bony margins of the sinus
is an associated allergic sinusitis, then acute inflamma-
may be normal, but over time, first the medial wall of the
tory cells and eosinophils may also be present (Meer and
sinus expands into the nasal passages, followed by progres-
Altini 2006).
sive expansion anteriorly and superiorly to raise the floor
of the orbit. As the lesion enlarges, there is often perfora-
tion of bone with extension into the soft tissues. With CT it
may be possible to demonstrate obstruction of the ostium. ­ etention Cyst and Pseudocyst of the
R
Maxillary Sinus

Retention cysts and pseudocysts are considered together


because they have similar behaviour and may be indistin-
guishable on radiological or clinical examination.
Together they are often referred to as a sinus cyst or
mucosal antral cyst, and many studies have not distin-
guished between them.

Clinical Features
Most studies use similar criteria for the definition of antral
cysts and illustrate ‘dome-­shaped’ soft-­tissue opacities
or  shadows arising from the sinus wall (MacDonald-­
Jankowski  1994; Myall et  al.  1974), and none used com-
plete opacification or ballooning expansion as a definition
of these cysts. It appears therefore that these studies did
not include true mucoceles, but investigated either reten-
Figure 15.13  Mucocele of the right maxillary sinus. There is
complete opacification of the sinus, with medial bulging into tion cysts or pseudocysts as distinctive lesions as defined by
the nose. Meer and Altini (2006) (Box 15.2).
254 Cysts of the Salivary and Minor Mucous Glands

Frequency that 48 (34.3%) had pain or swelling that might be of dental


Antral cysts and pseudocysts are probably quite common. origin. However, similar symptoms were seen 23.5% of
Myall et al. (1974) surveyed 1469 orthopantomographs and patients who did not have a mucosal antral cyst.
made a radiological diagnosis of mucosal antral cyst in 75
cases (5.1%). Allard et al. (1981b) found 94 examples in a
Radiological Features
series of 1080 radiographs (8.7%), and in their literature
review quoted the frequency in 11 other publications, It is on the basis of the radiological or CT features that
which ranged from 1.4% to 9.6%. In a review of 5021 these lesions are most clearly distinguished from sinus
sequential radiographs, Vallo et  al. (2010) found a fre- mucoceles. The cysts appear as spherical, ovoid, or dome-­
quency of 7.0%. A similar study by Casamassimo and Lilly shaped radiopacities that have a smooth and uniform out-
(1980) on 4546 panoramic radiographs showed a lower line (Figure 15.14). They may have a narrow or broad base
­frequency of 1.6%, while MacDonald-­Jankowski (1994) but, unlike mucoceles, the lesions are not destructive and
found a prevalence of 14% in a survey of 1000 consecutive the bony walls of the sinus maintain the normal contours.
panoramic radiographs in a South London dental casualty They vary in size from very small to large, but are distinct
department. from sinus mucoceles because they never completely fill
the sinus (MacDonald-­Jankowski 1994). Yeung et al. (2018)
Age measured 40 antral cysts and found an average size of
Antral cysts are found in young adults, with a peak in 6.28 mm (range 3–16.2 mm).
the  third decade and a reported mean age of 35.0 years Pseudocysts, which are often associated with odonto-
(Myall et  al.  1974; Casamassimo and Lilly  1980; genic infections, are located on the floor of the sinus, but
MacDonald-­Jankowski 1994). retention cysts may be located elsewhere, with up to 40%
located on the lateral walls or roof (Yeung et  al.  2018).
Sex They are otherwise indistinguishable. Follow-­up radiologi-
There is a slight predilection for males. Of the total of cal examination has shown that the cyst may persist with-
382  cases reported by Myall et  al. (1974), Casamassimo out a change in size for a long time, and may eventually
and  Lilly (1980), Allard et  al. (1981b), and MacDonald-­ disappear spontaneously.
Jankowski (1994), 225 (58.9%) occurred in males. The pro-
portion of males in each of these studies was roughly
Pathogenesis
the same.
The pathogenesis of mucosal cysts of the sinus has not
Site been definitely determined, but a previous infection is fre-
Most lesions are found on the floor of the sinus, with quently implicated. Studies have shown that there is a sig-
between 50% (Bhattacharyya  2000) and 100% at this site nificant association between dental infections and changes
(Casamassimo and Lilly 1980; Berg et al. 1989; MacDonald-­ to the mucosal lining of the maxillary sinus. However,
Jankowski  1994). Pseudocysts are always located on the
floor of the sinus, but retention cysts may be found attached
to the walls (Yeung et  al.  2018). In the majority of cases
only single cysts occur, but up to 20% of patients have bilat-
eral lesions (Casamassimo and Lilly  1980; MacDonald-­
Jankowski 1994; Bhattacharyya 2000).

Clinical Presentation
Retention cysts and pseudocysts are characterised by the
absence of symptoms, with the majority of lesions discov-
ered in the course of routine radiological examination.
Although many patients have a history of chronic sinusitis
or symptoms associated with dental pathology, these are
not specific and are no more common than in patients
without cysts (MacDonald-­Jankowski  1994; Vallo
Figure 15.14  Radiograph of a mucosal cyst of the maxillary
et  al.  2010; Yeung et  al.  2018). MacDonald-­Jankowski sinus. The lesion appears as a dome-­shaped radiopacity rising
(1994) reviewed 140 patients with mucosal cysts and found from the floor of the sinus.
­Lymphoepithelial Cyst 255

there is no distinction between nonspecific mucosal thick- are similar to mucous retention cysts of the oral mucosa
ening and localised dome-­shaped cystic change, both of (see Figures  15.5, 15.10, and  15.11) and are lined by flat-
which are equally associated with sinusitis or dental or tened, duct-­like cuboidal or columnar epithelium, or by
periodontal pathology (MacDonald-­Jankowski 1994; Vallo pesudostratified columnar epithelium. There may be little
et al. 2010; Yeung et al. 2018; Pérez-­Sayáns et al. 2020). inflammation other than related to the associated sinusitis
Few studies distinguish between pseudocysts due to and the cyst is filled with mucus. A calculus or mucus plug
accumulation of fluid and retention cysts associated with may be evident.
mucus retention. Meer and Altini (2006) agreed with
Gardner and Gullane (1986) that pseudocysts arise as a
Treatment
result of accumulation of inflammatory and mucus exu-
dates below the sinus mucosa, which is lifted from the Retention cysts and pseudocysts are not destructive and
underlying bone to form the characteristic dome-­shaped usually remain static, although many appear to regress
swelling on the floor of the sinus. Thus, the lesion is formed spontaneously. Because they usually cause little discom-
by pools of ‘mucoid’ material surrounded by inflamed fort, surgical intervention is often unnecessary. Removal is
fibrous and granulation tissue, with periosteum on the only needed if there are persistent and pertinent clinical
deep aspect. Meer and Altini (2006) reviewed the findings symptoms. Hadar et al. (2000) limited treatment to lesions
of earlier studies and suggested that odontogenic infection that occupied at least 50% of the sinus and used an endo-
of the maxillary teeth is the most common cause. scopic approach, which is now accepted as an effective
Pseudocysts are to be distinguished from antral polyps, treatment. In their series of 60 patients there were only two
which may be multiple, affect any part of the antral lining, cases that recurred and there were no complications.
and are a result of inflammation and accumulation of exu- Pseudocysts associated with a dental infection usually
dates in the hyperplastic and oedematous fibrous tissue of resolve when the cause has been removed.
the mucosa. By contrast, sinus mucoceles grow to a large size and can
With regard to retention cysts, it is generally agreed that be destructive. The basis of management is to decompress
these are caused by blockage of the ducts of the small the cyst, either by complete removal by curettage through
mucous glands in the sinus mucosa, resulting in a mucus-­ a  Caldwell–Luc approach or by an endoscopic approach
filled cyst lined by epithelium. Most of these lesions are through the middle meatus with marsupialisation of
small and clinically silent and are found as incidental find- the cyst.
ings in thickened and inflamed sinus mucosa removed for
chronic sinusitis, or within antral polyps. The cause of the
duct blockage may be an inspissated mucus plug or a small ­Lymphoepithelial Cysts
calculus.
The term lymphoepithelial cyst is often used as a generic
term for an epithelial-­lined cyst embedded in, or sur-
Histopathology
rounded by, lymphoid tissue. This description would
When explored surgically, it is possible to demonstrate the include branchial cleft cysts, intraoral lymphoepithelial
intact and undisturbed cyst. It has a smooth blue surface, is cysts, and lymphoepithelial cysts of the parotid gland. It
thin-­walled, and contains mucinous material. Allard et al. has also been used to include cystic lymphoepithelial
(1981b) stated that pseudocysts are almost always found lesions of the sort associated with Sjögren syndrome or
in an otherwise healthy-­looking sinus, whereas antral pol- with HIV. Lymphoepithelial cysts of the parotid gland are
yps will usually be seen in groups on inflamed oedema- probably developmental in origin, although some may be
tous mucosa. salivary duct cysts that arise as a result of cystic change in
Microscopically, a pseudocyst shows inflamed fibrous or intranodal salivary gland inclusions. Most intraoral lym-
granulation tissue containing pools of mucoid material. phoepithelial cysts arise in association with oral tonsils,
Sinus mucosa may be seen on one aspect and periosteum although an origin from the epithelium of minor salivary
on the other. Ducts or glands are not seen and the mucoid gland ducts, especially in the floor of the mouth where ton-
material appears to be primarily an inflammatory infil- sillar tissue is scarce, is a possibility. Intraoral lymphoepi-
trate, because mucin stains are usually negative (Meer and thelial cysts and lymphoepithelial cysts of the parotid gland
Altini 2006). are considered in this chapter. They are regarded as distinct
Retention cysts are usually small and are found within from branchial cleft cysts, which are true developmental
an inflamed sinus mucosa or an inflammatory polyp. They cysts and are discussed in Chapter 18.
256 Cysts of the Salivary and Minor Mucous Glands

­Intraoral Lymphoepithelial Cysts Site


The most common sites are the floor of the mouth and the
Clinical Features ventral surface of the tongue, with about 70% of all lesions
Reports of oral lymphoepithelial cysts have mostly been at these two sites (Table 15.4). In the largest series (Yang
single case reports, although there have been a number et al., 2012), 50% of lesions were located on the tongue and
of case series. Sykara et al. (2017) reported 25 cases and 38.3% in the floor of the mouth. Most tongue lesions were
reviewed the literature up to 2017. They found a total of found on the ventral surface (34.2% of the total), with 9.2%
45 papers that had documented 316 cases, including a at the base and 5.8% on the lateral border. The next most
series of 120 cases by Yang et al. (2012). Subsequently, da common site was the palate (5.0%), with rare lesions on the
Silva et  al. (2020) have reported a further 77 cases. buccal mucosa. In their review, da Silva et al. (2020) found
Table  15.4 summarises data from four of the larg- that 50.6% of cases were reported to be in the floor of the
est series. mouth, but we have noted that a number of papers do not
make a clear distinction between floor of the mouth and
ventral tongue. Cases have been reported on the anterior
Frequency pillar of the fauces (Buchner and Hansen 1980) and in the
Oral lymphoepithelial cysts are rare. In their review of oropharynx (da Silva et al. 2020).
more than 48 000 biopsies received from adults and chil-
dren over a 30-­year period, Jones and Franklin (2006a,b) Clinical Presentation
found 49 lymphoepithelial cysts representing only 0.10% of Lymphoepithelial cysts are usually symptomless and are
all biopsies. As a comparison, mucous extravasation and detected on routine examination, but a few patients com-
retention cysts represented 4.6% and 0.49%, respectively, of plain of swelling or discharge. Their size ranges from 1 to
all biopsies. 20 mm, but 80–90% of lesions are less than 10 mm in diam-
These proportions are similar to those reported in other eter (Yang et al. 2012; Sykara et al. 2017). The usual find-
series: 0.16% (Daley et al. 1994), 0.10% (Nonaka et al. 2011), ing is of a round or oval swelling of the oral mucosa, with
0.08% (Sykara et al. 2017), and 0.05% (da Silva et al. 2020). most lesions (50–60%) being yellow/white in colour (Yang
et al. 2012; da Silva et al. 2020). The swellings are submu-
cosal and freely mobile. Some were described as firm, but
Age most lesions are soft and painless. The clinical diagnosis is
Lymphoepithelial cysts occur across a wide age range from rarely correct, with most being diagnosed as mucoceles,
2 to 85 years with an average age of about 40 years and a ‘tumours’, lipomas, or ‘irritation fibromas’.
peak in the fourth to sixth decades (Table  15.4). At least
70% of lesions occur in adults over the age of 40 years (Yang Pathogenesis
et al. 2012; da Silva et al. 2020).
Most authors suggest that the intraoral lymphoepithelial
cyst arises from oral tonsillar tissue. Oral tonsils were first
Sex described in detail by Knapp (1970a,b), and are small accu-
Studies have shown variable frequencies in males and mulations of normal lymphoid tissue that resemble the
females, but suggest a slight predilection for females tonsils in Waldeyer’s ring. They are 1–3 mm in diameter
(Table  15.4). The largest review, of 316 cases, found that and are found in varying numbers on the soft palate, ven-
169 (53.5%) arose in females (Sykara et al. 2017). tral surface of the tongue, and floor of the mouth. Knapp

Table 15.4  Intraoral lymphoepithelial cysts: age, sex, and site distribution.

References n Male (%) Age, mean (range) FOM (%) Vent tongue (%)

Buchner and Hansen (1980) 38 60.5 39.8 (14–81) 50.0 18.4


Yang et al. (2012) 120 30.8 44.1 (2–75) 38.3 34.2
Sykara et al. (2017) 25 44.0 33.0 (15–51) 30.8 36.0
a
da Silva et al. (2020) 77 29.9 46.5 (13–85) 19.5 10.4

FOM, floor of mouth; n, number of patients.


a
 da Silva et al. included oropharynx (22% of lesions).
­Intraoral Lymphoepithelial Cyst 257

first suggested that lymphoepithelial cysts develop in oral evidence of mucus. On the other hand, a small number of
tonsils in which the crypt opening becomes plugged, caus- lymphoepithelial cysts are lined by duct-­like or pseu-
ing dilation of the crypt and a cystic structure. Buchner and dostratified epithelium, with mucus in the lumen and sali-
Hansen (1980) agreed that most lymphoepithelial cysts vary tissue in the wall (Buchner and Hansen 1980; Yang
probably arise from tonsillar crypts, but they noted that et al. 2012; Sykara et al. 2017; da Silva et al. 2020). In their
very few lesions showed any continuity between the lining report of 177 cases of oral mucous retention cysts, Stojanov
epithelium of the cyst and the surface epithelium. They et  al. (2017) described and illustrated a number of cases
postulated that some cysts may arise from epithelial inclu- that were surrounded by a dense lymphocytic infiltrate
sions or salivary gland tissue that may be located in close and resembled lymphoepithelial cysts.
proximity to tonsillar tissue.
The studies of Toto et  al. (1982) found that cyst-­lining
Histopathology
epithelium could be of tonsillar crypt or of salivary duct
origin. An origin from salivary duct epithelium, especially Lymphoepithelial cysts in the oral cavity show similar
in the floor of the mouth where tonsillar tissue is scarce, is ­histological features to the lymphoepithelial cysts of the
likely. Nair and Schroeder (1986, 1987) undertook a num- parotid gland and to branchial cleft cysts. They show an
ber of studies of the anatomy of the minor salivary glands epithelial-­lined cystic cavity surrounded by lymphoid tis-
in monkeys and described aggregates of normal lymphoid sue, usually with well-­formed and normal germinal cen-
tissue embracing the ducts of minor glands close to the tres (lymphoid follicles; Figure 15.15). Overall about 95%
orifice. Such tissue is termed duct-­associated lymphoid tis- are lined by stratified squamous epithelium that is usu-
sue (DALT) and was thought to be analogous to similar ally parakeratinised, and about half of these may show
tissues in the lungs or gastrointestinal tract. DALT may be hyperplasia (Buchner and Hansen  1980; Sykara
found associated with minor salivary gland ducts through- et  al.  2017; da Silva et  al. 2020). Occasional cases have
out the oral mucosa, and Nair and Schroeder (1986) con- been orthokeratinised (Buchner and Hansen,  1980).
tend that some of the oral tonsils illustrated by Knapp Pseudostratified ciliated (respiratory-­type) epithelium or
(1970a) are in fact ducts with accumulations of lym- simple cuboidal duct-­like epithelium has been reported in
phoid tissue. less than 5% of cases and mucous or goblet cells are occa-
These studies suggest that intraoral lymphoepithelial sionally seen. In almost all cases the lumen contains des-
cysts may arise through a number of mechanisms. Some quamated parakeratotic cells and debris (Figure 15.15),
may arise as a result of cystic dilatation of tonsillar crypts, and some contain inflammatory cell or amorphous eosin-
while others may be retention cysts arising from the super- ophilic material resembling mucus.
ficial ducts of minor salivary glands. Histological exami- In some cases, the lumen of the cyst communicates with
nation of lesions suggests that the majority probably arise the oral cavity through an epithelial-­lined tract. In their
from tonsillar tissue, since most lesions are lined by par- series of 77 cases, da Silva et  al. (2020) found 23 (29.9%)
akeratinised stratified squamous epithelium and the that communicated with the oral epithelium and 3 (3.9%)
lumen is filled with desquamated epithelial cells without that communicated with an adjacent salivary duct.

Figure 15.15  Lymphoepithelial cyst of the floor


of the mouth. The cyst lies just below the oral
epithelium and is surrounded by lymphoid tissue
with normal germinal centres (arrows).
258 Cysts of the Salivary and Minor Mucous Glands

The cyst is closely enveloped by lymphoid tissue, which


in most cases surrounds the entire cyst, but in a few it is
present in only part of the wall. In most cysts the lymphoid
tissue shows normal germinal centres (Figure 15.15), but
in up to half of cases there may be only a diffuse infiltrate
of lymphocytes.

Treatment
The treatment of choice for oral lymphoepithelial cysts is
complete surgical excision. The precise technique and sur-
gical approach depend on the location of the cyst and the
proximity of adjacent vital structures.

­ ymphoepithelial Cysts of the


L Figure 15.16  A parotid lymphoepithelial cyst is lined by thin
epithelium overlying dense lymphoid tissue.
Parotid Gland

Lymphoepithelial cysts of the parotid gland are essentially Type I cysts (14.1%; 9 cases) were simple cysts formed
salivary duct cysts associated with a dense lymphoid stroma. from dilated ducts surrounded by diffuse infiltrates of
Some regard parotid lymphoepithelial cysts to be branchial inflammatory cells, but without germinal centres. The epi-
cleft cysts that have arisen from branchial cleft remnants thelial lining was most often simple columnar and duct-­
sequestered in the parotid gland. Although this may be pos- like. Epimyoepithelial islands were seen in 8 of the 9 cases,
sible, we would regard the two as distinct and separate enti- and 7 cysts in this category were multilocular.
ties. Branchial cleft cysts are discussed in Chapter  18. Type II cysts (42.2%; 27 cases) were partially encapsu-
Parotid cysts with associated dense lymphoid tissue are also lated and separated from the surrounding parotid tissue.
seen in association with HIV disease and may be a feature These cysts were lined by squamous epithelium and half
of benign lymphoepithelial lesions (lymphoepithelial showed lymphoid follicles in the walls. Epimyoepithelial
sialadenitis) associated with Sjögren syndrome. We would islands were seen in only 5 cases.
also regard these as separate and distinctive lesions and Type III (43.8%; 28 cases) cysts were well encapsulated,
would use the terms HIV-­associated salivary gland disease lined by squamous epithelium, and showed prominent
and cystic lymphoepithelial lesion, respectively. ­germinal centres. Epimyoepithelial islands were seen in
Parotid gland lymphoepithelial cysts are uncommon and 5  cases. These cysts had a fibrous capsule resembling a
there are very few reported cases. Camilleri and Lloyd lymph node, but although lymphatic vessels were seen, no
(1990) found about 70 cases in the literature up to 1990, but subcapsular sinus structures could be identified.
since then there have been very few cases reported that Weidner et  al. (1986) described 5 cases and had found
have not been associated with HIV. The largest case series, similar features, although in 3 cases he found stratified or a
which forms the basis for this discussion, comprises 64 single layer of cuboidal cells with occasional mucous cells.
cases reported in 2009 (Wu et al. 2009). With regard to pathogenesis, Bernier and Bhaskar (1958)
The lesions are found over a wide age range (3–76 years), were the first to introduce the term lymphoepithelial cyst,
but the majority are encountered in adults, with a mean and made a clear distinction between these and branchial
age of 52 years. There is a slight predilection for females cleft cysts. They felt that parotid lesions were not derived
(56.3%; 36 of 64 cases). The cysts are unilateral and present from embryonic epithelial remnants, but probably arose
as slow-­growing, soft painless swellings with an average from glandular inclusions in intraparotid lymph nodes.
size of 30 mm (range 5–65 mm). An origin in lymph nodes is suggested by the finding that
Histologically, the cysts show similar features to oral many lesions are encapsulated (e.g. Types II and III) and
lymphoepithelial cysts and are composed of an epithelial-­ contain well-­formed germinal centres typical of normal
lined cystic space surrounded by, or embedded in, dense lymphoid tissue. On the other hand, germinal centres
lymphoid tissue (Figures 15.15 and 15.16). Most lesions are are not seen in all cases, and some show diffuse lympho-
unilocular, but about 25% were found to be multilocular. cytic infiltrates and epimyoepithelial islands similar to
Wu et al. (2009) described three distinctive histomorpho- the  ­features of benign lymphoepithelial lesions or lym-
logical patterns that they called Types I, II, and III. phoepithelial sialadenitis. Wu et al. (2009) interpreted their
­Polycystic (Dysgenetic) Disease of the Parotid Gland 259

findings to propose that the lesions start as ductal dila-


tion in association with sialadenitis (Type I histology).
Subsequently a cyst develops and is accompanied by reac-
tive lymphoid hyperplasia and gradual encapsulation as
peripheral granulation tissue matures (progression to Type
II and III histology). It is not possible to definitively state
which mechanism is correct and it is probable that a num-
ber of different pathogenic processes may be responsible
for a common end result – an epithelial-­lined cyst embed-
ded in lymphoid tissue.

HIV-­Associated Salivary Gland Disease


Disorders of the salivary glands are a common manifesta-
tion of HIV infection and may be increasing as HIV infec-
tion persists following the advent of effective therapies
such as highly active antiretroviral therapy (HAART; Figure 15.17  A multinucleated giant cell (arrow) is seen within
the dense lymphoid infiltrate of HIV-­associated cystic lymphoid
reviewed by Meer 2019). A particular disorder that affects
hyperplasia.
the parotid glands and is associated with cystic change has
been called HIV-­associated lymphoepithelial cysts, but is
now usually referred to simply as HIV-­associated salivary
gland disease or more precisely as cystic lymphoid hyperpla- ­ olycystic (Dysgenetic) Disease
P
sia. This was first fully characterised in the early 1990s and of the Parotid Glands
is the subject of a large literature. The lesions are character-
ised by lymphoepithelial cysts and a brief description is This is a rare developmental disorder that arises in the
relevant to allow distinction from non-­HIV-­related cysts of parotid glands and is not related to similar polycystic disor-
the parotid gland. ders affecting other organs. Seifert et al. (1981) and Batsakis
Cystic lymphoid hyperplasia affects up to about 20% of et al. (1988) first drew attention to this condition, but up to
patients with HIV, occurs primarily in adult males, and 2017 fewer than 20 cases had been reported (Srikant
may be seen before the onset of AIDS. It almost always et  al.  2017). Although the lesion is associated with the
affects the parotid glands and is bilateral in over 60% of parotid glands, 3 cases have been reported in the subman-
cases. It presents as diffuse enlargement of the glands and dibular and minor glands (Koudounarakis et  al.  2016;
may be accompanied by Sjögren-­like symptoms, including Srikant et al. 2017).
dry mouth and eyes, and arthralgia. Clinically, the disorder occurs almost always in females,
Histologically, the glands show changes similar to the with only 6 reported cases in males, and with a wide age
lymphoepithelial lesions of Sjögren syndrome. However, range at presentation of between 6 and 85 years. It presents
lesions in HIV patients are associated with diffuse lym- as bilateral parotid involvement, with a history of fluctuat-
phoid infiltrates with prominent follicular hyperplasia and ing, non-­tender parotid gland swelling, often with a long
lysis of germinal centres. Another distinguishing feature is history before diagnosis. A number of cases have been
the presence of multinucleated giant cells (Figure 15.17), reported with a familial background in which parents and
which have been shown to contain p24 (HIV-­1) protein in their offspring have been affected (Seifert et al. 1981; Smyth
immunocytochemical studies (Vicandi et  al.  1999). The et al. 1993; Srikant et al. 2017).
most characteristic feature is the finding of large and mul- Histology shows that the normal acinar parenchyma of
tiple cystic spaces lined by duct-­like epithelium. the glands is almost completely replaced by a honeycombed,
Although these lesions are superficially similar to parotid net-­like pattern of thin-­walled interlinked cysts that appear
lymphoepithelial cysts, they can be differentiated on the to arise from the intercalated ducts (Figure 15.18). The lob-
basis of the distinctive clinical presentation and by the ular architecture of the gland is maintained, however.
­histology that shows that the cysts are almost always mul- Small clusters of serous acini and occasional ducts may be
tiple and often bilateral, and are embedded in diffuse lym- retained in the intercystic fibrous septa. In places, the lin-
phoid infiltrates with prominent follicles. In contrast, ing cells may be cuboidal and show apocrine change. The
lymphoepithelial cysts tend to be single, unilocular, and cyst lumina contain amorphous eosinophilic mucus-­like
surrounded by a zone of lymphoid tissue, often with few material and may occasionally contain mucus plugs or
or no follicles. sialoliths The lesions are not inflamed.
260 Cysts of the Salivary and Minor Mucous Glands

Figure 15.19  Sclerosing polycystic adenoma. Cystically dilated


Figure 15.18  Polycystic disease of the parotid gland. The gland
ducts, with a cribriform pattern, are embedded in a fibrosed and
is largely replaced by a network of thin-­walled cysts of variable
sclerosing collagenous stroma. Source: Courtesy of Prof.
size and shape. Source: Courtesy of Dr K. Donath.
John Eveson.

Familiarity with this disease should prevent a histologi- carcinoma in situ in the ductal epithelium (Gnepp  2003;
cal misdiagnosis of a cystic neoplasm. No surgery is nec- Gnepp et al. 2006).
essary except for diagnosis and for cosmetic reasons if Immunohistochemical and molecular studies have
required. shown good evidence that the lesion is a true neoplasm.
Skálová et  al. (2006) examined polymorphisms of the
human androgen receptor (HUMARA) and showed that
­Sclerosing Polycystic Adenoma lesions were monoclonal, which was the first evidence of a
neoplastic nature. More recently, studies have shown con-
This lesion was first reported as a reactive fibrosing pseu- sistent mutations in PTEN and alterations in the PI3 kinase
doneoplastic lesion in the major salivary glands and was pathway, as well as loss of PTEN protein expression in
called sclerosing polycystic adenosis (Smith et al. 1996). It is ductal and acinar cells (Bishop et  al.  2020; Skálová
now known that the lesion is a neoplasm and it has been et  al.  2021). This profile is similar to salivary duct carci-
classified as sclerosing polycystic adenoma in the latest noma and confirms that sclerosing polycystic adenoma is a
World Health Organization (WHO) classification of head neoplasm (Bishop et al. 2022; Bishop and Thompson 2021).
and neck tumours (Bishop et  al.  2022) The lesion is not The histological features are characteristic and should
primarily a cystic lesion, but it is multicystic and has been not cause confusion with polycystic disease or with other
included in this chapter for completion and to ensure that salivary cysts. The main area of diagnostic difficulty is dif-
it is properly distinguished from polycystic disease of the ferentiation from other benign or malignant salivary gland
parotid glands. tumours (Bishop and Thompson 2021).
Sclerosing polycystic adenoma is a rare neoplasm, with
fewer than 100 cases reported. Almost all cases have arisen
in the parotid gland, but occasional lesions have been ­Keratocystoma
found in the submandibular gland and about 15 have been
reported in the minor glands (Das et al. 2020). Lesions have Another multicystic lesion that may be encountered in the
a slight predilection for females and are found over a wide parotid gland is the keratocystoma. The true nature of
age range (7–84 years), but with a peak in the fifth decade the  lesion is unknown and the literature has variously
(Bishop et  al.  2020; Bishop et  al.  2022). Clinically, the described it as a benign cyst, a choristoma, or more fre-
lesions present as a solitary swelling. quently as a benign neoplasm. It was first reported in the
Histologically the tumour shows an unencapsulated parotid gland of an 8-­year-­old girl and was thought to be a
mass of sclerotic and fibrosed collagenous connective choristoma with adnexal features resembling a trichoade-
­tissue containing accumulations of ductal and acinar epi- noma (Seifert et  al.  1999). Subsequently, only 10 further
thelial elements, with cystic spaces (Figure  15.19) form- cases have been reported, arising in adults from 18 to
ing  a cribriform pattern. Areas of apocrine or squamous 51 years (Nagao et  al.  2002; Zhang et  al.  2010; Huang
metaplasia and mucous cell may be seen. About half of et  al.  2012; Hirata et  al.  2014; Wang et  al.  2015; Ahuja
cases have shown features of epithelial dysplasia or even et al. 2016; Aresta et al. 2019; Komatsu et al. 2020). Most
­Keratocystom 261

authors consider the lesion to be a benign neoplasm, with and from polycystic disease or sclerosing polycystic ade-
evidence that it arises from intercalated ducts that have noma, and the multicystic nature distinguishes it from rare
undergone squamous metaplasia (Nagao et al. 2002; Wang epidermoid cysts that might arise at this site. The main
et al. 2015). area of diagnostic difficulty is differentiation from kerati-
Keratocystoma arises as a tumour-­like mass in the nising squamous cell carcinoma and from salivary gland
parotid gland and is composed of multiple cystic spaces tumours that may have a similar cystic morphology. This
lined by stratified squamous epithelium showing ortho-­ might include mucoepidermoid carcinoma and pleomor-
and parakeratinisation, but without a prominent granular phic adenoma, which may on occasion show prominent
cell layer. The cystic spaces may be filled with lamellated squamous metaplasia with prominent cyst formation
keratin to form solid masses. Solid islands of squamous (Goulart et al. 2011).
epithelium are also seen. The cysts are often irregular and Keratocystoma also shows remarkably similar histologi-
multilocular. cal features to the solid or multicystic odontogenic kerato-
Many more cases need to be reported before this lesion cyst (see Chapter 7). However, it has only been reported in
is  fully understood. It appears to be completely benign, the parotid gland and cannot be confused with intraosse-
with no evidence of recurrence after adequate surgical ous keratocysts, but may cause diagnostic difficulties when
removal (Wang et  al.  2015; Aresta et  al.  2019; Komatsu faced with a multicystic soft-­tissue lesion lined by kerati-
et al. 2020). The lesion is unlikely to be confused with other nised stratified squamous epithelium. Such lesions located
cysts that may arise in the salivary glands. The prominent in the parotid gland should be diagnosed as keratocystoma,
keratinisation distinguishes it from lymphoepithelial cysts not as soft-­tissue keratocyst.
262

16

Surgical Ciliated Cyst

CHAPTER MENU
­Clinical Features, 263
•• Frequency, 263
•• Age, 264
•• Sex, 264
•• Site, 264
•• Clinical Presentation,  264
­Radiological Features, 265
•• Radiological Differential Diagnosis,  266
­Pathogenesis, 267
­Histopathology, 268
­Treatment, 268

The surgical ciliated cyst is an implantation cyst that arises A similar ciliated cyst has also been reported in the man-
as a result of entrapment of fragments of respiratory epi- dible (reviewed by Theofilou et al. 2021), but all cases have
thelium into a wound following maxillary sinus surgery or resulted from implantation of sinus epithelium into a man-
facial trauma (Nelson and Speight 2022). It can be regarded dibular wound during simultaneous surgery on the maxilla.
as a type of mucous retention cyst of iatrogenic origin. With regard to terminology, the cyst has been called
It  was first reported in Japan in 1927 (Kubo  1927) as ­surgical ciliated cyst of the maxilla, but in Japan the favoured
a  ­maxillary cyst occurring after radical sinus surgery. name is postoperative maxillary cyst. However, these terms do
Subsequently, Gregory and Shafer (1958) drew attention to not account for lesions that arise in the mandible. The lesions
the development of these cysts in the maxilla of patients have also been included as a type of antral mucocele or have
whose sinuses had been opened surgically and first pro- been called respiratory implantation cyst or surgical implan-
posed the term ‘surgical ciliated cyst of the maxilla’. tation cyst, but these names have not been widely adopted.
This was followed by a small number of reports in the The name surgical ciliated cyst remains the most widely
German literature, but it was not until the 1980s that many used, and we will use this term here, specifying as appro-
reports began to record a high incidence in Japan. These priate mandible or maxilla as the site of occurrence.
cases followed treatment of chronic sinusitis by a maxillary Because the surgical ciliated cyst is a simple implanta-
antrostomy procedure that entered the sinus through tion cyst, similar to lesions that may arise randomly at
the  antero-­lateral wall (Caldwell-­Luc surgery; Kaneshiro other anatomical sites, it has usually been regarded as an
et al. 1981; Maeda et al. 1987; Yamamoto and Takagi 1986; oddity and has rarely been included in studies of jaw cysts
Nishioka et al. 2005). or in published classifications. However, it has a distinctive
The Caldwell-­Luc procedure has become less common aetiology and clinicopathological features and has been
as sinusitis is now treated more conservatively. considered as a distinct entity in Japan for many years. For
Increasingly, therefore, cases are reported following max- the first time, it has now been included as an entity in the
illary osteotomy procedures, although they may occasion- latest (fifth) edition of the World Health Organization (WHO)
ally arise after midfacial (le Fort) fractures or traumatic classification of head and neck tumours (WHO  2022a;
tooth extractions. Nelson and Speight 2022).

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
­Clinical Features 263

­Clinical Features biopsies (0.03%), while others have recorded no cases at all.
In the series recorded in Table 1.3, only two papers reported
Most descriptions of the surgical ciliated cyst are case reports surgical ciliated cysts, with frequencies of 0.08% (Daley
recording a single or a few cases. Niederquell et al. (2016) et  al.  1994; Canada) and 0.5% (Ali  2011; Kuwait) of all
reviewed the literature and found 23 articles recording jaw cysts.
details of 284 patients between 1978 and 2014. In addition, Nishioka et  al. (2005) reviewed the worldwide preva-
however, there are many papers, especially from Japan, lence over a 20-­year period and found only 56 cases from
recording a high prevalence of this cyst and experience with outside of Japan compared to 1141 from just their own
many hundreds of cases (Kaneshiro et  al.  1981; Maeda department. They also asked colleagues from 31 university
et  al.  1987; Yamamoto and Takagi  1986; Nakamura departments in 16 countries to report on the number of
et al. 1995; Maruyama et al. 2002). One paper from Japan cysts encountered between 1976 and 1996. The largest
recorded 1141 cases from one department alone (Nishioka number (80 cases) were reported in South Korea, followed
et al. 2005). There is only one case series from Europe or the by one department in Germany with 42 cases, 23 cases in
USA, and this reports only 21 patients (Basu et  al.  1988; Birmingham (England), and 16  in Sao Paulo (Brazil). No
England). Table  16.1 summarises data from representative cases at all were reported in 20 departments, and the
selected series and reviews where details have been provided. remainder reported only 2 or 3.
Outside Japan, the only case series suggesting the lesion
may not be rare is from Birmingham (England; Basu et al.
Frequency
1988), where 21 patients were diagnosed over a period of
Globally the surgical ciliated cyst appears to be rare, but three years (Table 16.1).
a  very high prevalence has been recorded in Japan. The variability in frequency is intriguing, but is clearly
Yamamoto and Takagi (1986) found that surgical ciliated associated with the number of surgical procedures carried
cysts constituted 19.5% of all oral cysts in their department. out in different settings. The high number of cases in Japan
Nakamura et al. (1995) reviewed 1234 jaw cysts and found were mostly reported between about 1970 and 1995, and is
that 21.6% (n  =  267) were surgical ciliated cysts. More thought to be due to a high prevalence of chronic sinusitis
recently, Nishioka et al. (2005) reported 1141 surgical cili- among young people and the very common use of the
ated cysts, representing 16.1% of all jaw cysts. Caldwell-­Luc procedure in the immediate postwar period.
Outside of Japan, the cyst shows a much lower frequency. Outside of Japan radical sinus surgery has been less com-
In Shear’s department at the University of the Witwatersrand, mon, and more recent cases have been associated with
only 5 cases were diagnosed over a 32-­year period (see osteotomies. The Caldwell-­Luc procedure is used less fre-
Table  1.1), representing 0.1% of 3498  jaw cysts, and in quently now and a reduction in cases from Japan is to be
Sheffield we have seen only 7 cases since 1996. Nonaka expected. The different causes of the cyst are discussed
et  al. (2011) diagnosed only 3 cases among over 10 300 later in this chapter (see ‘Pathogenesis’).

Table 16.1  Age, sex, and causation from selected case series or reviews of surgical ciliated cysts.

References n Age mean (range) Male (%) Type of surgery Delaya mean (range)

Case series
Yamamoto and Takagi (1986) 60 39.1 (21–72) 50.0 Sinusitisb 18.3 (4–49)
Basu et al. (1988) 21 43.7 (25–74) 42.9 Sinusitis 20.0 (7–39)
Pe et al. (1990) 22 49.0 (33–68) 72.7 Sinusitis 26.5 (10–48)
Nishioka et al. (2005) 1141 48.5 (21–80) 58.7 Sinusitis 31.0 (3–61)
Reviews
Niederquell et al. (2016) 284 47.0 (19–85) 57.0 Mixedc 22.0 (0.5–60)
Kahn et al. (2021) 9 59.4 (41–76) 44.4 Sinus lift (implants) 3.9 (0.5–10)
Theofilou et al. (2021) 15 32.4 (15–53) 53.3 Osteotomy 9.1 (3–21)

n, number of patients.
a
 Delay is years between surgery and diagnosis.
b
 Surgery for sinusitis was almost always a Caldwell-­Luc procedure.
c
 Six cases were after osteotomy. The remainder followed sinus surgery.
264 Surgical Ciliated Cyst

Age Sex
The age of presentation is, to some extent, determined The sex distribution varies between studies, with no clear
by the type of surgery that precedes the lesion. The evidence of a predilection for males or females (Table 16.1).
majority of reported cases follow a Caldwell-­Luc proce-
dure and diagnosis is made about 20–30 years after the Site
surgery (Table 16.1). The surgical ciliated cyst is there-
The surgical ciliated cyst has always been reported as an
fore seen in adults, with very few cases reported below
implantation cyst that arises in the maxilla, following sinus
the age of 20 years. The average age of presentation is
surgery. In 1994, however, the first case of a ciliated cyst
about 45–50 years (Table  16.1). Figure  16.1 shows the
was reported in the mandible following implantation of
age distribution of 1141 patients in the study of Nishioka
sinus epithelium from a nasal osteocartilagenous graft
et al. (2005). The majority of their patients were in the
used for chin augmentation (Nastri and Hookey  1994).
fourth to sixth decades, with a clear peak (34.7% of
Subsequently 14 cases have been reported in the mandible
cases) in the fifth decade and an age range of 21–80 years.
as a result of chin augmentation surgery or bimaxillary
The vast majority of the cases in this study (98.7%) arose
osteotomies (reviewed by Theofilou et al. 2021).
following surgery for sinusitis and there were no
Maxillary lesions are located in the posterior, molar/pre-
osteotomies.
molar region of the maxilla. The cysts are always found
More recent studies suggest that cysts following osteoto-
towards the floor of the sinus. Occasional bilateral cysts
mies occur in a younger age group. Theofilou et al. (2021)
have been reported in patients who have had sinus surgery
reviewed the literature and found 15 cases that followed
on both sides.
maxillary osteotomies, with an average age of 32.4 years
The site of mandibular cases depends on the type of sur-
(range 15–53) and a delay between surgery and diagnosis
gery. Those associated with chin augmentation are found in
of between 3 and 21 years.
the anterior mandible, while those that follow a mandibular
Conversely, cases that have followed sinus lift surgery
osteotomy may be located in the premolar/molar region.
(sinus floor augmentation) appear to arise in older age
groups, reflecting the fact that the purpose of this surgery
Clinical Presentation
is to facilitate the insertion of implants. In a systematic
review, Kahn et al. (2021) found 9 cases, with an average Surgical ciliated cysts are postoperative implantation cysts
age at presentation of 59.4 years and a delay following sur- and patients must always have a previous history of sur-
gery of only 3.9 years (Table 16.1) gery involving the maxillary sinus. The period between

400 Figure 16.1  Age distribution of 1141


patients with surgical ciliated cyst.
Males Source: Data from Nishioka et al. (2005).
350
Females

300
222

250
No of cases

180
200
139

150

100
174 80
125
50 102
32
43 14
16 11 3
0
0-9 10-19 20-29 30-39 40-49 50-59 60-69 70-79 80-89 90+
Age
­Radiological Features 265

surgery and onset of symptoms varies from 6 months to ­Radiological Features


60 years, with an average of about 20 years (Table 16.1). The
majority of patients (about 90%) present between 10 and Radiographs show a well-­defined radiolucent lesion in the
30 years after surgery (Kaneshiro et  al.  1981; Yamamoto alveolar bone, closely related to the floor of the maxillary
and Takagi  1986). Cysts associated with osteotomies may sinus. The cyst is usually rounded and unilocular and may
present at a younger age and with a shorter delay since sur- be at least partially corticated (Figures 16.2–16.4).
gery (Theofilou et al. 2021; Table 16.1). In the series of 60 cases reported by Yamamoto and
Occasionally a cyst may be silent and symptomless and Takagi (1986), 56 (93.3%) were unilocular lesions, with
found incidentally on radiographs taken for other reasons. only 4 reported as multilocular. In 42 of their 60 cases buc-
In most cases, however, patients complain of swelling with cal cortical bone was present and 58% (35 cases) had a cor-
pain or tenderness. ticated margin in at least part of the cyst. Most cases (43:
Basu et al. (1988) found that 20 of their 21 patients pre- 71.6%) were located below the antral floor, although 24
sented with pain and intraoral swelling and 13 (61.9%) also (40%) raised the floor and occupied half of the sinus, and
showed extraoral swelling. In their systematic review, 17 cases (28.3%) filled the entire sinus. Kaneshiro et  al.
Niederquell et al. (2016) found that clinical symptoms were (1981) found that 68% of their 71 cases had evidence of cor-
recorded for 219 patients. Of these, 142 (64.8%) complained tication. Most (61.7%) were between 21 and 30 mm in
of pain, 54 (24.6%) of pain and swelling, and 14 (6.5%) of diameter, with only 7 (9.9%) cases greater then 31 mm.
swelling alone. Thus almost all patients (95.6%) presented The cystic area appears to encroach on the sinus itself,
with a combination of pain and/or swelling. Nishioka et al. but lack of communication between the two has been dem-
(2005) found that only 2 of their 1141 patients were onstrated by injecting the sinus with a radiopaque material
symptomless. (Kaneshiro et al. 1981; Basu et al. 1988).
Cases reported in the mandible have usually presented As the cysts enlarge, ballooning expansion of the buccal
as a swelling with pain or tenderness. Theofilou et  al. and palatal cortical plates may become apparent and the
(2021) found that only 2 of the 14 reported cases were sinus wall may become thinned and eventually perforated
­incidental findings. Moreover, 11 cases presented with (Basu et al. 1988; Leung et al. 2012). Gradually, a cyst may
swelling, of which 4 also showed pain or tenderness. One expand beyond the original boundaries of the sinus
case presented with mental nerve paraesthesia. One man- (Figure  16.3). Expansion is best seen on examination by
dibular case was found to be extraosseous and presented computed tomography (CT) or magnetic resonance imag-
as a fluctuant subcutaneous swelling on the chin (Kelly ing (MRI). The cysts show a ballooning pattern of expan-
et al. 2000). sion with corticated margins, and T2-­weighted MRI shows
hypointense contents suggesting low water and high

Box 16.1  Surgical Ciliated Cyst: Key Features


●● It is an implantation cyst caused by entrapment of
sinus epithelium into a wound
●● Patients always have previous history of surgery or
trauma involving the maxillary sinus
●● Arise in the alveolar bone of the posterior maxilla
●● Rare cases in the mandible follow implantation of
sinus epithelium during bimaxillary surgery
●● Globally, they are rare – less than 0.5% of jaw cysts
●● Common in Japan, representing up to 20% of jaw cysts
●● Mean age is about 45 years, with a peak in the
fifth decade
●● No sex predilection
●● Most patients complain of swelling and pain or
tenderness
●● Radiology shows a well-­demarcated unilocular Figure 16.2  Surgical ciliated cyst. Radiograph shows a
lesion that is at least partially corticated well-­defined radiolucency at the site of a previously extracted
●● Histology shows a cyst lined by pseudostratified cili- tooth. The cyst is clearly corticated along the inferior and
ated columnar (respiratory) epithelium anterior margin. Note that the lamina dura and periodontal
space of the adjacent teeth are intact (arrows).
266 Surgical Ciliated Cyst

Figure 16.3  Surgical ciliated cyst


located in the posterior maxilla
associated with a Le Fort I osteotomy
(metal plates can be seen towards the
top right of the figure). The cyst is well
demarcated and corticated (arrows).

protein content, consistent with mucus (Leung et al. 2012; of the surgical ciliated cyst to a mucosal cyst of the sinus.
Theofilou et al. 2021). In both cases a tooth has been extracted and if a respiratory
Pe et  al. (1990) evaluated 34 cysts using CT and found epithelial lining is present the lesions may be indistin-
that they were round and expansile and many showed evi- guishable. A history of sinus surgery or a difficult trau-
dence of fluid contents. All but 1 of their 34 cases showed matic extraction may suggest a diagnosis of surgical ciliated
some evidence of perforation of the sinus wall: 28 of the cyst. Note, however, that retention cysts arise in the floor of
antero-­lateral wall, 27 of the medial or nasal wall, and 14 of the sinus and are not intraosseous, whereas the surgical
the posterior wall. Furthermore, 13 cases (38.2%) filled the ciliated cyst is within the alveolar bone and has a well-­
sinus and 21 (61.8%) partially filled the sinus. The largest defined bone margin that is at least partially corticated
case in their series also perforated the orbital floor. (Figure 16.2). Mucosal retention cysts can never be corti-
Surgical ciliated cysts in the mandible usually present as cated on the superior surface. On occasion it may not in
a well-­defined unilocular radiolucency with a corticated fact be possible to distinguish between a surgical ciliated
margin (Nastri and Hookey  1994; Bourgeois and Nelson cyst and a retention cyst of the maxillary sinus.
2005; Lazar et al. 2006; Li et al. 2014; Cai et al. 2015). Most Surgical ciliated cysts may also fill the sinus and may
lesions are solitary, but multiple cysts have been described, cause bony expansion or perforation of the sinus walls
adjacent to inserted plates and to the wound margins (Basu et al. 1988; Pe et al. 1990). Such features are an omi-
(Bourgeois and Nelson 2005; Cai et al. 2015). Mandibular nous sign and may be indistinguishable from a sinus
lesions have also been described that are multilocular mucocele, but must also be differentiated from destructive
(Koutlas et al. 2002) or have an irregular scalloped outline neoplasms. The surgical ciliated cyst is ballooning and
(Ragsdale et al. 2009). One mandibular case was found to expansile, and even when filling the sinus, a corticated mar-
be extraosseous and presented entirely in the soft tissue of gin may be visible and the cyst can be shown to be separate
the chin (Kelly et al. 2000). from the sinus by CT or using contrast medium. Many sur-
gical ciliated cysts also show a fluid level. A final diagnosis,
however, may depend on histological examination.
Radiological Differential Diagnosis
A further consideration is differentiation from radicular or
Although the radiological features are characteristic, a residual cysts. The surgical ciliated cyst arises in the alveolar
diagnosis cannot be made on radiology alone. The key bone of the maxilla and may be in close proximity to the
diagnostic criteria are a history of previous surgical inter- roots of the maxillary teeth. On radiology the lamina dura
vention, and an epithelial lining of respiratory-­type epithe- and periodontal space of the associated teeth will be intact
lium (Box  16.1). On radiology alone the surgical ciliated (Figure 16.2), and this excludes a diagnosis of radicular cyst.
cyst may be indistinguishable from other cysts that arise in A radicular cyst can also be excluded if the teeth are vital.
the maxillary antrum (discussed in Chapter  15). A com- Figure 16.4 shows a well-­demarcated corticated cyst in an
parison of Figures 16.2 and 15.14 illustrates the similarity edentulous region of the maxilla. This radiological
­Pathogenesis 267

Figure 16.4  A surgical ciliated cyst located in the


edentulous maxilla. Such a lesion is
indistinguishable from a residual cyst and
diagnosis must depend on the histological
findings.

appearance is almost diagnostic of a residual cyst, but in this In 1990, Sugar et al. (1990) reported the first cases of sur-
case the cyst was lined entirely by respiratory epithelium gical ciliated cyst arising in the maxilla at the site of maxil-
and a diagnosis of surgical ciliated cyst was made, with an lary (Le Fort) osteotomies. Subsequently there have been
assumption that it had followed a previous difficult 10 reports of postosteotomy cysts, with a total of 15 cases
extraction. (reviewed by Theofilou et al. 2021). All have followed max-
Another maxillary cyst that can cause diagnostic confu- illary osteotomies, mostly advancement procedures of the
sion is the nasopalatine duct cyst, since this may also be Le Fort I type.
lined by respiratory epithelium (see Chapter 13). However, Nishioka et  al. (2005) reported 1141 of their own cases
nasopalatine duct cysts are found in the midline of the ante- from Japan and reviewed 54 cases from other parts of the
rior maxilla and surgical ciliated cysts are almost always world. Of their own cases, 98.7% were caused by surgery for
located in the posterior maxilla. A diagnosis of surgical cili- chronic sinusitis, while the rest were associated with removal
ated cyst in the midline anterior maxilla could only be made of polyps or dental extraction, or 1 case that followed a facial
if there was clear evidence of previous surgery at this site. fracture. Of the cases from other countries, only 67.3% were
caused by sinus surgery, 9.1% by trauma or fracture, and
5.5% followed osteotomies. In their systematic review of 23
published papers, Niederquell et al. (2016) found 210 cases
­Pathogenesis where the cause was documented: 190 (90.1%) had had
Caldwell-­Luc surgery, 14 (6.7%) had had other types of sinus
The surgical ciliated cyst is caused by implantation of sinus surgery, and 6 cases followed a Le Fort osteotomy.
mucosal epithelium into a wound following surgery involv- More recently, cases have been reported after sinus floor
ing the maxillary sinus. The vast majority of cases have augmentation surgery for insertion of implants. Kahn et al.
been reported in Japan and have followed surgery for (2021) reported 4 cases and in a systematic review found a
chronic sinusitis using the Caldwell-­Luc procedure, which further 5 cases. The cysts arose from 6 months to 10 years
involves perforation of the antero-­lateral wall of the sinus after surgery (Table  16.1) and were located in the aug-
by an intraoral approach. The high frequency of cysts in mented bone adjacent to implants.
Japan has been attributed to the large number of cases of Cases reported in the mandible have all been associated
maxillary sinusitis that occurred in Japan during and just with implantation of sinus epithelium during surgery that
after the Second World War, which were treated by the has involved simultaneous interventions in the maxilla and
Caldwell-­Luc procedure because antibiotics were not avail- mandible. There have been 12 reports of mandibular cili-
able. Most large series in Japan were reported between ated cysts, with a total of 14 cases (Nastri and Hookey 1994;
about 1970 and 1990 and there have been few since, sug- Anastassov and Lee  1999; Kelly et  al.  2000; Imholte and
gesting that the frequency of this postoperative cyst is now Schwartz 2001; Koutlas et al. 2002; Bourgeois and Nelson
declining. This is probably caused in part by conservative 2005; Lazar et al. 2006; Ragsdale et al. 2009; Li et al. 2014;
management of sinusitis with antibiotics, and to the Cai et al. 2015; Seifi et al. 2016; Syyed et al. 2019; reviewed
increasing use of endoscopy to treat sinus disease as an by Theofilou et al. 2021). Of the 14 cases, 8 were caused by
alternative to the Caldwell-­Luc procedure. implantation of sinus or nasal epithelium into the anterior
268 Surgical Ciliated Cyst

mandible during chin augmentation using an autologous ­Histopathology


graft from the maxilla or an osteocartilaginous graft from
the anterior nose. The remaining 6 cases followed bimaxil- The cysts may be removed intact and will be seen as a sac-­
lary osteotomies and were probably caused by transfer of like cystic lesion with a thick fibrous wall. On dissection
epithelial fragments on contaminated instruments. the lumen contains a brown, watery or mucoid liquid.
The pathogenesis of the cyst must be due to proliferation Many lesions are removed piecemeal or will be ruptured,
of the implanted epithelium with subsequent cyst forma- and the pathologist will receive fragments of cyst wall. The
tion. This may follow a similar process to that seen in radic- luminal surface is covered with epithelium. Thickenings or
ular cysts, where chronic inflammation causes epithelial papillary process are not seen.
proliferation followed by formation of a cyst cavity and Histologically, the surgical ciliated cyst is lined in whole
growth by hydrostatic pressure. However, this does not or in part by typical respiratory-­type, pseudostratified cili-
explain why implanted cysts of the jaws only follow sinus ated columnar epithelium (Figure  16.5; Yamamoto and
surgery and are always lined by respiratory epithelium. Takagi 1986; Maeda et al. 1987; Basu et al. 1988; Maruyama
During all the surgical procedures in and around the jaws, et al. 2002; Nishioka et al. 2005). About 30% of cysts may
implantation of epithelial fragments must be quite com- also show focal areas of stratified squamous epithelium
mon and must involve stratified squamous as well as sinus that is often thin and flattened, or of cuboidal cells only two
epithelium. Although it is known that implanted stratified to five cell layers thick (Figure 16.6).
squamous epithelium can result in epidermal implant Maruyama et  al. (2002) examined 360 cysts and found
cysts in the skin, this does not appear to occur in the mouth. that all cases showed pseudostratified ciliated columnar
We must assume therefore that implanted oral epithe- epithelium that occupied 66% or more of the lining, with
lium may not survive, but that respiratory epithelium can frequent goblet cells. Up to 28% of the lining was simple
retain the ability to persist and to form cysts. This is likely cuboidal epithelium with goblet cells, and up to 6% may
to be due to secretion of mucus. Surgical ciliated cysts are be a flattened stratified squamous epithelium. In another
lined by respiratory-­type epithelium with frequent mucous study (Maeda et  al.  1987), 12.9% of cysts were lined
or goblet cells. Cyst formation is probably due to persistent ­predominantly by squamous epithelium, 2.9% by simple
secretion of mucus by the epithelial remnants, with accu- cuboidal epithelium, and the remainder (84.2%) by
mulation of the mucus creating a cyst lumen. Continuous ­pseudostratified ciliated columnar epithelium, either alone
secretion causes intraluminal pressure and gradual (67.1%) or with focal areas of squamous epithelium.
expansion. The delay between surgery and diagnosis of Mucous (goblet) cells were always a prominent feature.
10–30 years or more suggests that growth is slow. In  this The cyst wall is composed of loose, fibrous connective
respect, the surgical ciliated cyst can be regarded as a type tissue, often with focal areas of inflammation. A character-
of mucous retention cyst of iatrogenic origin. Although istic feature that has been noted by a number of workers
this process is rarely cited, the Japanese literature has long (Yamamoto and Takagi 1986; Basu et al. 1988; Maruyama
regarded the surgical ciliated cyst to be a form of retention et al. 2002; Nishioka et al. 2005) is that the fibrous tissue
cyst (Yamamoto and Takagi 1986). beneath the flattened or squamous epithelium is often
None of these mechanisms wholly explains the higher acellular and hyalinised (Figure  16.6). This is not seen
prevalence in Japanese patients. As mentioned previously, associated with the respiratory-­type epithelium.
the frequent use of Caldwell-­Luc surgery to treat chronic The cyst wall does not usually contain any mucous
sinusitis in the postwar period has been cited as the reason glands or ducts. This feature helps to distinguish the cyst
for the high prevalence of cysts in Japan. Nishioka et  al. from normal sinus mucosa, where subepithelial mucous
(2005), however, reported 1141 cases in their single depart- glands are often seen.
ment and noted that the annual rate had remained almost
constant, at 50 cases per year, right up until 2001. They sug-
gested therefore that the Caldwell-­Luc operation could not ­Treatment
be the only explanation for the high prevalence since, even
in Japan, the operation had become less common. They Surgical ciliated cysts are usually treated by enucleation or
suggested that age may be a factor, because antral surgery by marsupialisation into the sinus (Kaneshiro et al. 1981;
in Japan is often at a younger age and the high incidence of Yoshikawa et al. 1982). In both methods access to the sinus
cysts may be related to the fact that the facial skeleton is is required and this can be by an antrostomy through the
still growing. Although this is possible, there is no research antero-­lateral wall of the sinus via an intraoral approach
evidence to support it. (Caldwell-­Luc procedure), or by a meatal antrostomy via
­Treatment 269

Figure 16.5  Surgical ciliated cyst. The lining is


composed of pseudostratified ciliated columnar
epithelium.

Figure 16.6  Surgical ciliated cyst. In this field the


cyst is lined in part by pseudostratified ciliated
columnar epithelium (right side), but elsewhere the
lining is thin, flattened simple squamous or
cuboidal epithelium. In these areas the fibrous wall
shows a subepithelial band of acellular hyalinised
fibrous tissue (arrows).

nasal endoscopy. The exact approach may depend on the multilocular lesions, or cysts located in the antero-­lateral
site of the cyst. If the cyst is located close to the anterior or region of the sinus.
lateral wall, then a Caldwell-­Luc approach may be neces- A number of authors have emphasised the need to pre-
sary (Cho et al. 2019). Sometimes the cyst may be marsupi- vent the onset of surgical ciliated cysts. Theofilou et  al.
alised via a Caldwell-­Luc procedure, followed by insertion (2021) noted that postosteotomy cases most often arose in
of a drain through a meatal antrostomy. Cho et al. (2019) the gap created by a maxillary advancement procedure,
treated 67 patients by transnasal endoscopic marsupialisa- either with or without a concomitant graft. They suggested
tion and found an overall recurrence rate of 9.0% (6 cases), that the gap or the graft could be protected by placement of
but noted that 5 of the recurrences were associated with a protective membrane that would prevent the ingress of
cysts located at the antero-­lateral aspect of the sinus. They epithelial cells. Niederquell et  al. (2016) proposed that
concluded that over 90% of all cases could be treated chronic sinusitis should be treated whenever possible by
with  meatal antrostomy alone, but that a concomitant conservative methods, with avoidance of invasive surgery
Caldwell-­Luc procedure may be needed for larger lesions, of the maxillary sinus.
270

17

Pseudocysts of the Jaws: Simple Bone Cyst and Stafne Bone Cavity

CHAPTER MENU
Simple Bone Cyst, 271
●● Classification and Terminology,  271
●● Clinical Features,  272
–– Frequency, 272
–– Age, 273
–– Sex, 273
–– Site, 273
–– Clinical Presentation,  274
●● Radiological Features,  275
–– Multiple Simple Bone Cysts,  276
●● Simple Bone Cysts Associated with Cemento-­osseous Dysplasia,  276
–– Summary and Conclusions,  278
●● Pathogenesis, 278
–– Summary and Conclusions,  280
●● Histopathology, 280
●● Treatment, 281
Stafne Bone Cavity, 281
●● Clinical Features,  281
–– Frequency, 281
–– Age, 281
–– Sex, 282
–– Site, 282
–– Clinical Presentation,  282
●● Radiological Features,  282
–– Radiological Differential Diagnosis,  284
●● Pathogenesis, 284
●● Histopathology, 285
●● Treatment, 285
Focal Osteoporotic Bone Marrow Defect, 285
●● Clinical Features,  285
●● Radiological Features,  286
–– Radiological Differential Diagnosis,  286
●● Pathogenesis, 286
●● Histopathology, 286
●● Cavitational Osteonecrosis,  286
Aneurysmal Bone Cyst, 287

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
­Simple Bone Cys  271

This chapter considers a number of lesions of the jaws that Classification and Terminology
present clinically or radiologically as cystic lesions, but are
A notable feature of all the publications is uncertainty
empty or fluid-­filled bony cavities without an epithelial
about the nature of this lesion and this is reflected in the
­lining. These pseudocysts are rare in the jaws, but are
large number of different names that have been used. This
important in the differential diagnosis of cystic lesions. The
has led to inconsistency and confusion in the literature,
most commonly encountered are the simple bone cyst and
meaning that readers cannot always be certain that the
the Stafne bone cavity, and these will be considered in
same lesion is being discussed. The earliest reports used
some detail. Other lesions  –  focal osteoporotic bone
the term traumatic bone cyst (Blum  1932; Hansen
­marrow defect and aneurysmal bone cyst – will be briefly
et  al.  1974), while at the same time others used haemor-
reviewed.
rhagic bone cyst (Howe  1965) or solitary bone cyst
(Rushton  1946). In the long bones, the lesion is often
referred to as unicameral bone cyst. Although this term is
­Simple Bone Cyst an accurate description,1 it is not widely used and is very
rarely applied to jaw lesions.
Simple bone cysts are fluid-­filled or empty intraosseous
Other terms that appear in the literature include idiopathic
cavities found most commonly in the proximal metaphy-
bone cavity, extravasation bone cyst, solitary bone cyst, and
seal region of the long bones in children and adolescents.
simple bone cyst. Most of these terms imply a causation or a
Similar lesions are found in the mandible and very sel-
clinical feature and are thus inaccurate or potentially mis-
dom in the maxilla. The first report of a jaw lesion is
leading. Traumatic bone cyst is often used, but should prob-
attributed to C.D. Lucas, who mentioned it during a dis-
ably be avoided because it assumes a traumatic aetiology that
cussion following a presentation to the American Dental
has not been fully substantiated. Similarly, the terms extrava-
Association by Theodor Blum, who reported his realisa-
sation cyst and haemorrhagic cyst emphasise the possible
tion that not all jaw cysts were of dental origin
role of haemorrhage, but this also is not fully substantiated
(Blum 1929). Later, Blum described three cases in detail
and these terms should not be used. ‘Idiopathic’ is a good
and called them traumatic bone cysts (Blum  1932).
term and is accurate in that it means ‘of unknown cause’.
Subsequently, Rushton (1946) described more cases in
Unfortunately, this name is also applied to the Stafne bone
detail and suggested diagnostic ­criteria that are similar to
cavity (discussed later in this chapter) and therefore this term
those used today (Hansen et al. 1974; Harnet et al. 2008;
should also be avoided. The most widely used names are soli-
Raubenheimer et al. 2017; Chrcanovic and Gomez 2019c;
tary bone cyst and simple bone cyst and these can be regarded
Box 17.1).
as synonymous. However, ‘solitary bone cyst’ implies that
lesions are solitary or single, but multiple or bilateral lesions
have been reported, so this term also lacks accuracy.
In the World Health Organization (WHO) classifications
Box 17.1  Simple Bone Cyst: Definition and
of jaw lesions, this cyst has been included under the cate-
Diagnostic Criteria
gory of ‘bone cysts’. In the first edition it was called simple
Simple bone cyst is a cystic radiolucency that on gross bone cyst (Pindborg and Kramer 1971), but in the second
or histological examination does not have an epithelial edition it was solitary bone cyst (Kramer et  al.  1992).
lining, but shows a cavity that is empty or contains Subsequent editions (Barnes et  al.  2005; El-­Naggar
small amounts of fluid. et al. 2017), including the latest (fifth) edition (WHO 2022a),
have consistently called this lesion simple bone cyst with
Diagnostic Criteria
‘solitary’, ‘traumatic’, and ‘haemorrhagic’ as synonyms.
●● A well-­demarcated cystic radiolucency, usually
Furthermore, the latest WHO classification of soft-­tissue
(>98%) in the mandible
and bone tumours also ­prefers the term simple bone cyst
●● The cavity is empty or contains small amounts of fluid
and only recommends ­‘solitary’ as an acceptable alternative
●● There is no epithelial lining
term (WHO 2022b).
●● Diagnosis is by exclusion  –  examination during

surgical intervention or histology excludes any


­
other known diagnosis
1
Unicameral derives from ‘uni-­’, meaning one, and the Latin ‘camera’,
Cystic lesions that meet these criteria may be found meaning chamber. It is therefore accurate as a term for a unilocular
in association with fibro-­ osseous lesions, especially or unicystic lesion. Interestingly, the term unicameral is more often
florid cemento-­osseous dysplasia. used to describe a system of government that uses only a single
legislative or parliamentary chamber.
272 Pseudocysts of the Jaws: Simple Bone Cyst and Stafne Bone Cavity

For these reasons we suggest that simple bone cyst should Gomez (2019c) identified 284 papers that reported 1253
always be used. This reflects the simple nature of the lesion cysts. Table  17.1 summarises a number of selected larger
and makes no presumptions regarding pathogenesis or case series (>20 cases) with a broad geographical distribu-
clinical findings. Although it is not a true cyst, it presents tion, as well as the findings of the review by Chrcanovic
clinically and radiologically as a cystic lesion and the term and Gomez (2019c). The simple bone cyst shows a number
‘cyst’ remains acceptable and widely used. Simple bone of characteristic features (Box 17.2) that assist in making a
cyst is also the preferred term for all lesions, no matter diagnosis, but ultimately a final diagnosis is made by exclu-
where in the skeleton they are encountered. sion of any known cause of a cystic cavity in the jaws
There is some evidence, discussed in detail in the follow- (Box 17.1).
ing sections, that the simple bone cyst is not a single clin-
icopathological entity, but may be a common end result of Frequency
a number of causative factors (see ‘Pathogenesis’). The vast Simple bone cysts account for about 3% of bone lesions and
majority of lesions are single or solitary, affecting the man- are most common in children and adolescents. Most
dible, but about 5.0% of patients may have multiple cysts lesions (about 80%) are found in the proximal metaphyseal
(Chrcanovic and Gomez  2019c). In addition, there are a regions of the humerus (50%) or femur (30%), with most of
number of reports of lesions similar to simple bone cysts the remainder found in the proximal tibia or other long
arising in association with cemento-­osseous dysplasia (e.g. bones. Jaw lesions are rare and represent less than 5% of all
Chadwick et al. 2011; Yeom and Yoon 2020). Shimoyama skeletal lesions and about 1% of all jaw cysts.
et al. (1999) suggested that the simple bone cyst may take In his review of cases from the University of the
three distinctive clinicopathological forms: simple solitary Witwatersrand, Shear found only 35 specimens during the
lesions, multiple lesions that may progress or recur, and 46-­year period under review, constituting 1% of all jaw
cysts associated with other lesions, primarily cemento-­ cysts, and 7.2% of non-­odontogenic cysts (see Table 1.1). In
osseous dysplasia. In the following sections solitary and a review of Sheffield specimens there were only 36 cases
multiple lesions will be considered together as variants of reported over a 30-­year period, representing 0.5% of 6862
the same entity, but lesions associated with cemento-­ cysts of the jaws (Jones and Franklin  2006a,b) and only
osseous dysplasia will be discussed separately (see later 0.07% of all oral biopsies. Other studies have shown similar
Box 17.3 and ‘Simple Bone Cysts Associated with Cemento-­ low frequencies, with only 4 cases reported among 2030 jaw
osseous Dysplasia’). cysts (0.2%) in a study from Italy (Lo Muzio et al. 2017) and
9 cases among 5295 cysts in Greece (0.2%; Tamiolakis
et al. 2019).
Clinical Features
Most simple bone cysts are found below the age of
Most reports of simple bone cyst in the jaws are single case 20 years and so they are relatively more common in chil-
reports or small series. Lima et  al. (2020) found 29 pub- dren. Jones and Franklin found an overall frequency of
lished case series (5 cases or more) between 1951 and 2019, 0.5% of jaw cysts, but the frequency in their paediatric pop-
reporting almost 800  lesions. In a detailed systematic ulation was 3.4% (19 cases among 556 jaw cysts; Jones and
review, including single case reports, Chrcanovic and Franklin  2006b), which was more than 10 times greater

Table 17.1  Age, sex, and site distribution, and frequency of scalloping, of simple bone cysts from selected case series and the
systematic review of Chrcanovic and Gomez (2019c).

References Country n Age mean (range) Male (%) Mandible (%) Scalloped (%)

Suei et al. (1998) Japan 52 23.0 (10–56) 48.1 98.1 NR


Cortell-­Ballester et al. (2009) Spain 21 26.5 (4–45) 33.3 100 NR
Chadwick et al. (2011) Canada 68 18.5 (11–58) 50.0 98.5 54.4
You et al. (2017) S Korea 27 29.5 (10–63) 29.6 96.6 56.7
Flores et al. (2017) Brazil 42 19.6 (7–66) 47.6 100 30.8
Lima et al. (2020) Brazil 60 16.9 (7–74) 50.0 98.4 72.4
Roma et al. (2021) Brazil 30 22.2 (11–52) 50.0 100 NR
Chrcanovic and Gomez (2019c) SR 1253 20.4 (2–79) 48.3 96.4 41.7

NR, not reported; SR, systematic review.


­Simple Bone Cys  273

Box 17.2  Simple Bone Cyst: Key Features

Clinical
●● 60–70% arise in the second decade

●● Average age is 20 years

●● Equal sex distribution

●● Over 98% are found in the mandible, often premolar/molar region

●● About 5% of patients may have multiple lesions – usually two and often bilateral

●● Usually symptomless

●● Cysts are occasionally seen in older females (fifth decade), associated with cemento-­
osseous dysplasia
Radiology
●● A well-­
demarcated radiolucency
●● 50% show areas of cortication

●● Up to 70% are scalloped around the tooth roots, which ‘hang’ into the cavity

●● A cone shape is characteristic, but only seen in about 10%

Histopathology
●● Gross examination at surgery shows an empty cavity

●● About 50% contain blood or a serosanguinous fluid

●● Do not have an epithelial lining

●● About 10% have a thin membranous lining of loose fibrovascular tissue

●● May be scattered osteoclasts and inflammatory cells

than the frequency of 0.3% in adults (Jones and a slight female predilection. In their large systematic
Franklin 2006a). They also found that in children the simple review, Chrcanovic and Gomez (2019c) found that 51.7% of
bone cyst represented 51.4% of non-­odontogenic cysts (19 cases arose in females and 48.3% in males. However, it has
cases among 37), compared with only 6.7% in adults. Other been shown that multiple lesions and lesions associated
but smaller studies have found higher frequencies in children. with cemento-­osseous dysplasia arise more commonly in
In a study from Israel, Manor et al. (2012) encountered 95 jaw females and in older age groups. This is discussed later in
cysts in children over a 20-­year period and found 17 (17.9%) this chapter.
simple bone cysts. Over the same period, they recorded no
cases in adults (≥17 years). Resnick et al. (2016) reported 45 Site
cases of simple bone cyst diagnosed over a 15-­year period at Simple bone cysts involving the jaws are almost always
the Boston Children’s Hospital (Boston, MA, USA). found in the mandible, with each case series only reporting
one or two cases in the maxilla (Table 17.1). Studies have
Age consistently shown that almost all the maxillary cases have
The simple bone cyst occurs in young individuals, with the involved the anterior regions and the majority of the man-
vast majority of cases occurring below the age of 20 years. dibular cases have been reported in the body (about 65%)
There is a wide age range, with cases reported in patents up or symphyseal areas (about 25%) (Copete et  al.  1998;
to 74 years and with an overall average of between about 20 Cortell-­Ballester et  al.  2009; Resnick et  al.  2016; Flores
and 25 years (Table 17.1). In all studies, however, the peak et al. 2017; You et al. 2017; Lima et al. 2020).
age is always in the second decade, with reported frequen- In their detailed analysis, Chrcanovic and Gomez (2019c)
cies in this decade of 78% (Howe 1965), 66% (Chrcanovic identified 1237 cases where the site was recorded and only
and Gomez 2019c), 60% (Roma et al. 2021), 58% (Hansen 45 (3.6%) were found in the maxilla. Of the mandibular
et al. 1974), and 56% (You et al. 2017). lesions, 26% were found anterior to the canines, and 42.8%
were located in the premolar/molar area. A further 10.3%
Sex extended from the molar region into the angle or ramus,
Studies have shown a variable sex distribution (Table 17.1). and 8.4% occupied most of the body of the mandible, from
A small number of studies show a predilection for females the canine region to the molars. Overall, therefore, more
(Cortell-­Ballester et al. 2009; You et al. 2017), but overall than 60% of lesions occupied the posterior body (molar/
the sex distribution appears to be about equal or with only premolar area) of the mandible, with a small number
274 Pseudocysts of the Jaws: Simple Bone Cyst and Stafne Bone Cavity

extending further posteriorly or anteriorly. They also found Lima et al. (2020) identified 23 published case series that
that 129 lesions (16.5%) crossed the midline of the mandi- had recorded any history of trauma and found that 5
ble and 6 (0.8%) involved the coronoid process. reported that no patients could recollect a traumatic event.
Occasional cases have been reported in the mandibular The remaining 18 reports recorded trauma in between 5.0%
condyle (Persson  1985; Rubin and Murphy  1989; Telfer and 87.5% of patients, with an overall average of 22.2%
et al. 1990; Magliocca et al. 2007). (range 0.0–87.5%).
It seems that early case reports may have been more fas-
Clinical Presentation tidious in recording possible traumatic events. In his review
Most simple bone cysts are found incidentally when a of 60 cases reported between 1929 and 1963, Howe (1965)
patient undergoes radiological examination for another found details of trauma in 33 (55%) cases. Of these, 11 were
purpose. In the case series shown in Table 17.1, three of the sporting injuries, 5 were road accidents (including 1 hit by
studies reported that all their cases were incidental find- a trolley car), 4 were due to falls (1 after an explosion), and
ings with no clinical signs or symptoms (Cortell-­Ballester 4 followed dental extraction. A further 10 cases were
et al. 2009: You et al. 2017; Roma et al. 2021). In the studies recorded as miscellaneous, including a gunshot to the face,
that recorded clinical findings, the frequency of symptoms a blow with an axe, and being trapped in swing doors.
was 18.3% (Lima et al. 2020) and 9.5% (Flores et al. 2017). A number of authors have noted an association with
In their review of the literature, Lima et al. (2020) identi- orthodontic treatment (Copete et al. 1998; Velez et al. 2010;
fied 25 case series that had recorded clinical findings and Resnick et  al.  2016). Velez et  al. (2010) found that 10
found that in 9, all the cases were incidental findings on (22.7%) of their patients had undergone orthodontic treat-
radiology, with no clinical signs or symptoms. Of the ment and considered this to be an important factor in the
remaining 16 reports, signs and symptoms were found in pathogenesis of the lesion. They suggested that the ‘micro-
between 2.2% and 30.4% of patients, with an overall aver- trauma’ associated with tooth movement may increase vas-
age of 15.2% (range 0.0–30.4%). Chrcanovic and Gomez cular activity and osteoclast stimulation, or that a
(2019c) found that clinical findings were recorded for 836 piezoelectric affect may promote bone cavity formation
patients and that only 88 (10.5%) reported symptoms. (Velez et al. 2010). In support of this hypothesis, they did
In almost all reports, the only clinical symptoms are provide some evidence of a true association with orthodon-
complaints of swelling associated with pain or tenderness. tic treatment. They compared the incidence of orthodontic
Flores et al. (2017) found that 4 of their 42 patients were treatment in their group of patients with simple bone cysts
aware of swelling, but none had pain, whereas Lima et al. to the incidence in an age-­and sex-­matched control group
(2020) stated that 11 of their patients complained of pain or without cysts. Of the cyst patients 23% had undergone
swelling. Hansen (1974) detailed clinical findings in 61 orthodontic treatment, compared with only 13% of the
patients and found that 44 (72.1%) were incidental findings matched controls.
and only 8 (13.1%) complained of pain or tenderness. In Others suggest, however, that the association with ortho-
the remaining 9 patients the symptoms were vague and dontic treatment is entirely coincidental, since the peak
non-­specific. No patients complained of swelling, although age of presentation of the simple bone cyst (the second dec-
on clinical or radiological examination 14 (23.0%) cases ade) is the same as the age at which most adolescents have
showed some evidence of bone expansion. Howe (1965) orthodontic treatment and have radiographs taken (Copete
reviewed 60 early cases and found that 20 (33.3%) patients et al. 1998). Resnick et al. (2016) noted that all the cases in
reported symptoms. Of these, 16 (26.7%) had noted swell- their series were found incidentally on radiographs taken
ing and in 2 cases this was accompanied by pain. The during orthodontic treatment or routine dental care.
remaining 4 cases reported pain alone (2 cases) or pain Routine radiology of the jaws at a young age may also be
with paraesthesia (2 cases). the reason why the peak age of presentation is in the sec-
The simple bone cyst has been thought to be caused by ond decade. It is possible that many lesions may remain
trauma (see ‘Pathogenesis’), but overall only about 25% of symptomless and undiscovered until later in life when
patients report an episode of trauma (Chrcanovic and radiographs are taken to investigate other disorders. This
Gomez  2019c). In the series in Table  17.1, trauma was would explain the much lower prevalence in older age
recorded in 4 of the reports. The proportion of patients groups, when routine panoramic radiographs are much
reporting an episode of trauma was 28.6% (Flores less common. This same premise may also account for the
et  al.  2017), 23.8% (Cortell-­Ballester et  al.  2009), 11.7% association with trauma. Young people are more likely to
(Lima et al. 2020), and 7.4% (You et al. 2017). In their series sustain a traumatic injury and undergo radiological exami-
of 45 paediatric patients, Resnick et al. (2016) found that nation, whereupon an asymptomatic lesion may be
only 6 (13.3) patients had a history of trauma. discovered.
­Simple Bone Cys  275

Although simple bone cysts often embrace the teeth, the 30.8% (Flores et al. 2017), and 25.0% (Velez et al. 2010). In
teeth are found to be vital in about 95% of cases (Chrcanovic their systematic review, Chrcanovic and Gomez (2019c)
and Gomez 2019c; Lima et al. 2020). found that 41.7% of 561 cases where the feature was
Simple bone cysts of the long bones are one of the most recorded showed scalloping. MacDonald-­Jankowski (1995)
common causes of pathological fracture in children and regarded this feature as typical of the simple bone cyst and
adolescents, and about 80% of cases present with a fracture used the wonderfully descriptive term of the tooth roots or
of the humerus or femur. In the jaws, however, pathologi- apices ‘hanging’ within the cavity of the cyst (Figure 17.1).
cal fractures are very rare, and have only been encountered In most cases the lamina dura and periodontal space are
in 0.6% of cases (Chrcanovic and Gomez 2019c). intact and normal, but scalloped lesions may closely
embrace the teeth, and the lamina dura may be lost
(Figure  17.1). Suei et  al. (2010) reviewed the radiological
Radiological Features
findings of 30 cases and found that 9 (30%) had a scalloped
Most simple bone cysts are symptomless and a final diag- margin, and that in all 9 there was some loss of the lamina
nosis is made on the basis of radiological examination and dura. Conversely, 21 cases had a smooth margin and only 4
a lack of specific pathological features. This means that the of these (19.0%) showed any loss of lamina dura. Cases
diagnosis is always made on the basis of radiology followed with loss of lamina dura were also larger, may show root
by exclusion of other causes of a radiolucent cystic cavity resorption, and were more likely to have recurred. Overall,
(Box 17.1). however, root resorption or displacement of the teeth is
The cyst appears as a radiolucent area that is well demar- very rare and has been noted in only about 1.0% of cases
cated, with an irregular but definite margin and slight cor- (Chrcanovic and Gomez 2019c).
tication (Figure 17.1). The majority of lesions (85–90%) are Another characteristic radiological feature that is almost
unilocular (Velez et al. 2010; Chrcanovic and Gomez 2019c; diagnostic of the simple bone cyst is the presence of a cone-­
Roma et al. 2021), but a scalloped margin is common and is shaped margin. Copete et al. (1998) described this charac-
regarded as a characteristic feature. teristic cone-­shaped morphology in 28 (63.6%) of their 44
The scalloped margin is at the superior aspect of the cases. The typical shape was of a lesion that was oval or
lesion, where extensions of the cyst rise up between the rounded towards the posterior aspect of the mandible, but
tooth roots (Figure 17.1). This feature is so characteristic as with a cone shape at the anterior margin, with two planes
to be regarded as almost diagnostic of the simple bone cyst. converging at a 45° angle (Figure 17.1, arrows). This mor-
However, it is not seen in all cases (Table 17.1). The reported phology was present in 23 (52.3%) of their cases. Two cases
frequencies of a scalloped margin have been 72.4% (Lima showed a double cone shape, with cones projected at the
et al. 2020), 71.7% (Howe 1965), 68.2% (Copete et al. 1998), anterior and posterior margins, and three cases had ‘half-­
56.7% (You et al. 2017), 31.6% (MacDonald-­Jankowski 1995), cones’, with a single straight edge interfacing with the
curved margin. Their remaining cases were oval (7 cases:
15.9%), irregular (7 cases: 15.9%), or round (2 cases: 4.5%).
Copete et al. (1998) also found that 22 (50%) of their cases
had a corticated margin, but of these only 6 were corticated
around the whole periphery. Lima et  al. (2020) recorded
the shape of 49 of their cases and found that 40 (81.6%)
were oval and 9 (18.4%) were cone shaped. In their system-
atic review, Chrcanovic and Gomez (2019c) found that
only 8.2% (28 of 340) of cases where the shape was recorded
were cone shaped. These data suggest that although the
cone shape may be specific to the simple bone cyst, it is
only seen in up to 10% of cases.
A number of studies have recorded the size of the lesions
and have found that it ranges from 5 to 140 mm, with an
Figure 17.1  Simple bone cyst. Radiograph shows a cystic cavity average of 31 mm (Copete et al. 1998; Resnick et al. 2016;
involving an extensive area in the right body of the mandible. Flores et  al.  2017; You et  al.  2017; Roma et  al.  2021;
This example has a well-­defined margin with cortication. Chrcanovic and Gomez 2019c). Although they may grow to
Inter-­radicular scalloping is a prominent feature, and the tooth
a large size, most lesions appear to expand through the
roots appear to ‘hang’ into the cystic cavity. The anterior margin
of the cyst is cone shaped (arrows). Source: Courtesy of Prof. cancellous bone, often with minimal buccal or lingual
Christoffel Nortjé. expansion. In the larger reviews or case series, the
276 Pseudocysts of the Jaws: Simple Bone Cyst and Stafne Bone Cavity

proportion that have shown expansion has been between Simple Bone Cyst Associated
12% and 25% (Velez et  al.  2010; Flores et  al.  2017; with Cemento-­osseous Dysplasia
Chrcanovic and Gomez  2019c; Lima et  al.  2020), but the
In 1976, Melrose et  al. described a series of cases of
images and descriptions suggest that expansion is minimal
cemento-­osseous dysplasia that were extensive, with mul-
and tends to be smooth and fusiform (Suei et  al.  2010).
tiple lesions affecting more than one quadrant, mostly in
Thinning of the cortical plates is commonly seen (73.6% of
the mandible. They introduced the term ‘florid osseous
cases), but perforation of the cortical plates is rare and has
dysplasia’ for these lesions. They presented 34 patients and
been reported in less than 4% of cases (Chrcanovic and
in 14 noted cystic radiolucencies that they regarded as sim-
Gomez 2019c).
ple bone cysts. This was the first report of a possible asso-
Multiple Simple Bone Cysts ciation of simple bone cysts with florid cemento-­osseous
It has been reported that in about 5.0% of patients, sim- dysplasia. Subsequently there have been a number of small
ple bone cysts may be multiple or bilateral. In the case case series and single case reports (Table 17.2; Horner and
series shown in Table  17.1, bilateral lesions were Forman  1988; Higuchi et  al.  1988; Tong et  al.  2003;
reported in 2 cases (3.4%) by Lima et al. (2020) and in 2 Mahomed et  al.  2005; Velez et  al.  2010; Zillo Martini
cases (7.4%) by You et al. (2017). Howe (1965) reviewed et al. 2010; Peacock et al. 2015; Yeom and Yoon 2020).
60 patients and found 1 (1.7%) who had two lesions. A It is uncertain whether these lesions should be regarded
number of studies have reported multiple lesions that as simple bone cysts or as cystic change that may occur as
have arisen in association with cemento-­osseous dyspla- part of the natural history and development of cemento-­
sia, but these may be a separate entity and are discussed osseous dysplasia. In this respect, it is well documented
in the next section. that a number of fibro-­osseous lesions, including cemento-­
It is important to note that Chrcanovic and Gomez osseous dysplasia, show cystic change both on radiology
(2019c), in their systematic review, excluded cases that and on histological examination. Su et al. (1997a,b) under-
were associated with cemento-­osseous or fibrous dysplasia took a detailed analysis of 241 cemento-­osseous dysplasias
on the basis that they behave differently. They found a total and found that about 30% showed radiolucent lesions with
of 1253 cysts in 1187 patients. There were 58 patients (4.9%) well-­demarcated margins that may resemble simple bone
with multiple cysts, of which 45 patients had two cysts, cysts. On histological examination, they noted that large
11 had three cysts, and 2 had many cysts. They also found blood-­filled spaces are common. In a more recent review of
that patients with multiple cysts were more often female fibro-­osseous lesions, Eversole et  al. (2008) noted that
(71.3% vs 49.5% for solitary cysts) and older (26.6 years vs empty bone cavities and circumscribed radiolucent areas
19.7 years) and that multiple cysts were more often scal- are a characteristic feature of fibro-­osseous lesions, includ-
loped (67.1% vs 37.4%), were more likely to cause bone ing ossifying fibroma and cemento-­osseous dysplasia. They
expansion (36.2% vs 24.6%) and to recur or persist (12.0% noted that in florid cemento-­osseous dysplasia, these cavi-
vs 3.3%). ties may be empty, without a lining, and resemble simple
These data support the views of Shimoyama et al. (1999) bone cyst. In large case series of cemento-­osseous lesions,
that multiple or bilateral lesions could be regarded as a however, cystic change has not been prominent or has not
separate type of simple bone cyst that is distinctive from been noted as a specific feature. Cavalcanti et  al. (2018)
solitary lesions (Box 17.3). reviewed the cone beam computed tomography (CBCT)

Table 17.2  Age and sex distribution of simple bone cysts that have been reported to be associated
with cemento-­osseous dysplasia, from selected case series and the review of Yeom and Yoon (2020).

References Country n Age mean (range) Female (%)

Horner and Forman (1988) UK 4 36.0 (28–43) 100


Higuchi et al. (1988) Japan 4 40.5 (31–49) 100
Mahomed et al. (2005) S Africa 7 42.3 (26–54) 85.7
Tong et al. (2003) China 3 57.3 (31–49) 100
Zillo Martini et al. (2010) Brazil 3 49.7 (11–66) 100
Yeom and Yoon (2020) Review 45 43.5 (11–66) 93.3

Note: The review by Yeom and Yoon (2020) incorporates some of the included case series.
­Simple Bone Cys  277

features of 82 cemento-­osseous dysplasias, and although stroma of heavily mineralised lesions. Histological examina-
their images show a number of well-­demarcated cystic tion showed a variable pattern of mineralisation, but the
radiolucencies, they did not regard any cases as being asso- authors noted areas of cellular osteoid associated with resorp-
ciated with simple bone cyst. Many of the hypodense tion that created fluid-­or blood-­filled spaces that coalesced to
(lucent) lesions that they illustrated contained areas of form empty, smooth-­surfaced cystic spaces. The authors pro-
opacity typical of cemento-­osseous dysplasia. posed that this process represented the formation of simple
Kato et al. (2020) undertook a detailed CBCT analysis of bone cysts and may explain the association of bone cysts and
60 cases of cemento-­osseous dysplasia involving 244 areas cemento-­osseous dysplasia. However, their illustrations sug-
of the jaws. They identified five areas that they described as gest cystic spaces within the stroma of the lesions surrounded
simple bone cyst, characterised as hypodense (lucent) cor- or lined by lesional tissue, rather than a cavity within the
ticated lesions that caused expansion and perforation of bone, which is a feature of the simple bone cyst.
the cortical bone. However, the case that they illustrated We would suggest that this process represents a form of
showed a corticated hypodense area containing the roots of cystic degeneration that might be a common feature of
a molar tooth with hypercementosis and irregular deposits fibro-­osseous lesions generally, rather than a specific exam-
of mineralisation typical of cemento-­osseous dysplasia. ple of an association of two different lesions. A similar pro-
In a similar review, also of 82 cases, Pereira et al. (2016) cess also explains the often-­cited association between
did not find any cases that they described as being associ- fibro-­osseous lesions and blood-­filled cystic spaces that
ated with simple bone cysts. They did find, however, that have been called secondary aneurysmal bone cyst. These
florid cemento-­osseous dysplasia typically shows multiple almost certainly represent degenerative change and are not
radiolucent and mixed radiolucent/radiopaque areas. true aneurysmal bone cysts, which are now known to be a
Their illustrations show radiolucencies that resemble sim- neoplasm. The term aneurysmal bone cyst should no
ple bone cysts, with scalloping of the margins and the longer be used in the context of these secondary, reactive
‘hanging’ roots feature. Furthermore, they showed that changes (WHO 2022b). Aneurysmal bone cyst is discussed
early and intermediate lesions showed multiple well-­ in more detail later in this chapter.
demarcated radiolucencies and suggested that cemento-­ Table  17.2 summarises demographic data from five
osseous dysplasia is a progressive disease that becomes reports of simple bone cysts associated with cemento-­
increasingly mineralised over time. osseous dysplasia, and from a review of 45 cases by Yeom
This progression was also shown in the original series and Yoon (2020). In addition, there are reports of cysts aris-
described by Melrose (1976). He found that 14 of his ing within cemento-­osseous dysplasia in some of the larger
patients had cystic radiolucencies that proved to be empty series of simple bone cysts, including 6 cases included in a
on surgical exploration or histological examination. series of 44 cysts by Velez et al. (2010), 6 cases among 20
Although he called these simple bone cysts, he also sug- cysts by Peacock et  al. (2015), and 1 case in each of the
gested that they were a stage in the progression of the series of 60 and 44 cysts reported by Lima et al. (2020) and
cemento-­osseous dysplasia. He described and illustrated Copete et  al. (1998), respectively. In total about 50 cases
progressive mineralisation of the radiolucencies over a have been reported.
period of 1–29 years. On histological examination, he also The data in Table 17.2 show a different demographic of
found that cases often showed ‘aneurysmally’ dilated ves- patients to those summarised in Table  17.1. Patients
sels and blood-­filled spaces that he thought might repre- reported as having cysts associated with cemento-­osseous
sent early cyst development. Other cystic spaces were lined dysplasia were almost all female and had an average age of
by a thin layer of fibrous tissue with scattered osteoclasts. about 45 years, with a peak in the fifth and sixth decades,
These features are in keeping with the concept that fibro-­ compared with patients with simple bone cysts alone who
osseous lesions show cystic degeneration of the stroma, show an equal sex distribution and an average age of about
either in early lesions before mineralisation (Pereira 20 years, with a peak in the second decade. In reports where
et al. 2016) or in the stroma of later mineralised lesions (Su the ethnicity is recorded, cases associated with cemento-­
et al. 1997a,b; Eversole et al. 2008). Such degeneration may osseous dysplasia have, almost without exception, been
result in empty or blood-­filled spaces, often resembling sim- reported in black females. This demographic is the same as
ple bone cyst or aneurysmal bone cyst (van Heerden for cemento-­osseous dysplasia generally (Su et  al.  1997b;
et al. 1989; Raubenheimer et al. 2016). Raubenheimer et al. Eversole et  al.  2008; Pereira et  al.  2016). A review of the
(2016) described 18 cases (in 14 patients) of large expansile papers and of the illustrations shows that most cases have
osseous dysplasia. Of these cases, 5 were described as con- presented as typical florid cemento-­osseous dysplasia with
taining simple bone cysts, but these were illustrated as multiple radiolucencies, many of which are mixed with
hypodense areas on computed tomography (CT) within the focal areas of mineralisation.
278 Pseudocysts of the Jaws: Simple Bone Cyst and Stafne Bone Cavity

A diagnosis of simple bone cyst is by exclusion (Box 17.1) Summary and Conclusions


and most reports have recorded surgical intervention with These studies present a rather complex account of the
or without histological examination, which has demon- relationship between cystic radiolucencies in cemento-­
strated an empty bone cavity (Horner and Forman  1988; osseous dysplasia and simple bone cysts. On the one hand,
Higuchi et al. 1988; Tong et al. 2003; Mahomed et al. 2005; cystic areas and well-­demarcated radiolucencies are a
Velez et  al.  2010; Zillo Martini et  al.  2010; Peacock well-­established feature of fibro-­osseous lesions, includ-
et al. 2015). Taken together, the findings of multiple radio- ing cemento-­osseous dysplasia (van Heerden et  al.  1989;
lucencies and an empty cavity have been taken to confirm Su et al. 1997a,b; Eversole et al. 2008; Pereira et al. 2016;
a diagnosis of simple bone cyst. However, in many of the Raubenheimer et  al.  2016; Cavalcanti et  al.  2018; Kato
reports, the radiolucent cystic spaces often contained foci et al. 2020). These may be seen in early lesions that have
of mineralisation and the associated teeth showed hyperce- not yet mineralised or in later or large lesions that may
mentosis. These features have been reported in most cases undergo cystic degeneration. In this respect, therefore,
of cemento-­osseous dysplasia and radiolucent, empty these cystic spaces may not be simple bone cysts, but
cystic spaces are a consistent feature, especially in early merely part of the spectrum of radiological and histologi-
and developing lesions (Su et al. 1997b; Pereira et al. 2016; cal features that are seen in a variety of fibro-­osseous
Kato et al. 2020). lesions.
The largest case series of simple bone cysts associated An alternative explanation is that they represent a variant
with cemento-­osseous dysplasia is of 23 cases reported by of simple bone cyst that may be seen in older individuals in
Chadwick et al. (2011). This series is widely cited and often association with cemento-­osseous dysplasia as a result of a
used to confirm the association between the two lesions similar pathogenic mechanism (see ‘Pathogenesis’) with a
and to suggest that up to one-­third of simple bone cysts common end result. This explanation is supported by the
may be associated with florid cemento-­osseous dysplasia observation that most of the cysts described in cemento-­
(e.g. Raubenheimer et  al.  2017). However, care must be osseous dysplasia have been empty cavities with character-
taken in interpreting this report. First, it is not clear istic radiological features similar to simple bone cysts in
whether Chadwick et al. (2011) reported a sequential series young people (Horner and Forman  1988; Higuchi
of cases, or rather reported and compared selected cohorts et  al.  1988; Tong et  al.  2003; Mahomed et  al.  2005; Velez
of 68 solitary and 23 cemento-­osseous dysplasia-­associated et  al.  2010; Zillo Martini et  al.  2010; Peacock et  al.  2015).
cysts. Secondly, the 23 cases recorded as simple bone cysts This concept supports the proposal of Shimoyama et  al.
in cemento-­osseous dysplasia did not meet the criteria for (1999) that the simple bone cyst takes three clinicopatho-
diagnosis (Box 17.1), but were diagnosed only on the radio- logical forms: simple solitary lesions, multiple lesions that
logical findings, with no exclusion of other pathologies by may progress or recur, and cysts in older patients associated
surgical intervention or histological analysis. It is possible, with cemento-­osseous dysplasia (Box 17.3).
therefore, that many of the multiple radiolucent lesions A number of authors regard simple bone cysts associated
reported as simple bone cysts were in fact early lesions of with cemento-­osseous dysplasia as distinctive and separate
cemento-­osseous dysplasia. The authors stated that histo- lesions that have different clinicopathological presentation
logical examination was not necessary for the diagnosis of and behaviour and should not be included in the spectrum
cemento-­osseous dysplasia, but overlooked the fact that of ‘classic’ simple bone cyst (Chrcanovic and Gomez 2019c).
surgical or histological examination is necessary to con-
firm a diagnosis of simple bone cyst.
Pathogenesis
Chadwick et al. (2011) suggested that simple bone cysts
associated with cemento-­osseous dysplasia are larger and Neither the aetiology nor the pathogenesis of the simple
more extensive than solitary lesions because they present bone cyst is known, but there are a number of theories that
later in older individuals, but they conclude that they are have been examined. As discussed previously (see ‘Clinical
similar lesions that arise in different biological circum- Presentation’), it has often been stated that the lesion is
stances. In both cases, however, they propose that the caused by trauma or may be associated with orthodontic
underlying pathogenesis is related to a disturbance of treatment. Although about 25% of patients report an asso-
the normal balance of osteoblast–osteoclast function. In ciated traumatic episode (Chrcanovic and Gomez 2019c) or
the young this may be associated with remodelling of the are undergoing orthodontic treatment (Velez et al. 2010),
alveolar bone during growth and development, while in these are a minority and the associations cannot be inter-
adults with cemento-­osseous dysplasia there may be a preted as causation. In particular, no studies have com-
pathological imbalance with a common end result (see pared the incidence of trauma among patients with simple
‘Pathogenesis’). bone cysts with the incidence of trauma in a matched
­Simple Bone Cys  279

cohort of young people without cysts. It is quite possible at operation to be empty, the fact that some contain
that the association is entirely coincidental due to the blood or serosanguineous fluid tends to support the
­frequent radiological examination of young people follow- concept of a ­h aematoma breaking down. As noted pre-
ing trauma to the face. viously, cystic degeneration is a common finding in
With regard to orthodontic treatment, Velez et al. (2010) fibro-­o sseous lesions and is often associated with
did provide some evidence of a true association by showing haemorrhage to produce empty or blood-­f illed cavities
that 23% of their cyst patients had undergone orthodontic resembling simple bone cyst or aneurysmal bone cyst
treatment compared to only 13% of matched controls. (van Heerden et  al.  1989; Raubenheimer et  al.  2016).
Olech et  al. (1951) first suggested that trauma was the Raubenheimer et al. (2016) in particular noted areas of
cause and proposed that trauma to a bone results in cellular osteoid that were associated with resorption
intramedullary haemorrhage, which on occasion may fail that created fluid-­ or blood-­f illed spaces that coalesced
to organise and can resorb to leave an empty cystic cavity. to form empty, smooth-­s urfaced cystic spaces. They
They suggested that cysts seem to develop only after injury proposed that this process represented the formation of
to those areas of the skeleton where cancellous bone is simple bone cysts, and it is possible that a similar
enclosed in a heavy compact cortical layer. This would mechanism may occur within an organising haema-
explain the most frequent sites in the metaphyses of long toma following trauma.
bones and in the mandible. It would also explain the fact Shimoyama et  al. (1999) reviewed the literature and
that most simple bone cysts develop in young individuals. found that there had been eight different theories about the
This proposal by Olech et  al. (1951) has never been pathogenesis of the simple bone cyst, but these can be con-
substantiated nor refuted and most authors still regard solidated into three potential mechanisms (Harnet
some form of trauma as the most likely cause. The et al. 2008): trauma, a disorder or imbalance in bone growth
breakdown of haematomas and their failure to organ- or turnover, and degeneration of a bone lesion or tumour.
ise, particularly if they are large, is a well-­known prob- Harnet et al. (2008) regarded trauma as the most likely
lem in surgery and it is perfectly conceivable that this and the most widely accepted cause of the simple bone
could occur following intramedullary haemorrhage. cyst. They suggested that the process first proposed by
Although the majority of simple bone cysts are found Olech et  al. (1951), of intramedullary haemorrhage

Box 17.3  Simple Bone Cyst: Clinicopathological Variants

Solitary Cysts
●● Represent more than 90% of all simple bone cysts

●● Average age about 20 years

●● Males and females affected equally

●● More than 98% are found in the mandible

Multiple Cysts
●● Represent 5% or less of all simple bone cysts

●● Usually only two lesions and may be bilateral

●● Average age about 27 years

●● 70% are found in females

●● More than 90% found in the mandible

Cysts Associated with Florid Cement-­osseous Dysplasia


●● Frequency uncertain, but probably less than 5% of total

●● May be multiple lesions

●● Average age about 45 years

●● More than 90% are found in females

●● The vast majority reported in black females

●● Usually in the mandible, but involve more than one quadrant

●● The cysts are empty or contain fluid, but often contain focal mineralisation

●● The cysts may represent secondary degenerative changes


280 Pseudocysts of the Jaws: Simple Bone Cyst and Stafne Bone Cavity

followed by breakdown of the haematoma, is the primary Histopathology


pathogenic mechanism. They further proposed that this is
Pathological examination cannot be used to diagnose a sim-
facilitated by venous stasis that causes aseptic necrosis and
ple bone cyst, but is essential to confirm the radiological
activation of osteoclasts. Although trauma is less likely to
diagnosis and to exclude any other cause for the cystic lesion.
be involved in lesions of the long bones, Abdel-­Wanis and
When the cyst cavities are opened at operation, they are
Tsuchiya (2002) suggested that the primary event in lesions
usually found to be empty or to contain small amounts of
of the humerus and femur was obstruction of venous
blood or serosanguineous fluid. Howe (1965) reviewed 60
drainage, resulting in accumulation of interstitial fluid and
cysts and found that where the contents were recorded,
cavity formation. In the metaphyseal regions of long bones
37.5% were completely empty and 78% showed no evidence
they proposed that venous obstruction may be secondary to
of a lining. In a similar study, Hansen et al. (1974) found
a developmental error, but at other sites the aetiological
that 50% of their cysts were completely empty, 38.3% con-
factors were unknown.
tained fluid, and the remainder (11.7%) contained frag-
A developmental error, or disorder of bone growth, is the
ments of connective tissue.
second of the three possible mechanisms. This would cer-
In their systematic review, Chrcanovic and Gomez
tainly explain the common location of long-­bone lesions
(2019c) found that of 706 cases where the contents were
adjacent to the growth plates. In the mandible, Harnet et al.
recorded, 54.0% were completely empty cavities and 46.0%
(2008) suggested that this is related to development of the
contained some fluid.
mandible, but it is more likely that the alveolar bone, which
Most surgeons, when they encounter the empty cavity,
has a high rate of bone turnover and remodelling, is highly
undertake curettage of the underlying bone, which often
susceptible to an imbalance in osteoblast/osteoclast func-
appears firm and normal. If a specimen is taken for histo-
tion. Velez et al. (2010) proposed that the tooth movement
logical examination, the pathologist may receive only frag-
associated with orthodontic treatment may increase vascu-
ments of soft tissue and bone.
lar activity and osteoclast stimulation. Raubenheimer et al.
When a lining is present, histological examination shows
(2016) noted increased osteoclast activity as a factor in the
a loose, vascular fibrous tissue of variable thickness
initiation of cystic cavities in fibro-­osseous lesions, and
(Figure  17.2). There is no epithelial lining. Haemorrhage
Chadwick et  al. (2011) supported the suggestion that an
is usually present and haemosiderin pigment, foamy histi-
imbalance in osteoblast/osteoclast function initiated cyst
ocytes, and scattered small multinucleate cells may be
formation, and would explain the occurrence of simple bone
seen. The adjacent bone, when included in the specimen, is
cysts in older individuals associated with cemento-­osseous
usually normal, but may show areas of osteoid, or of
lesions. Furthermore, they and others (Mahomed et al. 2005;
resorption with osteoclasts.
Velez et  al.  2010; Peacock et  al.  2015) suggested that the
A common finding is accumulation of amorphous, often
association with older females may be due to impaired oste-
fibrillar eosinophilic material that is usually described as
oblastic activity associated with hormonal changes.
fibrin (Figure  17.3). Although it resembles fibrin,
The third proposed mechanism is degeneration of a bone
Baumhoer et  al. (2011) have shown that this material is
lesion or tumour. The idea that simple bone cysts arise as a
actually composed of collagen or the bone matrix protein
result of necrosis of an intraosseous neoplasm is no longer
considered tenable, since residual tumour or epithelial ele-
ments are never seen histologically. However, as already
discussed, cystic degeneration in fibro-­osseous lesions may
result in an empty cystic cavity that shows features of sim-
ple bone cyst.

Summary and Conclusions
As can be seen from this discussion, these three pathogenic
mechanisms have similar elements and are not mutually
exclusive. Overall, it seems that the simple bone cyst is not
a single clinicopathological entity, but can be regarded as a
spectrum with different causes although with a similar
pathogenesis and a common end result – an intraosseous
cystic cavity. This is in keeping with the concept that there
Figure 17.2  A simple bone cyst is lined by loose vascular
are three clinicopathological variants of the simple bone fibrous tissue. The underlying bone is normal, but areas of
cyst (Box 17.3). cellular bone and osteoid can be seen.
­Stafne Bone Cavit  281

really be called a pseudocyst, because the cystic appearance


is essentially an illusion caused by an anatomical anomaly
that produces an indentation of the lingual aspect of the
mandible. Although not strictly pathological in nature, it is
important because the cystic appearance on radiology must
be correctly interpreted and considered in the differential
diagnosis of radiolucent lesions in the mandible.
The condition was first described in 1942 by Stafne, who
reported 35 bone cavities at the angle of the mandible.
Since then the features have become well documented and
many hundreds of cases have been reported. Philipsen
et al. (2002) undertook a comprehensive review of the lit-
erature and identified 103 publications reporting 316 clini-
cal cases (including 69 of their own), and an additional 16
papers reporting 267 cases found on archaeological or
Figure 17.3  Simple bone cyst. Here the lining is thickened and museum specimens of dried skulls. Subsequently there
inflamed, with accumulations of macrophages and eosinophilic
have been many further single case reports and a number
fibrin-­like material. Studies have shown that this is not fibrin, but
is composed of collagens and matrix proteins (see text for details). of series that have examined the radiological or imaging
features (Quesada-­Gómez et al. 2006; Schneider et al. 2014;
decorin. These authors undertook a histological and immu- Hisatomi et al. 2019; Morita et al. 2021).
nohistochemical analysis of 51 simple bone cysts, includ- With regard to terminology, a number of names have
ing 20 from the mandible. They found that 12 (23.5%) been used for this lesion, including static bone cavity, static
contained deposits of hypocellular eosinophilic fibrin-­like bone cyst, static bone defect, idiopathic bone cavity or cyst,
material. Immunohistochemistry demonstrated that in all latent bone cavity or cyst, lingual mandibular bone defect
cases this material was negative for fibrin, but showed or depression, and mandibular concavity. However, the
expression of collagens I or III and decorin. most widely used term pays homage to Stafne, and most
authors call it the Stafne bone cavity.

Treatment
Clinical Features
Simple bone cysts are usually treated as part of the diagnos-
tic process. To determine the nature of the radiolucency, the Frequency
lesion is opened to reveal an empty cavity, as described Stafne bone cavity is almost always encountered inciden-
above. The cyst wall is then curetted causing haemorrhage tally during radiological examination for another purpose.
into the cavity, and in the vast majority of cases this results It is rarely classified as a jaw cyst and is not usually included
in uneventful healing. It is presumed that granulation tis- in reports of jaw lesions from pathology departments.
sue and eventually new bone proliferate and replace the Overall the condition is not uncommon and has been
haemorrhage caused as a result of the surgery. In their sys- found as an incidental finding in about one in every 1000
tematic review, Chrcanovic and Gomez (2019c) found that radiographs.
99.2% of all cases had been treated by surgical access only Philipsen et al. (2002) reported 69 new cases from Japan that
(32.7%) or by surgical access and curettage (66.5%). had been found on examination of 42 600 consecutive radio-
Recurrence is unusual, but many papers have reported graphs, giving a frequency of 0.16%. Others have reported
lesions that persist, and often do not differentiate between similar frequencies. Oikarinen and Julku (1974) examined
recurrence and persistence. Overall, however, Chrcanovic 10 000 orthopantomograms and found 10 examples, a fre-
and Gomez (2019c) found that of 691 lesions that had been quency of 0.1%. MacDonald and Yu (2020) reviewed 6252 con-
followed up, only 32 (4.6%) persisted during a follow-­up secutive radiographs and found 3 cases (0.05%) and Assaf et al.
period of between 1 and 216 months. (2014) found 11 (0.08%) cases on review of 14 005 radiographs.

Age
­Stafne Bone Cavity The cavities are found in adults with an average age of
between 50 and 60 years (Stafne  1942; Schneider
The Stafne bone cavity is not a cyst, but it appears as an et  al.  2014; Hisatomi et  al.  2019; Aps et  al.  2020; Morita
intraosseous cystic lesion on a plain radiograph. Nor can it et al. 2021). About 65% of all cases are encountered in the
282 Pseudocysts of the Jaws: Simple Bone Cyst and Stafne Bone Cavity

fifth and sixth decades. The age range has been shown to
be 11–87 years. No cases have ever been reported in an
individual less than 11 years of age, and only about 2%
have been found in persons under 20 years (Philipsen
et al. 2002).

Sex
About 85% of cases occur in males (male : female ratio of
about 6 : 1). Philipsen et  al. (2002) reviewed 316 clinical
cases and found that 268 (84.8%) arose in males. A similar
sex distribution has been reported in other large series. In
the original report, Stafne (1942) found 28 of 34 (82.4%)
cases in men and Schneider et  al. (2014), Hisatomi et  al.
(2019), Aps et al. (2020), and Morita et al. (2021) found that
Figure 17.4  Stafne bone cavity. A dried bone specimen shows a
males were affected in 66.6%, 79.0%, 87.2%, and 70.0% of
well-­demarcated indentation in the lingual aspect of the
cases, respectively. posterior mandible. Source: Courtesy of Prof. Philip V. Tobias.

Site
Stafne bone cavities are only found in the mandible and the
vast majority are located on the lingual aspect, at the angle
of the mandible below the inferior dental (ID) canal
(Figures  17.4 and  17.5). However, cases are occasionally
seen in the anterior mandible, and very rarely in the ramus.
Philipsen et al. (2002) found that of 316 clinical cases, 270
(85.4%) were located on the lingual aspect of the posterior
mandible, 40 (12.7%) on the lingual aspect of the anterior
mandible, and 6 (1.9%) on the lingual aspect of the ramus.
Among the 267 archaeological or museum specimens that
they reviewed, they found a similar distribution (85.0%,
12.0%, and 1.5%, respectively), but also found a report of a
single case on the buccal aspect of the ascending ramus
(Shields 2000). Figure 17.5  Stafne bone cavity. A plain radiograph shows a
Posterior cases are always located below the ID canal and well-­demarcated and corticated oval radiolucency. The cavity is
below the inferior dental canal and impinges onto the lower
are associated with the submandibular gland below the
border of the mandible. Source: Courtesy of Dr Hilton Mirels.
mylohyoid muscle in the submandibular space. Anterior
cases, however, are associated with the sublingual gland
Radiological Features
and are located above the mylohyoid muscle. A more
detailed analysis of the location of the cavities and the rela- On plain radiographs the cavities appear as a round or oval
tionship to the teeth and ID canal is discussed later (see cystic radiolucency that is well demarcated, often with a
‘Radiological Features’). characteristic ‘punched-­out’ appearance (Figure  17.5).
Most cases have a smooth outline and in about 90% the
Clinical Presentation margin is partially or totally corticated (Hisatomi
Stafne bone cavities are symptomless and do not cause et  al.  2019). The appearance of an intraosseous lesion
swellings, and patients never present with clinical com- (Figure 17.5) is an illusion and on three-­dimensional anal-
plaints. However, a small number of cases in the posterior yses by CT or direct observation it can be seen that the cav-
mandible may disrupt the continuity of the lower border ity is an indentation in the lingual aspect of the mandible
and an indentation may be felt if the region is palpated (Figures  17.4 and  17.6). Mann (1992) studied 10 cases in
extraorally (Stafne  1942). Hisatomi et  al. (2019) showed dried bone archaeological specimens and prepared moulds
that 17% of cases caused a discontinuity of the lower using silicone impression material. He found that the max-
­border of the mandible, but they did not report on clinical imum dimensions ranged from 7 to 14 mm and that 7 of the
findings or whether these cases were detectable on 10 cases had a ‘mushroom’ shape, in that the internal
palpation. dimensions were greater than the opening. This feature is
­Stafne Bone Cavit  283

clearly visible when the cavities are examined by CT or plate. Type II showed the bottom of the cavity touching the
magnetic resonance imaging (MRI) (Figure 17.6), but may buccal cortical plate, and in Type III the buccal cortical
also be appreciated on plain radiographs when, apart from plate was expanded. Six cases were Type I, 7 were Type II,
the outer distinct cortication, a second inner ring can and 3 were Type III. In a similar study, Morita et al. (2021)
sometimes be seen that encircles an area of more marked examined 40 cases and found that 60% were Type I and 40%
radiolucency (Figure 17.5). were Type II. They did not find any cases with buccal
Examination by plain radiographs may lead to an errone- expansion (Type III).
ous diagnosis of an intraosseous lesion, resulting in unnec- Aps et  al. (2020) have undertaken a detailed review of
essary surgical intervention. The true nature of the cavity is the radiology of the Stafne bone cavity in order to accu-
therefore best appreciated by examination using CT or rately map the location and relationships to adjacent struc-
MRI, which will show that the margins are continuous tures (Figure 17.7). They identified 64 papers reporting 109
with the lingual cortical plate and the lumen opens into the cases where there was sufficient detailed information to
soft tissues at the lingual aspect of the mandible (Ariji accurately locate the defects. They found 8 patients who
et al. 1993; MacDonald 2016; Friedrich et al. 2020; Morita had multiple cavities and in 6 cases these were bilateral. Of
et al. 2021). MRI also enables soft tissues to be visualised the remaining 101 solitary cases, they found that 76 (75.2%)
and shows that in most cases the defects contain exten- were located in the posterior mandible, 11 (10.9%) in the
sions of the adjacent submandibular (posterior cases) or premolar region, 9 (8.9%) in the canine/incisor region, and
sublingual gland (anterior cases) (Probst et  al.  2014; 5 (5.0%) in the ramus. Three cases in the anterior region
Friedrich et al. 2020). Earlier studies have used sialography crossed the midline.
to confirm the presence of salivary tissue and in this way it Of the 76 cases that affected the posterior mandible, 29
has been shown that a Stafne bone cavity located in the (28.7% of the total) involved only the angle and 37 (36.6%)
posterior aspect of the ascending ramus contained lobes of only the molar region; 10 (9.9%) extended from the molar
the parotid gland (Barker 1988). region to the angle. All the posterior cases were located
CT analysis has also shown that the cavities may extend below the ID canal, but about half of cases (50.5%)
across the full width of the mandible and may impinge on, encroached on or were contiguous with the canal. Only 3
or even expand, the buccal cortical plate. Ariji et al. (1993) (3.0%) cases, all in the anterior region, were seen to be asso-
examined 15 cases by CT and classified them into three ciated with a root apex and might mimic a periapical lesion.
types. In Type I the bottom of the concavity was located in The authors concluded that the bone cavities can arise at
cancellous bone and did not contact the buccal cortical any site in the mandible and drew attention to fact that on
a plain radiograph many can mimic other types of radiolu-
cent lesions. Figure  17.7 summarises the distribution of
Stafne bone cavities based on the data of Aps et al. (2020).

5%

30%
10%
35% 10%
10%

Figure 17.6  Stafne bone cavity. Computed tomography (CT)


shows the cavity to be a well-­demarcated indentation into the Figure 17.7  Stafne bone cavity. Site distribution of cavities
lingual aspect of the mandible. Note that the internal superior– within the mandible. Source: Data from Aps et al. (2020), rounded
inferior dimension is greater than than the opening. to the nearest 5% for clarity and to aid recall.
284 Pseudocysts of the Jaws: Simple Bone Cyst and Stafne Bone Cavity

Radiological Differential Diagnosis


Box 17.4  Stafne Bone Cavity: Key Features
Stafne bone cavities are found incidentally on radiological
investigation, but because they are simple radiolucencies, a ●● The cavity is an indentation on the lingual aspect of
definitive diagnosis cannot be made on plain radiographs the mandible
alone. In early studies, the finding of a radiolucency, ●● Not uncommon: frequency is about 1 in every 1000
­presumed to represent an intraosseous cystic lesion, led to radiographs (0.1%)
surgical intervention, whereupon the surgeon would find ●● Predominantly found in males (85%)
an empty cavity that was continuous with the soft tissues at ●● Average age about 55  years. Peak in fifth and
the lingual aspect of the mandible. This would confirm the sixth decades
diagnosis, but had involved an unnecessary surgical ●● Symptomless and found as an incidental finding on
procedure. radiographs
The majority of lesions are well-­demarcated radiolucen- ●● Well-­demarcated, usually corticated radiolucency
cies in the molar/angle region of the mandible and are ●● More than 80% are found in the posterior mandible
located below the ID canal. This appearance is typical of ●● Always below the ID canal
the Stafne bone cavity and excludes a lesion of odontogenic ●● About 12% are found in the anterior mandible, above
origin, since these are located in the tooth-­bearing alveolar the mylohyoid muscle
bone above the ID canal. When such a radiolucency is seen ●● Rarely found (<5.0%) in the ramus
incidentally on a routine radiograph, the diagnosis should ●● They contain lobules of normal salivary gland
be confirmed by CT or MRI. Schneider et  al. (2014) pre-
sented an algorithm for diagnosis of the Stafne bone cavity,
in which they suggest that a unilocular radiolucency in the proposal seems extremely unlikely given that the cavities
posterior mandible with corticated margins and below the are never seen in children and have a peak age of presenta-
ID canal is diagnostic. If these four criterion are not met, tion in the fifth and sixth decades. It is now generally
then CBCT or MRI can be used to confirm the diagnosis by accepted that the defect is acquired as a result of resorption
demonstrating a lingual opening or presence of soft tissue of the bone due to pressure from a lobe of the submandibu-
in the cavity. Schneider et al. state that surgical exploration lar or sublingual glands. This is supported by the observa-
can be avoided in almost all cases by careful consideration tion that the cavities almost always contain salivary tissue.
of the radiological findings. If such a mechanism were to apply, then it would be
If the radiolucency does prove to be intraosseous, then a expected that the cavities might grow over time. This has
bone lesion is the likely diagnosis. A central giant cell been shown by Friedrich et  al. (2020), who reported and
lesion is one of the most commonly encountered non-­ illustrated four cases that gradually increased in size over
odontogenic radiolucent lesions of the jaws, but even these periods ranging from 1 to 12 years. These authors also pro-
are rare at this site (Chrcanovic et al. 2018). Other possibili- posed that very early lesions might manifest only as depres-
ties include tumour metastases, ossifying fibroma, osteoma, sions of the lingual cortex that would not be visible on
or osteoblastoma, but all are uncommon and some will radiological examination. This is supported by direct exam-
show a mixed radiolucent/radiopaque lesion. Note that the ination of dried mandibles from museum or archaeological
simple bone cyst arises above the ID canal and should not collections that show a higher incidence of bone defects
be confused with a posteriorly located Stafne bone cavity. than seen in clinical cases detected on radiology (Harvey
Anterior bone cavities are more problematic because they and Noble  1968; Philipsen et  al.  2002). It has also been
arise in the tooth-­bearing areas (above the mylohyoid mus- shown that larger lesions are seen in older individuals
cle) and occasionally may be located at the apex of a tooth. (Oikarinen and Julku 1974).
For anterior radiolucencies, therefore, a provisional diagno- Although pressure resorption is regarded as the most
sis of an odontogenic cyst is most likely to be the first likely cause, there have been no explanations as to how this
­consideration and a final diagnosis will depend on further might happen. Some have proposed that it may be associ-
imaging, but will often necessitate surgical intervention. ated with vascular changes resulting in increased pressure
from the facial artery that results in cavitation into which
salivary tissue might proliferate (Lello and Makek  1985).
Pathogenesis
There is very little evidence for this, however, and it does
Early papers suggested that the bone defect was congenital not explain cavities at sites away from vascular areas.
and of developmental origin, caused by entrapment or pro- The ‘glandular hypothesis’ is therefore the most widely
trusion of salivary tissue into the lingual aspect of the man- accepted. Philipsen et al. (2002) suggested that inflamma-
dible during ossification (Fordyce  1956). However, this tion and fibrosis in a salivary gland might increase the
­Focal Osteoporotic Bone Marrow Defec  285

pressure on the mandibular cortex. This theory also seems made by careful radiological examination alone (Schneider
unlikely. If it were the case that an altered texture of the et al. 2014; see ‘Radiological Features’) and surgical inter-
gland was necessary, then it might be expected that bone vention is rarely needed. Although most cavities remain
cavities would be associated with salivary gland tumours, static, progression has been reported, especially in younger
and might be more commonly encountered in the ramus individuals (Friedrich et  al.  2020). Periodic radiological
adjacent to the parotid gland, where tumours are most follow-­up may be advisable until it can be confirmed that
common. Nevertheless, local resorption over time is still the defect is static.
the most accepted theory and is given credence by the
observation that the surface of the bone in the depth of the
defects is irregular and shows pitting consistent with osteo- ­Focal Osteoporotic Bone
clastic activity (Harvey and Noble 1968; Mann 1992). Marrow Defect

Histopathology The focal osteoporotic bone marrow defect is a cystic radi-


olucency associated with a focal proliferation or hyper-
On gross examination during surgical exploration, the plasia of normal haematopoietic or fatty marrow. It is
cavities contain soft tissue or may appear empty if the con- usually regarded as an anatomical anomaly that is an
tents are displaced by instrumentation. In either case, the incidental finding and does not require treatment.
cavities are usually approached from a buccal aspect and However, it is important in the differential diagnosis of
the surgeon will find that there is no lingual cortex and cystic radiolucencies of the jaws, and a final diagnosis can
they may have inadvertently entered the submandibu- only be made on histological examination of the contents.
lar space. The defect was first reported by Cahn in 1954 and has
Histological examination of the contents does not become well recognised. Most reports are single case
­provide any diagnostic information, but may be helpful to reports, but there have been a number of early case series
exclude another cause for the cystic cavity if radiological of about 20 cases (Lipani et al. 1982; Makek and Lello 1986;
examination has not been diagnostic. Schneider et  al.  1988). The largest series, however, was
Many early reports have reported on the contents of the reported by Barker et al. (1974), who recorded 197 cases.
cavities and have shown that the vast majority contain lob- Reichart and Philipsen (2004) reviewed 277 cases and
ules of normal salivary gland tissue, although occasionally Shankland and Bouquot (2004) added a further 100 new
only fibrous tissue or fat has been found (Philipsen cases. Table 17.3 summarises data from these three larger
et al. 2002). Buchner et al. (1991) reviewed 24 anterior lin- studies.
gual cases and found salivary tissue in 21 (87.5%), fat or
fibrous tissue in 2, and 1 case that was empty. The salivary
Clinical Features
tissue is submandibular gland in posterior cavities and sub-
lingual gland in anterior cavities. Parotid gland has been About 90% of the defects are found in the mandible, with
reported in the ramus (Barker 1988). most (80%) in the molar region. Between about 70%
(Shankland and Bouquot 2004) and 85% (Lipani et al. 1982;
Schneider et  al.  1988) have been located in edentulous
Treatment
areas at the site of a previous tooth extraction. They are
The most appropriate management of the Stafne bone cav- more frequent in females (Table  17.3) and are found in
ity is to establish a correct diagnosis, after which no further adults with an average age of 40–50 years. Over 70% are
treatment is needed. In most cases the diagnosis can be encountered in the fourth to sixth decades.

Table 17.3  Age, sex, and site distribution of focal osteoporotic bone marrow defect from the two largest case series and the review
of Reichart and Philipsen (2004).

Reference n Age mean (range) Female (%) Mandible (%)

Barker et al. (1974) 197 41.8 (7-73) 72.9 90.9


Shankland and Bouquot (2004) 100 50.1 (NR) 77.0 65.0
Reichart and Philipsen (2004) 277 40.2 (25–71) 76.9 90.5

NR, not recorded.


286 Pseudocysts of the Jaws: Simple Bone Cyst and Stafne Bone Cavity

The vast majority of cases have been reported as symp- Shankland and Bouquot (2004) discussed the possible
tomless and have been diagnosed after a chance finding pathogenic mechanisms and proposed that the defect may
during radiological examination for other reasons. represent an early manifestation of osteoporosis and bone
However, Shankland and Bouquot (2004) noted that 57% of marrow oedema, associated with ischaemia and a local
their 100 patients reported tenderness on palpation, and malfunction of blood flow. They suggest that local trauma
Makek and Lello (1986) found that 25% of their patients may be one factor that could initiate these changes.
had experienced pain.
Histopathology
Radiological Features When explored surgically, the cavities contain soft tissue
The focal osteoporotic bone marrow defect is a round or with small amounts of residual bone. In most cases histo-
oval cystic radiolucency that may be well demarcated, logical examination shows normal haematopoietic marrow
but only infrequently shows evidence of cortication. The with varying amounts of fat. Barker et al. (1974) examined
degree of radiolucency is variable and some cases may the histology of 181 defects and found that normal marrow
be difficult to visualise on a standard plain radiograph. occupied more than half the contents in 61.3% of cases. In
Cases in the mandible are always located in the alveolar 38 (21.0%) cases the contents were almost entirely fat.
bone above the ID canal. Schneider et al. (1988) exam- Shankland and Bouquot (2004) found that all cases con-
ined 20 cases and found corticated margins in only 2 tained viable trabeculae of bone, with marrow that was
(10.0%). They also found that in all cases, the normal essentially normal, although 79% was fatty and only 21%
bony trabeculae were still visible and in 55% there were contained red (haematopoietic) elements. They also
areas of radiopacity. Barker et  al. (1974) reported that showed that 55% of cases showed evidence of inflamma-
most cases had ill-­defined borders and occasionally had tion and 51% had small focal areas of ischaemia or necrosis.
a ‘moth-­eaten’ appearance. They illustrated a number of
cases with variable shapes and borders, but most also
Cavitational Osteonecrosis
showed evidence of normal ­trabeculae. Most cases are
between 10 and 20 mm in maximum diameter, but Two research groups have reported bone cavities of the
defects up to 60 mm have been reported (Schneider jaws, similar to osteoporotic bone marrow defects, that are
et  al.  1988). Shankland and Bouquot (2004) measured associated with a type of aseptic osteonecrosis. Bouquot
100 cases and found an average diameter of 16.2 mm et al. (1992) described bone cavities associated with neural-
(range 5–35 mm). gia and used the term neuralgia inducing cavitational oste-
onecrosis (NICO), and Lechner et  al. (2017) described
Radiological Differential Diagnosis similar lesions that they called fatty degenerative osteone-
The radiological features are so variable that the bone mar- crosis in the jawbone (FDOJ).
row defect may resemble any other type of radiolucency of Bouquot et  al. (1992) reported 135 patients who pre-
the jaws. The only reported characteristic features are the sented with bone cavities (224  in total) associated with
association with a previous tooth extraction, and that the facial neuralgia. In their detailed analysis they found that
normal trabecular structure of the cancellous bone might the lesions were composed of fibrosed bone marrow, with
still be visible within the radiolucency. Nevertheless, in evidence of bone or marrow necrosis and inflammation.
most cases surgical exploration with histological examina- The average age of the patients was 49.1 years (range
tion of the contents of the cavity is necessary to exclude 24–84 years) and 66.6% were female. Cases were distrib-
known pathological entities and to confirm the diagnosis. uted equally between the mandible and maxilla, but the
most common sites were the mandibular molar region
(32.3% of cases) or the maxillary canine region (18.5%). The
Pathogenesis
authors regarded NICO as a localised form of chronic
The cause of these defects is not known, but the most com- osteomyelitis of the jaws, and believed that it is a form of
pelling explanation is of an accumulation of hyperplastic ischaemic osteonecrosis similar to that associated with
marrow in an area of bone healing. This would account for bone pain in other parts of the skeleton, especially the
the fact that the defects are often found at a site of a previ- femur or humerus (Bouquot and McMahon  2003).
ous tooth extraction. Other theories have suggested an Although Bouquot et al. (1992) introduced the term NICO,
association with anaemias, where there is an increased they were not the first to note an association between facial
demand for haematopoiesis, or persistence of haematopoi- neuralgia and bone cavities. In their paper, they recorded
etic marrow into adulthood. almost 2000 cases that had been reported between 1976
­Aneurysmal Bone Cys  287

and 1991, and credited Ratner et  al. (1979) with the first of long bones, especially the femur, tibia, humerus, and verte-
definitive description. brae. Only about 1.5% arise in the jaw bones.
More recently, members of a German research group Aneurysmal bone cysts, including jaw lesions, have been
have described similar lesions that they have called FDOJ divided into primary and secondary types, where the sec-
(Lechner et  al.  2017,  2019,  2020). They first described 24 ondary type arises in association with other lesions, most
patients with bone cavities associated with systemic immu- often fibro-­osseous or giant cell lesions (Arora et al. 2014).
nological disorders or facial pain (Lechner et al. 2017). In In the previous edition of this book and in the 2005 WHO
this and a later study, they demonstrated that the bone classification of head and neck tumours (Barnes
cavities showed chronic inflammation and fatty necrosis et al. 2005), the aneurysmal bone cyst was defined as pri-
and were almost always aseptic and associated with mary or secondary and it was thought that most lesions
increased expression of the pro-­inflammatory chemokine were reactive in nature. It is now known that this concept
CCL5 (RANTES; Lechner et  al.  2019). The lesions were is wrong and outdated.
similar to the cavities described by Bouquot et  al. (1992) Aneurysmal bone cyst has been shown to be a neoplasm
and the authors suggested that NICO and FDOJ are the associated with rearrangements of the USP6 gene with a
same lesion. In a later paper, Lechner et al. (2020) stated wide range of fusion partners (Oliveira et  al.  2004,  2005;
that FDOJ is also synonymous with osteoporotic bone mar- Oliveira and Chou  2014; Warren et  al.  2017; Šekoranja
row defect. Since the authors have provided few details et  al.  2020). Specifically, primary aneurysmal bone cysts
about the clinical context of this lesion, further research is are neoplasms that show USP6 gene rearrangements, but
needed to establish the relationship between osteoporotic so-­called secondary aneurysmal bone cysts do not show
bone marrow defects and these reported osteonecrotic USP6 rearrangements and should be regarded as secondary
cavities. changes in pre-­existing lesions (Oliveira et al. 2004; Arora
In the absence of further research, these lesions have et  al.  2014; Flanagan and Speight  2014). True neoplastic
never been widely accepted and, shamefully, the original aneurysmal bone cyst does occur in the jaw bones, but very
authors who described NICO have been the subject of ridi- few cases have yet been reported (Oliveira et  al.  2004;
cule, personal abuse, and litigation (Bouquot and Brooks et al. 2019; McMullen et al. 2019; Cleven et al. 2020).
McMahon 2003). A number of authors have reviewed the In the fourth (2017) and fifth (2022) editions of the WHO
controversy around NICO (Zuniga  2000; Sciubba  2009) classification of head and neck tumours, the aneurysmal
and have suggested that further research and clinical trials bone cyst is defined as a cystic or multicystic, osteolytic
are needed. neoplasm associated with blood-­filled spaces lined by
fibrous septae that contain osteoclast-­type giant cells (El-­
Naggar et al. 2017; WHO 2022a; Jordan and Koutlas 2022).
­Aneurysmal Bone Cyst Furthermore, the classification does not encompass sec-
ondary changes in other lesions and the term ‘secondary
In previous editions of this book and in many textbooks of aneurysmal bone cyst’ is no longer acceptable and should
oral and maxillofacial pathology, the aneurysmal bone cyst not be used (WHO 2022a,b). Changes resembling aneurys-
has been included in the classification of jaw cysts, usually mal bone cyst, seen in a variety of jaw lesions, represent
categorised as a non-­odontogenic cyst or pseudocyst. It has haemorrhagic cystic change and should be described sim-
been described as a multicystic lesion composed of variably ply as cystic degeneration or cystic change within the
sized blood-­filled spaces surrounded by vascular fibrous tis- lesion. Any reference to these features as ‘aneurysmal bone
sue, rich in osteoclast-­type giant cells. Aneurysmal bone cysts cyst-­like’ is probably best avoided, since it may cause
can arise at any site, but have a predilection for the metaphysis ­confusion with rare but truly neoplastic lesions.
288

18

Developmental Cysts

CHAPTER MENU
Classification and Terminology, 289
Dermoid and Epidermoid Cysts, 291
●● Clinical Features,  291
–– Frequency, 292
–– Age, 292
–– Sex, 292
–– Site, 292
–– Clinical Presentation,  293
●● Radiological Features,  293
●● Pathogenesis, 294
●● Histopathology, 294
●● Treatment, 295
Developmental Cysts of Foregut Origin, 295
●● Classification and Terminology,  295
Heterotopic Gastrointestinal Cyst, 296
●● Clinical Features,  296
–– Frequency, 296
–– Age, 297
–– Sex, 297
–– Site, 297
–– Clinical Presentation,  298
●● Histopathology, 298
●● Treatment, 298
Bronchogenic Cyst, 298
●●Clinical Features,  299
●●Histopathology, 299
●● Treatment, 300
Branchial Cleft Anomalies, 300
Branchial Cleft Cysts, 300
●● Clinical Features,  300
–– Frequency, 300
–– Age and Sex,  300
–– Site, 300
–– Clinical Presentation,  301
●● Histopathology, 301

●● Differential Diagnosis,  303


–– Differentiation of Branchial Cleft Cyst from Cystic Metastatic Carcinoma,  305
●● Treatment, 305

Thyroglossal Duct Cyst, 305

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
­Classification and Terminolog  289

●● Clinical Features,  305


–– Frequency, 305
–– Age, 305
–– Sex, 306
–– Site, 306
–– Clinical Presentation,  306
●● Histopathology, 307
–– Differential Diagnosis,  307
●● Treatment, 308
Nasopharyngeal Cysts, 308
Thymic Cyst, 308

The development of the head and neck commences at especially in adult patients when they must be distin-
about the third week of intrauterine life, when the three guished clinically and histologically from metastatic carci-
germ cell layers form the trilaminar embryonic disk that noma. In adults, most neck swellings are caused by lymph
subsequently undergoes a complex process of three-­ node enlargement of one sort or another, with neoplastic
dimensional folding to form the developed embryo disease being more likely with increasing age. Of cystic
(Nanci 2017). In this way, the three germ cell layers become neck lesions metastatic carcinoma is, overall, probably the
orientated to form the internal and external structures of most common lesion encountered in adults. The relative
the fully developed human. A crucial process in the head frequency and differential diagnosis of cysts of the neck
and neck region is the formation of the branchial or phar- have been reviewed by Luna and Pfaltz (2009).
yngeal arches, which give rise to the facial processes that
eventually migrate and fuse to form the eyes, ears, jaws and
mouth, and associated structures. Aberrations in these pro- ­Classification and Terminology
cesses give rise to many facial developmental anomalies.
Developmental cysts of the head and neck are mostly The literature and terminology relating to the develop-
congenital and are present at birth, although some grow mental cysts of the oral and maxillofacial regions are com-
slowly and remain occult for many years, with clinical plex and reflect their rarity and our lack of understanding
presentation in childhood or adolescence, and occasionally about their pathogenesis. A particular area of confusion is
in later adulthood. The majority of these cysts are formed the terminology relating to dermoid cysts, epidermoid
due to entrapment of ectoderm or endoderm during fusion cysts, and teratomas. Much of the head and neck literature
of the facial processes, or incomplete obliteration of the uses ‘dermoid’ as a generic term to refer to developmental
branchial clefts or pouches. All developmental cysts are cysts in the floor of the mouth that are lined by stratified
rare, but the most commonly encountered cyst, constitut- squamous epithelium. The ‘sublingual dermoid’ is often
ing about 50%, is the thyroglossal duct cyst, followed by divided into three variants, as originally suggested by
branchial cleft cysts (about 20%). Other lesions are very Meyer (1955), who had recognised the confusion around
rarely encountered. these lesions and noted that many different names and
The classification of developmental cysts of the head and classifications had been used. He suggested that, as a
neck is difficult and complex (see next section), but in the group, the developmental cysts in the floor of the mouth
maxillofacial regions they cause clinical problems in two should be called ‘dysontogenic cysts’, which literally
main areas related to their site of origin. Many arise in the means cysts formed by a disorder of development. He
midline and often present clinically as swellings in the divided them into three types based on the histological
upper neck, in the sublingual space, or in the tongue. In features: epidermoid cyst, lined by stratified squamous epi-
neonates these cysts may be associated with obstruction of thelium with no evidence of appendages; dermoid cyst,
the airway and require urgent attention, while in children lined by epithelium with skin appendages including hair
and adolescents they must be considered in the differential follicles and sebaceous or sweat glands; and teratoid cyst,
diagnosis of swellings affecting the floor of the mouth lined by stratified squamous epithelium with appendages,
and tongue. but also with connective tissue elements and respiratory-­
The second area of difficulty is cysts presenting in the or gastrointestinal-­type epithelium.
lateral neck (branchial cleft cysts), which are usually Although this terminology is still widely used, many
benign but can cause considerable diagnostic problems, papers still refer to the sublingual dermoid cyst as
290 Developmental Cysts

encompassing cystic lesions with any of these features. regarded as teratomas. Most reports of teratoid cysts illus-
However, the teratoid cyst is often also interpreted to mean trate cysts lined by a mixture of stratified squamous epithe-
teratoma. Although Meyer (1955) defined the teratoid cyst lium and respiratory or gastrointestinal epithelium.
as having elements from all three germ cell layers, the case Because these are of ectodermal and endodermal origin,
he illustrated was lined by skin-­type and respiratory-­type respectively, it has often been stated that these cysts show
epithelium, but the connective tissue element was normal tissue derived from two germ cell layers and thus represent
for the site and not heterotopic. A true teratoma is defined a type of mature teratoma. For this reason, the terms tera-
as a germ cell neoplasm (Smirniotopoulos and toid and teratoma have on occasion been used synony-
Chiechi 1995), and the vast majority (about 90%) arise in mously. Similarly, the dermoid cyst, which shows skin-­type
the gonads or sacral regions and show lines of differentia- epithelium with appendages at a heterotopic site (e.g. the
tion representing all three germ cell layers (ectoderm, mes- tongue), has been called a type of mature teratoma. A num-
oderm, and endoderm). They are often multicystic and are ber of reports have also shown that a dermoid cyst may be
lined by epithelium of ectodermal (skin-­type) and endo- lined by more than one type of epithelium. Furthermore,
dermal (respiratory-­ or gastrointestinal-­type) origin, but foregut cysts, which take their origin from foregut epithe-
also show mesodermal structures, including cartilage, lium of endodermal origin, may also show a mixed epithe-
bone, neural elements, and sometimes fully formed teeth. lial lining. For these reasons, some regard all these cyst
Although they are often well formed, the connective tissue types as variants of mature teratoma and use ‘dermoid’ as a
elements are heterotopic in that they are located away generic term to encompass all cysts in the sublingual region.
from their normal anatomical site. Teratomas can be A further cause of confusion is the epidermoid cyst.
divided into mature types that are usually benign, and This cyst is lined by stratified squamous epithelium with-
immature malignant types (Smirniotopoulos and out appendages and in the context of the oral cavity it is
Chiechi 1995). clinically indistinguishable from a dermoid cyst. Both are
True teratomas do arise in the head and neck regarded as developmental cysts, but the epidermoid cyst
(Smirniotopoulos and Chiechi  1995; Lopes et  al.  2005; is histologically similar to the common type of cutaneous
Shah et al. 2009; Tonni et al. 2010; Morlino et al. 2015) and cyst that is often called the ‘sebaceous cyst’. Clinicians fre-
are found within the cranium, oropharynx, nasopharynx, quently use ‘sebaceous’ as a descriptive name for any type
or in the midline of the neck. Most are diagnosed in utero of simple skin cyst, but the term should not be used by
or at birth and the cervical region is most commonly pathologists or as a diagnosis. There are many types of
affected. Oropharyngeal and jaw lesions are the second cutaneous (adnexal) cyst, the most common of which is
most common and are often referred to as ‘epignathus’. an inclusion cyst that arises in the superficial or infun-
Morlino et al. (2015) recorded reports of only 48 oropharyn- dibular part of the hair follicle and should be called the
geal (including the nasopharynx) cases between 1931 and epidermal cyst, epidermal inclusion cyst, or infundibular
2013; 32 (66.6%) of the cases were diagnosed in utero or at cyst (Kaya and Saurat  2018). Other common types of
birth. Lopes et  al. (2005) reported a mass in the buccal adnexal cyst are the tricholemmal cyst (arising from the
mucosa in a 9-­year-­old girl, which comprised stratified outer root sheaf of the hair follicle) and the true seba-
squamous epithelium with appendages, respiratory epithe- ceous cyst, which is properly called the steatocystoma.
lium, and heterotopic mesodermal elements, including Confusion arises because the common epidermal cyst is
bone and neuroglial tissue. In a review of the literature the often called epidermoid cyst, and a number of early pub-
authors found only 21 previously recorded cases within the lications have included the ­common cutaneous acquired
oral cavity; 13 (61.9%) arose in the tongue and the remain- epidermal cyst as a developmental cyst of the oral regions
der (38.1%) were found in the palate, although two showed and have occasionally ­categorised it alongside der-
pharyngeal extension. All cases were diagnosed in utero or moid cysts.
at birth and all were located in the tongue or palate and in Branchial cleft cysts are more clearly defined, since they
the midline. Their own case was the only case reported take their origin from the branchial clefts and are always in
arising in childhood and was the first in the buccal mucosa. the lateral neck. Although histologically they may be simi-
Subsequently a second case arising in the cheek of a neo- lar to other developmental cysts, their distinctive clinical
nate has been reported (Singh et al. 2012). features mean that they are unlikely to be confused with
From this brief discussion it can be seen that true terato- other cyst types.
mas of the maxillofacial regions do occur, but they are rare In this chapter we make a clear distinction between the
and are very unlikely to be encountered after birth. Teratoid developmental cysts that arise most commonly in the
cysts, on the other hand, have often been reported as part tongue or floor of the mouth and the common epidermal
of the spectrum of ‘dermoid’ cysts, but these should not be inclusion cysts of the skin. Although these commonly arise
­Dermoid and Epidermoid Cyst  291

on the skin of the face, they are acquired and are not con- ­Dermoid and Epidermoid Cysts
sidered here. Readers are referred to reviews (e.g. Kaya and
Saurat  2018) or to standard dermatopathology texts for Dermoid and epidermoid cysts are developmental cysts
details of these common skin lesions. that can occur at any site in the body. The majority are con-
We also make a distinction between dermoid and epider- genital and are found in the gonads or the sacral region. In
moid cysts, and teratomas. We regard teratomas as germ one of the largest studies, Taylor et al. (1966) reviewed 514
cell neoplasms that occasionally may arise in the head and cases from the Mayo Clinic and found 184 (35.8%) in the
neck or oral cavity, but are distinct from the developmental head and neck region, of which only 35 (6.8% of the total)
cysts discussed here. Teratoid cysts are poorly defined, but were located in the floor of the mouth. Most head and neck
most reports describe a cyst lined by two types of epithe- lesions are found at the lateral aspect of the eyebrow at the
lium, and we would regard these as histological variants of site of fusion of the zygomatofrontal suture (Taylor
dermoid cysts or foregut cysts. Although occasionally these et  al.  1966; Pollard et  al.  1976; Smirniotopoulos and
may be histologically similar to a mature teratoma, they are Chiechi 1995). Dermoid and epidermoid cysts are develop-
distinctive in their clinical presentation, and teratoma mental cysts arising from entrapped ectodermal tissue and
should only be diagnosed if there is clear evidence of tis- are lined by stratified squamous epithelium with (dermoid
sues derived from all three germ cell layers, including het- cyst) or without (epidermoid cyst) skin appendages. Apart
erotopic connective tissue elements. True teratomas in the from these histological differences, the cysts are indistin-
head and neck usually include well-­formed cartilage, bone, guishable and are usually considered together (Box 18.1).
and neuroectodermal or neuroglial tissues (Smirniotopoulos Because they represent a developmental disorder they are
and Chiechi 1995; Lopes et al. 2005; Shah et al. 2009; Tonni sometimes referred to as dysontogenic cysts.
et al. 2010).
From the forgoing discussion it is clear that there is great
Clinical Features
overlap in the clinical and histological features of the
developmental cysts and a simple classification is not pos- Most reports of these cysts are single case reports or small
sible. Here we distinguish the cysts primarily by their clini- case series. King et  al. (1994) reviewed the literature and
cal characteristics and histological features. Boxes 18.1–18.4 found 195 fully documented floor-­of-­mouth cases. More
summarise the key features of the most commonly encoun- recently, MacNeil and Moxham (2010) found 198 floor-­of-­
tered lesions. mouth cysts, but only included cases where there was

Box 18.1  Dermoid and Epidermoid Cysts: Key Features

Significance and
Pathogenesis Clinical features Histopathology differential diagnosis

Epidermoid cyst Entrapment of Third most common (12%) Lined by stratified Sublingual swellings
ectoderm during developmental cyst in the squamous epithelium may cause airway
fusion of the man- head and neck without appendages obstruction and
Dermoid cyst dibular processes Only 0.2% of oral lesions Lined by stratified difficulties with
arising from the first
Mean age about 35 years, squamous epithelium feeding and speech
branchial arch with peak in second to with appendages, Must be differenti-
fourth decades including sebaceous ated from other
Equal sex distribution glands, sweat glands, or lesions that may
Located in the floor of hair follicles arise in the sublin-
the mouth gual/tongue region,
Teratoid cyst A variant of dermoid Lined by stratified
Cause sublingual (50%) or including ranula,
and epidermoid cyst, squamous and
submental (45%) swelling, foregut cysts,
but with concurrent respiratory or gastroin-
occasionally in anterior thyroglossal duct
entrapment of testinal epithelium
tongue or submandibu- cyst, and salivary
endodermal elements.
lar region gland tumours
Some may be fore-
gut cysts
292 Developmental Cysts

information about treatment. Santos et  al. (2020) and Sex


Misch et al. (2020) added 27 and 12 new cases, respectively, Overall, dermoid and epidermoid cysts seem to have a
to the literature. Lesions have also been reported in the slight predilection for males, but there are varied reports.
tongue. Campbell and Walker (2010) found 12 reports of King et al. (1994) recorded 61% of lesions in males and 39%
dermoid cyst in the tongue, and Ueno et al. (2018) reviewed in females (n  =  124), and MacNeil and Moxham (2010)
13 cases of epidermoid cyst. found 58.0% in males (n = 188). Longo et al. (2003) reported
that 12 (75.0%) of their 16 cases were in males. In their
Frequency series of 27 cases, however, Santos et  al. (2020) found 18
Dermoid and epidermoid cysts are the third most common (66.6%) in females. They also noted a higher female predi-
congenital cyst affecting the head and neck region, consti- lection for dermoid cysts, where 10 of 14 (71.4%) cases
tuting about 12% of the total (Al-­Khateeb and Al Zoubi arose in females. Among paediatric cases, Misch et  al.
2007; Brucoli et  al.  2020). Thyroglossal duct cyst and (2020) also showed a female predilection of 66.6% (8 of 12
branchial cleft cyst are the first and second most common, cases). For cases in the tongue, Campbell and Walker
respectively. (2010) found that 58.3% were female, while Ueno et  al.
Nevertheless, they are still relatively rare and represent (2018) reported that 61.5% were male.
less than 0.2% of maxillofacial lesions. Santos et al. (2020)
found 27 (0.17%) cases among a total of 15 387 maxillofa- Site
cial biopsies. Of these, 14 (0.09%) were dermoid cysts and For oral lesions, the midline of the floor of the mouth is the
13 (0.08%) were epidermoid. In the Sheffield series, Jones most common location (Figure 18.1). At this site they may
and Franklin (2006a,b) found only 27 (0.06%) dermoid cause an intraoral sublingual swelling or an extraoral sub-
cysts reported from a total of more than 48 000 head and mental swelling or both. Meyer (1955) noted that floor-­of-­
neck specimens over a 30-­year period. The lesion was mouth lesions are always located above (deep or lingual to)
more common in children, with 8 (0.2%) cases identified the mylohyoid muscle, but may be found above or below
among 4406 specimens (Jones and Franklin 2006b) com- the geniohyoid muscle. If above the geniohyoid, the cyst is
pared with only 19 (0.04%) cases in adults (n  =  44 007; located between the genioglossus and geniohyoid muscles
Jones and Franklin 2006a). In a review of 10 311 oral biop- and presents primarily as a sublingual swelling in the floor
sies, Nonaka et  al. (2011) found 12 (0.1%) epidermoid of the mouth. Conversely, if the cyst is below the geniohy-
cysts and 6 (0.05%) dermoid cysts. For comparison, these oid, it is located between the geniohyoid and mylohyoid
frequencies are similar to nasolabial cyst and simple muscles and presents primarily as an extraoral swelling in
bone cyst. the submental region.
King et  al. (1994) reviewed 195 case reports and noted
Age that some cases might penetrate the mylohyoid muscle and
Although these cysts are congenital, they grow slowly and occupy the sublingual and submandibular spaces concur-
may enlarge over years or decades and not become clini- rently. They found that 52% of cases were recorded as
cally apparent until late adolescence or adulthood ­sublingual, 26% as submental, and 6% as submandibular.
(Smirniotopoulos and Chiechi  1995). Most dermoid and The remaining cases (16%) occupied more than one space.
epidermoid cysts occur in the second to fourth decades. In their similar review of 198 case reports, MacNeil and
Santos et al. (2020) found that dermoid cysts presented at Moxham (2010) showed that 52.5% were reported as sub-
an average age of 37.2 years (n = 14; range 1–81 years) and lingual, 40.5% as submental, 18.2% as submandibular, and
epidermoid cysts at 38.2 years (n  =  13; range 11.6% as mixed.
11 months–82 years). Longo et al. (2003) reported 16 cases Almost all cases have been reported in the midline of the
with an average age of 27.8 years (range 5–51 years). In floor of the mouth or submental region, but occasionally a
their review of 198 cases, MacNeil and Moxham (2010) cyst may have a lateral location. Teszler et  al. (2007)
found that 66.0% arose in adults over the age of 16, 20% reported two cases that presented as submandibular swell-
were in children, and 14% were in neonates (<1 year). ings in the lateral aspect of the upper neck. One case was
There is some evidence that lesions in the body of the located in the lateral floor of the mouth deep to the mylo-
tongue may present at an earlier age than floor-­of-­mouth hyoid muscle in the sublingual space, but close to the deep
lesions. Campbell and Walker (2010) reviewed 12  lingual lobe of the submandibular gland. The second case was
dermoid cysts and found an average age of 8.8 years (range located superficial to the mylohyoid muscle in the subman-
2 months–17 years), and Ueno et al. (2018) reported 13 lin- dibular space. Lateral cysts appear to be rare and probably
gual epidermoid cysts with an average age of 18.0 years represent less than 5% of the total reported. Teszler et al.
(range 4 months-­45 years). (2007) noted only 12 reported cases, but MacNeil and
­Dermoid and Epidermoid Cyst  293

Moxham (2010) suggest that there have been about 36 cells were focal and few in number and the authors inter-
cases reported. However, most authors consider that later- preted it as an odontogenic cyst with sebaceous metaplasia.
ally located cysts are midline cysts that have become dis- The second cyst was unusually large and had prominent
placed and bulge over the posterior aspect of the mylohyoid sebaceous glands, as well as other dermal appendages
muscle to enter the submandibular space. It is also possible including hair follicles and sweat glands. This cyst was
that some lesions reported in the upper neck are diagnosed as an intraosseous dermoid cyst.
branchial cysts.
As suggested above, the vast majority of these develop- Clinical Presentation
mental (dysontogenic) cysts arise in the sublingual region, The most common presentation is of an intraoral sublingual
but cases have been reported at other sites in the oral cavity. swelling that lifts the tongue (Figure 18.1) and may lead to
Most of these have been reported in the body of the tongue difficulty in speaking, eating, breathing, or closing the
(Campbell and Walker  2010; Ueno et  al.  2018). They are mouth. Deeper lesions between the geniohyoid and mylohy-
found in the mobile anterior two-­thirds of the tongue and oid muscles produce a submental swelling in the neck, giv-
present as firm swellings about 15 mm in diameter, ing the patient a ‘double-­chin’ appearance. The swelling
although cases up to 70 mm have been reported (Campbell may feel doughy or fluctuant. Lesions presenting at birth can
and Walker 2010). Ueno et al. (2018) reviewed 13 cases of obstruct the airway and be life threatening. However, this is
epidermoid cyst in the body of the tongue, but noted that rare, and most cysts grow slowly and present in childhood or
some cases might be acquired as a result of trauma and adolescence. Santos et  al. (2020) found that patients had
implantation of surface epithelium into deeper tissues. been aware of a lesion for up to 25 years, but with an average
This might apply especially to cysts on the ventral tongue duration of 53.3 months (range 1–300) for dermoid cysts and
in older individuals. 45.7 months (range 2–120) for epidermoid cysts.
Santos et al. (2020) presented 27 new cases and showed As noted previously (see ‘Site’), the cyst may occasionally
that only 10 (37.0%) were located in the floor of the mouth. present as a swelling in the anterior, mobile part of the
These were 6 dermoid cysts and 4 epidermoid cysts. They tongue or in the submandibular region. Lesions at other
also found 6 dermoid cysts in the lip and 2 on the gingiva or sites, including the buccal mucosa, palate, and gingiva,
alveolus. Of the 9 epidermoid cysts, 2 were found on the have been reported but are rare.
gingivae, 3 on the plate, 2 in the tongue, and 2 in the buc-
cal mucosa.
Radiological Features
There have been a number of reports of intraosseous der-
moid cysts in the mandible. Issa and Davies (1971) recorded Radiology plays little role in diagnosis, but imaging is
an exceptionally rare example of a dermoid cyst that important for surgical planning and to confirm the cystic
occurred in the coronoid region of the mandible in a nature of the lesion. Ultrasound can be used to identify the
26-­year-­old woman, and Craig et  al. (1980) reported an lesions as cystic, but they are best visualised using mag-
intraosseous dermoid cyst in the midline of the mandible netic resonance imaging (MRI), which shows the extent of
in a 28-­year-­old man. However, the diagnosis was made on the lesion and the relationships to mylohyoid and
the basis of finding sebaceous glands associated with a lin-
ing of ortho-­ and parakeratinised stratified squamous epi-
thelium. It is possible that these cysts actually represented
sebaceous metaplasia in an odontogenic cyst. Sebaceous
glands have been found in occasional odontogenic kerato-
cysts (Brannon, 1977; discussed in Chapter 7) and Chi et al.
(2007) reported 5 cases of orthokeratinised odontogenic
cyst with focal areas of sebaceous glands (discussed in
Chapter 12). Although these might be regarded as dermoid
cysts, Chi et al. (2007) found that all 5 showed clinical and
radiological features typical for an odontogenic cyst, and
they strongly argued that they should be regarded as
orthokeratinised odontogenic cysts with focal sebaceous
differentiation. Vuhahula et al. (1993), however, suggested
that a true dermoid cyst can be found within the bone.
They examined 12 orthokeratinised jaw cysts and found 2 Figure 18.1  Dermoid cyst. The cyst fills the floor of the mouth
that contained sebaceous cells. In the first the sebaceous and has raised the tongue.
294 Developmental Cysts

geniohyoid muscles and adjacent structures. The cyst Histopathology


appears as a well-­demarcated, round or oval, hypointense
When removed intact, the cysts appear as firm, rounded
(lucent) lesion. T2-­weighted images show a markedly
masses. On dissection they often have a thick wall and con-
hyperintense (white) lesion, suggesting fluid contents. The
tain keratinaceous material similar to epidermal cysts of
cysts range in size from 5 to 70 mm, but the overall average
the skin. Dermoid cysts contain appendages and may show
is about 30 mm (Santos et al. 2020).
areas of thickening.
Both dermoid and epidermoid cysts are lined by kerati-
nised stratified squamous epithelium resembling epider-
Pathogenesis mis, but are distinguished by the fact that dermoid cysts
The origin of dermoid and epidermoid cysts, like other have skin appendages.
developmental cysts, is controversial. They are lined by Dermoid cysts are lined by stratified squamous epithe-
epithelium of ectodermal origin, suggesting that they arise lium, usually without rete pegs, but with prominent
due to abnormal sequestration of epithelium during fusion orthokeratinisation. By definition, the dermoid cyst con-
of the facial processes (Smirniotopoulos and Chiechi 1995). tains skin appendages and sebaceous glands, sweat glands,
With regard to oral lesions, these are found in the anterior or hair follicles may be seen (Figures 18.2 and 18.3). Santos
aspect of the oral cavity in the floor of the mouth or ante- et al. (2020) described the histopathological features of 14
rior tongue, consistent with entrapment of epithelium of dermoid cysts and found that 12 (85.7%) contained hair fol-
ectodermal origin during fusion of the mandibular pro- licles, 12 (85.7%) had sebaceous glands, and 4 (28.6%) had
cesses, which take their origin from the first branchial sweat glands.
arches (Adams et al. 2016; Nanci 2017). Epidermoid cysts are lined by flattened stratified squa-
A number of cysts have been described that are lined by mous epithelium without adnexal structures. Oral lesions
epithelium of ectodermal (skin-­type) and endodermal (res- are similar to epidermal cysts of the skin and may be filled
piratory-­ or gastrointestinal-­type) origin. These are often with lamellae of desquamated keratin.
referred to as teratoid cysts. As the primitive mouth (sto- Occasionally, cysts may be encountered that are lined by
modeum) develops, it is separated from the developing two types of epithelium. These are termed teratoid cysts
foregut by a bilaminar membrane (the buccopharyngeal and are the rarest of the developmental cysts of the oral
membrane) formed by apposed ectoderm and endoderm. cavity. These were originally described by Meyer (1955)
At about 32 days of intrauterine life, the membrane breaks and were defined as showing two types of epithelium and
down so that the oral cavity communicates directly with connective tissue elements. King et  al. (1994), in their
the foregut and there is probably a considerable amount of review of 195 case reports of ‘dermoid’ cysts of the floor of
intermingling of ectoderm and endoderm. It is possible
therefore that occasional dermoid or epidermoid cysts may
include heterotopic elements of endoderm derived from
the foregut, and this may explain why some cysts are lined
in part by respiratory, gastric, or intestinal epithelium. It is
these cysts that have been referred to as ‘teratoid’. Foregut
cysts, discussed later in this chapter, also take their origin
from heterotopic endodermal elements within the tongue.
Cysts similar to the developmental epidermoid cysts
under discussion may also be acquired and arise as a result
of inclusion or implantation of epithelium as a result of
trauma. Common cutaneous epidermal cysts arise as a
result of inclusion of epithelium following trauma, infec-
tion, or blocked hair follicles. It is possible that some epi-
dermoid cysts in the oral cavity may also be acquired as a
result of implantation of epithelium following trauma
(Smirniotopoulos and Chiechi 1995). Epidermal inclusion
cysts may arise on the lips, but Ueno et al. (2018) also sug-
gested that tongue lesions may be caused by trauma. They
presented a small epidermoid cyst on the ventral aspect of Figure 18.2  Low-­power view of a dermoid cyst. The cyst lies
just below the mucosa of the floor of the mouth (arrow) and is
the tongue, lateral to the midline, that they felt may have lined mostly by flattened stratified squamous epithelium. Skin
been acquired following trauma. appendages are noted at the deep aspect (see Figure 18.3).
­Developmental Cysts of Foregut Origi  295

Figure 18.3  High-­power of part of the cyst seen in Figure 18.2. The lining is orthokeratinised and well-­formed hair follicles and
sebaceous glands can be seen.

the mouth, found that 72.9% were diagnosed as dermoid not be possible to make a clear distinction between a dermoid
cysts, 22.2% as epidermoid cysts, and only 4.9% as teratoid. cyst with focal areas of another epithelial type and a benign
This suggests that cysts with multiple phenotypes are mature teratoma. It should be noted, however, that true tera-
rare. However, most cases reported as teratoid in the litera- tomas contain obvious heterotopic connective tissue ele-
ture have shown stratified squamous epithelium and ments and are diagnosed in utero or at birth.
another epithelium, but no evidence of heterotopic con- There are a number of reports of simultaneous occur-
nective tissue elements. Many cases show muscle or sali- rence of dermoid cysts in association with another type of
vary tissue in the wall, but these are normal for the site of developmental cyst. These appear to arise in the floor of
occurrence of the lesion. Crivelini et al. (2001) reported a the mouth as well as at other sites, including the pancreas,
cyst lined by gastrointestinal epithelium and typical der- spleen, and mediastinum. The entrapment during develop-
moid cyst epithelium, and in their report of 14 cases of der- ment of pluripotential cells may allow for different path-
moid cysts, Santos et al. (2020) found 2 (14.3%) cases with ways of differentiation at the same site, or multiple cysts
areas of respiratory epithelium and 1 (7.1%) with gut epi- may arise concurrently from entrapped epithelium from
thelium. Alsharif and Zhao (2009) reviewed 101 cysts in different sources. In the floor of the mouth, dermoid cysts
the floor of the mouth and found that 33  were dermoid have been described in association with gastrointestinal
cysts, 48 were epidermoid cysts, and 20 were described as cysts (Eppley et  al.  1985; Arcand et  al.  1988; Crivelini
teratoid. Histological details were not given for all lesions, et al. 2001; Ho and Crean 2003; Şimşek-­Kaya et al. 2018;
but they were described as being lined partly by stratified Robinson et  al.  2021), bronchogenic cysts (Obiechina
squamous epithelium and partly by gastric or intestinal et al. 1999; Gleizal et al. 2006), and lymphoepithelial cysts
epithelium. The authors also reviewed the literature and (Ahn et al. 1996; Epivatianos et al. 2005).
found 15 cases reported between 1959 and 2007. The major-
ity were described as simple cysts and only 3 of the total of Treatment
35 cases recurred.
We would regard these cysts as simple variants of dermoid Treatment of epidermoid and dermoid cysts of the soft tis-
or epidermoid cysts. As discussed previously (see sues is by surgical excision.
‘Classification and Terminology’), care must be taken not to
overdiagnose these as true teratomas. Nevertheless, true tera-
­Developmental Cysts of Foregut Origin
tomas do on occasion arise within the oral cavity (Lopes
et  al.  2005; Shah et  al.  2009; Tonni et  al.  2010; Morlino
Classification and Terminology
et al. 2015). There are also cases reported as teratoid cysts that
do include connective tissue elements and thus may be true Developmental cysts of the alimentary tract are relatively
mature teratomas (Harada et  al.  1995; Bonilla et  al.  1996). common, with an overall incidence of about 1  in 4500
From these reports, it can be seen that occasionally, it may births and a prevalence in children of 0.2% (reviewed by
296 Developmental Cysts

Sangüesa Nebot et  al.  2018). Mostly they are formed by would explain the varied epithelial linings that are found in
errors in the development of the notochord, when strands lingual cysts of foregut origin, as well as the occasional find-
or islands of endoderm are sequestered during folding of ing of endodermal elements in dermoid cysts.
the notochordal plate and become separated from the Intraoral cysts of foregut origin may show a variety of
developing gastrointestinal tract. These isolated cells then histological features, but the clinical features and mode of
give rise to alimentary tract or enteric duplication cysts, presentation are similar. For this reason, they have been
which are found in the midline along the axis of the noto- described under a number of names, including heterotopic
chord at any site along the gastrointestinal tract from the gastrointestinal cyst, oral alimentary tract cyst, anterior
mouth to the rectum (Sangüesa Nebot et al. 2018). Although median lingual cyst, bronchogenic cyst, foregut duplication
usually referred to as cysts, about 20% are tubular struc- cyst, and lingual foregut cyst. They can, however, be divided
tures and rarely they may present as diverticulae. Enteric into two groups based on the histological features: those
duplication cysts are characterised by three elements: a lin- lined primarily by mixed types of gastrointestinal epithe-
ing of epithelium of endodermal origin (gastrointestinal or lium and those lined primarily by respiratory epithelium
respiratory), an envelope of smooth muscle, and a close without gastric or intestinal epithelium. For this reason,
attachment to the associated gastrointestinal tract. In most case reports and series often define more accurately the
cases, the lining of the cyst is the same as the associated type of cyst being referred to, so terms such as lingual cyst
adjacent normal gastrointestinal structure. Most duplica- with respiratory epithelium and lingual cyst with gastrointes-
tion cysts (about 35%) affect the small intestines, but over- tinal epithelium may be seen in the literature.
all about one-­third are associated with the foregut. Despite the many terms used to describe these lesions,
The developing foregut is separated from the stomodeum we will use those most commonly used in the literature.
by the buccopharyngeal membrane, which breaks down at The first group are called heterotopic gastrointestinal cyst,
about 32 days of intrauterine life so that the oral cavity while those lined by respiratory epithelium are broncho-
communicates directly with the alimentary canal. The genic cyst. The key features of the two types are summa-
foregut gives rise to the pharynx, oesophagus, lower res- rised in Box 18.2.
piratory tract (trachea and lungs), stomach, liver, gallblad-
der, pancreas, and duodenum. Foregut duplication cysts
­Heterotopic Gastrointestinal Cyst
may be found associated with any of these structures, but
most often arise in the oesophagus (20%), followed by the
Clinical Features
bronchial apparatus (15%), stomach (7%), and duodenum
(5%) (Sangüesa Nebot et  al.  2018; Liu and Adler  2014). Heterotopic gastrointestinal cysts are rare and most cases
Cysts in the head and neck are rare and account for less in the literature have been single case reports. Morgan
than 0.5% of foregut lesions and less than 0.3% of all enteric et al. (1996) reviewed the literature from 1895 and found
duplication cysts. When they do arise, the vast majority are only 32 cases, and in a similar review Said-­Al-­Naief et al.
found in the tongue or floor of the mouth (Morgan (1999) found only 40 cases. In addition, there have been 9
et al. 1996; Kieran et al. 2010; Luo et al. 2015). Occasional cases reported by Wiersma et  al. (1992) and 6 by Eaton
cases are found in the palate, tonsils, epiglottis, orophar- et al. (2001). Subsequently there have been about 30 single
ynx, or anterior neck (Kieran et al. 2010). case reports and a large series of 22 cases reported by
The anatomical level of origin may determine the precise Kieran et  al. (2010). Luo et  al. (2015) reviewed 23 cases
nature of the lesion. Cysts in the oral cavity are almost reported between 1971 and 2014.
exclusively found in the tongue and the varied nature of the
epithelial lining suggests that they do not meet the three Frequency
defining characteristics of enteric duplication cysts that Oral cysts are very rare and most maxillofacial clinicians
arise in the hindgut. Rather, it seems that the entrapped are unlikely ever to encounter a case. Overall, enteric
endoderm in tongue lesions may be pluripotent, with the duplication cysts have an incidence of about 1  in every
ability to differentiate into any of the epithelial types of 4500 births and a prevalence in children of about 0.2%, but
endodermal origin: respiratory, gastric, or intestinal, as well only about 0.3% of these might be encountered in the oral
as stratified squamous. It is also probable that endoderm of cavity (Eaton et al. 2001; Okur et al. 2014; Sangüesa Nebot
the foregut and ectoderm of the stomodeum can become et al. 2018). They are very rarely included in series of oral
entrapped in the developing tongue at the sites of fusion of biopsies. In their reviews of almost 50 000 oral lesions,
the tuberculum impar (of first branchial arch origin, lined Jones and Franklin (2006a,b) did not record a single case.
by ectoderm) with the hyperbranchial eminence that forms In similar reports, covering over 70 000 maxillofacial biop-
the posterior one-­third of the tongue and is derived from sies, no cases were recorded (Daley et al. 1994; Grossmann
the second, third and fourth arches lined by endoderm. This et al. 2007; Nonaka et al. 2011).
­Heterotopic Gastrointestinal Cys  297

Box 18.2  Developmental Cysts of Foregut Origin: Key Features

Significance and differential


Pathogenesis Clinical features Histopathology diagnosis

Gastrointestinal Arise from the Very rare in the oral Lined by gastric and/ Cysts in neonates may
cyst endoderm of the cavity. Less than 0.3% of or intestinal obstruct the airway and be
develop- enteric duplication cysts epithelium life threatening
ing foregut 85% before 2 years Occasional cases also Must be differentiated from
65% in males contain squamous or other lesions that may arise in
70% in the tongue, respiratory the sublingual/tongue region,
usually anterior epithelium including ranula, dermoid and
30% in floor of epidermoid cysts, thyroglossal
the mouth duct cyst, and salivary gland
Asymptomatic tumours Histology allows
swellings differentiation from
other cysts
Bronchogenic Arise from the Very rare in the Lined by respiratory Must be differentiated from
cyst endoderm of the oral cavity epithelium without thyroglossal duct cyst. Both
develop- 70% occur in the midline evidence of gastro- are lined by respiratory
ing foregut neck, usually in the intestinal epithelium epithelium, but bronchogenic
suprasternal notch Stratified squamous cyst may contain cartilage
More common in epithelium is and smooth muscle in
males (75%) occasionally seen the wall
Only about 30 oral The wall may Thyroglossal cyst may have
cases reported contain cartilage or thyroid gland in the wall
75% located in smooth muscle
the tongue

Age 62% (n  =  42; Manor et  al.  1999), 64% (n  =  22; Kieran
Most cases are diagnosed in neonates and infants, with et al. 2010), and 62% (n = 21; Luo et al. 2015).
about 70% of cases diagnosed in the first year and 85%
before two years. Morgan et  al. (1996) reviewed 32 cases Site
and found that although the majority were diagnosed Almost all oral gastrointestinal cysts have been found in
before 2 years of age, cases had been reported up to 35 years. the tongue or the floor of the mouth. In the most detailed
Manor et  al. (1999) reported an age range of 0–42 years, analysis, Morgan et  al. (1996) found that 97% (31 of 32
with a mean age of 5.5 years and median age of 6 months. cases) arose at these sites, with 58% in the tongue and 39%
Kieran et al. (2010) reported 22 cases with an average age in the floor of the mouth. One case was reported in the
of 2.5 years and a median of 1.5 years. The age range was submandibular region. Of the 18 tongue lesions, 12 (66.6%)
from 5 days to 7 years; 9 cases were diagnosed in the first were located in the anterior dorsum, and 2 (11%) were at
year and 3 were in neonates. Luo et al. (2015) reviewed 22 the posterior junction; 1 case was at the lateral border and
cases and found an age range from birth to 41 years, but the 3 (16.6%) were found on the ventral surface. Luo et  al.
majority (16 cases: 73%) were diagnosed in the first year, (2015) reviewed 23 oral cases and found that 7 (30.4%)
including 9  in neonates. Of the 16 cases of foregut cysts were in the dorsum of the tongue, 9 (39.1%) on the ventral
reported by Wiersma et al. (1992), 14 presented in neonates surface, and 7 (30.4%) were located in the floor of
and the other two at 3 and 6 years. the mouth.
Manor et  al. (1999) reviewed a total of 54 cases, all of
Sex which were in the tongue. Of these, 45 (83.3%) affected the
There is a male predilection, with about 65% of cases in dorsum, of which 5 extended into the posterior tongue.
males. In the largest reviews or series, the proportion of There were 6 cases (11.0%) found on the ventral surface,
males has been reported as 70% (n = 32; Morgan et al. 1996), of which 3 extended into the floor of the mouth. Three
298 Developmental Cysts

cases were so large as to present with both dorsal and In the 32 cases reviewed by Morgan et al. (1996), 26 (81%)
­ventral swelling. contained gastric epithelium and 6 (19%) contained intesti-
nal epithelium. However, only 13 (41%) were lined entirely
Clinical Presentation by gastric epithelium and 5 (16%) entirely by intestinal epi-
The most frequent clinical presentation is an asympto- thelium. The remainder showed a mixed lining, with combi-
matic lingual swelling covered by clinically normal nations of gastric and intestinal with squamous or respiratory
mucosa. Larger cysts, especially in neonates and infants, epithelium. There were areas of squamous epithelium in 10
may cause obstruction of the airway or problems with cases, but only 2 had respiratory epithelium. However, this is
feeding. Of the 54 cases reviewed by Manor et al. (1999), a small series and the true frequency of different epithelial
only 5 recorded any disturbance in tongue function, eat- combinations is not known. Cysts with a mix of gastrointes-
ing, or speaking. Similarly, Morgan et  al. (1996) found tinal and respiratory epithelium do appear to be very rare.
that 69% of cases presented as symptomless swellings, Luo et  al. (2015) reviewed case reports of gastrointestinal
while the remainder caused functional problems associ- cysts that contained respiratory epithelium and found only
ated with feeding, swallowing, or breathing. Of the 23 21 reports between 1971 and 2014.
cases reviewed by Luo et  al. (2015), 13 (56.5%) were When lined by gastric mucosa, the epithelium is of the
symptomless. The remainder caused problems with type seen in the body and fundus of the stomach
speech or feeding, and 2 cases in neonates also obstructed (Figure 18.4). Both parietal and chief cells may be found,
the airway. and typical gastric glands may be present. Some cysts have
a muscularis mucosa (Lipsett et al. 1993; Eaton et al. 2001).
Cysts lined with intestinal mucosa may show Paneth cells,
Histopathology
goblet cells, and argentaffin cells (Gorlin and Jirasek 1970;
Gastrointestinal cysts are lined by epithelium derived from Lipsett et al. 1993).
the endoderm of the foregut, but, by definition, must con- As mentioned previously in this chapter, gastrointestinal
tain either gastric or intestinal epithelium or both. Cysts cysts are occasionally multiple or found in combination
may also show areas of squamous epithelium or respira- with dermoid or epidermoid cysts (Eppley et  al.  1985;
tory epithelium. The most commonly encountered epithe- Arcand et al. 1988; Wiersma et al. 1992; Crivelini et al. 2001;
lium is gastric (Figure  18.4). Note, however, that lingual Ho and Crean  2003; Şimşek-­Kaya et  al.  2018; Robinson
cysts lined only by squamous epithelium are dermoid or et al. 2021).
epidermoid cysts and those lined predominantly by respir-
atory epithelium without gastrointestinal epithelium are
Treatment
bronchogenic cysts.
Treatment consists of surgical excision. Recurrences are
rare, but have been reported.

­Bronchogenic Cyst

Bronchogenic cyst is the second type of developmental


cysts that may arise from the developing foregut. They are
distinguished from other types of duplication cyst because
they arise from the primitive foregut during the formation
of the tracheobronchial tree, and are lined predominantly
by respiratory epithelium without evidence of gastrointesti-
nal epithelium. They are also known as bronchial cysts or
descriptively as cysts with respiratory epithelium.
They are found most frequently within the mediastinum
or thorax, but may rarely be encountered in the neck
(McAdams et  al.  2000; Ustundag et  al.  2005; Luna and
Pfaltz  2009) and less often in the tongue or floor of the
mouth (Wiersma et  al.  1992; Obiechina et  al.  1999;
Figure 18.4  Gastrointestinal cyst, lined by gastric-­type
epithelium with surface mucous cells (foveolar cells) and glands. Benhammou et  al.  2006; Gleizal et  al.  2006; Aldawood
Source: Courtesy of Dr Jos Hille. et al. 2021).
­Bronchogenic Cys  299

Clinical Features (8 of 14 cases) were males. Of the 5 cases reviewed by


Aldawood et  al. (2021), 2  were located in the ventral
Bronchogenic cysts in the neck are most often encountered
tongue, 1 in the base of the tongue and 1 in the floor of
in the suprasternal notch region and over 70% are in the
the mouth. There was 1  lesion in the mandibular vesti-
midline (Ustundag et al. 2005). In a review of 70 cases in
bule extending into the lower lip. Of the 5 cases, 4 were in
the head and neck region reported up to 2005, Ustundag
males and the age of presentation ranged from 1  day to
et al. (2005) found only 5 lesions above the hyoid bone, 3 of
6 years.
which were submental and 2 in the tongue. The subman-
dibular region was the site of 2 cases, while others were
found in the upper neck close to the thyroid gland, or just Histopathology
lateral to the larynx or epiglottis. Most lesions are located
Bronchogenic cysts are lined by pseudostratified ciliated
in the skin or immediate subcutaneous tissues.
columnar (respiratory-­type) epithelium (Figure  18.5),
When present at a lateral site, they may be found overly-
although focal areas of simple cuboidal or stratified squa-
ing the clavicles or at the anterior border of the sternomas-
mous epithelium may be present. They do not contain gas-
toid muscle. At this site they must be considered in the
tric or intestinal epithelium. The cyst wall may contain
differential diagnosis of branchial cleft cysts and metastatic
cartilage, smooth muscle, and seromucous glands. Focal
head and neck carcinoma (Luna and Pfaltz 2009).
areas of inflammation are occasionally seen. Of the 21
Cervical bronchogenic cysts are found in males with a
cases reviewed by Peters et al. (2018), all were lined by typi-
M : F ratio of about 3 : 1 (Ustundag et al. 2005) and become
cal respiratory epithelium, but in addition 10 (47.6%)
clinically apparent just after birth. In the series of Ustundag
showed focal areas of squamous epithelium and 6 (28.6%)
et  al. the age range was from 0 to 54 years, but only 12
had areas of simple cuboidal epithelium. Wiersma et  al.
(17.1%) cases were found in adults. They usually present as
(1992) found 7 bronchogenic cysts in their series of 16 fore-
symptomless masses that slowly increase in size. Lesions
gut cysts and showed that 3 were lined by respiratory epi-
may communicate with the skin through a sinus tract.
thelium alone, 3 also had focal areas of squamous
Oral lesions are very rare, but the frequency may depend
epithelium, and 1 case showed foci of both squamous and
on the criteria for diagnosis. Peters et al. (2018) reviewed
simple columnar epithelium.
the literature from 1963 and found only 21 cases affecting
Aldawood et al. (2021) presented a case that was lined by
the tongue (including 2 of their own). They called the cysts
respiratory epithelium and had foci of cartilage and smooth
‘lingual cysts with respiratory epithelium’ and defined
muscle in the wall. They also found 4 similar cases in the
them as being lined by respiratory epithelium without evi-
literature. All had focal areas of stratified squamous epi-
dence of gastrointestinal epithelium. More recently,
thelium and cartilage in the wall; 1 case also showed areas
Aldawood et al. (2021) applied more stringent criteria and
of smooth muscle.
only included cases that also showed evidence of cartilage
in the wall. They reported 1  new case, but only found 4
previous cases in the literature since 1982.
Although cartilage and other mesodermal elements
(including smooth muscle) may be present in the wall, we
suggest that a diagnosis of bronchogenic cyst is appropri-
ate for a cyst lined predominantly by respiratory epithe-
lium without evidence of gastrointestinal epithelium.
Areas of stratified squamous epithelium may be found
and some cases will show cartilage or smooth muscle in
the wall.
Using these broader criteria, there are still only about 30
cases of oral lesions in the literature (Obiechina et al. 1999;
Ustundag et  al.  2005; Benhammou et  al.  2006; Gleizal
et al. 2006; Peters et al. 2018; Aldawood et al. 2021).
In the cases reviewed by Peters et al. (2018), 13 (61.9%)
were in the anterior dorsum of the tongue, 4 in the ventral
tongue, and 4 were located in the floor of the mouth. The
Figure 18.5  Bronchogenic cyst, lined by pseudostratified
average age of presentation was 9.4 years (range 6 months ciliated columnar epithelium with occasional mucous (goblet)
to 42 years) and in those where the sex was reported 57% cells. Source: Courtesy of Dr Colin Ades.
300 Developmental Cysts

Treatment referred to as lymphoepithelial cysts. Lymphoepithelial


cysts arise in the oral cavity or the parotid glands and arise
Treatment consists of complete surgical excision, after
from tonsil or salivary gland. They should be regarded as
which recurrence is rare.
distinct from developmental cysts of branchial arch origin.
Lymphoepithelial cysts are considered in Chapter 15. Note,
however, that true branchial cysts of first-­arch origin may
­Branchial Cleft Anomalies arise in association with the parotid gland and present as a
pre-­auricular swelling. The key features of branchial cleft
The branchial arches form between the fourth and seventh
cysts are summarised in Box 18.3.
weeks of gestation, and give rise to most of the structures
of the face, oral cavity, neck, and pharynx. The branchial
apparatus comprises six paired arches, but the fifth arch is Clinical Features
rudimentary in humans and the sixth arch is small and is
often considered as part of the fourth arch. For practical Frequency
purposes, therefore, the developing face and oral cavity are Branchial cleft cysts are rare, but are still the second most
derived from four branchial arches (Adams et  al.  2016; common cause of swellings in the neck in neonates and
Nanci 2017). These are composed of expansions of meso- children, after thyroglossal duct cyst. In their review of
derm forming bulges that are separated externally by more than 44 000  maxillofacial biopsies in adults, Jones
branchial clefts and internally by pharyngeal pouches. The and Franklin (2006a) found 24 branchial cysts (0.05%).
first arch develops in front of the buccopharyngeal mem- Balikci et al. (2013) reviewed 630 sequential neck masses in
brane and is lined externally and internally by ectoderm. all age groups and found that 119 (18.9%) were congenital
The second to fourth arches develop behind the membrane and of these only 26 (21.9% of congenital lesions and 4.1%
and are lined externally by ectoderm and internally by of the total) were branchial cysts. The most common con-
endoderm. genital lesion was thyroglossal duct cyst (39 cases). The
Branchial cleft anomalies occur due to incomplete oblit- overall most common cause of neck masses was neoplastic
eration of the branchial clefts or pharyngeal pouches caus- (300 cases: 47.6%), followed by inflammatory (211 cases:
ing persistence or entrapment of the epithelial linings. In 33.9%). They found, however, that congenital cysts were
this way, the anomalies may present as cysts, sinuses or significantly more likely to be diagnosed in people less
fistulas, or combinations of these defects. Detailed and than 20 years of age, where 45.1% of cases were congenital.
excellent reviews of branchial anomalies have been pro- Conversely, masses in patients over 40 years were more
vided by Luna and Pfaltz (2009), Adams et al. (2016), and likely to be malignant, accounting for 47.5% of cases. Of all
Wenig (2017a). congenital neck masses, Al-­Khateeb and Al Zoubi (2007)
The vast majority of anomalies (about 95%) derive from found that 22% (55 of 252 cases) were branchial cysts and
the second branchial arch and up to 4% from the first arch. in a review of 226 congenital developmental cysts, Brucoli
Anomalies of the third and fourth arches are very rare and et al. (2020) found that 44% were thyroglossal duct cysts,
usually present as sinuses or abscesses in the lower neck 43% branchial cysts, and 13% dermoid cysts.
that open either onto the skin at the lower third of the ster-
nocleidomastoid muscle, or internally into the piriform Age and Sex
sinus. Anomalies of the first and second arches are most Branchial cleft cysts affect males and females equally and
often cysts, although up to one-­third may present as a sinus most present in the third and fourth decades, with reported
or fistula. All the anomalies are congenital, but in general average ages of 18.6 years (Al-­Khateeb and Al Zoubi 2007),
sinuses and fistulas are diagnosed in neonates or infants, 34.0 years (Brucoli et  al.  2020), and 31.7 years (Pupić-­
while cysts grow slowly over a long period of time and most Bakrač et  al.  2021). In the largest series ever published
present in adults in the third to fifth decades. (n  =  468; Bhaskar and Bernier  1959), 61% of cases were
diagnosed in the third decade and 92.5% were diagnosed
under the age of 40 years. Only 35 cases (7.5%) were
­Branchial Cleft Cysts encountered over the age of 40 years.

Branchial cleft cysts arise in the lateral aspect of the neck. Site
When used without qualification, the term branchial cleft Because most branchial cysts arise from the second arch,
cyst is taken to refer to cysts found in the upper lateral neck the most common location is in the upper lateral aspect of
and derived from the second branchial arch. Because of the neck, just anterior to the sternocleidomastoid muscle
their similar histopathological features, they are often close to the angle of the mandible. Pupić-­Bakrač et  al.
­Branchial Cleft Cyst  301

Box 18.3  Branchial Cleft Cyst: Key Features

Pathogenesis
●● Entrapment of epithelium due to incomplete obliteration of the branchial clefts
●● 95% arise from the 2nd branchial arch
Clinical features
●● Second most common (22%) developmental cyst in the head and neck
●● Found in the lateral aspect of the neck
●● Most are in the submandibular region just anterior to the sternocleidomastoid muscle
●● Rare 1st arch cysts are pre- or post-auricular
●● Mean age about 30 years with peak in 3rd & 4th decades
●● Equal gender distribution
●● Wide age range but 90% diagnosed under 40 years.
●● Usually present as symptomless neck masses
Histopathology
●● Simple unilocular cysts lined by stratified squamous epithelium
●● Respiratory epithelium is occasionally (<5%) seen
●● The wall contains lymphoid tissue
Significance and differential diagnosis
●● Must be differentiated from cystic metastatic carcinoma.
●● Over age 35 years assume a metastatic carcinoma until proved otherwise.
●● Carcinomas show cytological evidence of malignancy and usually have solid areas or invasion of the wall.
●● HPV-associated carcinomas are strongly p16 positive

(2021) reported 46 patients with 48 branchial anomalies sternocleidomastoid muscle (Luna and Pfaltz 2009). There
and found that 77% were cysts of second-­arch origin and may be a mucoid or, if infected, a purulent discharge.
were located in the upper lateral neck. Anomalies of the third or fourth arches that open into the
Anomalies arising from the first arch are found in the piriform sinus may compromise the airway and present as
region of the parotid gland and present as pre-­auricular or stridor.
post-­auricular swellings. In the series presented by Pupić-­
Bakrač et al. (2021), 8 (16.7%) cases were of first-­arch origin
Histopathology
presenting in the parotid region, of which 4 (50%) pre-
sented as sinuses opening onto the skin. Most cysts are carefully excised and the pathologist will
Anomalies associated with the third or fourth branchial receive an intact round or spherical cystic mass. On dissec-
arches are most often sinuses and are found in the lower tion there is a thin fibrous wall, but thickenings are often
neck, adjacent to the lower third of the sternocleidomas- noted, representing areas where there is an accumulation
toid muscle and the suprasternal notch. of lymphoid tissue (Figure 18.6). The lumen may contain a
semi-­solid cheesy material (sometimes referred to as ‘gru-
Clinical Presentation mous’) or a mucoid or pale brown fluid. If the lumen con-
Branchial cysts have an average size of about 40 mm, but tains thickened or solid areas, then a diagnosis of branchial
can range from 10 to 140 mm (Wenig 2017a; Pupić-­Bakrač cyst is unlikely and solid areas should be carefully sampled.
et  al.  2021). The most frequent symptoms are swelling, Histological examination shows that most branchial cysts
which may be progressive or intermittent, with occasional are simple unilocular cysts lined by thin, regular stratified
pain. Sometimes lesions have been found to enlarge in squamous epithelium that may be para-­or orthokeratinised
response to an upper respiratory tract infection. The swell- (Figure 18.6). About 95% of cases are lined wholly by strati-
ings are soft and compressible and often fluctuant. fied squamous epithelium (Bhaskar and Bernier  1959;
Occasionally, there may be a sinus or fistula that opens Wenig 2017a; Pupić-­Bakrač et al. 2021), while the remainder
onto the skin at the anterior aspect of the also show areas of simple columnar, respiratory, or ciliated
302 Developmental Cysts

(a)

(b) (c)

Figure 18.6  Branchial cyst. Images from three different cysts. The cysts are lined by thin regular stratified squamous epithelium.
(a) Focal accumulations of lymphoid tissue can be seen (right and top). (b) Well-­formed normal germinal centres are seen. (c) In this
example a focal area shows respiratory-­type epithelium with surface cilia (arrows).

epithelium (Figure  18.6c). Occasional cases (<2%) may be usually empty or contains only sparse amounts of cellular
lined wholly by respiratory epithelium. Most cases arise debris and occasional inflammatory cells.
superficially from the branchial clefts and the lining is In virtually all cases, the cyst wall contains lymphoid tis-
derived from ectoderm, but it is possible that cysts showing sue with well-­formed germinal centres (Figure  18.6a, b).
respiratory epithelium arise at least in part from the endo- The lymphoid tissue may embrace the whole of the cyst or
derm of a pharyngeal pouch. The lumen of the cyst is may be focal. Focal areas of inflammation are seen in up to
­Branchial Cleft Cyst  303

50% of cases and may be accompanied by granulation tis- carcinoma) or are salivary gland tumours associated with
sue and fibrosis. If the lining is ruptured, foreign-­body the submandibular gland. Occasionally, however, a meta-
giant cells may be seen in the wall. The epithelial lining is static carcinoma may be cystic in nature both on clinical exami-
cytologically bland, but in inflamed cases there may be nation and histologically.
some evidence of reactive atypia.
There have been occasional reports of carcinomas aris- Differentiation of Branchial Cleft Cyst from Cystic
ing in branchial cysts (often referred to as bronchogenic Metastatic Carcinoma
carcinoma). These have shown an otherwise quite typical One of the greatest challenges for clinicians and patholo-
cyst, but with cytologically malignant epithelium. In the gists, when faced with a cystic mass in the neck, is to differ-
absence of clinical evidence of a primary carcinoma else- entiate between a branchial cleft cyst and a cystic metastasis
where, authors have assumed that these lesions represent from a carcinoma in the nasopharynx or oropharynx.
primary carcinomas arising in a developmental cyst. In a study of 135 patients from all age groups, with cysts
However, this is regarded as extremely rare or unlikely, and in the lateral neck (Grønlund et al. 2016), 19 (14.5%) were
it is generally accepted that a diagnosis of carcinoma aris- found to be metastatic carcinoma. Of these, 16 were meta-
ing in branchial cyst is not appropriate (Luna and static squamous cell carcinomas and 3 were papillary carci-
Pfaltz 2009; Wenig 2017a). All cases should be regarded as noma of the thyroid. The diagnosis for 89 cysts (65.9%) was
metastatic carcinoma, even in those rare cases where a pri- benign lateral neck cyst (branchial cleft cyst). However, it
mary lesion cannot be found. was found that all the malignant cases arose over the age of
35 years and that 33.3% (14 of 42) of cysts in the fourth and
fifth decades were metastases. Conversely, 87.0% of cysts in
Differential Diagnosis
patients under 31 years were branchial cysts. The authors
Branchial cysts present as neck masses and any cause of concluded that in patients over the age of 35 years, any
neck swelling must be considered in the clinical differential neck cyst should be regarded as malignant until proved
diagnosis. In infants and children, the majority of neck otherwise. In an analysis of 630  neck swellings, Balikci
swellings are congenital developmental lesions and the et al. (2013) found that in patients under the age of 20 years,
most likely cause is either thyroglossal duct cyst or branchial 85% of swellings are congenital or inflammatory, while
cleft cyst. The distinction is usually clear: thyroglossal duct over the age of 40 years, 85% are neoplastic or inflamma-
cysts arise in the midline of the upper neck, while branchial tory and about 15% are malignant.
cleft cysts are always lateral. Haemangioma or lymphangi- Up to about 50% of head and neck carcinomas may show
oma (cystic hygroma) may also arise in the lateral neck cervical lymph node metastases at first presentation and in
(Balikci et al. 2013), but has distinctive diagnostic features lesions of the nasopharynx and oropharynx a neck mass is
that should not cause confusion with branchial cysts. often the first clinical sign of disease. Furthermore, these
The cystic hygroma is a developmental abnormality that is metastases, especially from human papillomavirus (HPV)–
often present at birth and most cases are diagnosed before associated oropharyngeal squamous cell carcinoma, are
the age of 2 years. The lesion is more correctly designated as often cystic and must be differentiated from benign cystic
a cavernous type of lymphangioma, but cystic hygroma is a lesions. Cystic metastases usually show cytological evi-
commonly used clinical term. It most frequently involves dence of malignancy and have solid areas that resemble a
the face and lateral neck, where it presents as a large diffuse, typical conventional keratinising squamous carcinoma.
soft, and compressible swelling. The lesion is usually unilat- Non-­keratinising, HPV-­associated metastases, however,
eral, but the entire side of the neck and lower face may be may be more difficult to diagnose. Often the cyst lining will
involved. The dilated vessels are often superficial, the overly- show areas of thickening, with ribbon-­like proliferations of
ing skin may be blue, and the swelling transilluminates. carcinoma (Figure 18.7a), and tumour may invade into the
There may be a history of gradual or sudden enlargement. wall. Malignant cysts are also more likely to contain
Histologically, the cystic hygroma consists of multiple necrotic material, show inflammation, and have thicken-
dilated cystic spaces lined by endothelial cells, and often ing or fibrosis of the wall. Metastases are also within a
filled with pale eosinophilic lymph. lymph node, so the periphery of the lesion may show obvi-
In older children and adolescents (<20 years), inflamma- ous lymphatic vessels and normal subcapsular sinuses. In
tory masses may also be encountered (Balikci et al. 2013). the majority of cases, therefore, histological examination
These are not cystic, are usually firm or tender on palpa- should confirm the diagnosis.
tion, and represent enlarged lymph nodes. Occasionally a cystic metastasis may show histological
Neck masses in adults are more likely to be neoplastic, features of a simple cyst and a precise diagnosis may not be
but the majority of these are not cystic and most are associ- possible (Figure  18.7b). Furthermore, occasional cystic
ated with lymph nodes (lymphomas or metastatic metastases from HPV-­associated oropharyngeal carcinomas
304 Developmental Cysts

(a) (b)

(c) (d)

Figure 18.7  Cystic metastasis of a human papillomavirus (HPV)–associated oropharyngeal carcinoma. (a) In places the lining is
thickened with obvious cytological atypia. (b) Elsewhere the lining is thin and bland, with no evidence of malignancy (compare to
Figure 18.6b). (c, d) p16 is strongly expressed throughout the epithelium.

show focal areas with ciliated epithelium, leading to a misdi- a branchial cyst usually shows a hypocellular smear or
agnosis of branchial cyst (Bishop and Westra  2015). The cell block, with nucleated squamous cells and variable
presence of cilia cannot therefore be taken as a sign of benig- numbers of inflammatory cells. Fragments of keratin or
nity nor considered as a diagnostic feature of a developmen- parakeratotic cells with smooth hyperchromatic nuclei
tal cyst. may be seen, but are not prominent. An FNA from an
These data show that great care must be taken when try- inflamed case may show cells with some nuclear atypia,
ing to interpret the histology of cystic neck masses, espe- but this is usually accompanied by evidence of inflam-
cially in adults over the age of 35. Special care is needed mation and the nuclear changes appear reactive or
when presented with small incisional biopsies that may not degenerative. An FNA from a metastatic carcinoma will
be representative (e.g. compare Figures 18.6b and 18.7b). If usually show obvious malignant cells, but cytology of a
there is any doubt, a diagnosis should not be given and in cystic lesion may be less helpful and FNA cytology is
patients over 35 years of age a malignancy should be notoriously difficult, with sensitivities and specificities
assumed until proved otherwise. Although a primary of about 90% and 60%, respectively (Grønlund et al. 2016;
lesion may be occult, in 80–90% of cases a primary tumour Layfield et  al.  2016). In general, an FNA from a cystic
can be found on careful imaging or by blind biopsies in the metastasis will show a greater degree of cellularity, cell
oropharynx or upper aerodigestive tract. clusters, more necrotic debris, and clear evidence of
Fine needle aspiration (FNA) cytology is often used in cytological atypia and mitoses (Layfield et  al.  2016;
the differential diagnosis of neck masses. An FNA from Wenig 2017a).
­Thyroglossal Duct Cys  305

The majority of cystic metastases may come from HPV-­ in the anterior neck, just below the thyroid and cricoid car-
associated squamous cell carcinomas in the oropharynx. tilages. The thyroglossal duct disintegrates by about the
Immunocytochemistry for p16 is an acceptable surrogate eighth week, but remnants of the duct may remain at any
marker for high-­risk HPV and can be used as a test for both site along its line of descent. In an autopsy study, Sprinzl
primary or metastatic HPV-­associated carcinomas et al. (2000) found that 41% of children had retained rem-
(Lewis 2020). Metastatic carcinomas show positive expres- nants of the thyroglossal duct, or ectopic thyroid tissue.
sion of p16 that is diagnostic if greater than 70% of the cells These residual islands of epithelium give rise to the thy-
show strong nuclear and cytoplasmic staining (Figure 18.7c, roglossal duct cyst. Furthermore, up to 40% of the adult
d). However, up to 50% of branchial cysts may show expres- population may have an accessory, median lobe of the thy-
sion of p16 (Pai et al. 2009; Cao et al. 2010), although it is roid, the pyramidal lobe, which represents a vestigial rem-
usually weak, focal, and limited to the basal and parabasal nant of the thyroglossal duct.
layers (Cao et  al.  2010; Müller et  al.  2015). With careful
interpretation, p16 can be useful in tissue sections to dif-
Clinical Features
ferentiate between a branchial cyst and a metastatic HPV-­
associated carcinoma. Again, care is needed in small Frequency
biopsies and if there is any doubt, then HPV testing using The thyroglossal duct cyst is the most common of the
polymerase chain reaction (PCR) or in situ hybridisation developmental cysts of the neck, accounting for about 45%
should be carried out. of congenital abnormalities in the neck in children. In
p16 staining is not recommended in the assessment of their analysis of 630 neck masses in all age groups, Balikci
FNA cytology specimens, since it is unreliable and is diffi- et al. (2013) found 119 congenital cysts, of which 39 were
cult to interpret because few cases reach the 70% positive thyroglossal duct cysts, representing 32.7% of congenital
threshold (El-­Salem et al. 2019; Lewis 2020). If an FNA is cysts and 6.2% of all neck masses. Al-­Khateeb and Al Zoubi
equivocal, then it must be repeated or a biopsy is required. (2007) found that thyroglossal duct cysts constituted 55.6%
However, direct HPV testing by PCR on FNA specimens of all congenital cysts in the neck and about 63% of cysts
may be useful, and a number of centres now use liquid-­ diagnosed in the first two decades. In a similar study,
based HPV testing, similar to that used for cervical screen- Brucoli et al. (2020) reviewed 226 patients with congenital
ing (Lewis 2020). neck cysts and found that 100 (44%) were thyroglossal duct
Metastatic lesions from papillary thyroid carcinoma may cysts. Thompson et al. (2016) estimated a population inci-
occasionally be cystic, but usually show areas with a papil- dence of 2.23 cases per 100 000 persons per year in their
lary architecture, follicles with colloid, and typical nuclear catchment area of Southern California.
features that are diagnostic. Immunohistochemistry is also
helpful, since papillary thyroid carcinomas may express Age
thyroglobulin, TTF1, and PAX8, none of which is seen in Although the lesion is congenital, most cysts grow slowly
branchial cysts. and many are not diagnosed until adulthood, with an aver-
age age of presentation of about 30 years.
Thompson et al. (2016) reported a series of 685 thyroglos-
Treatment
sal duct cysts and found an average age at diagnosis of
Branchial cysts are removed by careful surgical excision. 31.3 years (range 10 months–87 years). Only 38% of cases
Recurrence is very rare (Papadogeorgakis et  al.  2009; were diagnosed in the first two decades, while 39.3% were
Brucoli et al. 2020; Pupić-­Bakrač et al. 2021). diagnosed over the age of 40 years. Thus, there is a bimodal
age distribution, with peaks in the first decade (26.1% of
cases) and in the fifth decade (15.0% of cases). De Tristan
­Thyroglossal Duct Cyst et al. (2015) also showed a bimodal distribution with peaks
in the first and fourth decades, but with an overall average
The thyroid gland develops at about the fourth week of age of 29 years (n = 282; range 1–78 years).
intrauterine life from the foramen caecum on the dorsal Cysts affecting the tongue may present in a younger
surface of the tongue, just posterior to the circumvallate age group, probably because they are more likely to
papillae. The developing gland descends in a caudal direc- cause symptoms. Burkart et  al. (2009) reviewed 16  lin-
tion, forming a hollow epithelial stalk, known as the thy- gual cysts and found an average age at presentation of
roglossal duct. The thyroglossal duct crosses the anterior 3.01 years (range 1 month–16 years), but with 50% of
surface of the hyoid bone, loops onto the posterior surface, cases presenting in the first year and only one case in the
and then descends to the site of the normal thyroid gland second decade.
306 Developmental Cysts

Box 18.4  Thyroglossal Duct Cyst: Key Features

Pathogenesis
●● Arises from remnants of the thyroglossal duct
Clinical features
●● The most common (45%) developmental cyst in the head and neck
●● Mean age about 30 years with bimodal peaks in the 1st and 5th decades
●● Equal gender distribution in adults
●● More common in males (60%) in children
●● Located in the midline of the upper neck close to the hyoid bone.
●● Rare in the tongue – about 3% of cases
●● Usually a symptomless swelling, but 25% may have pain or tenderness
●● May lift on swallowing
Histopathology
●● Most are lined by respiratory epithelium but squamous epithelium may be seen
●● Most (70%) contain ectopic thyroid gland in the wall
Significance and differential diagnosis
●● Tongue lesions may obstruct the airway, especially in neonates.
●● Very high recurrence rate (40% - 50%) if thyroglossal duct remnants are not removed with the cyst (Sistrunk
procedure)
●● Rare cases (3%) contain papillary thyroid carcinoma.
●● Must be differentiated from bronchogenic cyst. Both are lined by respiratory epithelium but thyroglossal duct
cyst has thyroid gland in the wall.
●● Bronchogenic cyst may contain cartilage and smooth muscle..

Sex connected to the bone at operation. Cysts are never located


Across all age groups, thyroglossal duct cyst shows an in the lateral neck and those few not in the midline are just
equal sex distribution, but there is some evidence that in to one side in a paramedial location. A number of studies
children males are more likely to be affected. Thompson have shown a slight predilection for the left side (Ren
et  al. (2016) found that overall, 50.2% of their 685 cases et al. 2011; de Tristan et al. 2015; Thompson et al. 2016).
presented in males and 49.8% in females, but in the first Cysts may be located in the oral cavity, either in the poste-
two decades 57.5% arose in males and over the age of rior floor of the mouth just above the hyoid bone or very rarely
20 years 54.2% arose in females. This distribution is sup- in the tongue. Burkart et al. (2009) reviewed 189 patients with
ported by the study of Geller et  al. (2014), who reported thyroglossal duct cysts and found 16 (8.5%) with cysts located
128 paediatric patients ( 14 years) and found that 60.9% in the tongue. In a systematic review, however, Gioacchini
were males, and also by Burkart et al. (2009), who found et al. (2015) found 24 studies reporting a total of 1371 cysts
that 13 of their 16 (81.2%) paediatric patients with tongue and noted that only 39 (2.8%) were located in the tongue.
cysts were male.
Clinical Presentation
Site Thyroglossal duct cysts present as a mobile swelling or
Thyroglossal duct cysts are always located in the upper cystic mass with or without associated pain or tenderness.
neck and the vast majority (98%) are located in the midline Thompson et al. (2016) found that 99.7% of their cases pre-
and below the hyoid bone (de Tristan et al. 2015; Thompson sented with a mobile mass, while the remaining 5 cases
et al. 2016). In their review of 685 cases, Thompson et al. (2.3%) were fixed. In addition, 164 cases (23.9%) also
(2016) found that the majority (75.9%) were located below showed pain or tenderness, and 123 (18.0%) showed signs
the hyoid bone and 24.1% were suprahyoid. Whether above of infection. There were 67 cases (9.8%) with a draining
or below the hyoid bone, most lesions are found to be sinus or fistula.
­Thyroglossal Duct Cys  307

In their systematic review, Gioacchini et al. (2015) found


that 75% of cases presented as a painless swelling, while C
34% showed evidence of infection or an abscess and 18%
had a fistula or sinus. In their series of 282 cases, de Tristan C
et al. (2015) showed that 280 (99.3%) presented as a swell-
ing, of which about 25% were growing and 25% were inter-
mittent. Only 30.0% of patients complained of pain and
20.6% had signs of infection. They also found that 23.4%
were associated with difficulty swallowing and a small
number caused problems with speech (1.4%) or breath-
ing (0.4%).
A classic sign of the thyroglossal duct cyst is that it lifts
when the patient swallows or protrudes the tongue, but
this is only seen in about 20% of cases (de Tristan
et al. 2015).
Cysts arising in the tongue are more likely to present
with symptoms associated with difficulty in eating or
breathing and large cysts in neonates may be life threaten-
ing. In Burkart et al.’s (2009) series of 16 tongue lesions, 7
cases (43.8%) presented with signs of airway obstruction.

Histopathology
Thyroglossal duct cysts are surgically excised, usually with Figure 18.8  Thyroglossal duct cyst (C) lined by
a portion of the hyoid bone and associated tissue contain- pseudostratified columnar epithelium. Thyroid tissue (arrows) is
ing residual duct (the ‘Sistrunk’ procedure). The cyst itself present in the wall; haematoxylin and eosin (H&E) stain.
has a fibrous wall and on dissection may be multilocular.
Cysts have an average size of about 25 mm (range 2–85 mm; and squamous cell carcinoma have also been reported
Thompson et al. 2016). Lingual cysts may be removed by (Lustmann et al. 1989; Thompson et al. 2017).
excision alone and the cysts are usually smaller, with a
range from 5 to 20 mm (Burkart et al. 2009). Differential Diagnosis
Thyroglossal duct cysts are lined by pseudostratified The major source of potential confusion is with other cystic
columnar (respiratory) epithelium, stratified squamous neck masses, but the thyroglossal duct cyst is always in the
epithelium, or both. Most often a lining of both respiratory midline (or paramedial) and arises high in the neck close to
and stratified squamous epithelium is seen (Figure 18.8). the hyoid bone. When upward displacement of the cyst
Thompson et al. (2016) undertook a detailed analysis of the occurs on swallowing, it is diagnostic. Most other neck
histology of their 685 cases. They found that 51% were masses arise in the lateral neck.
lined by both respiratory and stratified squamous epithe- Bronchogenic cysts are the main possible source of misdi-
lium, 38% by respiratory epithelium alone, and 10% by agnosis. These may arise in the midline, but are more often
stratified squamous epithelium alone. located in the lower neck close to the sternal notch.
The cyst wall is fibrous, but most cases (87%) showed evi- Histologically, bronchogenic cysts may also be lined by res-
dence of inflammation and 65% showed small areas of cyst piratory epithelium and stratified squamous epithelium,
rupture with associated foamy histiocytes. Most lesions but they usually show cartilage in the wall and may have
(71%) had ectopic thyroid gland in the wall (Figure  18.8) smooth muscle. Conversely, thyroglossal duct cysts do not
and 15.2% showed seromucous glands. Occasional cases show smooth muscle and usually have focal nodules of
may show well-­formed lymphoid tissue in the wall. thyroid gland in the wall. A small number (<1.0%) of thy-
There is a well-­recognised association between thyroglos- roglossal duct cysts, however, may have cartilage in the
sal duct cyst and papillary thyroid carcinoma (Thompson wall (Thompson et al. 2016).
et  al.  2016,  2017). In their series of 685 cases, Thompson The differential diagnosis of lingual cysts includes thy-
et  al. (2016) found 22 (3.2%) cysts that contained classic roglossal duct cyst, bronchogenic cyst, gastrointestinal
papillary thyroid carcinomas. The carcinomas are found in cyst, and dermoid and epidermoid cysts. Thyroglossal duct
the cyst wall and are usually associated with ectopic thyroid cyst is always located deep in the posterior tongue, whereas
gland. Occasional examples of follicular thyroid carcinoma the others are more often located in the anterior tongue or
308 Developmental Cysts

in the ventral tongue and anterior floor of the mouth. (Tornwaldt’s bursa) or from Rathke’s pouch. These cysts
Histologically each cyst has distinguishing features, except are very rare, rarely cause any symptoms, and are usually
for bronchogenic cyst, which as discussed can resemble incidental findings at autopsy or on imaging.
thyroglossal duct cyst. Tornwaldt cyst derives from entrapped ectoderm if
the notochord fails to detach from the pharyngeal wall.
The cyst forms a smooth, submucosal mass covered by
Treatment
normal mucosa, and is usually less than 20 mm in
Surgical excision is usually advised for the treatment of diameter. It is usually symptomless unless infected,
thyroglossal duct cysts. The recommended approach is when patients may experience headaches, dizziness,
called the Sistrunk operation (first described in detail by earache, halitosis, and sore throat. It is lined by respira-
Walter Sistrunk in 1920; Geller et  al.  2014; Gioacchini tory epithelium, unless infected when there may be
et al. 2015; Thompson et al. 2016). The procedure involves squamous metaplasia.
removal of the cyst along with the central part of the hyoid Rathke cleft cysts are found anterior or cranial to the
bone and a 1 cm block of tissue surrounding the duct. The usual site of origin of retention or Tornwaldt cysts, and
thyroglossal duct should be traced down to the pyramidal arise at the same site as craniopharyngiomas from part of
lobe of the thyroid gland and up to the foramen caecum at the craniopharyngeal canal. The cyst is formed by a failure
the base of the tongue. to obliterate Rathke’s pouch during formation of the pitui-
This method ensures that any residual remnants of the tary gland. Most cysts are intracranial, but occasional
duct are removed and reduces recurrence. In their system- lesions may occur in the nasopharynx. They are usually
atic review, Gioacchini et al. (2015) reported that 90.4% of symptomless and are found incidentally on imaging for
cases had been treated according to this procedure, with a other reasons. When symptoms do arise, patients may
recurrence rate of 5.3%. Of the few cases that had been complain of sudden-­onset headaches, visual disturbances,
treated by cystectomy alone, 47.6% recurred. In Thompson nausea, and pituitary symptoms. The cysts are lined by res-
et al.’s (2016) series, 94.4% of the patients had the Sistrunk piratory or squamous epithelium.
procedure, with a recurrence rate of about 1.0%; 31 patients Other cysts that can arise in the pharynx or nasopharynx
had a cystectomy alone, of which 14 (45.2%) recurred. include branchial cleft cysts, dermoid or epidermoid cysts,
Burkart et al. (2009) treated 16 lingual cases by a tran- and teratoid cysts.
soral route using endoscopy. The cysts were excised alone
without a Sistrunk procedure. None of their cases recurred.
­Thymic Cyst

­Nasopharyngeal Cysts Thymic cysts are rare clinical entities that arise in persis-
tent thymic tissue. The thymus develops from the pharyn-
Nasopharyngeal cysts are cystic entities that arise high in geal pouches of the third branchial arches and descends
the nasopharynx and can be classified as congenital or caudally into the upper chest. Thymic cysts arise from rem-
acquired. All are rare and do not present problems in the nants of thymic tissue, can be found in any location
differential diagnosis of the developmental cysts of the between the angle of the mandible and the sternal notch,
neck and maxillofacial regions discussed in this chapter. and can be midline or lateral. About 70% present in the first
Nasopharyngeal cysts have been well reviewed by Marom decade and the remainder are usually diagnosed before the
et  al. (2009) and Wenig (2017b). Acquired cysts are most age of 30 years.
common and are retention cysts associated with seromu- Thymic cysts are rarely diagnosed clinically, but are a
cous glands or tonsillar tissue. They may be located in the chance finding on histological examination of a cystic mass
midline or laterally. They are lined by ciliated or non-­ that has provisionally been diagnosed as another type of
ciliated columnar epithelium, with areas of squamous developmental cyst, usually a branchial or thyroglossal
metaplasia in response to inflammatory stimuli. Lymphoid duct cyst (Luna and Pfaltz 2009). On histological examina-
follicles are often present in the wall. The cyst lumen con- tion the cyst is lined by squamous or simple cuboidal epi-
tains mucoid material and epithelial debris. thelium, but by definition, and to make the diagnosis, the
Congenital cysts in the nasopharynx are found in the cyst wall contains thymic tissue including lymphoid tissue
midline and may arise either from the pharyngeal bursa and Hassall corpuscles.
309

Bibliography

Abaza, N.A., Gold, L., and Lally, E. (1989). Granular cell Agaram, N.P., Collins, B.M., Barnes, L. et al. (2004).
odontogenic cyst: a unicystic ameloblastoma with late Molecular analysis to demonstrate that odontogenic
recurrence as follicular ameloblastoma. Journal of Oral keratocysts are neoplastic. Archives of Pathology and
and Maxillofacial Surgery 47: 168–175. Laboratory Medicine 128: 313–317.
Abdel-­Wanis, M.E. and Tsuchiya, H. (2002). Simple bone cyst Aguilo, L., Cibrian, R., Bagan, J.V., and Gandia, J.L. (1998).
is not a single entity: point of view based on a literature Eruption cysts: retrospective clinical study of 36 cases.
review. Medical Hypotheses 58: 87–91. Journal of Dentistry for Children 65: 102–106.
Abdoola, I., Munzhelele, I.T., and Ibrahim, M. (2020). Augustine, D., Rao, R.S., Lakshminarayana, S., Prasad, K.,
Extensive mandibular odontogenic keratocysts associated Patil, S. (2021). Sub-epithelial hyalinization, incomplete
with basal cell nevus syndrome treated with Carnoy’s cystic lining, and corrugated surface could be a predictor of
solution versus marsupialization. Annals of Maxillofacial recurrence in Odontogenic Keratocysts. Journal of Oral
Surgery 10: 47–50. Biology and Craniofacial Research 11: 423–429.
Abé, T., Maruyama, S., Yamazaki, M. et al. (2014). Ahlfors, E., Larsson, A., and Sjogren, S. (1984). The
Intramuscular keratocyst as a soft tissue counterpart of odontogenic keratocyst: a benign cystic tumor? Journal of
keratocystic odontogenic tumor: differential diagnosis Oral and Maxillofacial Surgery 42: 10–19.
by immunohistochemistry. Human Pathology 45: Ahn, S.K., Won, J.H., Lee, S.H. et al. (1996). Lymphoepithelial
110–118. cyst associated with epithelial inclusion cyst. American
Abiko, Y., Murata, M., Ito, Y. et al. (2001). Immunohistochemical Journal of Dermatopathology 18: 424–426.
localization of amelogenin in human odontogenic tumors, Ahn, S.G., Kim, S.A., Kim, S.G. et al. (2008). Beta-­catenin
using a polyclonal antibody against bovine amelogenin. gene alterations in a variety of so-­called calcifying
Medical Electron Microscopy 34: 185–189. odontogenic cysts. APMIS 116: 206–211.
Abrams, A.M., Howell, F.V., and Bullock, W.K. (1963). Ahuja, G., Aulakh, K., Sethi, E. et al. (2016). Keratocystoma of
Nasopalatine cysts. Oral Surgery, Oral Medicine, and Oral minor salivary gland of the palate. The Saint’s International
Pathology 16: 300–332. Dental Journal 2: 21. https://doi.org/10.4103/2454-­3160.202125.
Abu-­Id, M.H., Kreusch, T., and Bruschke, C. (2005). Al-­Amery, S.M., Nambiar, P., Jamaludin, M. et al. (2015).
Glandular odontogenic cyst of the mandible. Mund-­, Cone beam computed tomography assessment of the
Kiefer-­und Gesichtschirurgie 9: 188–192. (English abstract maxillary incisive canal and foramen: considerations of
of article in German). anatomical variations when placing immediate implants.
Ackermann, G., Cohen, M., and Altini, M. (1987). The PLoS One 1: e0117251.
paradental cyst: a clinicopathologic study of 50 cases. Oral Aldawood, Z., Moyer, D.J., and Woo, S.B. (2021). A
Surgery, Oral Medicine, and Oral Pathology 64: 308–312. bronchogenic cyst masquerading as a tongue mass. Head
Adachi, P., Soubhia, A.M., Horikawa, F.K., and Shinohara, and Neck Pathology https://doi.org/10.1007/
E.H. (2011). Mucocele of the glands of Blandin-­ s12105-­021-­01303-­x. Epub ahead of print. PMID: 33638760.
Nuhnclinical, pathological, and therapeutical aspects. Oral Aldred, M.J., Talacko, A.A., Allan, P.G., and Shear, M. (2002).
and Maxillofacial Surgery 15: 11–13. Odontogenic cyst with verrucous proliferation. Journal of
Adams, A., Mankad, K., Offiah, C., and Childs, L. (2016). Oral Pathology and Medicine 31: 500–503.
Branchial cleft anomalies: a pictorial review of Ali, M.A. (2011). Biopsied jaw lesions in Kuwait: a six-­year
embryological development and spectrum of imaging retrospective analysis. Medical Principles and Practice
findings. Insights Into Imaging 7: 69–76. 20: 550–555.

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
310 Bibliography

Ali, M. and Baughman, R.A. (2003). Maxillary odontogenic Al-­Moraissi, E.A., Dahan, A.A., Alwadeai, M.S. et al. (2017).
keratocyst: a common and serious clinical misdiagnosis. What surgical treatment has the lowest recurrence rate
Journal of the American Dental Association (1939) following the management of keratocystic odontogenic
134: 877–883. tumor? A large systematic review and meta-­analysis.
Al-­Khateeb, T.H. and Al Zoubi, F. (2007). Congenital neck Journal of Cranio-­Maxillo-­Facial Surgery 45: 131–144.
masses: a descriptive retrospective study of 252 cases. Alsharif, M.J. and Zhao, Y. (2009). Teratoid cyst of floor of the
Journal of Oral and Maxillofacial Surgery 65: 2242–2247. mouth: a clinicopathologic study of 20 Chinese patients.
Al-­Khateeb, T.H. and Bataineb, A.B. (2006). Pathology International Journal of Surgical Pathology 17: 111–115.
associated with impacted mandibular third molars in a Al-­Talabani, N.G. and Smith, C.J. (1980). Experimental
group of Jordanians. Journal of Oral and Maxillofacial dentigerous cysts and enamel hypoplasia: their possible
Surgery 64: 1598–1602. significance in explaining the pathogenesis of human
Allard, R.H. (1982). Nasolabial cyst: review of the literature dentigerous cysts. Journal of Oral Pathology 9: 82–91.
and report of 7 cases. International Journal of Oral Surgery Altini, M. and Cohen, M. (1982). The follicular primordial
11: 351–359. cyst (odontogenic keratocyst). International Journal of Oral
Allard, R.H.B., van der Kwast, W.A.M., and van der WaaI, Surgery 11: 175–182.
I. (1981a). Nasopalatine duct cyst. Review of the literature Altini, M. and Cohen, M. (1987). Experimental extra-­
and report of 22 cases. International Journal of Oral follicular histogenesis of follicular cysts. Journal of Oral
Surgery 10: 447–461. Pathology 16: 49–52.
Allard, R.H.B., van der Kwast, W.A.M., and van der Waal, Altini, M. and Shear, M. (1992). The lateral periodontal cyst:
I. (1981b). Mucosal antral cysts. Review of the literature an update. Journal of Oral Pathology and Medicine
and report of a radiographic survey. Oral Surgery, Oral 21: 245–250.
Medicine, and Oral Pathology 51: 2–9. Altini, M., Coleman, H., Doglioni, C. et al. (2000). Calretinin
Allison, R.T. (1974). Electron microscopic study of ‘Rushton’ expression in ameloblastomas. Histopathology 37: 27–32.
hyaline bodies in cyst linings. British Dental Journal 137: Álvares, P.R., Arruda, J.A.A., Silva, L.P.D. et al. (2017).
102–104. Immunohistochemical expression of TGF-­β1 and
Allison, R.T. (1977a). Microprobe and microradiographic MMP-­9 in periapical lesions. Brazilian Journal of Oral
studies of hyaline bodies in odontogenic cysts. Journal of Research 31: e51.
Oral Pathology 6: 44–50. Alves, D.B.M., Tuji, F.M., Alves, F.A. et al. (2018). Evaluation
Allison, R.T. (1977b). Hyalin material bounding dystrophic of mandibular odontogenic keratocyst and ameloblastoma
calcification in the epithelial lining of odontogenic cysts. by panoramic radiograph and computed tomography.
Journal of Oral Pathology 6: 113–121. Dento Maxillo Facial Radiology 47: 20170288.
Allison, J. and Garlington, G. (2017). The value of cone beam Amin, M., Witherow, H., Lee, R., and Blenkinsopp, P. (2003).
computed tomography in the management of dentigerous Surgical ciliated cyst after maxillary orthognathic surgery:
cysts – a review and case report. Dental Update 44: 182–188. report of a case. Journal of Oral and Maxillofacial Surgery
Allmendinger, A., Gabe, M., and Destian, S. (2009). Median 61: 138–141.
palatine cyst. Journal of Radiology Case Reports 3: 7–10. Anastassov, G.E. and Lee, H. (1999). Respiratory mucocele
Almazyad, A., Li, C.C., and Woo, S.B. (2020). Benign alveolar formation after augmentation genioplasty with nasal
ridge keratosis: clinical and histopathologic analysis of 167 osteocartilaginous graft. Journal of Oral and Maxillofacial
cases. Head and Neck Pathology 14: 915–922. Surgery 57: 1263–1265.
Al-­Moraissi, E.A., Pogrel, M.A., and Ellis, E. (2016a). Anastassov, G.E., Haiavy, J., Solodnik, P. et al. (2000).
Enucleation with or without adjuvant therapy versus Submandibular gland mucocele. Diagnosis and
marsupialization with or without secondary enucleation in management. Oral Surgery, Oral Medicine, Oral Pathology,
the treatment of keratocystic odontogenic tumors: a Oral Radiology, and Endodontics 89: 159–163.
systematic review and meta-­analysis. Journal of Cranio-­ Andiran, N., Sarikayalar, F., Unal, O.F. et al. (2001). Mucocele
Maxillo-­Facial Surgery 44: 1395–1403. of the anterior lingual salivary glands: from extravasation
Al-­Moraissi, E.A., Pogrel, M.A., and Ellis, E. (2016b). Does to an alarming mass with a benign course. International
the excision of overlying oral mucosa reduce the Journal of Pediatric Otorhinolaryngology 61: 143–147.
recurrence rate in the treatment of the keratocystic Andrade, A.L., Santos, E.M., Carmo, A.F. et al. (2017).
odontogenic tumor? A systematic review and meta-­ Analysis of tryptase-­positive mast cells and
analysis. Journal of Oral and Maxillofacial Surgery immunoexpression of MMP-­9 and MMP-­13 in periapical
74: 1974–1982. lesions. International Endodontic Journal 50: 446–454.
Bibliography 311

Anneroth, G., Hall, G., and Stuge, U. (1986). Nasopalatine odontogenic cysts with mucous prosoplasia: a pilot study.
duct cyst. International Journal of Oral and Maxillofacial Histopathology 66: 791–797.
Surgery 15: 572–580. Argyris, P.P., Wetzel, S.L., Pambuccian, S.E. et al. (2016).
Anthonappa, R.P., Ekambaram, M., Neboda, C.N.G. et al. Primordial odontogenic cyst with induction phenomenon
(2018). Genetic basis of dentigerous cysts associated with (zonal fibroblastic hypercellularity) and dentinoid material
supernumerary teeth: a narrative review. Journal of versus archegonous cystic odontoma: you choose! Head
Investigative and Clinical Dentistry 9: e12267. and Neck Pathology 10: 237–244.
Antonoglou, G.N., Sándor, G.K., Koidou, V.P., and Ariji, E., Fujiwara, N., Tabata, O. et al. (1993). Stafne’s bone
Papageorgiou, S.N. (2014). Non-­syndromic and syndromic cavity. Classification based on outline and content
keratocystic odontogenic tumors: systematic review and determined by computed tomography. Oral Surgery, Oral
meta-­analysis of recurrences. Journal of Cranio-­Maxillo-­ Medicine, and Oral Pathology 76: 375–380.
Facial Surgery 42: e364–e371. Arora, S.S., Paul, S., Arora, S., and Kapoor, V. (2014).
Aps, J.K.M., Koelmeyer, N., and Yaqub, C. (2020). Stafne’s Secondary jaw aneurysmal bone cyst (JABC) – a possible
bone cyst revisited and renamed: the benign mandibular misnomer? A review of literature on secondary JABCs,
concavity. Dento Maxillo Facial Radiology 49: 20190475. their pathogenesis and oncogenesis. Journal of Oral
Aquilanti, L., Mascitti, M., Togni, L. et al. (2021). Non-­ Pathology and Medicine 43: 647–651.
neoplastic jaw cysts: a 30-­year epidemiological study of Arwill, T. and Heyden, G. (1973). Histochemical studies on
2150 cases in the Italian population. British Journal of Oral cholesterol formation in odontogenic cysts and
and Maxillofacial Surgery 59: 168–173. granulomas. Scandinavian Journal of Dental Research 81:
Aragaki, T., Michi, Y., Katsube, K. et al. (2010). Comprehensive 406–410.
keratin profiling reveals different histopathogenesis of Arwill, T. and Kahnberg, K.E. (1977). Odontogenic keratocyst
keratocystic odontogenic tumor and orthokeratinized associated with an intramandibular chondroma. Journal of
odontogenic cyst. Human Pathology 41: 1718–1725. Oral Surgery 35: 64–67.
Arashiyama, T., Kodama, Y., Kobayashi, T. et al. (2012). Ghost Assaf, A.T., Solaty, M., Zrnc, T.A. et al. (2014). Prevalence of
cell odontogenic carcinoma arising in the background of a Stafne’s bone cavity–retrospective analysis of 14,005
benign calcifying cystic odontogenic tumor of the panoramic views. In vivo 28: 1159–1164.
mandible. Oral Surgery, Oral Medicine, Oral Pathology, Ataoglu, H., Uckan, S., Oz, G., and Altinors, N. (2002).
Oral Radiology 114: e35–e40. Maxillofacial hydatid cyst. Journal of Oral and
Arcand, P., Granger, J., and Brochu, P. (1988). Congenital Maxillofacial Surgery 60: 454–456.
dermoid cyst of the oral cavity with gastric choristoma. Atkinson, M.E. (1972). A histological study of tooth grafts in an
Journal of Otolaryngology 17: 219–222. inbred strain of mice. Journal of Oral Pathology 1: 115–124.
Arendorf, T. (1981). Prostaglandin E2 and bone and root Atkinson, M.E. (1976). A histological study of odontogenic
resorption in hamsters. Journal of the Dental Association of cysts formed following mouse molar tooth transplantation.
South Africa 36: 317–322. Journal of Oral Pathology 5: 347–357.
Aresta, A., Girolami, I., Paolino, G. et al. (2019). Atkinson, M.E. (1977). An autoradiographic study of
Keratocystoma of the parotid gland is rare and requires an experimental odontogenic cyst formation in the mouse.
integrated multidisciplinary approach: report of a case and Journal of Oral Pathology 6: 382–386.
a literature review. Diagnostic Cytopathology 47: 614–616. August, M., Faquin, W.C., Troulis, M., and Kaban,
Argyris, P.P. and Koutlas, I.G. (2017). Orthokeratinized L.B. (2000). Differentiation of odontogenic keratocyst from
odontogenic cyst with an associated keratocystic nonkeratinizing cysts by use of fine-­needle aspiration
odontogenic tumor component and ghost cell biopsy and cytokeratin-­10 staining. Journal of Oral and
keratinization and calcifications in a patient with gardner Maxillofacial Surgery 58: 945–940.
syndrome. Head and Neck Pathology 11: 240–248. August, M., Faquin, W.C., Troulis, M.J., and Kaban,
Argyris, P.P., Nelson, A.C., and Koutlas, I.G. (2015a). L.B. (2003). Dedifferentiation of odontogenic keratocyst
Keratinizing odontogenic cyst with verrucous pattern epithelium after cyst decompression. Journal of Oral and
featuring negative human papillomavirus status by Maxillofacial Surgery 61: 678–683.
polymerase chain reaction. Oral Surgery, Oral Medicine, Azevedo, R.S., Cabral, M.G., dos Santos, T.C. et al. (2012).
Oral Pathology, Oral Radiology 119: e233–e240. Histopathological features of keratocystic odontogenic
Argyris, P.P., Wehrs, R.N., García, J.J., and Koutlas, tumor: a descriptive study of 177 cases from a Brazilian
I.G. (2015b). Fluorescence in-­situ hybridization identifies population. International Journal of Surgical Pathology
Mastermind-­like 2 (MAML2) rearrangement in 20: 154–160.
312 Bibliography

Azzie, G. and Beasley, S. (2003). Diagnosis and treatment of Barrett, A.W., Sneddon, K.J., Tighe, J.V. et al. (2017).
foregut duplications. Seminars in Pediatric Surgery Dentigerous cyst and ameloblastoma of the jaws:
12: 46–54. correlating the histopathological and clinicoradiological
Babal, P., Soler, P., Brozman, M. et al. (1987). In situ features avoids a diagnostic pitfall. International Journal of
characterization of cells in periapical granuloma by Surgical Pathology 25: 141–147.
monoclonal antibodies. Oral Surgery, Oral Medicine, and Barrett, A.W., Garg, M., Armstrong, D. et al. (2020). Cystic
Oral Pathology 64: 348–352. squamous cell carcinomas of the jaws: twelve cases
Bacci, C., Valente, M.L., Quadrio, M., and Berengo, M. (2011). highlighting histopathological pitfalls. International
Is the median palatine cyst a distinct entity? Journal of Journal of Surgical Pathology 28: 624–630.
Oral and Maxillofacial Surgery 69: 1385–1389. Barros, C.C.D.S., Santos, H.B.P., Cavalcante, I.L. et al. (2018).
Bale, A.E., Gailani, M.R., and Leffel, D.J. (1995). The Gorlin Clinical and histopathological features of nasopalatine
syndrome gene: a tumour suppressor active in basal cell duct cyst: a 47-­year retrospective study and review of
carcinogenesis and embryonic development. Proceedings of current concepts. Journal of Cranio-­Maxillo-­Facial Surgery
the Association of American Physicians 107: 253–257. 46: 264–268.
Balikci, H.H., Gurdal, M.M., Ozkul, M.H. et al. (2013). Neck Bascones, J. and Llanes, F. (2005). Clear cells in epithelial
masses: diagnostic analysis of 630 cases in Turkish rests of Malassez. Oral Oncology 41: 99–100.
population. European Archives of Oto-­Rhino-­Laryngology Bastos, V.C., Gomez, R.S., and Gomes, C.C. (2021). Revisiting
270: 2953–2958. the human dental follicle: from tooth development to its
Bando, Y., Henderson, B., Meghi, S. et al. (1993). association with unerupted or impacted teeth and
Immunocytochemical localization of inflammatory pathological changes. Developmental Dynamics https://doi.
cytokines and vascular adhesion receptors in radicular org/10.1002/dvdy.406. Epub ahead of print. PMID: 34324745.
cysts. Journal of Oral Pathology and Medicine 22: 221–227. Basu, M.K., Rout, P.G.J., Rippen, J.W., and Smith, A.J. (1988).
Baqain, Z.H., Jayakrishnan, A., Farthing, P.M., and Hardee, The post-­operative maxillary cyst Experience with 23 cases.
P. (2005). Recurrence of a solitary bone cyst of the International Journal of Oral and Maxillofacial Surgery 17:
mandible: case report. British Journal of Maxillofacial 282–284.
Surgery 43: 333–335. Batsakis, J.G. and Raymond, A.K. (1989). Sialocysts of the
Barker, G.R. (1988). A radiolucency of the ascending ramus parotid glands. Annals of Otology, Rhinology and
of the mandible associated with invested parotid salivary Laryngology 98: 487–489.
gland material and analogous with a Stafne bone cavity. Batsakis, J.G., Bruner, J.M., and Luna, M.A. (1988). Polycystic
British Journal of Oral and Maxillofacial Surgery (dysgenetic) disease of the parotid glands. Archives of
26: 81–84. Otolaryngology – Head and Neck Surgery 114: 1146–1148.
Barker, B.F., Jensen, J.L., and Howell, F.V. (1974). Focal Bauer, W.H. (1943). Histogenesis of the so-­called secondary
osteoporotic bone marrow defects of the jaws. An analysis enamel cuticle and dental cuticle. American Journal of
of 197 new cases. Oral Surgery, Oral Medicine, and Oral Orthodontics and Oral Surgery 29: 626–634.
Pathology 38: 404–413. Baumhoer, D., Smida, J., Nathrath, M., and Jundt, G. (2011).
Barnes, L., Eveson, J.W., Reichart, P., and Sidransky, D. (2005). The nature of the characteristic cementum-­like matrix
World Health Organization Classification of Tumours. deposits in the walls of simple bone cysts. Histopathology
Pathology and Genetics of Head and Neck Tumours. Lyon: 59: 390–396.
IARC Press. Baurmash, H. (2001). Treating oral ranula: another case
Barreto, D.C., Gomez, R.S., Bale, A.E. et al. (2000). PTCH against blanket removal of the sublingual gland. British
gene mutations in odontogenic keratocysts. Journal of Journal of Oral and Maxillofacial Surgery 39: 217–220.
Dental Research 79: 1418–1422. Baurmash, H. (2003). Mucoceles and ranulas. Journal of Oral
Barreto, D.C., De Marco, L., Castro, W.H., and Gomez, R.S. (2001). and Maxillofacial Surgery 61: 369–378.
Glandular odontogenic cyst: absence of PTCH gene mutation. Bautista, C.R.G., Milhan, N.V.M., Ankha, M.D.V.E.A. et al.
Journal of Oral Pathology and Medicine 30: 125–128. (2019). Bilateral mandibular buccal bifurcation cyst: a case
Barreto, D.C., Bale, A.E., De Marco, L., and Gomez, R.S. (2002). report emphasizing the role of imaging examination in the
Immunolocalization of PTCH protein in odontogenic cysts diagnosis. Autopsy and Case Reports 11: e2018073.
and tumors. Journal of Dental Research 81: 757–760. Beasley, J.D. (1976). Traumatic cyst of the jaws: report of
Barrett, A.W., Abdullakutty, A., Norris, P.M. et al. (2016). 30 cases. Journal of the American Dental Association 92:
Molecular diagnostics in the differential diagnosis of 145–152.
glandular odontogenic cysts and mucoepidermoid Belibasakis, G.N., Rechenberg, D.K., and Zehnder, M. (2013).
carcinoma-­case reports. Oral Surgery 9: 193–200. The receptor activator of NF-­κB ligand-­osteoprotegerin
Bibliography 313

system in pulpal and periapical disease. International at an oral pathology referral center during a 43 year period.
Endodontic Journal 46: 99–111. Brazilian Journal of Otorhinolaryngology 82: 536–542.
Bell, R.B. and Dierks, E.J. (2003). Treatment options for the Bhakhar, V.P., Shah, V.S., Ghanchi, M.J. et al. (2016). A
recurrent odontogenic keratocyst. In: The Odontogenic Comparative analysis of cytokeratin 18 and 19 expressions
Keratocyst, Oral and Maxillofacial Surgery Clinics of North in odontogenic keratocyst, dentigerous cyst and radicular
America, vol. 15 (eds. M.A. Pogrel and B.L. Schmidt), cyst with a review of literature. Journal of Clinical and
429–446. Philadelphia, PA: W.B. Saunders. Diagnostic Research 10: ZC85–ZC89.
Bell, R.C., Chauvin, P.J., and Tyler, M.T. (1997). Gingival cyst Bhargava, D., Deshpande, A., and Pogrel, M.A. (2012).
of the adult: a review and a report of eight cases. Journal of Keratocystic odontogenic tumour (KCOT)–a cyst to a
the Canadian Dental Association 63: 533–535. tumour. Oral and Maxillofacial Surgery 16: 163–170.
Bell, D., Holsinger, C.F., and El-­Naggar, A.K. (2010). Bhaskar, S.N. (1972). Nonsurgical resolution of radicular
CRTC1/MAML2 fusion transcript in central cysts. Oral Surgery, Oral Medicine, and Oral Pathology 34:
mucoepidermoid carcinoma of mandible–diagnostic and 458–468.
histogenetic implications. Annals of Diagnostic Pathology Bhaskar, S.N. and Bernier, J.L. (1959). Histogenesis of
14: 396–401. branchial cysts: a report of 468 cases. American Journal of
Bell, D., Lewis, C., El-­Naggar, A.K., and Weber, R.S. (2016). Pathology 35: 407–423.
Primary intraosseous mucoepidermoid carcinoma of the Bhaskar, S.N., Bolden, T.E., and Weinmann, J.P. (1956).
jaw: reappraisal of The MD Anderson Cancer Center Pathogenesis of mucoceles. Journal of Dental Research 35:
experience. Head and Neck 38 (Suppl. 1): e1312–e1317. 863–874.
Belli, E., Renzi, G., Balestra, F.M. et al. (2004). Bilateral Bhat, A., Mitra, S., Chandrashekar, C. et al. (2019).
parotid voluminous masses: a case report. Journal of Odontogenic cysts and odontogenic tumors in a large rural
Craniofacial Surgery 15: 165–169. area from India. A 10-­year reflection. Medicine and
Bello, I.O. (2016). Keratocystic odontogenic tumor: a biopsy Pharmacy Reports 92: 408–412.
service’s experience with 104 solitary, multiple and Bhattacharyya, N. (2000). Do maxillary sinus retention cysts
recurrent lesions. Medicina Oral, Patología Oral y Cirugía reflect obstructive sinus phenomena? Archives of
Bucal 21: e538–e546. Otolaryngology – Head and Neck Surgery 126: 1369–1371.
Benhammou, A., Benyoussef, N., Bencheikhe, R. et al. (2006). Bilodeau, E.A. and Seethala, R.R. (2019). Update on
Bronchogenic cyst of the tongue. Annales d’Oto-­ odontogenic tumors: proceedings of the North American
Laryngologie et de Chirurgie Cervico-­Faciale 123: 148–151. Head and Neck Pathology Society. Head and Neck
Benn, A. and Altini, M. (1996). Dentigerous cysts of Pathology 13: 457–465.
inflammatory origin. A clinicopathologic study. Oral Bilodeau, E.A., Acquafondata, M., Barnes, E.L., and Seethala,
Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and R.R. (2015). A comparative analysis of LEF-­1 in odontogenic
Endodontics 81: 203–209. and salivary tumors. Human Pathology 46: 255–259.
Berg, O., Carenfelt, C., and Sobin, A. (1989). On the diagnosis Binkley, G.W. and Johnson, H.H. (1951). Epithelioma
and pathogenesis of intramural maxillary cysts. Acta adenoides cysticum: basal cell nevi, agenesis of the corpus
Otolaryngology (Stockholm) 108: 464–468. callosum and dental cysts. American Medical Association
Berge, T.I., Helland, S.B., Sælen, A. et al. (2016). Pattern of Archives of Dermatology and Syphilology 63: 73–84.
recurrence of nonsyndromic keratocystic odontogenic Binnie, W.H. and Rowe, A.H.R. (1974). The incidence of
tumors. Oral Surgery, Oral Medicine, Oral Pathology, Oral epithelial rests, proliferations and apical periodontal cysts
Radiology 122: 10–16. following root canal treatment in young dogs. British
Bermejo, A., Aguirre, J.M., Lopez, P., and Saez, M.R. (1999). Dental Journal 137: 56–60.
Superficial mucocele: report of 4 cases. Oral Surgery, Oral Bishop, J.A. and Thompson, L.D.R. (2021). Sclerosing
Medicine, Oral Pathology, Oral Radiology, and Endodontics Polycystic Adenoma. Surgical Pathology Clinics 14: 17–24.
88: 469–472. Bishop, J.A. and Westra, W.H. (2015). Ciliated HPV-­related
Bernardi, L., Visioli, F., Nör, C., and Rados, P.V. (2015). carcinoma: a well-­differentiated form of head and neck
Radicular cyst: an update of the biological factors related carcinoma that can be mistaken for a benign cyst.
to lining epithelium. Journal of Endodontics 41: 1951–1961. American Journal of Surgical Pathology 39: 1591–1595.
Bernier, J.L. and Bhaskar, S.N. (1958). Lymphoepithelial Bishop, J.A., Yonescu, R., Batista, D. et al. (2014). Glandular
lesions of salivary glands; histogenesis and classification odontogenic cysts (GOCs) lack MAML2 rearrangements: a
based on 186 cases. Cancer 11: 1156–1179. finding to discredit the putative nature of GOC as a
Bezerra, T.M., Monteiro, B.V., Henriques, Á.C. et al. (2016). precursor to central mucoepidermoid carcinoma. Head
Epidemiological survey of mucus extravasation phenomenon and Neck Pathology 8: 287–290.
314 Bibliography

Bishop, J.A., Gagan, J., Baumhoer, D. et al. (2020). Sclerosing International Journal of Paediatric Otorhinolaryngology
polycystic “adenosis” of salivary glands: a neoplasm 38: 71–75.
characterized by PI3K pathway alterations more correctly Borghesi, A., Nardi, C., Giannitto, C. et al. (2018).
named sclerosing polycystic adenoma. Head and Neck Odontogenic keratocyst: imaging features of a benign
Pathology 14: 630–636. lesion with an aggressive behaviour. Insights into Imaging
Bishop, J.A., Losito, N.S., Petersson, B.F. et al. (2022). 9: 883–897.
Sclerosing polycystic adenoma. In: WHO Classification of Bornstein, M.M., Balsiger, R., Sendi, P., and von Arx,
Tumours Editorial Board. Head and Neck Tumours, T. (2011). Morphology of the nasopalatine canal and dental
Section 4.0.2.16 (WHO Classification of Tumours Series), implant surgery: a radiographic analysis of 100 consecutive
5e, vol. 9. Lyon: International Agency for Research on patients using limited cone-­beam computed tomography.
Cancer https://publications.iarc.fr/Book-­And-­Report-­ Clinical Oral Implants Research 22: 295–301.
Series/Who-­Classification-­Of-­Tumours. Borrás-­Ferreres, J., Sánchez-­Torres, A., and Gay-­Escoda,
Blackwell, K.A., Raisz, L.G., and Pilbeam, C.C. (2010). C. (2016). Malignant changes developing from odontogenic
Prostaglandins in bone: bad cop, good cop? Trends in cysts: a systematic review. Journal of Clinical and
Endocrinology and Metabolism 21: 294–301. Experimental Dentistry 8: e622–e628.
Blanas, N., Freund, B., Schwartz, M., and Furst, I.M. (2000). Bouckaert, M.M., Raubenheimer, E.J., and Jacobs, F.J. (2000).
Systematic review of the treatment and prognosis of the Maxillofacial hydatid cysts. Oral Surgery, Oral Medicine,
odontogenic keratocyst. Oral Surgery, Oral Medicine, Oral Oral Pathology, Oral Radiology, and Endodontics 89:
Pathology, Oral Radiology, and Endodontics 90: 553–558. 338–342.
Blum, T. (1929). Do all cysts in the jaws originate from the Bouquot, J.E. and McMahon, R.E. (2003). Charlatans in
dental system? Journal of the American Dental Association dentistry: ethics of the NICO wars. Journal of the American
16: 647–661. College of Dentists 70: 38–41.
Blum, T. (1932). Unusual bone cavities in the mandible: a Bouquot, J.E., Roberts, A.M., Person, P., and Christian,
report of three cases of traumatic bone cysts. Journal of the J. (1992). Neuralgia-­inducing cavitational osteonecrosis
American Dental Association 19: 281–301. (NICO). Osteomyelitis in 224 jawbone samples from
Bodin, I., Isacsson, G., and Julin, P. (1986). Cysts of the patients with facial neuralgia. Oral Surgery, Oral Medicine,
nasopalatine duct. International Journal of Oral and and Oral Pathology 73: 307–319.
Maxillofacial Surgery 15: 696–706. Bouquot, J.E., Gnepp, D.R., Dardick, I., and Hietanen,
Bodner, L. (2002). Cystic lesions of the jaws in children. J.H. (2000). Intraosseous salivary tissue: jawbone examples
International Journal of Pediatric Otorhinolaryngology of choristomas, hamartomas, embryonic rests, and
62: 25–29. inflammatory entrapment: another histogenetic source for
Bodner, L., Goldstein, J., and Sarnat, H. (2005). Eruption intraosseous adenocarcinoma. Oral Surgery, Oral Medicine,
cysts: a clinical report of 24 new cases. Journal of Clinical Oral Pathology, Oral Radiology, and Endodontics 90:
Pediatric Dentistry 28: 183–185. 205–217.
Bodner, L., Manor, E., Shear, M., and van der Waal, I. (2011). Bourgeois, S.L. and Nelson, B.L. (2005). Surgical ciliated cyst
Primary intraosseous squamous cell carcinoma arising in of the mandible secondary to simultaneous Le Fort I
an odontogenic cyst: a clinicopathologic analysis of 116 osteotomy and genioplasty: report of case and review of
reported cases. Journal of Oral Pathology and Medicine the literature. Oral Surgery, Oral Medicine, Oral Pathology,
40: 733–738. Oral Radiology, and Endodontics 100: 36–39.
Bodner, L., Manor, E., Joshua, B.Z., and Shaco-­Levy, Boussouni, S., Laporte, C., Gaudinat, M. et al. (2020).
R. (2015). Oral mucoceles in children—­analysis of 56 new Bilateral mandibular dentigerous cysts in a non-­syndromic
cases. Pediatric Dermatology 32: 647–650. patient: comprehensive review of the literature and case
Boffano, P., Ruga, E., and Gallesio, C. (2010). Keratocystic report. French Journal of Dental Medicine: 1–10.
odontogenic tumor (odontogenic keratocyst): preliminary Bouyssou, M. and Guilhem, A. (1965). Recherches
retrospective review of epidemiologic, clinical, and morphologiques et histochimiques sur les corps hyalins
radiologic features of 261 lesions from University of Turin. intrakystiques de Rushton. Bulletin du Groupement
Journal of Oral and Maxillofacial Surgery 68: 2994–2999. International pour la Recherche Scientifique en Stomatologie
Bolbaran, V., Martinez, B., and Rojas, R. (2000). Odontogenic et Odontologie (Bruxelles) 8: 81–104.
keratocysts. A retrospective study of 285 cases. (I. Clinical Bramley, P.A. (1971). Treatment of cysts of the jaws.
aspects). Medicina Oral 5: 331–337. Proceedings of the Royal Society of Medicine 64: 547–550.
Bonilla, J.A., Szeremeta, W., Yellon, R.F., and Nazif, Brannon, R.B. (1976). The odontogenic keratocyst. A
M.M. (1996). Teratoid cyst of the floor of the mouth. clinicopathologic study of 312 cases. Part 1. Clinical
Bibliography 315

features. Oral Surgery, Oral Medicine, and Oral Pathology Surgery, Oral Medicine, Oral Pathology, Oral Radiology
42: 54–72. 129: 192–199.
Brannon, R.B. (1977). The odontogenic keratocyst. A Buchner, A. (1973). Granular cell odontogenic cyst. Oral
clinicopathologic study of 312 cases. Part II. Histological Surgery, Oral Medicine, and Oral Pathology 36: 707–712.
features. Oral Surgery, Oral Medicine, and Oral Pathology Buchner, A. (1991). The central (intraosseous) calcifying
43: 233–255. odontogenic cyst: an analysis of 215 cases. Journal of Oral
Bree, A.F., Shah, M.R., and BCNS Colloquium Group (2011). and Maxillofacial Surgery 49: 330–339.
Consensus statement from the first international colloquium Buchner, A. and David, R. (1978). Lipopigment in
on basal cell nevus syndrome (BCNS). American Journal of odontogenic cysts. Journal of Oral Pathology 7: 311–317.
Medical Genetics. Part A 155A: 2091–2097. Buchner, A. and Hansen, L.S. (1979). The histomorphologic
Brooks, P.J., Chadwick, J.W., Caminiti, M. et al. (2019). spectrum of the gingival cyst in the adult. Oral Surgery,
Primary aneurysmal bone cyst of the mandibular condyle Oral Medicine, and Oral Pathology 48: 532–539.
with USP6-­CDH11 fusion. Pathology, Research and Practice Buchner, A. and Hansen, L.S. (1980). Lymphoepithelial cysts
215: 607–610. of the oral cavity. A clinicopathologic study of 38 cases. Oral
Brown, P. (1990). Unnecessary X-­rays blamed for cancer Surgery, Oral Medicine, and Oral Pathology 50: 441–449.
deaths. New Scientist 127 (1733): 27. Buchner, A., Carpenter, W.M., Merrell, P.W., and Leider,
Brown, L.H., Berkman, S., Cohen, D. et al. (1982). A A.S. (1991). Anterior lingual mandibular salivary gland
radiological study of the frequency and distribution of defect. Evaluation of twenty-­four cases. Oral Surgery, Oral
impacted teeth. Journal of the Dental Association of South Medicine, and Oral Pathology 71: 131–136.
Africa 37: 627–630. Buchner, A., Merrell, P.W., Hansen, L.S., and Leider,
Browne, R.M. (1970). The odontogenic keratocyst: clinical A.S. (1991). Peripheral (extraosseous) calcifying
aspects. British Dental Journal 128: 225–231. odontogenic cyst. A review of forty-­five cases. Oral Surgery,
Browne, R.M. (1971a). The odontogenic keratocyst-­ Oral Medicine, and Oral Pathology 72: 65–70.
histological features and their correlation with clinical Buchner, A., David, R., Carpenter, W., and Leider, A. (1996).
behaviour. British Dental Journal 131: 249–259. Pigmented lateral periodontal cyst and other pigmented
Browne, R.M. (1971b). The origin of cholesterol in odontogenic odontogenic lesions. Oral Diseases 2: 299–302.
cysts in man. Archives of Oral Biology 16: 107–113. Buckley, F.M., Huntley, P., and Speight, P.M. (1989). A lateral
Browne, R.M. (1972). Metaplasia and degeneration in periodontal cyst in association with a follicular cyst. British
odontogenic cysts in man. Journal of Oral Pathology Dental Journal 167: 26–28.
1: 145–158. Burkart, C.M., Richter, G.T., Rutter, M.J., and Myer, C.M. 3rd.
Browne, R.M. (1975). The pathogenesis of odontogenic cysts: (2009). Update on endoscopic management of lingual
a review. Journal of Oral Pathology 4: 31–46. thyroglossal duct cysts. Laryngoscope 119: 2055–2060.
Browne, R.M. (1976). Some observations on the fluids of Burke, G.W., Feagans, W.M., Elzay, R.P., and Schwartz,
odontogenic cysts. Journal of Oral Pathology 5: 74–87. L.D. (1966). Some aspects of the origin and fate of
Browne, R.M. (1991a). Animal models of odontogenic cysts. midpalatal cysts in human fetuses. Journal of Dental
In: Investigative Pathology of the Odontogenic Cysts (ed. Research 45: 159–164.
R.M. Browne), 111–122. Boca Raton, FL: CRC Press. Busaba, N.Y. and Salman, S.D. (1999). Maxillary sinus
Browne, R.M. (1991b). The classification of odontogenic mucoceles: clinical presentation and long-­term results of
cysts. In: Investigative Pathology of the Odontogenic Cysts endoscopic surgical treatment. The Laryngoscope 109:
(ed. R.M. Browne), 1–19. Boca Raton, FL: CRC Press. 1446–1449.
Browne, R.M. and Matthews, J.B. (1985). Intra-­epithelial Caetano Ados, S., Tjioe, K.C., Faustino, S.E. et al. (2013).
hyaline bodies in odontogenic cysts: an immunoperoxidase Immunolocalization of podoplanin in benign odontogenic
study. Journal of Oral Pathology 14: 422–428. tumours with and without ectomesenchyme. Archives of
Browne, R.M. and Smith, A.J. (1991). Pathogenesis of Oral Biology 58: 408–415.
odontogenic cysts. In: Investigative Pathology of the Cahn, L.R. (1954). Comment on hematopoietic marrow in
Odontogenic Cysts (ed. R.M. Browne), 88–109. Boca Raton, the jaws. Oral Surgery 7: 332–337.
FL: CRC Press. Cai, M., Shen, G., Lu, X., and Wang, X. (2015). Two
Browne, R.M., Rowles, S.L., and Smith, A.J. (1984). mandibular surgical ciliated cysts after Le Fort I osteotomy
Mineralised deposits in odontogenic cysts. IRCS Medical and genioplasty. British Journal of Oral and Maxillofacial
Science 12: 642–643. Surgery 53: 1040–1042.
Brucoli, M., Boffano, P., Benech, A. et al. (2020). Congenital Cairo, F., Rotunda, R., and Ficarra, G. (2002). A rare lesion of
nonvascular neck masses: a retrospective analysis. Oral the periodontium: the gingival cyst of the adult: a report of
316 Bibliography

three cases. International Journal of Periodontics and imaging features based on cone beam computed
Restorative Dentistry 22: 79–84. tomography scans. Brazilian Dental Journal 29: 99–104.
Calonius, P.E.B., Hakala, P., and Rapola, J. (1974). Congenital Chadwick, J.W., Alsufyani, N.A., and Lam, E.W. (2011).
cervical cysts and fistulas. Proceedings of the Finnish Dental Clinical and radiographic features of solitary and cemento-­
Society 70: 209–216. osseous dysplasia-­associated simple bone cysts. Dento
Camarda, A.J., Pham, J., and Forest, D. (1989). Mandibular Maxillo Facial Radiology 40: 230–235.
infected buccal cyst: report of two cases. Journal of Chaisuparat, R., Sawangarun, W., and Scheper, M.A. (2012).
Maxillofacial Surgery 47: 528–534. A clinicopathological study of malignant odontogenic
Camilleri, A.C. and Lloyd, R.E. (1990). Lymphoepithelial cyst tumours. Histopathology 61: 107–112.
of the parotid gland. British Journal of Oral and Chamda, R.A. and Shear, M. (1980). Dimensions of incisive
Maxillofacial Surgery 28: 329–332. fossae on dry skulls and radiographs. International Journal
Campana, F., Sibaud, V., Chauvel, A. et al. (2006). of Oral Surgery 9: 452–457.
Recurrent superficial mucoceles associated with Chan, H.L., Oh, T.J., Bashutski, J. et al. (2010). Cervical
lichenoid disorders. Journal of Oral and Maxillofacial enamel projections in unusual locations: a case report and
Surgery 64: 1830–1833. mini-­review. Journal of Periodontology 81: 789–795.
Campbell, R. and Walker, P. (2010). Management of Chao, W.C., Huang, C.C., Chang, P.H. et al. (2009).
congenital lingual dermoid cysts. International Journal of Management of nasolabial cysts by transnasal endoscopic
Pediatric Otorhinolaryngology 74: 567–571. marsupialization. Archives of Otolaryngology – Head and
Cannell, H., Kerawala, C., and Farthing, P. (1997). Stomatitis Neck Surgery 135: 932–935.
glandularis: two confirmed cases of a rare condition. Chapelle, K.A., Stoelinga, P.J., de Wilde, P.C. et al. (2004).
British Dental Journal 182: 222–225. Rational approach to diagnosis and treatment of
Cao, D., Begum, S., Ali, S.Z., and Westra, W.H. (2010). ameloblastomas and odontogenic keratocysts. British
Expression of p16 in benign and malignant cystic Journal of Oral and Maxillofacial Surgery 42: 381–390.
squamous lesions of the neck. Human Pathology 41: Chaudhry, A.P., Reynolds, D.H., La Chapelle, C.F., and
535–539. Vickers, R.A. (1960). A clinical and experimental study of
Cao, H., Chen, X., Hou, J. et al. (2020). The Shh/Gli signaling mucocele (retention cyst). Journal of Dental Research 39:
cascade regulates myofibroblastic activation of lung-­ 1253–1262.
resident mesenchymal stem cells via the modulation of Chavez, J.A. and Richter, K.J. (1999). Glandular odontogenic
Wnt10a expression during pulmonary fibrogenesis. cyst of the mandible. Journal of Oral and Maxillofacial
Laboratory Investigation 100: 363–377. Surgery 57: 461–464.
Carrillo, C., Peñarrocha, M., Peñarrocha, M. et al. (2010). Chehade, A., Daley, T.D., Wysocki, G.P., and Miller,
Immunohistochemical study of Langerhans cells in A.S. (1994). Peripheral odontogenic keratocyst. Oral
periapical lesions: correlation with inflammatory cell Surgery, Oral Medicine, and Oral Pathology 77: 494–497.
infiltration and epithelial cell proliferation. Medicina Oral, Cheng, Y.S., Liang, H., Wright, J., and Teenier, T. (2015).
Patología Oral y Cirugía Bucal 15: e335–e339. Multiple orthokeratinized odontogenic cysts: a case report.
Carter, K. and Worthington, S. (2016). Predictors of third Head and Neck Pathology 9: 153–157.
molar impaction: a systematic review and meta-­analysis. Chi, A.C., Owings, J.R., and Muller, S. (2005). Peripheral
Journal of Dental Research 95: 267–276. odontogenic keratocyst: report of two cases and review of
Carter, L.C., Carney, Y.L., and Perez-­Pudlewski, D. (1996). the literature. Oral Surgery, Oral Medicine, Oral Pathology,
Lateral periodontal cyst. Multifactorial analysis of a Oral Radiology, and Endodontics 99: 71–78.
previously unreported series. Oral Surgery, Oral Medicine, Chi, A.C., Neville, B.W., McDonald, T.A. et al. (2007). Jaw
Oral Pathology, Oral Radiology, and Endodontics 81: 210–216. cysts with sebaceous differentiation: report of 5 cases and a
Casamassimo, P.S. and Lilly, G.E. (1980). Mucosal cysts of the review of the literature. Journal of Oral and Maxillofacial
maxillary sinus: a clinical and radiographic study. Oral Surgery 65: 2568–2574.
Surgery, Oral Medicine, and Oral Pathology 50: 282–286. Chi, A.C., Haigney, R.J. 2nd, Spagnoli, D.B. et al. (2010).
Castro-­Núñez, J. (2016). Decompression of odontogenic Papillary synovial metaplasia-­like change in oral
cystic lesions: past, present, and future. Journal of Oral and mucoceles: a rare and previously undescribed
Maxillofacial Surgery 74: 104.e1–104.e9. histopathologic variant of a common oral lesion. Oral
Cataldo, E. and Mosadomi, A. (1970). Mucoceles of the oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and
mucous membrane. Archives of Otolaryngology 91: 360–365. Endodontics 109: 268–273.
Cavalcanti, P.H.P., Nascimento, E.H.L., Pontual, Chi, A.C., Lambert, P.R. 3rd, Richardson, M.S., and Neville,
M.L.D.A. et al. (2018). Cemento-­osseous dysplasias: B.W. (2011). Oral mucoceles: a clinicopathologic review of
Bibliography 317

1,824 cases, including unusual variants. Journal of Oral Chrcanovic, B.R., Gomes, C.C., and Gomez, R.S. (2018).
and Maxillofacial Surgery 69: 1086–1093. Central giant cell lesion of the jaws: an updated analysis of
Chinellato, L.E.M. and Damante, J.H. (1984). Contribution 2270 cases reported in the literature. Journal of Oral
of radiographs to the diagnosis of naso-­alveolar cyst. Oral Pathology and Medicine 47: 731–739.
Surgery, Oral Medicine, and Oral Pathology 58: 729–735. Chung, Y.S., Cho, Y., and Kim, B.H. (2019). Comparison of
Chirapathomsakul, D., Sastravaha, P., and Jansisyanont, outcomes of treatment for ranula: a proportion meta-­analysis.
P. (2006). A review of odontogenic keratocysts and the British Journal of Oral and Maxillofacial Surgery 57: 620–626.
behavior of recurrences. Oral Surgery, Oral Medicine, Oral Chuong, R., Donoff, R.B., and Guralnick, W. (1982). The
Pathology, Oral Radiology, and Endodontics 101: 5–9. odontogenic keratocyst. Journal of Oral and Maxillofacial
Cho, S.W., Lim, H.J., Song, Y. et al. (2019). Principal clinical Surgery 40: 797–802.
factors predicting therapeutic outcomes after surgical Cipriani, N.A., Lusardi, J.J., McElherne, J. et al. (2019).
drainage of postoperative cheek cysts: experience from a Mucoepidermoid carcinoma: a comparison of histologic
single center. Clinical and Experimental grading systems and relationship to MAML2
Otorhinolaryngology 12: 79–85. rearrangement and prognosis. American Journal of
Choi, J.H., Cho, J.H., Kang, H.J. et al. (2002). Nasolabial cyst: Surgical Pathology 43: 885–897.
a retrospective analysis of 18 cases. Ear, Nose, and Throat Cleven, A.H.G., Schreuder, W.H., Groen, E. et al. (2020).
Journal 81: 94–96. Molecular findings in maxillofacial bone tumours and its
Chow, H.T. (1998). Odontogenic keratocyst: a clinical diagnostic value. Virchows Archiv 476: 159–174.
experience in Singapore. Oral Surgery, Oral Medicine, Oral Cohen, M.A. (1979). Pathways of inflammatory cellular
Pathology, Oral Radiology, and Endodontics 86: 573–577. exudate through radicular cyst epithelium: a light and
Chrcanovic, B.R. and Gomez, R.S. (2016). Peripheral scanning electron microscope study. Journal of Oral
calcifying cystic odontogenic tumour and peripheral Pathology 8: 369–378.
dentinogenic ghost cell tumour: an updated systematic Cohen, M.A. and Hertzanu, Y. (1985). Huge growth potential
review of 117 cases reported in the literature. Acta of the nasolabial cyst. Oral Surgery, Oral Medicine, and
Odontologica Scandinavica 74: 591–597. Oral Pathology 59: 441–445.
Chrcanovic, B.R. and Gomez, R.S. (2017). Recurrence Cohen, D.A., Neville, B.W., Damm, D.D., and White,
probability for keratocystic odontogenic tumors: an D.K. (1984). The lateral periodontal cyst. Journal of
analysis of 6427 cases. Journal of Cranio-­Maxillo-­Facial Periodontology 55: 230–234.
Surgery 45: 244–251. Coleman, H., Altini, M., Ali, H. et al. (2001). Use of calretinin
Chrcanovic, B.R. and Gomez, R.S. (2018a). Squamous in the differential diagnosis of unicystic ameloblastomas.
odontogenic tumor and squamous odontogenic tumor-­like Histopathology 38: 312–317.
proliferations in odontogenic cysts: an updated analysis of Coleman, H., Meer, S., Altini, M. et al. (2005). Maxillary sinus
170 cases reported in the literature. Journal of Cranio-­ pathology in 119 patients: a histopathologic study. South
Maxillo-­Facial Surgery 46: 504–510. African Dental Journal 60: 142–145.
Chrcanovic, B.R. and Gomez, R.S. (2018b). Glandular Colgan, C.M., Henry, J., Napier, S.S., and Cowan, C.G. (2002).
odontogenic cyst: an updated analysis of 169 cases Paradental cysts: a role for food impaction in the
reported in the literature. Oral Diseases 24: 717–724. pathogenesis? A review of cases from Northern Ireland.
Chrcanovic, B.R. and Gomez, R.S. (2019a). Gingival cyst of British Journal of Oral and Maxillofacial Surgery 40: 163–168.
the adult, lateral periodontal cyst, and botryoid Colmenero, C., Patron, M., and Colmenero, B. (1990).
odontogenic cyst: an updated systematic review. Oral Odontogenic ghost cell tumours. Journal of Cranio-­
Diseases 25: 26–33. Maxillo-­Facial Surgery 18: 215–218.
Chrcanovic, B.R. and Gomez, R.S. (2019b). Adenomatoid Conceição, J.G., Gurgel, C.A., Ramos, E.A. et al. (2014). Oral
odontogenic tumor: an updated analysis of the cases mucoceles: a clinical, histopathological and
reported in the literature. Journal of Oral Pathology and immunohistochemical study. Acta Histochemica 116: 40–47.
Medicine 48: 10–16. Contos, J.G., Corcoran, J.F., LaTurno, S.A. et al. (1987).
Chrcanovic, B.R. and Gomez, R.S. (2019c). Idiopathic bone Langerhans cells in apical periodontal cysts: an
cavity of the jaws: an updated analysis of the cases immunohistochemical study. Journal of Endodontics
reported in the literature. International Journal of Oral and 13: 52–55.
Maxillofacial Surgery 48: 886–894. Copete, M.A., Kawamata, A., and Langlais, R.P. (1998).
Chrcanovic, B.R., Reis, B.M., and Freire-­Maia, B. (2011). Solitary bone cyst of the jaws. Radiographic review of
Paradental (mandibular inflammatory buccal) cyst. Head 44 cases. Oral Surgery, Oral Medicine, Oral Pathology, Oral
and Neck Pathology 5: 159–164. Radiology, and Endodontics 85: 221–225.
318 Bibliography

Correll, R.W., Jensen, J.L., and Rhyne, R.R. (1980). Lingual identifying odontogenic keratocysts. Pathology Oncology
cortical mandibular defects: a radiographic incidence Research 26: 2613–2620.
study. Oral Surgery, Oral Medicine, and Oral Pathology Cunha, J.F., Gomes, C.C., de Mesquita, R.A. et al. (2016).
50: 287–291. Clinicopathologic features associated with recurrence of
Cortell-­Ballester, I., Figueiredo, R., Berini-­Aytés, L., and the odontogenic keratocyst: a cohort retrospective analysis.
Gay-­Escoda, C. (2009). Traumatic bone cyst: a retrospective Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology
study of 21 cases. Medicina Oral, Patología Oral y Cirugía 121: 629–635.
Bucal 14: E239–E243. Cunha, J.L.S., Silva, A.F., Silva, J.V.R. et al. (2019). Botryoid
Costa, F.W., Viana, T.S., Cavalcante, G.M. et al. (2014). A odontogenic cyst: report of case found in routine
clinicoradiographic and pathological study of pericoronal imaginological examinations and an update of the
follicles associated to mandibular third molars. Journal of literature. Journal of Oral Diagnosis 4 (1) www.jordi.com.br.
Craniofacial Surgery 25: e283–e287. Curran, A.E., Damm, D.D., and Drummond, J.F. (2002).
Cottom, H.E., Bshena, F.I., Speight, P.M. et al. (2012). Pathologically significant pericoronal lesions in adults:
Histopathological features that predict the recurrence of histpopathologic evaluation. Journal of Oral and
odontogenic keratocysts. Journal of Oral Patholology and Maxillofacial Surgery 60: 613–617.
Medicine 41: 408–414. Da Silva, M.J., de Sousa, S.O., Correa, L. et al. (2002).
Courage, G.R., North, A.F., and Hansen, L.S. (1974). Median Immunohistochemical study of the orthokeratinized
palatine cysts. Review of the literature and report of case. odontogenic cyst: a comparison with the odontogenic
Oral Surgery, Oral Medicine, and Oral Pathology 37: 745–753. keratocyst. Oral Surgery, Oral Medicine, Oral Pathology,
Cousin, T., Bobek, S., and Oda, D. (2017). Glandular Oral Radiology, and Endodontics 94: 732–737.
odontogenic cyst associated with ameloblastoma: case da Silva, K.D., Coelho, L.V., do Couto, A.M. et al. (2020).
report and review of the literature. Journal of Clinical and Clinicopathological and immunohistochemical features of
Experimental Dentistry 9: e832–e836. the oral lymphoepithelial cyst: a multicenter study. Journal
Craig, G.T. (1976). The paradental cyst. A specific of Oral Pathology and Medicine 49: 219–226.
inflammatory odontogenic cyst. British Dental Journal da Silva, L.P., Gonzaga, A.K., Severo, M.L. et al. (2018).
141: 9–14. Epidemiologic study of odontogenic and non-odontogenic
Craig, G.T., Holland, C.S., and Hindle, M.O. (1980). Dermoid cysts in children and adolescents of a Brazilian population.
cyst of the mandible. British Journal of Oral Surgery 18: Medicina Oral, Patología Oral y Cirugía Bucal 23: e49–e53.
230–237. da Silva, T.A., Batista, A.C., Mendonça, E.F. et al. (2008).
Crane, H., Da Forno, P., Kyriakidou, E. et al. (2020). Multiple Comparative expression of RANK, RANKL, and OPG in
orthokeratinized odontogenic cysts: a report of two cases keratocystic odontogenic tumors, ameloblastomas, and
and review of the literature. Head and Neck Pathology dentigerous cysts. Oral Surgery, Oral Medicine, Oral
14: 381–385. Pathology, Oral Radiology, and Endodontics
Crivelini, M.M., Soubhia, A.M., Biazolla, E.R., and Neto, 105: 333–341.
S.C. (2001). Heterotopic gastrointestinal cyst partially lined D’addazio, G., Artese, L., Piccirilli, M., and Perfetti,
with dermoid cyst epithelium. Oral Surgery, Oral Medicine, G. (2014). Role of matrix metalloproteinases in radicular
Oral Pathology, Oral Radiology, and Endodontics 91: cysts and periapical granulomas. Minerva Stomatologica
686–688. 63: 411–420.
Crowley, T.E., Kaugars, G.E., and Gunsolley, J.C. (1992). Daley, T. (1993). Pathology of intraoral sebaceous glands.
Odontogenic keratocysts: a clinical and histologic Journal of Oral Pathology and Medicine 22: 241–245.
comparison of the parakeratin and orthokeratin variants. Daley, T.D. and Wysocki, G.P. (1995). The small dentigerous
Journal of Oral and Maxillofacial Surgery 50: 22–26. cyst. A diagnostic dilemma. Oral Surgery, Oral Medicine,
Crusz, S.M. and Balkwill, F.R. (2015). Inflammation and Oral Pathology, Oral Radiology, and Endodontics
cancer: advances and new agents. Nature Reviews. Clinical 79: 77–81.
Oncology 12: 584–596. Daley, T.D., Wysocki, G.P., and Pringle, G.A. (1994). The
Cserni, D., Zombori, T., Stájer, A. et al. (2020a). relative incidence of odontogenic tumors: oral and jaw cysts
Immunohistochemical characterization of reactive in a Canadian population. Oral Surgery, Oral Medicine, Oral
epithelial changes in odontogenic keratocysts. Pathology Pathology, Oral Radiology, and Endodontics 77: 276–280.
Oncology Research 26: 1717–1724. Daley, T.D., Multari, J., and Darling, M.R. (2007). A case
Cserni, D., Zombori, T., Vörös, A. et al. (2020b). A report of a solid keratocystic odontogenic tumor: is it the
clinicopathological approach to odontogenic cysts: the role missing link? Oral Surgery, Oral Medicine, Oral Pathology,
of cytokeratin 17 and bcl2 immunohistochemistry in Oral Radiology, and Endodontics 103: 512–515.
Bibliography 319

Damante, J.H. and Fleury, R.N. (2001). A contribution to the cohort. European Archives of Oto-Rhino-Laryngology 278:
diagnosis of the small dentigerius cyst or the paradental 1223–1231.
cyst. Pesquisa Odontológica Brasileira 15: 238–246. de Moraes, M., de Lucena, H.F., de Azevedo, P.R. et al. (2011).
Dammer, U., Driemel, O., Mohren, W. et al. (2014). Comparative immunohistochemical expression of RANK,
Globulomaxillary cysts–do they really exist? Clinical Oral RANKL and OPG in radicular and dentigerous cysts.
Investigations 18: 239–246. Archives of Oral Biology 56: 1256–1263.
Das, S.N., Jyoti, K., Rath, R., and Pattnaik, B. (2020). Sclerosing de Oliveira, A.J., Silveira, M.L., Duarte, D.A., and Diniz,
polycystic adenosis of minor salivary glands: report of a rare M.B. (2018). Eruption Cyst in the Neonate. International
case with diagnostic approach and review of literature. Journal of Clinical Pediatric Dentistry 11: 58–60.
Journal of Oral and Maxillofacial Pathololgy 24: 590. de Oliveira, M.D., de Miranda, J.L., de Amorim, R.F. et al.
Dave, M., Thomson, F., Barry, S. et al. (2020). The use of (2004). Tenascin and fibronectin expression in odontogenic
localised CBCT to image inflammatory collateral cysts: a cysts. Journal of Oral Pathology and Medicine 33: 354–359.
retrospective case series demonstrating clinical and de Oliveira, M.G., Lauxen Ida, S., Chaves, A.C. et al. (2011).
radiographic features. European Archives of Paediatric Odontogenic epithelium: immunolabeling of Ki-67, EGFR
Dentistry 21: 329–337. and survivin in pericoronal follicles, dentigerous cysts and
David, L.A., Sándor, G.K., and Stoneman, D.W. (1998). The keratocystic odontogenic tumors. Head and Neck
buccal bifurcation cyst: in non-­surgical treatment an Pathology 5: 1–7.
option? Journal of the Canadian Dental Association 64: de Paula, A.M.B., Carvalhais, J.N., Domingues, M.G. et al.
712–716. (2000). Cell proliferation markers in the odontogenic
Dayan, D., Buchner, A., Gorsky, M., and Harel-­Raviv, M. (1988). keratocyst: effect of inflammation. Journal of Oral
The peripheral odontogenic keratocyst. International Journal Pathology and Medicine 29: 477–482.
of Oral and Maxillofacial Surgery 17: 81–83. de Sousa, S.F., Moreira, R.G., Gomez, R.S., and Gomes,
de Andrade, M., Silva, A.P., de Moraes Ramos-Perez, C.C. (2016). Interrogation of cancer hotspot mutations in
F.M. et al. (2012). Lateral periodontal cyst: report of case 50 tumour suppressor genes and oncogenes in calcifying
and review of the literature. Oral and Maxillofacial Surgery cystic odontogenic tumour. Oral Oncology 57: e1–e3.
16: 83–87. de Sousa, S.O., Cabezas, N.T., de Oliveira, P.T., and de Araújo,
de Arruda, J.A.A., Monteiro, J.L.G.C., Abreu, L.G. et al. V.C. (1997). Glandular odontogenic cyst. Report of a case
(2018a). Calcifying odontogenic cyst, dentinogenic ghost with cytokeratin expression. Oral Surgery, Oral Medicine,
cell tumor, and ghost cell odontogenic carcinoma: a Oral Pathology, Oral Radiology, and Endodontics 83: 478–483.
systematic review. Journal of Oral Pathology and Medicine de Sousa, S.O., Corrêa, L., Deboni, M.C., and de Araújo,
47: 721–730. V.C. (2001). Clinicopathologic features of 54 cases of
de Arruda, J.A.A., Schuch, L.F., Abreu, L.G. et al. (2018b). A paradental cyst. Quintessence International 32: 737–741.
multicentre study of 268 cases of calcifying odontogenic de Souza, L.L., Pontes, F.S.C., Pontes, H.A.R. et al. (2018).
cysts and a literature review. Oral Diseases 24: 1282–1293. Central mucoepidermoid carcinoma: an up-to-date
de Brito Monteiro, B.V., Bezerra, T.M., da Silveira, É.J. et al. analysis of 147 cases and review of prognostic factors.
(2016). Histopathological review of 667 cases of oral Journal of Cranio-Maxillo-Facial Surgery 46: 162–167.
mucoceles with emphasis on uncommon histopathological de Souza, P.E., Barreto, D.C., Fonseca, L.M. et al. (2000).
variations. Annals of Diagnostic Pathology 21: 44–46. Cysticercosis of the oral cavity: report of seven cases. Oral
de Camargo Moraes, P., Bönecker, M. et al. (2009). Mucocele Diseases 6: 253–255.
of the gland of Blandin-Nuhn: histological and clinical de Tristan, J., Zenk, J., Künzel, J. et al. (2015). Thyroglossal
findings. Clinical Oral Investigations 13: 351–353. duct cysts: 20 years’ experience (1992–2011). European
de Castro, M.S., Caixeta, C.A., de Carli, M.L. et al. (2018). Archives of Oto-Rhino-Laryngology 272: 2513–2519.
Conservative surgical treatments for nonsyndromic De Villiers, P., Liu, H., Suggs, C. et al. (2008). Calretinin
odontogenic keratocysts: a systematic review and meta- expression in the differential diagnosis of human
analysis. Clinical Oral Investigations 22: 2089–2101. ameloblastoma and keratocystic odontogenic tumor.
de Fátima Bernardes, V., de Lacerda, J.C., de Aguiar, M.C., American Journal of Surgical Pathology 32: 256–260.
and Gomez, R.S. (2008). Calcifying odontogenic cyst De Visscher, J.G.A.M., van der Wal, K.G.H., and Vogel,
associated with an orthokeratinized odontogenic cyst. P.L. (1989). The plunging ranula. Pathogenesis, diagnosis
Head and Neck Pathology 2: 324–327. and management. Journal of Cranio-Maxillo-Facial
de França, G.M., da Silva, L.B.A., Mafra, R.P. et al. (2020). Surgery 17: 182–185.
Recurrence-free survival and prognostic factors of DeGould, M.D. and Goldberg, J.S. (1991). Recurrence of an
odontogenic keratocyst: a single-center retrospective odontogenic keratocyst in a bone graft. Report of a case.
320 Bibliography

International Journal of Oral and Maxillofacial Surgery Drazić, R., Sopta, J., and Minić, A.J. (2010). Mast cells in
20: 9–11. periapical lesions: potential role in their pathogenesis.
Devi, P., Thimmarasa, V.B., Mehrotra, V., and Agarwal, Journal of Oral Pathology and Medicine 39: 257–262.
M. (2015). Multiple dentigerous cysts: a case report Dutra, S.N., Pires, F.R., Armada, L., and Azevedo, R.S. (2017).
and review. Journal of Maxillofacial Oral Surgery 14 Immunoexpression of Wnt/β-­catenin signaling pathway
(Suppl. 1): 47–51. proteins in ameloblastoma and calcifying cystic
Dewey, K.W. (1918). Cysts of the dental system. Dental odontogenic tumor. Journal of Clinical and Experimental
Cosmos 60: 555–570. Dentistry 9: e136–e140.
Dias, G., Marques, T., and Coelho, P. (2016). Treatment Duverger, O., Beniash, E., and Morasso, M.I. (2016). Keratins
options for keratocyst odontogenic tumour (KCOT): a as components of the enamel organic matrix. Matrix
systematic review. Oral Surgery 10: 193–209. Biology 52-­54: 260–265.
Díaz-­Belenguer, Á., Sánchez-­Torres, A., and Gay-­Escoda, Earl, P.D. and Ward-­Booth, R.P. (1985). A case of branchial
C. (2016). Role of Carnoy’s solution in the treatment of (lympho-­epithelial) cyst, illustrating the value of
keratocystic odontogenic tumor: a systematic review. ultrasound in diagnosis of cervical swellings. British
Medicina Oral, Patología Oral y Cirugía Bucal 21: e689–e695. Journal of Oral and Maxillofacial Surgery 23:
Difiore, P.M. and Hartwell, G.R. (1987). Median mandibular 292–297.
lateral periodontal cyst. Oral Surgery, Oral Medicine, and Eaton, D., Billings, K., Timmons, C. et al. (2001). Congenital
Oral Pathology 63: 545–550. foregut duplication cysts of the anterior tongue. Archives
Diniz, M.G., Borges, E.R., Guimarães, A.L. et al. (2009). of Otolaryngology – Head and Neck Surgery 127:
PTCH1 isoforms in odontogenic keratocysts. Oral Oncology 1484–1487.
45: 291–295. Ecker, J., Horst, R.T., and Koslovsky, D. (2016). Current role
Diniz, M.G., Galvão, C.F., Macedo, P.S. et al. (2011). Evidence of Carnoy’s solution in treating keratocystic odontogenic
of loss of heterozygosity of the PTCH gene in tumors. Journal of Oral and Maxillofacial Surgery 74:
orthokeratinized odontogenic cyst. Journal of Oral 278–282.
Pathology and Medicine 40: 277–280. Economopoulou, P. and Patrikiou, A. (1995). Glandular
Diniz, M.G., Gomes, C.C., de Sousa, S.F. et al. (2017). odontgenic cyst of the maxilla: report of a case. Journal of
Oncogenic signalling pathways in benign odontogenic Oral and Maxillofacial Surgery 53: 834–837.
cysts and tumours. Oral Oncology 72: 165–173. Edamatsu, M., Kumamoto, H., Ooya, K., and Echigo,
Dominguez, F.V. and Keszler, A. (1988). Comparative study S. (2005). Apoptosis-­related factors in the epithelial
of keratocysts, associated and non-­associated with nevoid components of dental follicles and dentigerous cysts
basal cell carcinoma syndrome. Journal of Oral Pathology associated with impacted third molars of the mandible.
17: 39–42. Oral Surgery, Oral Medicine, Oral Pathology, Oral
Donatsky, O., Hjørting-­Hansen, E., Philipsen, H.P., and Radiology, and Endodontics 99: 17–23.
Fejerskov, J. (1976). Clinical, radiologic and histopathologic EI Murtadi, A., Grehan, D., Toner, M., and Mc Cartan,
aspects of 13 cases of nevoid basal cell carcinoma B.E. (1996). Proliferation cell nuclear antigen staining in
syndrome. International Journal of Oral Surgery 5: 19–28. syndrome and nonsyndrome odontogenic keratocysts. Oral
Dong, Q., Pan, S., Sun, L.S., and Li, T.J. (2010). Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and
Orthokeratinized odontogenic cyst: a clinicopathologic Endodontics 81: 217–220.
study of 61 cases. Archives of Pathology and Laboratory Elias, F.M., Martins, M.T., Foronda, R. et al. (2005). Oral
Medicine 134: 271–275. cysticercosis: case report and review of the literature.
Donoff, R.B., Harper, E., and Guralnick, W.C. (1972). Revista do Instituto de Medicina Tropical de São Paulo
Collagenolytic activity in keratocysts. Journal of Oral 47: 95–98.
Surgery 30: 879–884. Eliasson, S., Isacsson, G., and Kondell, P.A. (1989). Lateral
Douglas, C.W.I. and Craig, G.T. (1986). Recognition of protein periodontal cysts. Clinical, radiographical and
apparently specific to odontogenic keratocyst fluids. histopathological findings. International Journal of Oral
Journal of Clinical Pathology 39: 1108–1115. and Maxillofacial Surgery 18: 191–193.
Douglas, C.W.I. and Craig, G.T. (1987). Evidence for the El-­Labban, N.G. (1979). Electron microscopic investigation of
presence of lactoferrin in odontogenic keratocyst fluids. hyaline bodies in odontogenic cysts. Journal of Oral
Journal of Clinical Pathology 40: 914–921. Pathology 8: 81–93.
Douglas, C.W.I. and Craig, G.T. (1989). Quantitation of Elliott, K.A., Franzese, C.B., and Pitman, K.T. (2004).
lactoferrin in odontogenic cyst fluids. Journal of Clinical Diagnosis and surgical management of nasopalatine duct
Pathology 42: 180–183. cysts. Laryngoscope 114: 1336–1340.
Bibliography 321

Ellis, G.L. and Auclair, P.L. (2008). Tumours of the Salivary Eversole, L.R. (1987). Oral sialocysts. Archives of
Glands, AFIP Atlas of Tumor Pathology. Series 4. Fascicle Otolaryngology – Head and Neck Surgery 113: 51–56.
9. Washington, DC: American Registry of Pathology and Eversole, L.R., Sabes, W.R., and Rovin, S. (1975). Aggressive
the Armed Forces Institute of Pathology. growth and neoplastic potential of odontogenic cysts: with
Ellis, G.L. and Shmookler, B.M. (1986). Aggressive special reference to central epidermoid and
(malignant?) epithelial odontogenic ghost cell tumor. Oral mucoepidermoid carcinomas. Cancer 35: 270–282.
Surgery, Oral Medicine, and Oral Pathology 61: 471–478. Eversole, R., Su, L., and ElMofty, S. (2008). Benign fibro-­
El-­Naggar, A.K., Chan, J.K.C., Grandis, J.R. et al. (2017). osseous lesions of the craniofacial complex. A review.
WHO Classification of Head and Neck Tumours, 4e. Head and Neck Pathology 2: 177–202.
Lyon: IARC. Eveson, J.W. (1988). Superficial mucoceles: pitfall in clinical
Eloy, P., Poirrier, A.L., Nicoli, T. et al. (2012). Cystic dilation and microscopic diagnosis. Oral Surgery, Oral Medicine,
of the distal end of the nasolacrimal duct: underrated and Oral Pathology 66: 318–322.
cause of epiphora in adults and its endoscopic treatment. Ewing, J. (1940). Neoplastic Diseases: A Treatise on Tumours,
Rhinology 50: 436–441. 4e. Philadelphia, PA: W.B. Saunders.
El-­Salem, F., Mansour, M., Gitman, M. et al. (2019). Real-­ Faisal, M., Ahmad, S.A., and Ansari, U. (2015). Glandular
time PCR HPV genotyping in fine needle aspirations of odontogenic cyst – literature review and report of a
metastatic head and neck squamous cell carcinoma: paediatric case. Journal of Oral Biology and Craniofacial
exposing the limitations of conventional p16 Research 5: 219–225.
immunostaining. Oral Oncology 90: 74–79. Falci, S.G., Verli, F.D., Consolaro, A., and Santos, C.R. (2013).
Emerson, T.G., Whitlock, R.I.H., and Jones, J.H. (1972). Morphological characterization of the nasopalatine region
Involvement of soft tissue by odontogenic keratocysts in human fetuses and its association to pathologies.
(primordial cysts). British Journal of Oral Surgery 9: 181–185. Journal of Applied Oral Science 21: 250–255.
Endo, M., Fujii, K., Sugita, K. et al. (2012). Nationwide survey Fantasia, J.E. (1979). Lateral periodontal cyst. An analysis of
of nevoid basal cell carcinoma syndrome in Japan revealing 46 cases. Oral Surgery, Oral Medicine, and Oral Pathology
the low frequency of basal cell carcinoma. American 48: 237–243.
Journal of Medical Genetics. Part A 158: 351–357. Fardal, O. and Johannessen, A.C. (1994). Rare case of
Epivatianos, A., Zaraboukas, T., and Antoniades, D. (2005). keratin-­producing multiple gingival cysts. Oral Surgery,
Coexistence of lymphoepithelial and epidermoid cysts on Oral Medicine, and Oral Pathology 77: 498–500.
the floor of the mouth: report of a case. Oral Diseases Farndon, P.A., Del Mastro, R.G., Evans, D.G., and Kilpatrick,
11: 330–333. M.W. (1992). Location of gene for Gorlin syndrome. Lancet
Eppley, B.L., Bell, M.J., and Sclaroff, A. (1985). Simultaneous 339 (8793): 581–582.
occurrence of dermoid and heterotopic intestinal cysts in Faustino, S.E., Pereira, M.C., Rossetto, A.C., and Oliveira,
the floor of the mouth of a newborn. Journal of Oral and D.T. (2008). Recurrent peripheral odontogenic keratocyst:
Maxillofacial Surgery 43: 880–883. a case report. Dento Maxillo Facial Radiology 37: 412–414.
Ertaş, Ü., Büyükkurt, C., Güngörmüş, M., and Kaya, O. (2003). Fell, H. (1957). The effect of excess vitamin A on cultures of
A large glandular odontogenic cyst of the mandible: report embryonic chicken skin explanted at different stages of
of a case. Journal of Contemporary Dental Practice 4: 53–58. differentiation. Proceedings of the Royal Society, London
Escoda Francolí, J., Almendros Marqués, N., Berini Aytés, L., (Biology) 146: 242–256.
and Gay, E.C. (2008). Nasopalatine duct cyst: report of 22 Fejerskov, O. and Krogh J. (1972). The calcifying ghost cell
cases and review of the literature. Medicina Oral, Patología odontogenic tumor: or the calcifying odontogenic cyst.
Oral y Cirugía Bucal 13: E438–E443. Journal of Oral Pathology 1: 273–287.
Etemad-­Moghadam, S., Baghaee, F., Dadafarid, Z., and Ferenczy, K. (1958). The relationship of globulomaxillary
Alaeddini, M. (2014). A 44-­year analysis of ghost cell cysts to the fusion of embryonal processes and to cleft
odontogenic tumour subtypes in an Iranian population. palates. Oral Surgery, Oral Medicine, and Oral Pathology
Journal of Cranio-­Maxillo-­Facial Surgery 42: 1154–1159. 11: 1388–1393.
Etoz, M. and Sisman, Y. (2014). Evaluation of the nasopalatine Ferreira, J.C.B., Vêncio, E.F., de Sá, R.T., and Gasperini,
canal and variations with cone-­beam computed G. (2019). Glandular odontogenic cyst in dentigerous
tomography. Surgical and Radiologic Anatomy 36: 805–812. relationship: an uncommon case report. Case Reports in
Evans, D.G., Ladusans, E.J., Rimmer, S. et al. (1993). Dentistry 2019: 8647158.
Complications of the naevoid basal cell carcinoma Ficarra, G., Chou, L., and Panzoni, E. (1990). Glandular
syndrome: results of a population based study. Journal of odontogenic cyst (sialo-­odontogenic cyst). International
Medical Genetics 30: 460–464. Journal of Oral and Maxillofacial Surgery 19: 331–333.
322 Bibliography

Fidele, N.B., Yueyu, Z., Zhao, Y. et al. (2019). Recurrence of and immunohistochemical profile of 10 cases. Journal of
odontogenic keratocysts and possible prognostic factors: Oral Pathology and Medicine 32: 163–170.
review of 455 patients. Medicina Oral, Patología Oral y Friedrich, R.E., Laumann, F., Zrnc, T., and Assaf, A.T. (2015).
Cirugía Bucal 24: e491–e501. The Nasopalatine Canal in Adults on Cone Beam
Fisher, A.D. (1976). Bone cavities in libro-­osseous lesions. Computed Tomograms—­A Clinical Study and Review of
British Journal of Oral Surgery 14: 120–127. the Literature. In vivo 29: 467–486.
Flanagan, A.M. and Speight, P.M. (2014). Giant cell lesions Friedrich, R.E., Barsukov, E., Kohlrusch, F.K. et al. (2020).
of the craniofacial bones. Head and Neck Pathology 8: Lingual Mandibular Bone Depression. In vivo 34: 2527–2541.
445–453. Fromm, A. (1967). Epstein’s pearls, Bohn’s nodules and
Flores, E.R., Tsai, K.Y., Crowley, D. et al. (2000). p63 and p73 inclusion cysts of the oral cavity. Journal of Dentistry for
are required for p53-­dependent apoptosis in response to Children 34: 275–287.
DNA damage. Nature 416 (6880): 560–564. Fujiwara, K. and Watanabe, T. (1988). Mucus-­producing cells
Flores, I.L., Hamilton, M.E., Zanchin-­Baldissera, E. et al. and ciliated epithelial cells in mandibular radicular cyst:
(2017). Simple and aneurysmal bone cyst: aspects of jaw an electron microscopic study. Journal of Oral and
pseudocysts based on an experience of Brazilian pathology Maxillofacial Surgery 46: 149–151.
service during 53 years. Medicina Oral, Patología Oral y Gadipelly, S., Reddy, V.B., Sudheer, M. et al. (2015). Bilateral
Cirugía Bucal 22: e64–e69. calcifying odontogenic cyst: a rare entity. Journal of
Fonseca, F.P., Pontes, H.A., Pontes, F.S., and al. (2013). Oral Maxillofacial and Oral Surgery 14: 826–831.
carcinoma cuniculatum: two cases illustrative of a Gaitán-­Cepeda, L.A., Quezada-­Rivera, D., Tenorio-­Rocha, F.,
diagnostic challenge. Oral Surgery, Oral Medicine, Oral and Leyva-­Huerta, E.R. (2010). Reclassification of
Pathology, Oral Radiology 116: 457–463. odontogenic keratocyst as tumour. Impact on the
Fordyce, G.L. (1956). The probable nature of so-­called latent odontogenic tumours prevalence. Oral Diseases 16: 185–187.
haemorrhagic cysts of the mandible. British Dental Journal Gao, Z., Mackenzie, I.C., Rittman, B.R. et al. (1988a).
101: 40–42. Immunocytochemical examination of immune cells in
Fornatora, M.L., Reich, R.F., Chotkowski, G., and Freedman, periapical granulomata and odontogenic cysts. Journal of
P.D. (2001). Odontogenic keratocyst with mural Oral Pathology 17: 84–90.
cartilagenous metaplasia: a case report and a review of the Gao, Z., Mackenzie, I.C., Williams, D.M. et al. (1988b).
literature. Oral Surgery, Oral Medicine, Oral Pathology, Oral Patterns of keratin-­expression in rests of Malassez and
Radiology, and Endodontics 92: 430–434. periapical lesions. Journal of Oral Pathology 17: 178–185.
Forssell, K. (1980). The primordial cyst. A clinical and Gardner, A.F. (1969). The odontogenic cyst as a potential
radiographic study. Proceedings of the Finnish Dental carcinoma: a clinicopathologic appraisal. Journal of the
Society 76: 129–174. American Dental Association 78: 740–755.
Forssell, K. and Sainio, P. (1979). Clinicopathological study of Gardner, D.G. (1988). An evaluation of reported cases of
keratinized cysts of the jaws. Proceedings of the Finnish median mandibular cysts. Oral Surgery, Oral Medicine, and
Dental Society 75: 36–45. Oral Pathology 65: 208–213.
Forssell, K., Sorvari, T.E., and Oksala, E. (1974). An analysis Gardner, D.G. and Gullane, P.J. (1986). Mucoceles of the
of the recurrence of odontogenic keratocysts. Proceedings maxillary sinus. Oral Surgery, Oral Medicine, and Oral
of the Finnish Dental Society 70: 135–140. Pathology 62: 538–543.
Fowler, C.B. and Brannon, R.B. (1989). The paradental cyst: a Gardner, D.G., Sapp, J.P., and Wysocki, G.P. (1978).
clinicopathologic study of six new cases and review of the Odontogenic and ‘fissuraI’ cysts of the jaws. Pathology
literature. Journal of Oral and Maxillofacial Surgery 47: Annual 13: 177–200.
243–248. Gardner, D.G., Kessler, H.P., Morency, R., and Schaffner,
Fowler, C.B., Brannon, R.B., Kessler, H.P. et al. (2011). D.L. (1988). The glandular odontogenic cyst: an apparent
Glandular odontogenic cyst: analysis of 46 cases with entity. Journal of Oral Pathology 17: 359–366.
special emphasis on microscopic criteria for diagnosis. Garlock, J.A., Pringle, G.A., and Hicks, M.L. (1998). The
Head and Neck Pathology 5: 364–375. odontogenic keratocyst: a potential endodontic
Freedman, P.D., Lumerman, H., and Gee, J.K. (1975). misdiagnosis. Oral Surgery, Oral Medicine, Oral Pathology,
Calcifying odontogenic cyst. A review and analysis of Oral Radiology, and Endodontics 85: 452–456.
seventy cases. Oral Surgery, Oral Medicine, and Oral Gebhardt, P. and Lenz, W. (1985). Zur Frage der Induktion
Pathology 40: 93–106. follikuIirer Zysten im Wechselgebiss durch
Fregnani, E.R., Pires, F.R., Quezada, R.D. et al. (2003). Wunelbehandlung von Milchzahnen. Deutsche
Calcifying odontogenic cyst: clinicopathological features Zahntirzdiche Zeitschrift 40: 541–543.
Bibliography 323

Geller, K.A., Cohen, D., and Koempel, J.A. (2014). odontogenic tumors, dentinogenic ghost cell tumor, and
Thyroglossal duct cyst and sinuses: a 20-­year Los Angeles ghost cell odontogenic carcinoma. Oral Surgery, Oral
experience and lessons learned. International Journal of Medicine, Oral Pathology, Oral Radiology 117: 626–633.
Pediatric Otorhinolaryngology 78: 264–267. Gomes, C.C. and Gomez, R.S. (2007). Odontogenic
Geng, N., Lv, D., Chen, Q.M. et al. (2012). Solid variant of keratocyst: a benign cystic neoplasm? Oral Oncology
keratocystic odontogenic tumor with ameloblastomatous 43: 619–620.
transformation: a case report and review of the literature. Gomes, C.C. and Gomez, R.S. (2011). PTCH1 gene
Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology inactivation is not a Keratocystic odontogenic tumour
114: 223–229. exclusive alteration. Oral Oncology 47: 226–227.
Gillette, R. and Weinmann, J.P. (1958). Extrafollicular stages Gomes, C.C., Diniz, M.G., and Gomez, R.S. (2009). Review of
in dentigerous cyst development. Oral Surgery, Oral the molecular pathogenesis of the odontogenic keratocyst.
Medicine, and Oral Pathology 11: 638–645. Oral Oncology 45: 1011–1014.
Gingell, J.C., Levy, B.A., and DePaola, L.G. (1985). Median Gomes, C.C., Guimarães, L.M., Diniz, M.G., and Gomez,
palatine cyst. Journal of Oral and Maxillofacial Surgery R.S. (2017). Molecular alterations in odontogenic
43: 47–51. keratocysts as potential therapeutic targets. Journal of Oral
Gioacchini, F.M., Alicandri-­Ciufelli, M., Kaleci, S. et al. Pathology and Medicine 46: 877–882.
(2015). Clinical presentation and treatment outcomes of Gomes, C.C., de Sousa, S.F., and Gomez, R.S. (2019).
thyroglossal duct cysts: a systematic review. International Craniopharyngiomas and odontogenic tumors mimic
Journal of Oral and Maxillofacial Surgery 44: 119–126. normal odontogenesis and share genetic mutations,
Gisakis, I.G., Palamidakis, F.D., Farmakis, E.T. et al. (2011). histopathologic features, and molecular pathways
Prevalence of impacted teeth in a Greek population. activation. Oral Surgery, Oral Medicine, Oral Pathology,
Journal of Investigative and Clinical Dentistry 2: 102–109. Oral Radiology 127: 231–236.
Giunta, J.L. (2002). Gingival cysts in the adult. Journal of Gomes, C.C., Diniz, M.G., Bastos, V.C. et al. (2020). Making
Periodontology 73: 827–831. sense of giant cell lesions of the jaws (GCLJ): lessons
Giunta, J. and Cataldo, E. (1973). Lymphoepithelial cysts of learned from next-­generation sequencing. Journal of
the oral mucosa. Oral Surgery, Oral Medicine, and Oral Pathology 250: 126–133.
Pathology 35: 77–84. Gomez, R.S. and De Marco, L. (2005). Possible molecular
Gleizal, A., Abouchebel, N., Lebreton, F., and Beziat, approach to the treatment of odontogenic keratocyst. Oral
J.L. (2006). Dermoid cyst of the tongue: an association of Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and
dermoid cyst with bronchogenic epithelium. Journal of Endodontology 99: 527–528.
Craniofacial Surgery 34: 113–116. Gomez, R.S., de Oliveira, J.R., and Castro, W.H. (2001).
Glynis, A., Foschi, F., Kefalou, I. et al. (2021). Regenerative Spontaneous regression of a paradental cyst. Dento Maxillo
endodontic procedures for the treatment of necrotic Facial Radiology 30: 296.
mature teeth with apical periodontitis: a systematic review González-­Alva, P., Tanaka, A., Oku, Y. et al. (2008).
and meta-­analysis of randomized controlled trials. Journal Keratocystic odontogenic tumor: a retrospective study of
of Endodontia 47: 873–882. 183 cases. Journal of Oral Science 50: 205–212.
Gnepp, D.R. (2003). Sclerosing polycystic adenosis of the González-­Alva, P., Inoue, H., Miyazaki, Y. et al. (2011).
salivary gland: a lesion that may be associated with Podoplanin expression in odontomas: clinicopathological
dysplasia and carcinoma in situ. Advances in Anatomic study and immunohistochemical analysis of 86 cases.
Pathology 10: 218–222. Journal of Oral Science 53: 67–75.
Gnepp, D.R., Wang, L.J., Brandwein-­Gensler, M. et al. (2006). Gorlin, R.J. and Goltz, R.W. (1960). Multiple nevoid basal cell
Sclerosing polycystic adenosis of the salivary gland: a epithelioma, jaw cysts and bifid rib: a syndrome. New
report of 16 cases. American Journal of Surgical Pathology England Journal of Medicine 262: 908–912.
30: 154–164. Gorlin, R.J. and Jirasek, J.E. (1970). Oral cysts containing gastric
Gold, L. and Christ, T. (1970). Granular cell odontogenic cyst. or intestinal mucosa: unusual embryologic accident or
Oral Surgery, Oral Medicine, and Oral Pathology 29: heterotopia. Journal of Oral Surgery 28: 9–11.
437–442. Gorlin, R.J., Pindborg, J.J., Clausen, F.P., and Vickers, R.A. (1962).
Goldenberg, D., Sciubba, J., and Tufano, R.P. (2004). Odontogenic The calcifying odontogenic cyst – a possible analogue of the
ghost cell carcinoma. Head and Neck 26: 378–381. cutaneous calcifying epithelioma of Malherbe. Oral Surgery,
Gomes da Silva, W., Ribeiro Bartholomeu Dos Santos, T.C., Oral Medicine, and Oral Pathology 15: 1235–1243.
Cabral, M.G. et al. (2014). Clinicopathologic analysis and Gorlin, R.J., Yunis, J.J., and Tuna, W. (1963). Multiple nevoid
syndecan-­1 and Ki-­67 expression in calcifying cystic basal cell carcinoma, odontogenic keratocysts and skeletal
324 Bibliography

anomalies syndrome. Acta Dermatologica Venereologica Gu, X.M., Zhao, H.S., Sun, L.S., and Li, T.J. (2006). PTCH
(Stockholm) 43: 39–55. mutations in sporadic and Gorlin-­syndrome-­related
Gorlin, R.J., Pindborg, J.J., Redman, R.S. et al. (1964). The odontogenic keratocysts. Journal of Dental Research
calcifying odontogenic cyst. A new entity and possible 85: 859–863.
analogue of the cutaneous calcifying epithelioma of Guo, J., Simon, J.H., Sedghizadeh, P. et al. (2013). Evaluation
Malherbe. Cancer 17: 723–729. of the reliability and accuracy of using cone-­beam
Goto, M., Ueda, S., Miyabe, S. et al. (2020). A rare case of computed tomography for diagnosing periapical cysts
odontogenic keratocyst extending into the sphenoid bone from granulomas. Journal of Endodontics 39: 1485–1490.
from the maxilla. International Journal of Surgery Case Guo, Y.Y., Zhang, J.Y., Li, X.F. et al. (2013). PTCH1 gene
Reports 71: 132–138. mutations in Keratocystic odontogenic tumors: a study of
Goulart, M.C., Freitas-­Faria, P., Goulart, G.R. et al. (2011). 43 Chinese patients and a systematic review. PLoS One
Pleomorphic adenoma with extensive squamous 8 (10): e77305.
metaplasia and keratin cyst formations in minor salivary Gurgel, C.A., Buim, M.E., Carvalho, K.C. et al. (2014).
gland: a case report. Journal of Applied Oral Science Transcriptional profiles of SHH pathway genes in
19: 182–188. keratocystic odontogenic tumor and ameloblastoma.
Grand, N.G. and Marwah, A.S. (1964). Pigmented gingival Journal of Oral Pathology and Medicine 43: 619–626.
cyst. Oral Surgery, Oral Medicine, and Oral Pathology Gurol, M.E., Burkes, J., and Jacoway, J. (1995). Botryoid
17: 635–639. odontogenic cyst: analysis of 33 cases. Journal of
Graunaite, I., Lodiene, G., and Maciulskiene, V. (2012). Periodontology 66: 1069–1073.
Pathogenesis of apical periodontitis: a literature review. Habibi, A., Saghravanian, N., Habibi, M. et al. (2007).
Journal of Oral and Maxillofacial Research 2: e1.1–e1.15. Keratocystic odontogenic tumor: a 10-­year retrospective
Graves, D.T., Oates, T., and Garlet, G.P. (2011). Review of study of 83 cases in an Iranian population. Journal of Oral
osteoimmunology and the host response in endodontic Science 49: 229–235.
and periodontal lesions. Journal of Oral Microbiology Hadar, T., Shvero, J., Nageris, B.I., and Yaniv, E. (2000).
17: 1–15. Mucus retention cyst of the maxillary sinus: the
Greer, R.O. and Johnson, M. (1988). Botryoid odontogenic endoscopic approach. British Journal of Oral and
cyst: clinicopathologic analysis of ten cases with three Maxillofacial Surgery 38: 227–229.
recurrences. Journal of Oral and Maxillofacial Surgery Hadi, U., Younes, A., Ghosseini, S., and Tawil, A. (2001).
46: 574–579. Median palatine cyst: an unusual presentation of a rare
Greer, R.O., Eskendri, J., Freedman, P. et al. (2018). entity. British Journal of Oral and Maxillofacial Surgery 39:
Assessment of biologically aggressive, recurrent glandular 278–281.
odontogenic cysts for mastermind-­like 2 (MAML2) Hammad, H.M., Nagrash, O.M., and Safadi, R.A. (2020).
rearrangements: histopathologic and fluorescent in situ Maspin, syndecan-­1, and Ki-­67 in the odontogenic
hybridization (FISH) findings in 11 cases. Journal of Oral keratocyst: an immunohistochemical analysis.
Pathology and Medicine 47: 192–197. International Journal of Dentistry 14: 7041520.
Gregory, G.T. and Shafer, W.G. (1958). Surgical ciliated cysts Han, M.H., Chang, K.H., Lee, C.H. et al. (1995). Cystic
of the maxilla. Journal of Oral Surgery 16: 251–253. expansile masses of the maxilla: differential diagnosis with
Gröbe, A., Hanken, H., Blessmann, M. et al. (2012). An CT and MR. American Journal of Neuroradiology 16:
odontogenic keratocystic tumor in the buccal space: an 333–338.
unusual site of origin and a review of the literature. In vivo Han, P.P., Nagatsuka, H., Siar, C.H. et al. (2007). A pigmented
26: 847–851. calcifying cystic odontogenic tumor associated with
Grønlund, S., Mey, K., Andersen, E., and Rasmussen, compound odontoma: a case report and review of
E.R. (2016). The true malignancy rate in 135 patients with literature. Head and Face Medicine 3: 35.
preoperative diagnosis of a lateral neck cyst. Laryngoscope Hansen, J. (1967). Keratocysts in the jaws. In: Oral Surgery II
Investigative Otolaryngology 1: 78–82. (Transactions of the Second International Conference on
Grossmann, S.M., Machado, V.C., Xavier, G.M. et al. (2007). Oral Surgery) (eds. E. Husted and E. Hjørting-­Hansen),
Demographic profile of odontogenic and selected 128–134. Copenhagen: Munksgaard.
nonodontogenic cysts in a Brazilian population. Oral Hansen, L.S. and Allard, R.H.B. (1984). Encysted parasitic
Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and larvae in the mouth. Journal of the American Dental
Endodontics 104: e35–e41. Association 108: 632–636.
Grundy, G.E., Adkins, K.F., and Savage, N.W. (1984). Cysts Hansen, L.S., Sapone, J., and Sproat, R.C. (1974). Traumatic
associated with deciduous molars following pulp therapy. bone cysts of jaws. Oral Surgery, Oral Medicine, and Oral
Australian Dental Journal 29: 249–256. Pathology 37: 899–910.
Bibliography 325

Hara, H., Ohishi, M., and Higuchi, Y. (1990). Fibrous dysplasia Hedin, M., Klämfeldt, A., and Persson, G. (1978). Surgical
of the mandible associated with large solitary bone cyst. treatment of nasopalatine ducts cysts. A follow-­up study.
Journal of Oral and Maxillofacial Surgery 48: 88–91. International Journal of Oral Surgery 7: 427–433.
Harada, H., Kusukawa, J., and Kameyama, T. (1995). Heikinheimo, K., Happonen, R.-­P., Forssell, K. et al. (1989). A
Congenital teratoid cyst of the floor of the mouth: a case botryoid odontogenic cyst with multiple recurrences.
report. International Journal of Oral and Maxillofacial International Journal of Oral and Maxillofacial Surgery
Surgery 24: 361–362. 18: 10–13.
Harnet, J.C., Lombardi, T., Klewansky, P. et al. (2008). Heikinheimo, K., Jee, K.J., Morgan, P.R. et al. (2007). Genetic
Solitary bone cyst of the jaws: a review of the changes in sporadic keratocystic odontogenic tumors
etiopathogenic hypotheses. Journal of Oral and (odontogenic keratocysts). Journal of Dental Research
Maxillofacial Surgery 66: 2345–2348. 86: 544–549.
Harris, M. (1978). Odontogenic cyst growth and Heikinheimo, K., Kurppa, K.J., Laiho, A. et al. (2015). Early
prostaglandin-­induced bone resorption. Annals of the dental epithelial transcription factors distinguish
Royal College of Surgeons of England 60: 85–91. ameloblastoma from keratocystic odontogenic tumor.
Harris, M. and Goldhaber, P. (1973). The production of a Journal of Dental Research 94: 101–111.
bone resorbing factor by dental cysts in vitro. British Henley, J., Summerlin, D.J., Tomich, C. et al. (2005).
Journal of Oral Surgery 10: 334–338. Molecular evidence supporting the neoplastic nature of
Harris, M. and Toller, P. (1975). The pathogenesis of dental odontogenic keratocyst: a laser capture microdissection
cysts. British Medical Bulletin 31: 159–163. study of 15 cases. Histopathology 47: 582–586.
Harris, M., Jenkins, M.V., Bennett, A., and Wills, M.R. (1973). Henry, C.R., Nace, M., and Helm, K.F. (2008). Collagenous
Prostaglandin production and bone resorption by dental spherulosis in an oral mucous cyst. Journal of Cutaneous
cysts. Nature 245: 213–215. Pathology 35: 428–430.
Harrison, J.D. (1975). Salivary mucoceles. Oral Surgery, Oral High, A.S. and Hirschmann, P.N. (1986). Age changes in residual
Medicine, and Oral Pathology 39: 268–278. radicular cysts. Journal of Oral Pathology 15: 524–528.
Harrison, J.D. (2010). Modern management and High, A.S. and Hirschmann, P.N. (1988). Symptomatic
pathophysiology of ranula: literature review. Head and residual radicular cysts. Journal of Oral Pathology 17: 70–72.
Neck 32: 1310–1320. High, A.S., Robinson, P.A., and Klein, C.E. (1993).
Harrison, J.D. and Garrett, J.R. (1972). Mucocele formation in Discrimination of parakeratinised odontogenic keratocysts
cats by glandular duct ligation. Archives of Oral Biology 17: from other odontogenic and non-­odontogenic cyst types by
1403–1414. expression of a 38kd cell-­surface glycoprotein. Journal of
Harrison, J.D. and Garrett, J.R. (1975). Experimental salivary Oral Pathology and Medicine 22: 363–367.
mucoceles in a cat. A histochemical study. Journal of Oral High, A.S., Main, D.M.G., Khoo, S.P. et al. (1996). The
Pathology 4: 297–306. polymorphous odontogenic cyst. Journal of Oral Pathology
Harrison, J.D., Kim, A., Al-­Ali, S., and Morton, R.P. (2013). and Medicine 25: 25–31.
Postmortem investigation of mylohyoid hiatus and hernia: Higuchi, Y., Nakamura, N., and Tashiro, H. (1988).
aetiological factors of plunging ranula. Clinical Anatomy Clinicopathologic study of cemento-­osseous dysplasia
26: 693–699. producing cysts of the mandible. Report of four cases. Oral
Harvey, W. and Noble, H.W. (1968). Defects on the lingual Surgery, Oral Medicine, and Oral Pathology 65: 339–342.
surface of the mandible near the angle. British Journal of Hills, A., Holden, A., and McGurk, M. (2016). Evolution of
Oral Surgery 6: 75–83. the management of ranulas: change in a single surgeon’s
Hassanein, A.M., Glanz, S.M., Kessler, H.P. et al. (2003). practice 2001–14. British Journal of Oral and Maxillofacial
Beta-­catenin is expressed aberrantly in tumors expressing Surgery 54: 992–996.
shadow cells. Pilomatricoma, craniopharyngioma, and Hirata, K., Fujima, N., Mizumachi, T. et al. (2014).
calcifying odontogenic cyst. American Journal of Clinical Radiological images of keratocystoma: a rare tumor of the
Pathology 120: 732–736. parotid gland. Acta Radiologica Short Reports 3:
Hauer, L., Seidlová, P., Merglová, V. et al. (2020). Complete 204798161454949.
removal of dentigerous cysts with preservation of Hirota, J., Maeda, Y., Ueta, E., and Osaki, T. (1989).
associated teeth as an alternative to marsupialization in Immunohistochemical and histologic study of cervical
children and preadolescents. Journal of Cranio-­ lymphoepithelial cysts. Journal of Oral Pathology and
Maxillofacial Surgery 48: 808–814. Medicine 18: 202–205.
Hayashida, A.M., Zerbinatti, D.C., Balducci, I. et al. (2010). Hirshberg, A., Dayan, D., and Horowitz, I. (1987).
Mucus extravasation and retention phenomena: a 24-­year Dentinogenic ghost cell tumor. International Journal of
study. BMC Oral Health 10: 15. Oral and Maxillofacial Surgery 16: 620–625.
326 Bibliography

Hirshberg, A., Kaplan, I., and Buchner, A. (1994). Calcifying Howe, G.L. (1965). ‘Haemorrhagic cysts’ of the mandible.
odontogenic cyst associated with odontoma: a possible British Journal of Oral Surgery 3: 55–75; 77–91.
separate entity (odontocalcifying odontogenic cyst). Howell, C.J.T. (1985). The sublingual dermoid cyst. Report of
Journal of Oral and Maxillofacial Surgery 52: 555–558. five cases and review of the literature. Oral Surgery, Oral
Hirshberg, A., Sherman, S., Buchner, A., and Dayan, D. (1999). Medicine, and Oral Pathology 59: 578–580.
Collagen fibres in the wall of odontogenic keratocysts: a Hoyos Cadavid, A.M., Kaminagakura, E., Rodrigues,
study with picrosirius red and polarizing microscopy. M.F.S.D. et al. (2019). Immunohistochemical evaluation of
Journal of Oral Pathology and Medicine 28: 410–412. Sonic Hedgehog signaling pathway proteins (Shh, Ptch1,
Hirshberg, A., Tsesis, I., Metzger, Z., and Kaplan, I. (2003). Ptch2, Smo, Gli1, Gli2, and Gli3) in sporadic and
Periapical actinomycosis: a clinicopathologic study. Oral syndromic odontogenic keratocysts. Clinical Oral
Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and Investigations 23: 153–159.
Endodontics 95: 614–620. Huang, G.T. (2010). Apical cyst theory: a missing link. Dental
Hisatomi, M., Munhoz, L., Asaumi, J., and Arita, E.S. (2019). Hypotheses 5: 76–84.
Stafne bone defects radiographic features in panoramic Huang, X.F., Li, W., Yang, X.D. et al. (2012). Keratocystoma of
radiographs: assessment of 91 cases. Medicina Oral, the parotid gland:a clinicopathological study and literature
Patología Oral y Cirugía Bucal 24: e12–e19. review. International Journal of Oral and Maxillofacial
Hjørting-­Hansen, E., Andreasen, J.O., and Robinson, Surgery 41: 256–260.
L.H. (1969). A study of odontogenic cysts with special Huang, G., Moore, L., Logan, R.M., and Gue, S. (2019).
reference to location of keratocysts. British Journal of Oral Histological analysis of 41 dentigerous cysts in a paediatric
Surgery 7: 15–23. population. Journal of Oral Pathology and Medicine 48: 74–78.
Ho, M.W. and Crean, S.J. (2003). Simultaneous occurrence of Hume, W.J., Moore, J.K., and Main, D.M.G. (1990).
sublingual dermoid cyst and oral alimentary tract cyst in Differences in in vitro growth of epithelium from
an infant: a case report and review of the literature. inflammatory and developmental odontogenic cysts.
International Journal of Paediatric Dentistry 13: 441–446. British Journal of Oral and Maxillofacial Surgery 28: 25–28.
Hodson, J.J. (1956). Muco-­epidermoid odontogenic cysts of Hunter, K.D. and Speight, P.M. (2014). The diagnostic
the jaws with special reference to those in the mandible. usefulness of immunohistochemistry for odontogenic
Proceedings of the Royal Society of Medicine 49: 637–639. lesions. Head and Neck Pathology 8: 392–399.
Hodson, J.J. (1962). Epithelial residues of the jaw with special Hussain, K., Edmonson, H.D., and Browne, R.M. (1995).
reference to the edentulous jaw. Journal of Anatomy Glandular odontogenic cysts. Diagnosis and treatment.
96: 16–24. Oral Surgery, Oral Medicine, Oral Pathology, Oral
Hofrath, H. (1930). Über das Vorkommen von Talgdrüsen in Radiology, and Endodontics 79: 593–602.
der Wandung einer Zahncyste, zugleich ein Beitrag zur Hyomoto, M., Kawakami, M., Inoue, M., and Kirita, T. (2003).
Pathogenese der Kiefer-­Zahncysten. Deutsche Monatsschr Clinical conditions for eruption of maxillary canines and
Zahnheilk 48: 65–76. mandibular premolars associated with dentigerous cysts.
Hong, S.S., Ogawa, Y., Yagi, T. et al. (1990). Benign American Journal of Orthodontics and Dentofacial
lymphoepithelial lesion with large cysts: case report. Orthopedics 124: 515–520.
Journal of Oral Pathology and Medicine 19: 266–270. Ide, F. and Kusama, K. (2002a). Myxoglobulosis-­like change
Hong, S.P., Ellis, G.L., and Hartman, K.S. (1991). Calcifying in a lower lip mucocele. Histopathology 40: 575–576.
odontogenic cyst. A review of ninety-­two cases with Ide, F. and Kusama, K. (2002b). Basal cell hamartia: a
reevaluation of their nature as cysts or neoplasms, the rudiment no longer. International Journal of Oral and
nature of ghost cells, and subclassification. Oral Surgery, Maxillofacial Surgery 31: 461–462.
Oral Medicine, and Oral Pathology 72: 56–64. Ide, F., Shimoyama, T., and Horie, N. (1996). Glandular
Horner, K. and Forman, G.H. (1988). Atypical simple bone odontogenic cyst with hyaline bodies: an unusual
cysts of the jaws. II: a possible association with benign dentigerous presentation. Journal of Oral Pathology and
fibroosseous (cemental) lesions of the jaws. Clinical Medicine 25: 401–404.
Radiology 39: 59–63. Ide, F., Shimoyama, T., and Horie, N. (2002). Peripheral
Horner, K., Forman, G.H., and Smith, N.J. (1988). Atypical odontogenic keratocyst: a report of 2 cases. Journal of
simple bone cysts of the jaws. I: recurrent lesions. Clinical Periodontology 73: 1079–1081.
Radiology 39: 53–57. Ide, F., Mishima, K., and Saito, I. (2003). Solid-­cystic tumor
Hovorakova, M., Lesot, H., Peterka, M., and Peterkova, R. variant of odontogenic keratocyst: an aggressive but benign
(2018). Early development of the human dentition lesion simulating keratoameloblastoma. Virchows Archiv
revisited. Journal of Anatomy 233: 135–145. 442: 501–503.
Bibliography 327

Ide, F., Mishima, K., Saito, I., and Kusama, K. (2008). Rare Iida, S., Aikawa, T., Kishino, M. et al. (2006b). Spheric mass
peripheral odontogenic tumors: report of 5 cases and beneath the alar base: MR images of nasolabial cyst and
comprehensive review of the literature. Oral Surgery, Oral schwannoma. American Journal of Neuroradiology 27:
Medicine, Oral Pathology, Oral Radiology, and Endodontics 1826–1829.
106: e22–e28. Imholte, M. and Schwartz, H.C. (2001). Respiratory
Ide, F., Horie, N., Shimoyama, T. et al. (2009). Infrequent implantation cyst of the mandible after chin augmentation:
clinicopathologic features of keratocystic odontogenic report of case. Otolaryngology and Head and Neck Surgery
tumour: a 29-­year multi-­institutional retrospective review. 124: 586–587.
Oral Surgery 2: 1–9. Irani, S. and Foroughi, F. (2017). Histologic variants of
Ide, F., Kikuchi, K., and Kusama, K. (2010). Is papillary calcifying odontogenic cyst: a study of 52 cases. Journal of
synovial metaplasia-­like change a hitherto undescribed Contemporary Dental Practice 18: 688–694.
histologic pattern of lip mucoceles? Oral Surgery, Oral Issa, M.A. and Davies, J.D. (1971). Dermoid cyst of the jaw.
Medicine, Oral Pathology, Oral Radiology, and Endodontics British Dental Journal 131: 543–546.
109: 800–801. Jackson, I.T., Potparic, Z., Fasching, M. et al. (1993).
Ide, F., Kikuchi, K., Miyazaki, Y. et al. (2010). Keratocyst of Penetration of the skull base by dissecting keratocyst.
the buccal mucosa: is it odontogenic? Oral Surgery, Oral Journal of Craniofacial Surgery 21: 319–325.
Medicine, Oral Pathology, Oral Radiology, and Endodontics Jacob, S., Zelano, B., Gungor, A. et al. (2000). Location and
110: 42–47. gross morphology of the nasopalatine duct in human
Ide, F., Mishima, K., Kikuchi, K. et al. (2011). Development adults. Archives of Otolaryngology – Head and Neck Surgery
and growth of adenomatoid odontogenic tumor related to 126: 741–748.
formation and eruption of teeth. Head and Neck Pathology Jaeger, F., de Noronha, M.S., Silva, M.L. et al. (2017).
5: 123–132. Prevalence profile of odontogenic cysts and tumors on
Ide, F., Ito, Y., Muramatsu, T. et al. (2012). Histogenetic Brazilian sample after the reclassification of odontogenic
relations between keratoameloblastoma and solid variant keratocyst. Journal of Craniofacial Surgery 45: 267–270.
of odontogenic keratocyst. Oral Surgery, Oral Medicine, Jaffe, H.L. (1953). Giant cell reparative granuloma, traumatic
Oral Pathology, Oral Radiology 114: 812–813. bone cyst and fibrous (fibro-­osseous) dysplasia of the
Ide, F., Kikuchi, K., Miyazaki, Y. et al. (2015). The early jawbones. Oral Surgery, Oral Medicine, and Oral Pathology
history of odontogenic ghost cell lesions: from Thoma to 6: 159–175.
Gorlin. Head and Neck Pathology 9: 74–78. Jaffe, H.L. and Lichtenstein, L. (1942). Solitary unicameral
Ide, F., Muramatsu, T., Miyazaki, Y. et al. (2019). Calcifying bone cyst with emphasis on the roentgen picture, the
odontogenic cyst showing a varied epithelial lining: an pathologic appearance, and the pathogenesis. Archives of
additional case with implications for the divergent Surgery 44: 1004–1025.
differentiation capacity of the cyst epithelium. Head and Jain, R., Morton, R.P., and Ahmad, Z. (2012). Diagnostic
Neck Pathology 13: 251–254. difficulties of plunging ranula: case series. Journal of
Ide, F., Ito, Y., Muramatsu, T. et al. (2020). The advent of Laryngology and Otology 126: 506–510.
studies on jaw cysts with keratinization: a review of Jain, M., Mittal, S., and Gupta, D.K. (2013). Primary
overlooked papers on odontogenic keratocyst and intraosseous squamous cell carcinoma arising in
orthokeratinized odontogenic cyst. Head and Neck odontogenic cysts: an insight in pathogenesis. Journal of
Pathology 14: 785–791. Oral and Maxillofacial Surgery 71: e7–e14.
Idowu, B.D., Al-­Adnani, M., O’Donnell, P. et al. (2007). A Janardhanan, M., Rakesh, S., Savithri, V., Aravind, T., Mohan,
sensitive mutation-­specific screening technique for M. (2021). Carcinoma Cuniculatum of Mandible
GNAS1 mutations in cases of fibrous dysplasia: the first Masquerading as Odontogenic Keratocyst: Challenges in
report of a codon 227 mutation in bone. Histopathology the Histopathological Diagnosis. Head and Neck Pathology
50: 691–704. 15 (4): 1313–1321.
Iezzi, G., Rubini, C., Zizzi, A. et al. (2011). Solid variant of Jattan, R., De Silva, H.L., De Silva, R.K. et al. (2011). A case
keratocystic odontogenic tumour: report of a case. Minerva series of odontogenic keratocysts from a New Zealand
Stomatologica 60: 133–138. population over a 20-­year period. New Zealand Dental
Iida, S., Fukuda, Y., Ueda, T. et al. (2006a). Calcifying Journal 107: 112–116.
odontogenic cyst: radiologic findings in 11 cases. Oral Jay, A., Dhanda, J., Chiodini, P.L. et al. (2005). Oral
Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and cysticercosis. British Journal of Oral and Maxillofacial
Endodontics 101: 356–362. Surgery 45 (4): 331–334.
328 Bibliography

Jensen, J.L. (1990). Superficial mucoceles of the oral mucosa. population over a 30-­year period. Journal of Oral Pathology
American Journal of Dermatopathology 12: 88–92. and Medicine 35: 500–507.
Jensen, J.L. and Erickson, J.O. (1974). Hyaline bodies in Jordan, R.C. and Koutlas, I.G. (2022). Aneurysmal bone cyst.
odontogenic cysts: electron microscopic observations. In: WHO Classification of Tumours Editorial Board. Head
Journal of Oral Pathology 3: 1–6. and Neck Tumours, Section 7.7.0.4 (WHO Classification of
Jeon, J.Y., Park, C.J., Cho, S.H., and Hwang, K.G. (2016). Bilateral Tumours Series), 5e, vol. 9. Lyon (France): International
dentigerous cysts that involve all four dental quadrants: a case Agency for Research on Cancer https://publications.iarc.
report and literature review. Journal of the Korean Association fr/Book-­And-­Report-­Series/Who-­Classification-­Of-
of Oral and Maxillofacial Surgeons 42: 123–126. ­Tumours.
Jeyaraj, P. (2019). The dilemma of extensive unilocular Jose, M., Rao, N.N., and Solomon, M.C. (2000). Glandular
radiolucent lesions of the jaws – value of odontogenic cyst. A rare entity with aggressive biological
immunohistochemistry as a diagnostic marker and behaviour: a case report. Indian Journal of Dental Research
prognostic Indicator. Annals of Diagnostic Pathology 11: 107–110.
40: 105–135. Joseph, J., Dungarwalla, M.M., and Jones, J. (2021). A case
Jia, M.Q., Jia, J., Wang, L., and Zou, H.X. (2019). Ghost cell report and literature review of multiple orthokeratinizing
odontogenic carcinoma of the jaws: report of two cases and odontogenic cysts: the great mimicker. Oral Surgery 14 (4):
a literature review. World Journal of Clinical Cases 7 (3): 407–411.
357–365. Kaczmarzyk, T., Mojsa, I., and Stypulkowska, J. (2012). A
Jinbu, Y., Kusama, M., Itoh, H. et al. (2003). Mucocele of systematic review of the recurrence rate for keratocystic
the glands of Blandin-­Nühn: clinical and histopathologic odontogenic tumour in relation to treatment modalities.
analysis of 26 cases. Oral Surgery, Oral Medicine, International Journal of Oral and Maxillofacial Surgery
Oral Pathology, Oral Radiology, and Endodontics 41: 756–767.
95: 467–470. Kahn, A., Matalon, S., Salem, R.B. et al. (2021). Sinus floor
Jing, W., Xuan, M., Lin, Y. et al. (2007). Odontogenic augmentation – associated surgical ciliated cysts: case
tumours: a retrospective study of 1642 cases in a Chinese series and a systematic review of the literature. Applied
population. International Journal of Oral and Maxillofacial Sciences 11: 1903.
Surgery 36: 20–25. Kahraman, D., Gunhan, O., and Celasun, B. (2018). A series
Johannessen, A.C. (1986). Esterase-­positive inflammatory of 240 odontogenic keratocysts: should we continue to use
cells in human periapical lesions. Journal of Endodontics the terminology of �keratocystic odontogenic tumour’ for
12: 284–288. the solid variant of odontogenic keratocyst? Journal of
Johnson, N.R., Batstone, M.D., and Savage, N.W. (2013). Cranio-­Maxillo-­Facial Surgery 46: 942–946.
Management and recurrence of keratocystic odontogenic Kaminagakura, E., Almeida, J.D., Carvalho, Y.R. et al. (2013).
tumor: a systematic review. Oral Surgery, Oral Medicine, Keratocyst of the buccal mucosa: case report and
Oral Pathology, Oral Radiology 116: e271–e276. immunohistochemical comparative study with sporadic
Johnson, N.R., Savage, N.W., Kazoullis, S., and Batstone, intraosseous keratocystic odontogenic tumor. Oral Surgery,
M.D. (2013). A prospective epidemiological study for Oral Medicine, Oral Pathology, Oral Radiology 116:
odontogenic and non-­odontogenic lesions of the maxilla e387–e392.
and mandible in Queensland. Oral Surgery, Oral Medicine, Kaminagakura, E., Domingos, P.L., da Rosa, M.R. et al.
Oral Pathology, Oral Radiology 115: 515–522. (2013). Detection of cytokeratins in ghost cells of calcifying
Johnson, N.R., Gannon, M., Savage, N.W., and Batstone, cystic odontogenic tumor indicates an altered
M.D. (2014). Frequency of odontogenic cysts and tumors: a keratinization and hair follicle differentiation for their
systematic review. Journal of Investigative and Clinical development. Annals of Diagnostic Pathology 17: 514–517.
Dentistry 5: 9–14. Kammer, P.V., Mello, F.W., and Rivero, E.R.C. (2020).
Jones, A.V. and Franklin, C.D. (2006a). An analysis of oral Comparative analysis between developmental and
and maxillofacial pathology found in adults over a 30-­year inflammatory odontogenic cysts: retrospective study and
period. Journal of Oral Pathology and Medicine 35: 392–401. literature review. Oral and Maxillofacial Surgery 24: 73–84.
Jones, A.V. and Franklin, C.D. (2006b). An analysis of oral Kaneshiro, S., Nakajima, T., Yoshikawa, Y. et al. (1981). The
and maxillofacial pathology found in children over a postoperative maxillary cyst: report of 71 cases. Journal of
30-­year period. International Journal of Paediatric Dentistry Oral Surgery 39: 191–198.
16: 19–30. Kaplan, I., Anavi, Y., Manor, R. et al. (2005a). The use of
Jones, A.V., Craig, G.T., and Franklin, C.D. (2006). Range and molecular markers as an aid in the diagnosis of glandular
demographics of odontogenic cysts diagnosed in a UK odontogenic cyst. Oral Oncology 41: 895–902.
Bibliography 329

Kaplan, I., Gal, G., Anavi, Y. et al. (2005b). Glandular Khandeparker, R.V., Khandeparker, P.V., Virginkar, A.,
odontogenic cyst: treatment and recurrence. Journal of and Savant, K. (2018). Bilateral maxillary dentigerous
Oral and Maxillofacial Surgery 63: 435–441. cysts in a nonsyndromic child: a rare presentation and
Kaplan, I., Anavi, Y., and Hirshberg, A. (2008). Glandular review of the literature. Case Reports in Dentistry 2018:
odontogenic cyst: a challenge in diagnosis and treatment. 7583082.
Oral Diseases 14: 575–581. Kichi, E., Enokiya, Y., Muramatsu, T. et al. (2005). Cell
Karabas, H.C., Ozcan, I., Tekkesin, M.S. et al. (2020). proliferation, apoptosis and apoptosis-­related factors in
Evaluation of radiolucent lesions associated with impacted odontogenic keratocysts and in dentigerous cysts. Journal
teeth: a retrospective study. Current Medical Imaging 16: of Oral Pathology and Medicine 34: 280–286.
1332–1339. Kieran, S.M., Robson, C.D., Nosé, V., and Rahbar, R. (2010).
Karhade, D.S., Afshar, S., and Padwa, B.L. (2019). What is Foregut duplication cysts in the head and neck:
the prevalence of undiagnosed nevoid basal cell presentation, diagnosis, and management. Archives of
carcinoma syndrome in children with an odontogenic Otolaryngology – Head and Neck Surgery 136: 778–782.
keratocyst? Journal of Oral and Maxillofacial Surgery 77: Kikuchi, K., Ito, S., Inoue, H. et al. (2012).
1389–1391. Immunohistochemical expression of podoplanin in
Kato, C.N.A.O., Barra, S.G., Amaral, T.M.P. et al. (2020). so-­called hard α-­keratin-­expressing tumors, including
Cone-­beam computed tomography analysis of cemento-­ calcifying cystic odontogenic tumor, craniopharyngioma,
osseous dysplasia-­induced changes in adjacent structures and pilomatrixoma. Journal of Oral Science 54:
in a Brazilian population. Clinical Oral Investigations 165–175.
24: 2899–2908. Kim, S. and Jeong, S. (2019). Mutation hotspots in the
Kaugars, G.E. (1986). Botryoid odontogenic cyst. Oral β-­catenin gene: lessons from the human cancer genome
Surgery, Oral Medicine, and Oral Pathology 62: 555–559. databases. Molecules and Cells 42: 8–16.
Kaugars, C.C., Kaugars, G.E., and DeBiasi, G.F. (1989a). Kim, S.-­G., Yang, B.-­E., Oh, S.-­H. et al. (2005). The differential
Extraosseous calcifying odontogenic cyst: report of case expression pattern of BMP-­4 between the dentigerous cyst
and review of literature. Journal of the American Dental and the odontogenic keratocyst. Journal of Oral Pathology
Association 119: 715–718. and Medicine 34: 178–183.
Kaugars, G.E., Miller, M.E., and Abbey, L.M. (1989b). Kim, S.A., Ahn, S.G., Kim, S.G. et al. (2007). Investigation of
Odontomas. Oral Surgery, Oral Medicine, and Oral the beta-­catenin gene in a case of dentinogenic ghost cell
Pathology 67: 172–176. tumor. Oral Surgery, Oral Medicine, Oral Pathology, Oral
Kawano, K., Okamura, K., Kashima, K. et al. (2013). Solid Radiology, and Endodontics 103: 97–101.
variant of keratocystic odontogenic tumor of the mandible: Kimonis, V.E., Goldstein, A.M., Pastakia, B. et al. (1997).
report of a case with a clear cell component and review of Clinical manifestations in 105 persons with nevoid basal
the literature. Oral Surgery, Oral Medicine, Oral Pathology, cell carcinoma syndrome. American Journal of Medical
Oral Radiology 116: e393–e398. Genetics 69: 299–308.
Kawashima, N., Suzuki, N., Yang, G. et al. (2007). Kinetics of Kimonis, V.E., Mehta, S.G., Digiovanna, J.J. et al. (2004).
RANKL, RANK and OPG expressions in experimentally Radiological features in 82 patients with nevoid basal cell
induced rat periapical lesions. Oral Surgery, Oral Medicine, carcinoma (NBCC or Gorlin) syndrome. Genetics in
Oral Pathology, Oral Radiology, and Endodontics Medicine 6: 495–502.
103: 707–711. Kimura, J.S., Wanderley, M.T., Pinto-­Junior Ddos, S., and
Kaya, G. and Saurat, J.H. (2018). Cutaneous adnexal cysts Zardetto, C.G. (2014). An unusual case of four
revisited: what we know and what we think we know. simultaneous eruption cysts in an infant. Journal of
Dermatopathology (Basel) 5: 79–85. Dentistry for Children 81: 38–41.
Keinan, D. and Cohen, R.E. (2013). The significance of King, R.C., Smith, B.R., and Burk, J.L. (1994). Dermoid cyst
epithelial rests of Malassez in the periodontal ligament. in the floor of the mouth. Review of the literature and case
Journal of Endodontics 39: 582–587. reports. Oral Surgery, Oral Medicine, and Oral Pathology
Kelly, J.P., Malik, S., and Stucki-­McCormick, S.U. (2000). 78: 567–576.
Tender swelling of the chin 40 years after genioplasty. Kitisubkanchana, J., Reduwan, N.H., Poomsawat, S. et al.
Journal of Oral and Maxillofacial Surgery 58: 203–206. (2021). Odontogenic keratocyst and ameloblastoma:
Khandelwal, P., Aditya, A., and Mhapuskar, A. (2015). radiographic evaluation. Oral Radiology 37: 55–65.
Bilateral calcifying cystic odontogenic tumour of mandible: Klestadt, W.D. (1953). Nasal cysts and the facial cleft cyst
a rare case report and review of literature. Journal of theory. Annals of Otology, Rhinology, and Laryngology
Clinical and Diagnostic Research 9: ZD20–ZD22. 62: 84–92.
330 Bibliography

Knapp, M.J. (1970a). Oral tonsils: location, distribution, and Koutlas, I.G., Gillum, R.B., Harris, M.W., and Brown,
histology. Oral Surgery, Oral Medicine, and Oral Pathology B.A. (2002). Surgical (implantation) cyst of the mandible
29: 155–161. with ciliated respiratory epithelial lining: a case report.
Knapp, M.J. (1970b). Pathology of oral tonsils. Oral Surgery, Journal of Oral and Maxillofacial Surgery 60: 324–325.
Oral Medicine, and Oral Pathology 29: 295–304. Kramer, I.R.H. (1963). Ameloblastoma: a clinicopathological
Knudson, A.G. (1971). Mutation and cancer: statistical study appraisal. British Journal of Oral Surgery 1: 13–28.
of retinoblastoma. Proceedings of the National Academy of Kramer, I.R.H. (1974). Changing views on oral disease.
Sciences USA 68: 820–823. Proceedings of the Royal Society of Medicine 67:
Knudson, A.G. (1996). Hereditary cancer: two hits revisited. 271–276.
Journal of Cancer Research and Clinical Oncology 122: Kramer, I.R.H. and Toller, P.A. (1973). The use of exfoliative
135–140. cytology and protein estimations in preoperative diagnosis
Koca, H., Esin, A., and Aycan, K. (2009). Outcome of of odontogenic keratocysts. International Journal of Oral
dentigerous cysts treated with marsupialization. Journal of Surgery 2: 143–151.
Clinical Pediatric Dentistry 34: 165–168. Kramer, I.R.H., Pindborg, J.J., and Shear, M. (1992).
Koivisto, T., Bowles, W.R., and Rohrer, M. (2012). Frequency Histological Typing of Odontogenic Tumours. Berlin:
and distribution of radiolucent jaw lesions: a retrospective Springer.
analysis of 9,723 cases. Journal of Endodontics 38: 729–732. Kratochvil, F.J. and Brannon, R.B. (1993). Cartilage in the
Kolář, Z., Geierova, M., Bouchal, J. et al. (2006). walls of odontogenic keratocysts. Journal of Oral Pathology
Immunohistochemical analysis of the biological potential and Medicine 22: 282–285.
of odontogenic keratocysts. Journal of Oral Pathology and Kubo, I. (1927). A buccal cyst occurred after a radical
Medicine 35: 75–80. operation of the maxillary sinus in Japanese. Z F OtolTokyo
Komatsu, H., Kumoi, K., Inokuchi, G. et al. (2020). 3: 896–897.
Keratocystoma of the parotid gland. Auris Nasus Larynx Kubota, Y., Ninomiya, T., Oka, S. et al. (2000). Interleukin
47: 481–484. IL-­1-­dependent regulation of matrix metalloproteinase-­9
Kontiainen, S., Ranta, H., and Lautenschlager, I. (1986). Cells (MMP-­9) secretion and activation in the epithelial cells of
infiltrating human periapical inflammatory lesions. odontogenic jaw cysts. Journal of Dental Research 79:
Journal of Oral Pathology 15: 544–546. 1423–1430.
Koppang, H.S., Johannessen, S., Haugen, L.K. et al. (1998). Kubota, Y., Yamashiro, T., Oka, S. et al. (2004). Relation
Glandular odontogenic cyst (sialo-­odontogenic cyst): between size of odontogenic jaw cysts and the pressure of
report of two cases and literature review of 45 previously fluid within. British Journal of Oral and Maxillofacial
reported cases. Journal of Oral Pathology and Medicine 27: Surgery 42: 391–395.
455–462. Kuczek, A., Beikler, T., Herbst, H., and Flemmig, T.F. (2003).
Koskimies, A.I., Ylipaavalniemi, P., and Tuompo, H. (1975). Eruption cyst formation associated with cyclosporin
Polyacrylamide gel electrophoresis of proteins in fluids A. Journal of Clinical Periodontology 30: 462–466.
from jaw cysts. Proceedings of the Finnish Dental Society Kular, J., Tickner, J., Chim, S.M., and Xu, J. (2012). An
71: 6–9. overview of the regulation of bone remodelling at the
Koudounarakis, E., Willems, S., and Karakullukcu, B. (2016). cellular level. Clinical Biochemistry 45: 863–873.
Dysgenetic polycystic disease of the minor and Kumchai, H., Champion, A.F., and Gates, J.C. (2021).
submandibular salivary glands. Head and Neck 38: Carcinomatous transformation of odontogenic keratocyst
E2437–E2439. and primary intraosseous carcinoma: a systematic review
Kouhsoltani, M., Halimi, M., and Jabbari, G. (2016). and report of a case. Journal of Oral and Maxillofacial
Immunohistochemical evaluation of myofibroblast density Surgery 79: 1081.e1–1081.e9.
in odontogenic cysts and tumors. Journal of Dental Kusama, K., Katayama, Y., Oba, K. et al. (2005). Expression of
Research, Dental Clinics, Dental Prospects 10: 37–42. hard alpha-­keratins in pilomatrixoma, craniopharyngioma,
Koutlas, I.G. and Sloan, P. (2022). Primary intraosseous and calcifying odontogenic cyst. American Journal of
carcinoma NOS. In: WHO Classification of Tumours Clinical Pathology 123: 376–381.
Editorial Board. Head and Neck Tumours, Section 7.2.2.2 Kusumi, A., Sakaki, H., Fukui, R. et al. (2004). High IL-­6
(WHO Classification of Tumours Series), 5e, vol. 9. Lyon: synthesis in cultured fibroblasts isolated from radicular
International Agency for Research on Cancer https:// cysts. Archives of Oral Biology 49: 643–652.
publications.iarc.fr/Book-­And-­Report-­Series/ Lake, S., Iwanaga, J., Kikuta, S. et al. (2018). The incisive
Who-­Classification-­Of-­Tumours. canal: a comprehensive review. Cureus 10: e3069.
Bibliography 331

Lalla, K., Mahomed, F., and Meer, S. (2016). Keratinizing mylohyoid muscle as the primary route of lesion
odontogenic cysts with a spectrum of verrucoid propagation. Korean Journal of Radiology 17: 264–270.
morphology: investigation of a potential role of human Lello, G.E. and Makek, M. (1985). Stafne’s mandibular lingual
papillomavirus. Oral Surgery, Oral Medicine, Oral cortical defect. Journal of Oral Maxillofacial Surgery 13:
Pathology, Oral Radiology 122: 625–630. 172–176.
Lam, K.Y. and Chan, A.C. (2000). Odontogenic keratocysts: a Lench, N.J., High, A.S., Markham, A.F. et al. (1996).
clinicopathological study in Hong Kong Chinese. Investigation of chromosome 9q22.3-­q31 DNA marker loss
Laryngoscope 110: 1328–1332. in odontogenic keratocysts. European Journal of Cancer
Laux, M., Abbott, P.V., Pajarola, G., and Nair, P.N.R. (2000). Oral Oncology 32B: 202–206.
Apical inflammatory root resorption: a correlative Lench, N.J., Telford, E.A.R., High, A.S. et al. (1997).
radiographic and histological assessment. International Characterisation of human patched germ line mutations in
Endodontic Journal 33: 483–493. naevoid basal cell carcinoma syndrome. Human Genetics
Layfield, L.J., Esebua, M., and Schmidt, R.L. (2016). Cytologic 100: 497–502.
separation of branchial cleft cyst from metastatic cystic Leonardi, R., Caltabiano, R., and Loreto, C. (2005).
squamous cell carcinoma: a multivariate analysis of Collagenase-­3 (MMP-­13) is expressed in periapical lesions:
nineteen cytomorphologic features. Diagnostic an immunohistochemical study. International Endodontic
Cytopathology 44: 561–567. Journal 38: 297–301.
Lazar, F., zur Hausen, A., Mischkowski, R., and Zoller, Leonardi, R., Perrotta, R.E., Loreto, C. et al. (2015). Toll-­like
J.E. (2006). Atypical cyst formation following chin receptor 4 expression in the epithelium of inflammatory
augmentation using a nasal osteocartilaginous graft. periapical lesions. An immunohistochemical study.
Journal of Cranio-­Maxillofacial Surgery 34: 107–112. European Journal of Histochemistry 59: 279–282.
Lechner, J., Schuett, S., and von Baehr, V. (2017). Aseptic-­ Leung, Y.Y., Wong, W.Y., and Cheung, L.K. (2012). Surgical
avascular osteonecrosis: local ‘silent inflammation’ in the ciliated cysts may mimic radicular cysts or residual cysts of
jawbone and RANTES/CCL5 overexpression. Clinical, maxilla: report of 3 cases. Journal of Oral and Maxillofacial
Cosmetic and Investigational Dentistry 9: 99–109. Surgery 70: e264–e269.
Lechner, J., Schulz, T., and von Baehr, V. (2019). Levanat, S., Gorlin, R.J., Fallet, S. et al. (1996). A two-­hit
Immunohistological staining of unknown chemokine model for developmental defects in Gorlin Syndrome.
RANTES/CCL5 expression in jawbone marrow Nature Genetics 12: 85–87.
defects-­osteoimmunology and disruption of bone Levanat, S., Pavelić, B., Crnić, I. et al. (2000). Involvement of
remodeling in clinical case studies targeting on PTCH gene in various noninflammatory cysts. Journal of
predictive preventive personalized medicine. EPMA Molecular Medicine 78: 140–146.
Journal 10: 351–364. Lewis, J.S. Jr. (2020). Human papillomavirus testing in head
Lechner, J., Zimmermann, B., Schmidt, M., and von Baehr, V. and neck squamous cell carcinoma in 2020: where are we
(2020). Ultrasound sonography to detect focal osteoporotic now and where are we going? Head and Neck Pathology 14:
jawbone marrow defects clinical comparative study with 321–329.
corresponding hounsfield units and RANTES/CCL5 Li, T.J. (2011). The odontogenic keratocyst: a cyst, or a cystic
expression. Clinical, Cosmetic and Investigational Dentistry neoplasm? Journal of Dental Research 90: 133–142.
12: 205–216. Li, T.J. and Yu, S.F. (2003). Clinicopathologic spectrum of the
Ledesma-­Montes, C., Hernándes-­Guerrero, J.C., and so-­called calcifying odontogenic cysts: a study of
Garcés-­Ortiz, M. (2000). Clinico-­pathologic study of 21 intraosseous cases with reconsideration of the
odontogenic cysts in a Mexican sample population. terminology and classification. American Journal of
Archives of Medical Research 31: 373–376. Surgical Pathology 27: 372–384.
Ledesma-­Montes, C., Gorlin, R.J., Shear, M. et al. (2008). Li, T., Browne, R.M., and Matthews, J.B. (1994).
International collaborative study on ghost cell odontogenic Quantification of PCNA+ cells within odontogenic jaw
tumours: calcifying cystic odontogenic tumour, dentinogenic cyst epithelium. Journal of Oral Pathology and Medicine
ghost cell tumour and ghost cell odontogenic carcinoma. 23: 184–189.
Journal of Oral Pathology and Medicine 37: 302–308. Li, T., Browne, R.M., and Matthews, J.B. (1995). Epithelial
Lee, J., Costandi, J., and Mandel, L. (2014). The residual cell proliferation in odontogenic keratocysts:
radicular cyst. New York State Dental Journal 80: 38–40. immunocytochemical study of Ki67 in simple, recurrent
Lee, J.Y., Lee, H.Y., Kim, H.J. et al. (2016). Plunging ranulas and basal cell naevus syndrome (BCNS)-­associated lesions.
revisited: a CT study with emphasis on a defect of the Journal of Oral Pathology and Medicine 24: 221–226.
332 Bibliography

Li, T.J., Browne, R.M., Prime, S.S. et al. (1996). p53 expression Lipani, C.S., Natiella, J.R., and Giteene, G.W. (1982). The
in odontogenic keratocyst epithelium. Journal of Oral hemopoietic defect of the jaws: a report of sixteen cases.
Patholgy and Medicine 25: 245–255. Journal of Oral Pathology 11: 411–416.
Li, T.J., Kitano, M., Yoshida, A. et al. (1997). Myxoglobulosis Lipsett, J., Sparnon, A.L., and Byard, R.W. (1993).
in an extravasation mucocele of the lower lip. Journal of Embryogenesis of enterocystomas-­enteric duplication cysts
Oral Pathology and Medicine 26: 342–344. of the tongue. Oral Surgery, Oral Medicine, and Oral
Li, T.-­J., Kitano, M., Chen, X.-­M. et al. (1998). Pathology 75: 626–630.
Orthokeratinized odontogenic cyst: a clinicopathological Liu, R. and Adler, D.G. (2014). Duplication cysts: diagnosis,
and immunocytochemical study of 15 cases. management, and the role of endoscopic ultrasound.
Histopathology 32: 242–251. Endoscopic Ultrasound 3: 152–160.
Li, Y., Li, L.J., Huang, J. et al. (2008). Central malignant Liu, M.H. and Huang, W.H. (2004). Oral abnormalities in
salivary gland tumors of the jaw: retrospective clinical Taiwanese newborns. Journal of Dentistry for Children 71:
analysis of 22 cases. Journal of Oral and Maxillofacial 118–120.
Surgery 66: 2247–2253. Liu, B., Yu, S.F., and Li, T.J. (2003). Multinucleated giant cells
Li, C.C., Feinerman, D.M., MacCarthy, K.D., and Woo, in various forms of giant cell containing lesions of the jaws
S.B. (2014). Rare mandibular surgical ciliated cysts: report express features of osteoclasts. Journal of Oral Pathology
of two new cases. Journal of Oral and Maxillofacial Surgery and Medicine 32: 367–375.
72: 1736–1743. Lizio, G., Corinaldesi, G., Bianchi, A., and Marchetti,
Liapatas, S., Nakou, M., and Rontogianni, D. (2003). C. (2011). Successful resolution of juvenile paradental cysts
Inflammatory infiltrate of chronic periradicular lesions: an after marsupialization in five consecutive patients.
immunohistochemical study. International Endododontic Australian Dental Journal 56: 427–432.
Journal 36: 464–471. Lo Muzio, L., Staibano, S., Pannone, G. et al. (1999).
Lima, L.P., Meira, H.C., Amaral, T.M.P. et al. (2019). Expression of cell cycle and apoptosis-­related proteins in
Mandibular buccal bifurcation cyst: case report and sporadic odontogenic keratocysts and odontogenic
literature review. Stomatologija 21: 57–61. keratocysts associated with the naevoid basal cell
Lima, L.B., de Freitas Filho, S.A., Barbosa de Paulo, L.F. et al. carcinoma syndrome. Journal of Dental Research 78:
(2020). Simple bone cyst: description of 60 cases seen at a 1345–1353.
Brazilian School of Dentistry and review of international Lo Muzio, L., Santarelli, A., Caltabiano, R. et al. (2005). p63
literature. Medicina Oral, Patología Oral y Cirugía Bucal expression in odontogenic cysts. International Journal of
25: e616–e625. Oral and Maxillofacial Surgery 34: 668–673.
Lin, S.K., Chiang, C.P., Hong, C.Y. et al. (1997). Lo Muzio, L., Mascitti, M., Santarelli, A. et al. (2017). Cystic
Immunolocalization of interstitial collagenase (MMP-­1) lesions of the jaws: a retrospective clinicopathologic study
and tissue inhibitor of metalloproteinases-­1 (TIMP-­1) in of 2030 cases. Oral Surgery, Oral Medicine, Oral Pathology,
radicular cysts. Journal of Oral Pathology and Medicine Oral Radiology 124: 128–138.
26: 458–463. Lombardi, T., Odell, E.W., and Morgan, P.R. (1995). p53
Lin, C.C., Chen, C.H., Lai, S. et al. (2000). Glandular immunohistochemistry of odontogenic keratocysts in
odontogenic cyst: a case report. Kaohsiung Journal of relation to recurrence, basal cell budding and basal-­cell
Medical Sciences 16: 53–58. naevus syndrome. Archives of Oral Biology 40:
Lin, C.C., Chen, C.H., Lin, L.M. et al. (2004). Calcifying 1081–1084.
odontogenic cyst with ameloblastic fibroma: report of Longo, F., Maremonti, P., Mangone, G.M. et al. (2003).
three cases. Oral Surgery, Oral Medicine, Oral Pathology, Midline (dermoid) cysts of the floor of the mouth: report
Oral Radiology and Endodontics 98: 451–460. of 16 cases and review of surgical techniques. Plastic and
Lin, L.M., Huang, G.T., and Rosenberg, P.A. (2007). Reconstructive Surgery 112: 1560–1565.
Proliferation of epithelial cell rests, formation of apical Lopes, M.A., Pereira, C.M., da Cruz Perez, D.E. et al. (2005).
cysts, and regression of apical cysts after periapical wound Benign teratoma of the buccal mucosa in a 9-­year-­old girl:
healing. Journal of Endodontics 33: 908–916. report of case and review of the literature. Oral Surgery,
Lin, H.P., Chen, H.M., Yu, C.H. et al. (2010). Oral Medicine, Oral Pathology, Oral Radiology, and
Clinicopathological study of 252 jaw bone periapical Endodontics 100: 598–602.
lesions from a private pathology laboratory. Journal of the Lopes-­Rocha, R., Dornela Verli, F., Lages Lima, N. et al.
Formosan Medical Association 109: 10–18. (2011). Nasolabial cyst: 18.5 year experience in a pathology
Lin, H.P., Wang, Y.P., Chen, H.M. et al. (2013). A laboratory. Minerva Stomatologica 60: 567–572.
clinicopathological study of 338 dentigerous cysts. Journal Loreto, C., Galanti, C., Leonardi, R. et al. (2013). Possible role
of Oral Pathology and Medicine 42: 462–467. of apoptosis in the pathogenesis and clinical evolution of
Bibliography 333

radicular cyst: an immunohistochemical study. MacDonald, D.S. and Yu, W. (2020). Incidental findings in a
International Endodontic Journal 46: 642–648. consecutive series of digital panoramic radiographs.
Lu, Y., Mock, D., Takata, T., and Jordan, R.C. (1999). Imaging Science in Dentistry 50: 53–64.
Odontogenic ghost cell carcinoma: report of four new MacDonald, D.S., Gu, Y., Zhang, L., and Poh, C. (2013). Can
cases and review of the literature. Journal of Oral clinical and radiological features predict recurrence in
Pathology and Medicine 28: 323–329. solitary keratocystic odontogenic tumors? Oral Surgery,
Lu, D.-­P., Tatemoto, Y., Kimura, T., and Osaki, T. (2002). Oral Medicine, Oral Pathology, Oral Radiology 115:
Expression of cytokeratins (CKs) 8, 13 and 18 and their 263–271.
mRNA in epithelial linings of radicular cysts: implication MacDonald-­Jankowski, D.S. (1994). Mucosal antral cysts
for the same CK profiles as nasal columnar epithelium in observed within a London inner-­city population. Clinical
squamous epithelial lining. Oral Diseases 8: 30–36. Radiology 49: 195–198.
Luna, M.A. and Pfaltz, M. (2009). Cysts of the neck, MacDonald-­Jankowski, D.S. (1995). Traumatic bone cysts in
unknown primary tumour and neck dissection. In: the jaws of a Hong Kong Chinese population. Clinical
Diagnostic Surgical Pathology of the Head and Neck, 2e Radiology 50: 787–791.
(ed. D. Gnepp), 839–881. Elsevier. Macdonald-­Jankowski, D.S. (2010). Orthokeratinized
Lund, V.J. (1985). Odontogenic keratocyst of the maxilla: a odontogenic cyst: a systematic review. Dento Maxillo Facial
case report. British Journal of Oral and Maxillofacial Radiology 39: 455–467.
Surgery 23: 210–215. MacDonald-­Jankowski, D.S. (2011). Keratocystic odontogenic
Luo, H.Y. and Li, T.J. (2009). Odontogenic tumors: a study of tumour: systematic review. Dento Maxillo Facial Radiology
1309 cases in a Chinese population. Oral Oncology 45: 40: 1–23.
706–711. MacDonald-­Jankowski, D.S. and Li, T.K. (2010). Keratocystic
Luo, Y., Shillingford, N., and Koempel, J.A. (2015). odontogenic tumour in a Hong Kong community: the
Histopathologic finding of both gastric and respiratory clinical and radiological features. Dento Maxillo Facial
epithelia in a lingual foregut cyst. Case Reports in Medicine Radiology 39: 167–175.
2015: 278376. Machado de Sousa, S.O., Campos, A.C., Satiago, J.L. et al.
Lustmann, J. and Bodner, L. (1988). Dentigerous cysts (1990). Botryoid odontogenic cyst: report of a case with
associated with supernumerary teeth. International clinical and histogenetic considerations. British Journal of
Journal of Oral and Maxillofacial Surgery 17: 100–102. Oral and Maxillofacial Surgery 28: 275–276.
Lustmann, J. and Copelyn, M. (1981). Oral cysticercosis. MacNeil, S.D. and Moxham, J.P. (2010). Review of floor of
Review of the literature and report of two cases. mouth dysontogenic cysts. Annals of Otology, Rhinology,
International Journal of Oral Surgery 10: 371–375. and Laryngology 119: 165–173.
Lustmann, J. and Shear, M. (1985). Radicular cysts arising Maeda, Y., Osaki, T., Yoneda, K., and Hirota, J. (1987).
from deciduous teeth. Review of the literature and report Clinicopathologic studies on postoperative maxillary cysts.
of 23 cases. International Journal of Oral Surgery 14: International Journal of Oral and Maxillofacial Surgery 16:
153–161. 682–681.
Lustmann, J., Benoliel, R., and Zeltser, R. (1989). Squamous Magliocca, K.R., Edwards, S.P., and Helman, J.I. (2007).
cell carcinoma arising in a thyroglossal duct cyst in the Traumatic bone cyst of the condylar region: report of 2
tongue. Journal of Oral and Maxillofacial Surgery cases. Journal of Oral and Maxillofacial Surgery 65:
47: 81–85. 1247–1250.
Lutz, J., Cimasoni, G., and Held, A.J. (1965). Histochemical Magnusson, B., Göransson, L., Ödesjö, B. et al. (1997).
observations on the epithelial lining of radicular cysts. Glandular odontogenic cyst. Report of seven cases. Dento
Acta Odontologica Scandinavica 9: 90–95. Maxillo Facial Radiology 26: 26–31.
Lv, K., Liu, J., Ye, W. et al. (2019). Multiple superficial Maheswaran, T., Ramesh, V., Oza, N. et al. (2014). Keratin
mucoceles concomitant with oral lichen planus: a case metaplasia in the epithelial lining of odontogenic cysts.
series. Oral Surgery, Oral Medicine, Oral Pathology, Oral Journal of Pharmacy and Bioallied Sciences 6:
Radiology 127: e95–e101. S110–S112.
MacDonald, D.S. (2015). A systematic review of the literature Mahomed, F., Altini, M., Meer, S., and Coleman, H. (2005).
of nevoid basal cell carcinoma syndrome affecting East Cemento-­osseous dysplasia with associated simple bone
Asians and North Europeans. Oral Surgery, Oral Medicine, cysts. Journal of Oral and Maxillofacial Surgery 63:
Oral Pathology, Oral Radiology 120: 396–407. 1549–1554.
MacDonald, D.S. (2016). Lesions of the jaws presenting as Main, D.M.G. (1970). Epithelial jaw cysts: a
radiolucencies on cone-­beam CT. Clinical Radiology 71: clinicopathological reappraisal. British Journal of Oral
972–985. Surgery 8: 114–125.
334 Bibliography

Main, D.M.G. (1985). Epithelial jaw cysts: 10 years of the decompression and later cystectomy: a long-­term follow-­up
WHO classification. Journal of Oral Pathology 41: 1–7. and a histologic study of 23 cases. Oral Surgery, Oral
Main, D.M.G. (1989). Follicular cysts of mandibular third Medicine, Oral Pathology, Oral Radiology, and Endodontics
molar teeth: radiological evaluation of enlargement. Dento 82: 122–131.
Maxillo Facial Radiology 18: 156–159. Markus, A.F. (1978–1979). Bilateral haemorrhagic bone cysts
Makarla, S., Bavle, R.M., Muniswamappa, S. et al. (2015). A of the mandible: a case report. British Journal of Oral
large extragnathic keratocystic odontogenic tumour. Case Surgery 16: 270–273.
Reports in Pathology 2015: 723010. Marom, T., Russo, E., Ben Salem, D., and Roth, Y. (2009).
Makek, M. and Lello, G.E. (1986). Focal osteoporotic bone Nasopharyngeal cysts. International Journal of Pediatric
marrow defects of the jaws. Journal of Oral and Otorhinolaryngology 73: 1063–1070.
Maxillofacial Surgery 44: 268–273. Marques, N.P., Marques, N.C.T., Sakai, V.T., and Hanemann,
Makhija, M. (2015). Cutaneous keratocyst and steatocystoma J.A.C. (2017). Inflammatory follicular cysts associated to
unified as sebaceous duct cyst, a hamartoma resembling necrotic primary teeth. European Archives of Paediatric
the sebaceous duct. American Journal of Dermatopathology Dentistry 18: 279–285.
37: 871–873. Martin, L., Speight, P.M. (2017). Odontogenic Cysts – an
Malcić, A., Jukić, S., Anić, I. et al. (2008). Alterations of FHIT update. Diagnostic Histopathology 23: 260–265.
and P53 genes in keratocystic odontogenic tumor, Martins, C.A., Rivero, E.R., Dufloth, R.M. et al. (2011).
dentigerous and radicular cyst. Journal of Oral Pathology Immunohistochemical detection of factors related to
and Medicine 37: 294–301. cellular proliferation and apoptosis in radicular and
Managutti, A., Managutti, S., Patel, H., and Menat, S. (2016). dentigerous cysts. Journal of Endodontia 37: 36–39.
Orthokeratinized odontogenic cyst (OOC) of condylar Márton, I.J. and Kiss, C. (2014). Overlapping protective and
head: a rare entity. Journal of Maxillofacial and Oral destructive regulatory pathways in apical periodontitis.
Surgery 15: 315–319. Journal of Endodontics 40: 155–163.
Manaktala, N., Boaz, K., Soni, K.M. et al. (2017). Maruyama, M., Onodera, K., and Ooya, K. (2002). A
Odontogenic cyst with verrucous proliferation exhibiting histopathological and lectin-­histochemical study of the
melanin pigmentation. Case Reports in Pathology 2017: lining epithelium in postoperative maxillary cysts. Oral
5079460. Diseases 8: 241–248.
Mann, R.W. (1992). Three-­dimensional representations of Maruyama, S., Yamazaki, M., Abé, T. et al. (2015).
lingual cortical defects (Stafne’s) using silicone Paradental cyst is an inclusion cyst of the junctional/
impressions. Journal of Oral Pathology and Medicine 21: sulcular epithelium of the gingiva: histopathologic and
381–384. immunohistochemical confirmation for its pathogenesis.
Manojlović, S., Grgurević, J., Knežević, G., and Kruŝlin, Oral Surgery, Oral Medicine, Oral Pathology, Oral
B. (1998). Glandular odontogenic cyst: a case report Radiology 120: 227–237.
and clinicopathological analysis of the relationship to Mascard, E., Gomez-­Brouchet, A., and Lambot, K. (2015).
central mucoepidermoid carcinoma. Head and Neck 19: Bone cysts: unicameral and aneurysmal bone cyst.
227–231. Orthopaedics and Traumatology, Surgery and Research 101:
Manor, E., Kachko, L., Puterman, M.B. et al. (2012). Cystic S119–S127.
lesions of the jaws – a clinicopathological study of 322 Mascitti, M., Santarelli, A., Sabatucci, A. et al. (2014).
cases and review of the literature. International Journal of Glandular odontogenic cyst: review of literature and report
Medical Science 9: 20–26. of a new case with cytokeratin-­19 expression. Open
Manor, R., Anavi, Y., Kaplan, I., and Calderon, S. (2003). Dentistry Journal 8: 1–12.
Radiological features of glandular odontogenic cyst. Dento Mass, E., Kaplan, I., and Hirshberg, A. (1995). A clinical and
Maxillo Facial Radiology 32: 73–79. histopathological study of radicular cysts associated with
Manor, Y., Buchner, A., Peleg, M., and Taicher, S. (1999). primary molars. Journal of Oral Patholology and Medicine
Lingual cyst with respiratory epithelium: an entity of 24: 458–461.
debatable histogenesis. Journal of Oral and Maxillofacial Matejka, M., Porteder, H., Kleinert, W. et al. (1985a).
Surgery 57: 124–127. Evidence that PGI2-­generation in human dental cysts is
Manzon, S., Graffeo, M., and Philbert, R. (2009). Median stimulated by leukotrienes C4 and D4. Journal of
palatal cyst: case report and review of literature. Journal of Maxillofacial Surgery 13: 93–96.
Oral and Maxillofacial Surgery 67: 926–930. Matejka, M., Porteder, H., Ulrich, W. et al. (1985b).
Marker, P., Brøndum, N., Clausen, P.P., and Bastian, Prostaglandin synthesis in dental cysts. British Journal of
H.L. (1996). Treatment of large odontogenic keratocysts by Oral and Maxillofacial Surgery 23: 190–194.
Bibliography 335

Matejka, M., Ulrich, W., Porteder, H. et al. (1986). Meghji, S., Harvey, W., and Harris, M. (1989). Interleukin
Immunohistochemical detection of 6-­oxo-­PGF1α and PGE2 I-­like activity in cystic lesions of the jaw. British Journal of
in radicular cysts. Journal of Maxillofacial Surgery 14: Oral and Maxillofacial Surgery 27: 1–11.
108–112. Meghji, S., Henderson, B., Bando, Y., and Harris, M. (1992).
Mathiesen, A. (1973). Preservation and demonstration of Interleukin-­1: the principal osteolytic cytokine produced
mast cells in human apical granulomas and radicular cysts. by keratocysts. Archives of Oral Biology 37: 935–943.
Scandinavian Journal of Dental Research 81: 218–229. Meghji, S., Qureshi, W., Henderson, B., and Harris, M.
Matthews, J.B. and Browne, R.M. (1987). An (1996). The role of endotoxin and cytokines in the
immunocytochemical study of the inflammatory cell pathogenesis of odontogenic cysts. Archives of Oral
infiltrate and epithelial expression of HLA-­DR in Biology 41: 523–531.
odontogenic cysts. Journal of Oral Pathology 16: 112–117. Mello, F.W., Melo, G., Kammer, P.V. et al. (2019). Prevalence
Matthews, J.B. and Browne, R.M. (1990). Diagnostic of odontogenic cysts and tumors associated with impacted
importance of cytokeratin expression in linings of third molars: a systematic review and meta-­analysis.
odontogenic cysts. Journal of Clinical Pathology Journal of Cranio-­Maxillo-­Facial Surgery 47: 996–1002.
43: 84–85. Melrose, R.J., Abrams, A.M., and Mills, B.G. (1976). Florid
Matthews, J.B., Mason, G.I., and Browne, R.M. (1988). osseous dysplasia. A clinical-­pathologic study of thirty-­four
Epithelial cell markers and proliferating cells cases. Oral Surgery, Oral Medicine, and Oral Pathology
in odontogenic jaw cysts. Journal of Pathology 41: 62–82.
156: 283–290. Mendes, R.A., Carvalho, J.F., and van der Waal, I. (2010).
Mazhari, N.J., Kumar, N., and Jain, S. (2001). Cysticercosis of Characterization and management of the keratocystic
the oral mucosa: aspiration cytologic diagnosis. Journal of odontogenic tumor in relation to its histopathological and
Oral Pathology and Medicine 30: 187–189. biological features. Oral Oncology 46: 219–225.
McAdams, H.P., Kirejczyk, W.M., Rosado-­de-­Christenson, Méndez, P., Junquera, L., Gallego, L., and Baladrón, J. (2007).
M.L., and Matsumoto, S. (2000). Bronchogenic cyst: Botryoid odontogenic cyst: clinical and pathological
imaging features with clinical and histopathologic analysis in relation to recurrence. Medicina Oral, Patología
correlation. Radiology 217: 441–446. Oral y Cirugía Bucal 12: E594–E598.
McGurk, M., Eyeson, J., Thomas, B., and Harrison, Menezes, R., Bramante, C.M., da Silva Paiva, K.B. et al.
J.D. (2008). Conservative treatment of oral ranula by (2006). Receptor activator NFkappaB-­ligand and
excision with minimal excision of the sublingual gland: osteoprotegerin protein expression in human periapical
histological support for a traumatic etiology. Journal of cysts and granulomas. Oral Surgery, Oral Medicine, Oral
Oral and Maxillofacial Surgery 66: 2050–2057. Pathology, Oral Radiology, and Endodontics 102: 404–409.
McMullen, P.D., Bridge, J.A., Blair, E.A. et al. (2019). Meningaud, J.-­P., Oprean, N., Pitak-­Arnnop, P., and
Aneurysmal bone cyst of the maxillary sinus with USP6 Bertrand, J.-­C. (2006). Odontogenic cysts: a clinical study
rearrangement: case report of a rare entity and review of of 695 cases. Journal of Oral Science 48: 59–62.
the literature. Head and Neck Pathology 13: 281–285. Meurman, J.H. and Ylipaavalniemi, P. (1982). Scanning
Meara, J.G., Li, K.K., Shah, S.S., and Cunningham, electron microscopy of the cavity surface in odontogenic
M.J. (1996). Odontogenic keratocysts in the pediatric jaw cysts. Proceedings of the Finnish Dental Society
population. Archives of Otolaryngology – Head and Neck 78: 194–200.
Surgery 122: 725–728. Meyer, A.W. (1914). Spolia Anatomica: a unique
Meara, J.G., Pilch, B.Z., Shah, S.S., and Cunningham, supernumerary para-­nasal sinus directly above the
M.J. (2000). Cytokeratin expression in the odontogenic superior incisors. Journal of Anatomy and Physiology
keratocyst. Journal of Oral and Maxillofacial Surgery 48: 118–120.
58: 862–865. Meyer, A.W. (1931). Median anterior maxillary cysts. Journal
Meer, S. (2019). Human immunodeficiency virus and salivary of the American Dental Association 18: 1851–1857.
gland pathology: an update. Oral Surgery, Oral Medicine, Meyer, I. (1955). Dermoid cysts (dermoids) of the floor of the
Oral Pathology, Oral Radiology 128: 52–59. mouth. Oral Surgery, Oral Medicine, and Oral Pathology
Meer, S., Altini, M. (2006) Cysts and pseudocysts of the 8: 1149–1164.
maxillary antrum revisited. South African Dental Journal Minami, M., Kaneda, T., Ozawa, K. et al. (1996). Cystic
61(1): 10–13. PMID: 16562612. lesions of the maxillomandibular region: MR imaging
Meerkotter, V.A. and Shear, M. (1964). Multiple primordial distinction of odontogenic keratocysts and ameloblastomas
cysts associated with bifid rib and ocular defects. Oral from other cysts. American Journal of Roentgenology 166:
Surgery, Oral Medicine, and Oral Pathology 18: 498–503. 943–949.
336 Bibliography

Mínguez-­Martinez, I., Bonet-­Coloma, C., Ata-­Ali-­Mahmud, literature. International Journal of Paediatric


J. et al. (2010). Clinical characteristics, treatment, and Otorhinolaryngology 36: 69–77.
evolution of 89 mucoceles in children. Journal of Oral and Mori, M., Kasai, T., Nakai, M. et al. (2000). Dentinogenic
Maxillofacial Surgery 68: 2468–2471. ghost cell tumor: histologic aspects,
Misch, E., Kashiwazaki, R., Lovell, M.A., and Herrmann, immunohistochemistry, lectin binding profiles, and
B.W. (2020). Pediatric sublingual dermoid and epidermoid biophysical studies. Oral Oncology 36: 134–143.
cysts: a 20-­year institutional review. International Journal Morimoto, Y., Tanaka, T., Nishida, I. et al. (2004).
of Pediatric Otorhinolaryngology 138: 110265. Inflammatory paradental cyst (IPC) in the mandibular
Mitchell, D.A. and Ward-­Booth, R.P. (1984). Atypical premolar region in children. Oral Surgery, Oral Medicine,
presentation of a solitary bone cyst. International Journal Oral Pathology, Oral Radiology and Endodontics 97:
of Oral Surgery 13: 256–259. 286–293.
Mohammed, M., Mahomed, F., and Ngwenya, S. (2019). A Morita, L., Munhoz, L., Nagai, A.Y. et al. (2021). Imaging
survey of pathology specimens associated with impacted features of Stafne bone defects on computed tomography:
teeth over a 21-­year period. Medicina Oral, Patología Oral y an assessment of 40 cases. Imaging Science in Dentistry
Cirugía Bucal 24: e571–e576. 51: 81–86.
Mokhtari, S., Mohsenifar, Z., and Ghorbanpour, M. (2013). Morlino, S., Castori, M., Servadei, F. et al. (2015).
Predictive factors of potential malignant transformation in Oropharyngeal teratoma, oral duplication, cervical
recurrent calcifying cystic odontogenic tumor: review of diplomyelia and anencephaly in a 22-­week fetus: a review
the literature. Case Reports in Patholology 2013: 853095. of the craniofacial teratoma syndrome. Birth Defects
Moleri, A.B., Moreira, L.C., and Carvalho, J.J. (2002). Research. Part A, Clinical and Molecular Teratology
Comparative morphology of 7 new cases of calcifying 103: 554–566.
odontogenic cysts. Journal of Oral and Maxillofacial Mortensen, H., Winther, J.E., and Bien, H. (1970).
Surgery 60: 689–696. Periapical granulomas and cysts. An investigation of
Monteagudo, B., Labandeira, J., Cabanillas, M. et al. (2012). 1600 cases. Scandinavian Journal of Dental Research
Prevalence of milia and palatal and gingival cysts in 78: 241–250.
Spanish newborns. Pediatric Dermatology 29: 301–305. Moskow, B.S. (1966). The pathogenesis of the gingival cyst.
Monteleone, L. and McLellan, M.S. (1964). Epstein’s pearls Periodontics 4: 23–28.
(Bohn’s nodules) of the palate. Journal of Oral Surgery Moskow, B.S. and Bloom, A. (1983). Embryogenesis of the
21: 301–304. gingival cyst. Journal of Clinical Periodontology
Moosavi, Z. and Hosseini, T. (2006). One-­year survey of 10: 119–130.
cutaneous lesions in 1000 consecutive Iranian newborns. Moskow, B.S., Siegel, K., Zegarelli, E.V. et al. (1970). Gingival
Pediatric Dermatology 23: 61–63. and lateral periodontal cysts. Journal of Periodontology
Moreillon, M.C. and Schroeder, H.E. (1982). Numerical 41: 249–260.
frequency of epithelial abnormalities, particularly Moskow, B.S. and Weinstein, M.M. (1975). Further
microkeratocysts, in the developing human oral mucosa. observations on the gingival cyst. Three case reports.
Oral Surgery, Oral Medicine, and Oral Pathology Journal of Periodontology 46: 178–182.
53: 44–55. Mosqueda-­Taylor, A., de la Piedra-­Garza, J.M., and Troncozo-­
Morgan, P.R. and Heyden, G. (1975). Enzyme histochemical Vázquez, F. (1998). Odontogenic keratocyst with chondroid
studies on the formation of hyalin bodies in the epithelium fibrous wall. A case report. International Journal of Oral
of odontogenic cysts. Journal of Oral Pathology 4: 120–127. and Maxillofacial Surgery 27: 58–60.
Morgan, P.R. and Johnson, N.W. (1974). Histological, Mosqueda-­Taylor, A., Irigoyen-­Camacho, M.E., Diaz-­Franco,
histochemical and ultrastructural studies on the nature of M.A., and Torres-­Tejero, M.A. (2002). Odontogenic cysts.
hyalin bodies in odontogenic cysts. Journal of Oral Analysis of 856 cases. Medicina Oral 7: 89–96.
Pathology 3: 127–147. Motamedi, M.H.K. and Talesh, K.T. (2005). Management of
Morgan, T.A., Burton, C.C., and Qian, F. (2005). A extensive dentigerous cysts. British Dental Journal 198:
retrospecive review of treatment of the odontogenic 203–206.
keratocyst. Journal of Oral and Maxillofacial Surgery 63: Moturi, K. and Kaila, V. (2018). Management of non-­
635–639. syndromic multiple impacted teeth with dentigerous cysts:
Morgan, W.E., Jones, J.K., Flaitz, C.M., and Hicks, a case report. Cureus 10: e3323.
M.J. (1996). Congenital heterotopic gastrointestinal cyst of Mourshed, F. (1964a). A roentgenographic study of
the oral cavity in a neonate: case report and review of dentigerous cysts. I. Incidence in a population sample.
Bibliography 337

Oral Surgery, Oral Medicine, and Oral Pathology Nair, P.N.R. (2006). On the causes of persistent apical
18: 47–53. periodontitis: a review. International Endodontic Journal
Mourshed, F. (1964b). A roentgenographic study of 39: 249–281.
dentigerous cysts. III. Analysis of 180 cases. Oral Surgery, Nair, P.N.R., Pajarola, G., and Luder, H.U. (2002). Ciliated
Oral Medicine, and Oral Pathology 18: 466–473. epithelium-­lined radicular cysts. Oral Surgery, Oral
Müller, S. (2021). Developmental odontogenic lesions Medicine, Oral Pathology, Oral Radiology, and Endodontics
associated with the crown of an impacted tooth: a guide to 94: 485–493.
the distinct histologic features required for classification. Nair, P.N.R., Pajarola, G., and Schroeder, H.E. (1996). Types
Head and Neck Pathology 15: 107–112. and incidence of human periapical lesions obtained with
Müller, S., Aiken, A., Magliocca, K., and Chen, A.Y. (2015). extracted teeth. Oral Surgery, Oral Medicine, Oral
Second Branchial Cleft Cyst. Head and Neck Pathology 9: Pathology, Oral Radiology, and Endodontics 81: 93–102.
379–383. Nair, P.N. and Schroeder, H.E. (1986). Duct-­associated
Munteanu, M.C., Petrescu, G.S., Sandu, A.M. et al. (2019). lymphoid tissue (DALT) of minor salivary glands and
Sialocyst of the parotid gland in a child. Current Health mucosal immunity. Immunology 57: 171–180.
Sciences Journal 45: 419–424. Nair, P.N.R. and Schroeder, H.E. (1987). Duct-­associated
Musa, N.J., Suresh, L., Hatton, M. et al. (2005). Multiple lymphoid tissue (DALT) of minor salivary glands in the
suppurative cystic lesions of the lips and buccal mucosa: a monkey, Cercopithecus aethiops. Archives of Oral Biology
case of suppurative stomatitis glandularis. Oral Surgery, 32: 311–313.
Oral Medicine, Oral Pathology, Oral Radiology, and Nair, P.N.R., Sundqvist, G., and Sjögren, U. (2008).
Endodontics 99: 175–179. Experimental evidence supports the abscess theory of
Myall, R.W.T., Eastep, P.B., and Silver, J.G. (1974). Mucous development of radicular cysts. Oral Surgery, Oral
retention cysts of the maxillary antrum. Journal of the Medicine, Oral Pathology, Oral Radiology, and Endodontics
American Dental Association 89: 1338–1342. 106: 294–303.
Myoung, H., Hong, S.P., Hong, S.D. et al. (2001). Odontogenic Nakamura, T., Ishida, J., Nakano, Y. et al. (1995). A study of
keratocyst: review of 256 cases for recurrence and cysts in the oral region. Cysts of the jaw. Journal of Nihon
clinicopathologic parameters. Oral Surgery, Oral Medicine, University School of Dentistry 37: 33–40.
Oral Pathology, Oral Radiology, and Endodontics 19: 328–333. Namgyel, T., Chaiyasamut, T., Suphangul, S., and
Nagao, T., Serizawa, H., Iwaya, K. et al. (2002). Wongsirichat, N. (2020). Histopathological features
Keratocystoma of the parotidgland: a report of two cases assessment of suspected pathologic pericoronal tissue of
of an unusual pathologic entity. Modern Pathology 15: embedded teeth. Journal of Oral and Maxillofacial Surgery,
1005–1010. Medicine, and Pathology 32: 316–322.
Nagasaki, A., Ogawa, I., Sato, Y. et al. (2018). Central Nanci, A. (ed.) (2017). Ten Cates’s Oral Histology: Development
mucoepidermoid carcinoma arising from glandular Structure and Function, 9e. St Louis, MO: Elsevier.
odontogenic cyst confirmed by analysis of MAML2 Nandakumar, H. and Sbankaramba, K.B. (1989). Hydatid cyst
rearrangement: a case report. Pathology International of the mandible: a case report. Journal of Oral and
68: 31–35. Maxillofacial Surgery 47: 759–761.
Nagata, T., Nomura, J., Matsumura, Y. et al. (2008). Radicular Naruse, T., Yamashita, K., Yanamoto, S. et al. (2017).
cyst in a deciduous tooth: a case report and review of Histopathological and immunohistochemical study in
literature. Journal of Dentistry for Children 75: 80–84. keratocystic odontogenic tumors: predictive factors of
Nair, P.N.R. (1987). Light and electron microscopic studies of recurrence. Oncology Letters 13: 3487–3493.
root canal flora and periapical lesions. Journal of Nastri, A.L. and Hookey, S.R. (1994). Respiratory epithelium
Endodontics 13: 29–39. in a mandibular cyst after grafting of autogenous bone.
Nair, P.N.R. (1998). Review. New perspectives on radicular International Journal of Oral and Maxillofacial Surgery 23:
cysts: do they heal? International Endodontic Journal 31: 372–373.
155–160. Nathan, H. and Luchanski, E. (1985). Sublingual gland
Nair, P.N.R. (2003). Non-­microbial etiology: periapical cysts herniation through the mylohyoid muscle. Oral Surgery,
sustain post-­treatment apical periodontitis. Endodontic Oral Medicine, and Oral Pathology 59: 21–23.
Topics 6: 96–113. Natkin, E., Oswald, R.J., and Carnes, L.I. (1984). The
Nair, P.N.R. (2004). Pathogenesis of apical periodontitis and relationship of lesion size to diagnosis, incidence, and
the causes of endodontic failures. Critical Reviews in Oral treatment of periapical cysts and granulomas. Oral Surgery,
Biology and Medicine 15: 348–381. Oral Medicine, and Oral Pathology 57: 82–94.
338 Bibliography

Natvig, K. and Larsen, T.E. (1978). Mucocele of the paranasal born in and around Meerut city. International Journal of
sinuses. A retrospective clinical and histological study. Clinical Pediatric Dentistry 13: 48–52.
Journal of Laryngology and Otology 92: 1075–1082. Nishioka, M., Pittella, F., Hamagaki, M. et al. (2005).
Nel, C., Robinson, L., Roza, A.L.O.C. et al. (2021). Calcifying Prevalence of postoperative maxillary cyst significantly
odontogenic cysts: a 20-­year retrospective clinical and higher in Japan. Oral Medicine and Pathology 10: 9–13.
radiological review. Dento Maxillo Facial Radiology 50: Noffke, C. and Raubenheimer, E.J. (2002). The glandular
20200586. odontogenic cyst: clinical and radiological features; review
Nel, C., Robinson, L., and van Heerden, W.F.P. (2020). of the literature and report of nine cases. Dento Maxillo
Ghost cell odontogenic carcinoma arising in the Facial Radiology 31: 333–338.
background of a calcifying odontogenic cyst. Oral Nonaka, C.F., Henriques, A.C., de Matos, F.R. et al. (2011).
Radiology 37: 537–542. Nonodontogenic cysts of the oral and maxillofacial region:
Nel, S., Hendrik, H.D., Boy, S.C., and Raubenheimer, demographic profile in a Brazilian population over a
E.J. (2015). Recent perspectives vis-­à-­vis the biological basis 40-­year period. European Archives of Oto-­Rhino-­
of tooth eruption. South African Dental Journal 70: Laryngology 268: 917–922.
238–241. Nortjé, C.J. and Wood, R.E. (1988). The radiologic features of
Nelson, B.L. and Speight, P.M. (2022). Surgical ciliated cyst. the nasopalatine duct cyst. An analysis of 46 cases. Dento
In: WHO Classification of Tumours Editorial Board. Head Maxillo Facial Radiology 17: 129–132.
and Neck Tumours, Section 7.1.0.11 (WHO Classification Nxumalo, T.N. and Shear, M. (1992). Gingival cyst in adults.
of Tumours Series), 5e, vol. 9. Lyon: International Agency Journal of Oral Pathology and Medicine 21: 309–313.
for Research on Cancer https://publications.iarc.fr/ Obiechina, A.E., Arotiba, J.T., and Ogunbiyi, J.O. (1999).
Book-­And-­Report-­Series/ Coexisting congenital sublingual dermoid and
Who-­Classification-­Of-­Tumours. bronchogenic cyst. British Journal of Oral and
Nelson, B.L. and White, S.J. (2022). Dentigerous cyst. In: Maxillofacial Surgery 37: 58–60.
WHO Classification of Tumours Editorial Board. Head and Ocak, A., Duman, S.B., Bayrakdar, I.S., and Cakur, B. (2017).
Neck Tumours, Section 7.1.0.3 (WHO Classification of Nasolabial cyst: a case report with ultrasonography and
Tumours Series), 5e, vol. 9. Lyon: International Agency magnetic resonance imaging findings. Case Reports in
for Research on Cancer https://publications.iarc.fr/ Dentistry 17: 4687409.
Book-­And-­Report-­Series/Who-­Classification-­Of-­Tumours. Ochiai, A. and Nakayama, M. (2015). Congenital mucocele of
Neville, B.W., Damm, D.D., Allen, C.M., and Ch, A.C. (2016). the gland of Blandin-­Nuhn. Oxford Medical Case Reports
Oral and Maxillofacial Pathology, 4e. St Louis, MO: 2015: 374–375.
Elsevier. Ochsenius, G., Escobar, E., Godoy, L., and Peñafiel,
Neville, B.W., Damm, D.D., and Brock, T. (1997). C. (2007). Odontogenic cysts: analysis of 2,944 cases in
Odontogenic keratocysts of the midline maxillary region. Chile. Medicina Oral, Patología Oral y Cirugía Bucal 12:
Journal of Oral and Maxillofacial Surgery 55: 340–344. E85–E91.
Niederquell, B.M., Brennan, P.A., Dau, M. et al. (2016). Öçok, O., Yaman, Z., Gunhan, O. et al. (2005). Botryoid
Bilateral postoperative cyst after maxillary sinus surgery: odontogenic cyst: report of a case with extensive epithelial
report of a case and systematic review of the literature. proliferation. International Journal of Oral and
Case Reports in Dentistry 2016: 6263248. Maxillofacial Surgery 34: 693–695.
Nigam, S., Singh, T., Mishra, A., and Chaturvedi, K.U. (2001). Oda, M., Miyamoto, I., Nishida, I. et al. (2016). A spatial
Oral cysticercosis: report of six cases. Head and Neck 23: association between odontomas and the gubernaculum
497–499. tracts. Oral Surgery, Oral Medicine, Oral Pathology, Oral
Nilsen, R., Johannessen, A.C., Skaug, N., and Matre, Radiology 121: 91–95.
R. (1984). In situ characterization of mononuclear cells in Oda, M., Nishida, I., Miyamoto, I. et al. (2018). Significance
human dental periapical inflammatory lesions using and usefulness of imaging characteristics of gubernaculum
monoclonal antibodies. Oral Surgery, Oral Medicine, and tracts for the diagnosis of odontogenic tumors or cysts.
Oral Pathology 58: 160–165. PLoS One 13: e0199285.
Ninomiya, T., Kubota, Y., Koji, T., and Shirasuna, K. (2002). Oehlers, F.A.C. (1970). Periapical lesions and residual dental
Marsupialization inhibits interleukin-­1α expression and cysts. British Journal of Oral Surgery 8: 103–113.
epithelial cell proliferation in odontogenic keratocysts. Ogden, G.R., Chisholm, D.M., Kiddie, R.A., and Lane,
Journal of Oral Pathology and Medicine 31: 526–533. D.P. (1992). p53 protein in odontogenic cysts: increased
Niranjan, M.M., Srivastava, N., Rana, V., and Chandna, expression in some odontogenic keratocysts. Journal of
P. (2020). Assessment of intraoral findings of neonates, Clinical Pathology 45: 1007–1010.
Bibliography 339

Oh, K.Y., Kim, J.E., Cho, S.D., Yoon, H.J., Lee, J.I., Hong, Olsen, D.B., Mostofi, R.S., and Lagrotteria, L.B. (1988).
S.D. (2022). Orthokeratinized odontogenic cyst: A large Steatocystoma simplex in the oral cavity: a previously
series and comprehensive literature review with emphasis undescribed condition. Oral Surgery, Oral Medicine, and
on synchronous multiple occurrence and neoplastic Oral Pathology 66: 605–607.
transformation. Oral Surgery, Oral Medicine, Oral Omura, S., Kawabe, R., Kobayashi, S., and Mizuki, N. (1997).
Pathology, and Oral Radiology 133: e72–e82. Odontogenic keratocyst appearing as a soap-­bubble or
O’Hara, A.J., Collins, T., and Howell, J.M. (2002). Gingival honeycomb radiolucency: report of a case. Journal of Oral
eruption cysts induced by cyclosporine administration to and Maxillofacial Surgery 55: 185–189.
neonatal dogs. Journal of Clinical Periodontology 29: Ordones, A.B., Neri, L., Oliveira, I.H. et al. (2013). Giant
507–513. nasolabial cyst treated using neumann incision: case
Ohki, K., Kumamoto, H., Ichinohasama, R. et al. (2004). PTC report. International Archives of Otorhinolaryngology 17:
gene mutations and expression of SHH, PTC, SMO, and 421–423.
GLI-­1 in odontogenic keratocysts. International Journal of Osny, F.J., Azevedo, L.R., Sant’Ana, E., and Lara, V.S. (2004).
Oral and Maxillofacial Surgery 33: 584–592. Glandular odontogenic cyst: case report and review of the
Oikarinen, V.J. and Julku, M. (1974). An literature. Quintessence International 35: 385–389.
orthopantomographic study of developmental mandibular Ostrofsky, M.K. and Baker, M.A.A. (1975). Oral cysticercosis.
bone defects (Stafne’s idiopathic bone cavities). Journal of the Dental Association of South Africa 30:
International Journal of Oral Surgery 3: 71–76. 535–537.
Oka, S., Kubota, Y., Yamashiro, T. et al. (2005). Effects of Packota, G.V., Hall, J.M., Lanigan, D.T., and Cohen,
positive pressure in odontogenic keratocysts. Journal of M.A. (1990). Paradental cysts on mandibular first molars
Dental Research 84: 913–918. in children: report of five cases. Dento Maxillo Facial
Okamoto, E., Kikuchi, K., Miyazaki, Y. et al. (2010). Radiology 19: 126–132.
Significance of podoplanin expression in keratocystic Padayachee, A. and Van Wyk, C.W. (1987). Two cystic lesions
odontogenic tumor. Journal of Oral Pathology and with features of both the botryoid odontogenic cyst and the
Medicine 39: 110–114. central mucoepidermoid tumour: sialo-­odontogenic cyst?
Okur, M.H., Arslan, M.S., Arslan, S. et al. (2014). Journal of Oral Pathology 16: 499–504.
Gastrointestinal tract duplications in children. European Pai, R.K., Erickson, J., Pourmand, N., and Kong, C.S. (2009).
Review for Medical and Pharmacological Sciences 18: p16(INK4A) immunohistochemical staining may be
1507–1512. helpful in distinguishing branchial cleft cysts from cystic
Olech, E., Sieber, H., and Weinmann, J.P. (1951). Traumatic squamous cell carcinomas originating in the oropharynx.
mandibular bone cysts. Oral Surgery, Oral Medicine, and Cancer 117: 108–119.
Oral Pathology 4: 1160–1172. Pan, S., Dong, Q., Sun, L.S., and Li, T.J. (2010). Mechanisms
Oliveira, A.M. and Chou, M.M. (2014). USP6-­induced of inactivation of PTCH1 gene in nevoid basal cell
neoplasms: the biologic spectrum of aneurysmal bone cyst carcinoma syndrome: modification of the two-­hit
and nodular fasciitis. Human Pathology 45: 1–11. hypothesis. Clinical Cancer Research 16: 442–450.
Oliveira, A.M., Perez-­Atayde, A.R., Dal Cin, P. et al. (2005). Pan, S. and Li, T.J. (2009). PTCH1 mutations in odontogenic
Aneurysmal bone cyst variant translocations upregulate keratocysts: are they related to epithelial cell proliferation?
USP6 transcription by promoter swapping with the ZNF9, Oral Oncology 45: 861–865.
COL1A1, TRAP150, and OMD genes. Oncogene 24: Pang, C.E., Lee, T.S., Pang, K.P., and Pang, Y.T. (2005).
3419–3426. Thoracic ranula: an extremely rare case. Journal of
Oliveira, A.M., Perez-­Atayde, A.R., Inwards, C.Y. et al. (2004). Laryngology and Otology 119: 233–234.
USP6 and CDH11 oncogenes identify the neoplastic cell in Pant, B., Carvalho, K., Dhupar, A., and Spadigam, A. (2019).
primary aneurysmal bone cysts and are absent in so-­called Bilateral nonsyndromic dentigerous cyst in a 10-­year-­old
secondary aneurysmal bone cysts. American Journal of child: a case report and literature review. International
Pathology 165: 1773–1780. Journal of Applied and Basic Medical Research 9: 58–61.
Oliveira, D.T., Consolaro, A., and Freitas, F.J. (1993). Papadogeorgakis, N., Petsinis, V., Parara, E. et al. (2009).
Histopathological spectrum of 112 cases of mucocele. Branchial cleft cysts in adults. Diagnostic procedures and
Brazilian Dental Journal 4: 29–36. treatment in a series of 18 cases. Oral and Maxillofacial
Oliveira, D.T., de Santis, L.P., Assao, A. et al. (2014). The Surgery 13: 79–85.
relationship between ezrin and podoplanin expressions in Paremala, K., Radhika, M.B., and Thambiah, L.J. (2011).
keratocystic odontogenic tumors. BMC Oral Health Myxoglobulosis of lower lip: report of two cases. Journal of
14: 150. Oral Maxillofacial Patholology 15: 232–235.
340 Bibliography

Parmar, R.M., Brannon, R.B., and Fowler, C.B. (2011). Perl, P., Perl, T., and Goldberg, B. (1972). Hydatid cyst in the
Squamous odontogenic tumor-­like proliferations in tongue. Oral Surgery, Oral Medicine, and Oral Pathology 33:
radicular cysts: a clinicopathologic study of forty-­two 579–581.
cases. Journal of Endodontia 37: 623–626. Perrini, N. and Fonzi, L. (1985). Mast cells in human
Partridge, M. and Towers, J.F. (1987). The primordial cyst periapical lesions: ultrastructural aspects and their possible
(odontogenic keratocyst): its tumour-­like characteristics physiopathological implications. Journal of Endodontics
and behaviour. British Journal of Oral and Maxillofacial 11: 197–202.
Surgery 25: 271–279. Persson, G. (1973). Remarkable recurrence of a keratocyst in
Patidar, M., Shetty, P., Patidar, N. et al. (2015). Biochemical a bone graft. International Journal of Oral Surgery
and cytological comparison of keratocystic odontogenic 2: 69–76.
tumours to nonkeratinising odontogenic cysts fluid. Persson, G. (1985). An atypical solitary bone cyst. Journal of
Journal of Clinical and Diagnostic Research 9: ZC34–ZC38. Oral and Maxillofacial Surgery 43: 905–907.
Patil, S., Halgatti, V., Khandelwal, S. et al. (2014). Prevalence Peters, S.M., Park, M., Perrino, M.A., and Cohen, M.D. (2018).
of cysts and tumors around the retained and unerupted Lingual cyst with respiratory epithelium: report of 2 cases
third molars in the Indian population. Journal of Oral and review of the literature. Oral Surgery, Oral Medicine,
Biology and Craniofacial Research 4: 82–87. Oral Pathology, Oral Radiology 126: e279–e284.
Patron, M., Colmenero, C., and Larrauri, J. (1991). Glandular Phelan, J.A., Kritchman, D., Fusco-­Ramer, M. et al. (1988).
odontogenic cyst: clinicopathological analysis of three Recurrent botryoid odontogenic cyst (lateral periodontal
cases. Oral Surgery, Oral Medicine, and Oral Pathology cyst). Oral Surgery, Oral Medicine, and Oral Pathology 66:
72: 71–74. 345–348.
Pavelić, B., Levanat, S., Crnić, I. et al. (2001). PTCH gene Philippou, S., Rühl, G.H., and Mandelartz, E. (1990).
altered in dentigerous cysts. Journal of Oral Pathology and Scanning electron microscopic studies and X-­ray
Medicine 30: 569–576. microanalysis of hyaline bodies in odontogenic cysts.
Pe, M.B., Sano, K., Kitamura, A., and Inokuchi, T. (1990). Journal of Oral Pathology and Medicine 19: 447–452.
Computed tomography in the evaluation of postoperative Philipsen, H.P. (1956). Om keratocyster (kolesteatom) i
maxillary cysts. Journal of Oral and Maxillofacial Surgery kæberne. Tandlaegebladet 60: 963–981.
48: 679–684. Philipsen, H.P. (2005). Keratocystic odontogenic tumor. In:
Peacock, M.E., Krishna, R., Gustin, J.W. et al. (2015). Head and Neck Tumours, WHO Classification of Tumours
Retrospective study on idiopathic bone cavity and its (eds. L. Barnes, J.W. Eveson, P. Reichart and D. Sidransky),
association with cementoosseous dysplasia. Oral Surgery, 306–307. Lyon: IARC Press.
Oral Medicine, Oral Pathology, Oral Radiology 119: Philipsen, H.P., Khongkhunthiang, P., and Reichart,
e246–e251. P.A. (2016). The adenomatoid odontogenic tumour: an
Peacock, Z.S., Cox, D., and Schmidt, B.L. (2010). Involvement update of selected issues. Journal of Oral Pathology and
of PTCH1 mutations in the calcifying epithelial Medicine 45: 394–398.
odontogenic tumor. Oral Oncology 46: 387–392. Philipsen, H.P., Reichart, P.A., Ogawa, I. et al. (2004). The
Pereira, D.L., Pires, F.R., Lopes, M.A. et al. (2016). Clinical, inflammatory paradental cyst: a critical review of 342 cases
demographic, and radiographic analysis of 82 patients from a literature survey, including 17 new cases from the
affected by florid osseous dysplasia: an international author’s files. Journal of Oral Pathology and Medicine 33:
collaborative study. Oral Surgery, Oral Medicine, Oral 147–155.
Pathology, Oral Radiology 122: 250–257. Philipsen, H.P., Samman, N., Ormiston, I.W. et al. (1992).
Pereira, F.A.C., Vidal, M.T.A., Campos, P.S.F. et al. (2012). Variants of the adenomatoid odontogenic tumor with a
Orthokeratinized odontogenic cyst: a report of two cases in note on tumor origin. Journal of Oral Pathology and
the mandible. Revista Odonto Ciência 27: 174–178. Medicine 21: 348–352.
Perez-­Aguirre, B., Soto-­Barreras, U., Loyola-­Rodriguez, Philipsen, H.P., Takata, T., Reichart, P.A. et al. (2002). Lingual
J.P. et al. (2018). Oral findings and its association with and buccal mandibular bone depressions: a review based
prenatal and perinatal factors in newborns. Korean Journal on 583 cases from a world-­wide literature survey, including
of Pediatrics 61: 279–284. 69 new cases from Japan. Dento Maxillo Facial Radiology
Pérez-­Sayáns, M., Suárez Peñaranda, J.M., Quintanilla, 31: 281–290.
J.A.S. et al. (2020). Clinicopathological features of Piatelli, A., Fiorini, M., Iezzi, G., and Rubini, C. (2003).
214 maxillary sinus pathologies. A ten-­year single-­centre Calretinin expression in odontogenic cysts. Journal of
retrospective clinical study. Head and Face Medicine 13: 24. Endodontics 29: 394–396.
Bibliography 341

Piatelli, A., Fioroni, M., Santinelli, A., and Rubini, C. (1998). outcomes in 44 sites. Oral Surgery, Oral Medicine, Oral
Expression of proliferating cell nuclear antigen in Pathology, Oral Radiology, and Endodontics 83: 215–221.
ameloblastomas and odontogenic cysts. Oral Oncology 34: Poomsawat, S., Punyasingh, J., and Weerapradist, W. (2006).
408–412. Expression of basement membrane components in
Piattelli, A., Rubini, C., Fioroni, M. et al. (2004). Expression odontogenic cysts. Oral Diseases 12: 290–296.
of transforming growth factor-­beta 1 (TGF-­beta 1) in Poyton, H.G. and Morgan, G.A. (1965). The simple bone cyst.
odontogenic cysts. International Endodontic Journal 37: 7–11. Oral Surgery, Oral Medicine, and Oral Pathology 20:
Precheur, H.V. and Krolls, S.O. (2009). An unusual 188–197.
presentation of an odontogenic keratocyst in the buccal Prado-­Ribeiro, A.C., Santos-­Silva, A.R., Faria, K.M. et al.
space: case report. Journal of Oral and Maxillofacial (2018). Radiation-­related superficial oral mucoceles: an
Surgery 67: 2513–2515. under-­recognized acute toxicity in head and neck cancer
Pimpalkar, R.D., Barpande, S.R., Bhavthankar, J.D., and patients. Medicina Oral, Patología Oral y Cirugía Bucal 23:
Mandale, M.S. (2014). Bilateral orthokeratinized e518–e523.
odontogenic cyst: a rare case report and review. Journal of Prætorius, F., Hjørting-­Hansen, E., Gorlin, R.J., and Vickers,
Oral and Maxillofacial Pathology 18: 262–266. R.A. (1981). Calcifying odontogenic cyst. Range, variations
Pindborg, J.J. and Hansen, J. (1963). Studies on odontogenic and neoplastic potential. Acta Odontologica Scandinavica
cyst epithelium. 2. Clinical and roentgenologic aspects of 39: 227–240.
odontogenic keratocysts. Acta Pathologica et Microbiologica Prætorius, F. and Ledesma-­Montes, C. (2005). Calcifying
Scandinavica. Section A 58: 283–294. cystic odontogenic tumour. In: Pathology and Genetics.
Pindborg, J.J. and Kramer, I.R.H. (1971). Histological Typing Head and Neck Tumours. World Health Organization
of Odontogenic Tumours, Jaw Cysts, and Allied Lesions, 1e. Classification of Tumours (eds. L. Barnes, J.W. Eveson,
Geneva: World Health Organization. P. Reichart and D. Sidransky), 313. Lyon: IARC Press.
Pindborg, J.J., Philipsen, H.P., and Henriksen, J. (1962). Premalatha, B.R., Roopa, R.S., Jude, J., and Prasad, K. (2012).
Studies on odontogenic cyst epithelium keratinization in Bilateral orthokeratinized odontogenic cyst: an unusual
odontogenic cysts. In: Fundamentals of Keratinization (eds. presentation and review. International Journal of
E.O. Butcher and R.F. Sognnaes), 151. Washington, DC: Contemporary Dentistry 3: 73–76.
American Association for the Advancement of Science. Preston, R.D. and Narayana, N. (2005). Peripheral
Pinswasdi, P. and Charoensiri, D.J. (1997). Cysticercosis in odontogenic keratocyst. Journal of Periodontology 76:
labial tissue. Case report. Australian Dental Journal 42: 2312–2315.
319–321. Probst, F.A., Probst, M., Maistreli, I.Z. et al. (2014). Imaging
Pires, F.R., Chen, S.-­Y., Perez, E.D. et al. (2004). Cytokeratin characteristics of a Stafne bone cavity–panoramic
expression in central mucoepidermoid carcinoma and radiography, computed tomography and magnetic resonance
glanular odontogenic cyst. Oral Oncology 40: 545–551. imaging. Oral and Maxillofacial Surgery 18: 351–353.
Pogrel, M.A. (2003a). The history of the odontogenic Pryor, S.G., Lewis, J.E., Weaver, A.L., and Orvidas,
keratocyst. In: The Odontogenic Keratocyst, Oral and L.J. (2005). Pediatric dermoid cysts of the head and
Maxillofacial Surgery Clinics of North America, vol. 15 (eds. neck. Otolaryngology and Head and Neck Surgery 132:
M.A. Pogrel and B.L. Schmidt), 311–315. Philadelphia, PA: 938–942.
W.B. Saunders. Pullon, P.A., Shafer, W.G., Elzay, R.P. et al. (1975). Squamous
Pogrel, M.A. (2003b). Decompression and marsupialization odontogenic tumor. Oral Surgery, Oral Medicine, and Oral
as a treatment for the odontogenic keratocyst. In: The Pathology 40: 616–630.
Odontogenic Keratocyst, Oral and Maxillofacial Surgery Pulver, W.H., Taubman, M.A., and Smith, D.J. (1978).
Clinics of North America, vol. 15 (eds. M.A. Pogrel and Immune components in human dental periapical lesions.
B.L. Schmidt), 311–315. Philadelphia, PA: W.B. Saunders; Archives of Oral Biology 23: 435–443.
415–427. Pupić-­Bakrač, J., Skitarelić, N., Pupić-­Bakrač, A. et al. (2021).
Pogrel, M.A. and Jordan, R.C. (2004). Marsupialization as a Branchial cleft anomalies: hybrid ‘branchial inclusion’
definitive treatment for the odontogenic keratocyst. theory. European Archives of Oto-­Rhino-­Laryngology 278:
Journal of Oral and Maxillofacial Surgery 62: 651–656. 2593–2601.
Pollard, Z.F., Harley, R.D., and Calhoun, J. (1976). Dermoid Qin, X.-­N., Li, J.-­R., Chen, X.-­M., and Long, X. (2005). The
cysts in children. Pediatrics 57: 379–382. glandular odontogenic cyst: clinicopathologic features and
Pompura, J.R., Sándor, G.K., and Stoneman, D.W. (1997). The treatment of 14 cases. Journal of Oral and Maxillofacial
buccal bifurcation cyst: a prospective study of treatment Surgery 63: 694–699.
342 Bibliography

Qu, J., Yu, F., Hong, Y. et al. (2015). Underestimated radiographic study of 32 cases. British Journal of Oral and
PTCH1 mutation rate in sporadic keratocystic odontogenic Maxillofacial Surgery 29: 54–57.
tumors. Oral Oncology 51: 40–45. Ratner, E.J., Person, P., Kleinman, D.J. et al. (1979). Jawbone
Quesada-­Gómez, C., Valmaseda-­Castellón, E., Berini-­Aytés, cavities and trigeminal and atypical facial neuralgias. Oral
L., and Gay-­Escoda, C. (2006). Stafne bone cavity: a Surgery, Oral Medicine, and Oral Pathology 48: 3–20.
retrospective study of 11 cases. Medicina Oral, Patología Raubenheimer, E.J., Noffke, C.E., and Boy, S.C. (2016).
Oral y Cirugía Bucal 11: E277–E280. Osseous dysplasia with gross jaw expansion: a review of
Rachanis, C.C., Altini, M., and Shear, M. (1979). A 18 lesions. Head and Neck Pathology 10: 437–443.
clinicopathological comparison of primordial cyst Raubenheimer, E.J., van Heerden, W.F.P., and Wright,
(keratocysts) in two different age groups. Journal of Dental J.M. (2017). Simple bone cyst. In: WHO Classification of
Research 58D: 2324. Head and Neck Tumours, 4e (eds. A.K. El-­Naggar, C. JKC,
Radden, B.G. and Reade, P.C. (1973). Odontogenic cysts. A J.R. Grandis, et al.), 259–260. Lyon: IARC Press.
review and clinicopathological study of 368 odontogenic Re Cecconi, D., Achilli, A., Tarozzi, M. et al. (2010).
cysts. Australian Dental Journal 18: 218–225. Mucoceles of the oral cavity: a large case series
Ragsdale, B.D., Laurent, J.L., Janette, A.J., and Epker, (1994–2008) and a literature review. Medicina Oral,
B.N. (2009). Respiratory implantation cyst of the mandible Patologia Oral, Cirugia Bucal 15: e551–e556.
following orthognathic surgery. Journal of Oral Reddy, R., Islam, M.N., Bhattacharyya, I. et al. (2019). The
Maxillofacial Pathology 13: 30–34. reliability of MAML2 gene rearrangement in
Ramachandra, P., Maligi, P., and Raghuveer, H. (2011). A discriminating between histologically similar glandular
cumulative analysis of odontogenic cysts from major odontogenic cysts and intraosseous mucoepidermoid
dental institutions of Bangalore city: a study of 252 carcinomas. Oral Surgery, Oral Medicine, Oral Pathology,
cases. Journal of Oral and Maxillofacial Pathology Oral Radiology 127: e136–e147.
15: 1–5. Redman, R.S., Whitestone, B.W., Winne, C.E. et al. (1990).
Ramer, M., Montazem, A., Lane, S.L., and Lumerman, Botryoid odontogenic cyst. Report of a case with histologic
H. (1997). Glandular odontogenic cyst. Report of a case evidence of multicentric origin. International Journal of
and review of the literature. Oral Surgery, Oral Medicine, Oral and Maxillofacial Surgery 19: 144–146.
Oral Pathology, Oral Radiology, and Endodontics Reeve, C.M. and Levy, B.P. (1968). Gingival cysts: a review of
84: 54–57. the literature and a report of four cases. Periodontks 6:
Ramer, M. and Valauri, D. (2005). Multicystic lateral 115–117.
periodontal cyst and botryoid odontogenic cyst. Regezi, J.A., Sciubba, J.J., and Jordan, R.C.K. (eds.) (2017).
Multifactorial analysis of previously unreported series and Oral Pathology: Clinical Pathologic Correlations, 7e. St
review of the literature. New York State Dental Journal Louis, MO: Elsevier.
71: 47–51. Rehefeldt-­Erne, S., Nägeli, M.C., Winterton, N. et al. (2016).
Ramos, L.M., Vargas, P.A., Coletta, R.D. et al. (2012). Bilateral Nevoid basal cell carcinoma syndrome: report from the
buccal bifurcation cyst: case report and literature review. Zurich nevoid basal cell carcinoma syndrome cohort.
Head and Neck Pathology 6: 455–459. Dermatology 232: 285–292.
Rangaswamy, S., Singh, M., and Yumnum, R. (2018). True Reichart, P.A. and Philipsen, H.P. (2004). Odontogenic Tumors
median palatal cyst; a rare case report. Journal of Oral and Allied Lesions. London: Quintessence.
Maxillofacial Pathology 22: 286. Reichart, P.A., Philipsen, H.P., and Sciubba, J.J. (2006). The
Rangiani, A. and Motahhary, P. (2009). Evaluation of bax and new classification of head and neck tumours (WHO)—­any
bcl-­2 expression in odontogenic keratocysts and changes? Oral Oncology 42: 757–758.
orthokeratinized odontogenic cysts: a comparison of two Reinders, M.G., van Hout, A.F., Cosgun, B. et al. (2018). New
cysts. Oral Oncology 45: e41–e44. mutations and an updated database for the patched-­1
Rao, R.S., Patil, S., Majumdar, B., and Oswal, R.G. (2015). (PTCH1) gene. Molecular Genetics and Genomic Medicine
Comparison of special stains for keratin with routine 6: 409–415.
hematoxylin and eosin stain. Journal of International Oral Ren, W., Zhi, K., Zhao, L., and Gao, L. (2011). Presentations
Health 7: 1–5. and management of thyroglossal duct cyst in children
Rapidis, A.D. and Langdon, J.D. (1982). Median cysts of the versus adults: a review of 106 cases. Oral Surgery, Oral
jaws–not a true clinical entity. International Journal of Oral Medicine, Oral Pathology, Oral Radiology, and Endodontics
Surgery 11: 360–363. 111: e1–e6.
Rasmusson, L.G., Magnusson, B.C., and Borrman, H. (1991). Resnick, C.M., Dentino, K.M., Garza, R., and Padwa,
The lateral periodontal cyst. A histopathological and B.L. (2016). A management strategy for idiopathic bone
Bibliography 343

cavities of the jaws. Journal of Oral and Maxillofacial Risnes, S. (1974). The prevalence and distribution of cervical
Surgery 74: 1153–1158. enamel projections reaching into the bifurcation on
Rho, M.H., Kim, D.W., Kwon, J.S. et al. (2006). OK-­432 human molars. Scandinavian Journal of Dental Research
sclerotherapy of plunging ranula in 21 patients: it can be a 82: 413–419.
substitute for surgery. American Journal of Neuroradiology Ritchey, B. and Orban, B. (1953). Cysts of the gingivae.
27: 1090–1095. Oral Surgery, Oral Medicine, and Oral Pathology 6:
Rich, A.M. and Neville, B.W. (2022). Nasopalatine duct cyst. 765–771.
In: WHO Classification of Tumours Editorial Board. Head Rivero, E.R., Grando, L.J., de Oliveira, R.G. et al. (2014).
and Neck Tumours, Section 7.1.0.10 (WHO Classification Utility of cell block in the cytological preoperative
of Tumours Series), 5e, vol. 9. Lyon: IARC Press https:// diagnosis of keratocystic odontogenic tumor. Pathology,
publications.iarc.fr/Book-­And-­Report-­Series/ Research and Practice 210: 224–227.
Who-­Classification-­Of-­Tumours. Robinson, H.B.G. (1945). Classification of cysts of the jaws.
Richardson, M.S. (2019). Non-­neoplastic lesions of the American Journal of Orthodontics and Oral Surgery 31:
salivary glands. In: Head and Neck Pathology, 3e (eds. 370–375.
L.D.R. Thompson and J.A. Bishop), 244–248. Philadelphia, Robinson, L., Sengoatsi, T., and van Heerden, W.F.P. (2021).
PA: Elsevier. Concomitant congenital intraoral dermoid cyst and
Richman, J. and Johnston, J. (2020). Gingival cyst of the adult heterotopic gastrointestinal cyst. Head and Neck Pathology
in a pediatric patient: report of a case. Clinical Case Reports 15: 298–302.
8: 359–362. Robinson, R.A. and Vincent, S.D. (2012). Tumours and Cysts
Ricucci, D. and Bergenholtz, G. (2004). Histologic features of of the Jaws, AFIP Atlas of Tumour Pathology. Series 4,
apical periodontitis in human biopsies. Endodontic Topics Fascicle 16. Silver Spring, MD: ARP Press.
8: 68–87. Rodini, C.O., Batista, A.C., Dionísio, T.J. et al. (2008).
Ricucci, D., Loghin, S., Siqueira, J.F. Jr., and Abdelsayed, Morphologic evaluation and expression of matrix
R.A. (2014). Prevalence of ciliated epithelium in metalloproteinases-­2 and 9 and nitric oxide during
apical periodontitis lesions. Journal of Endodontia 40: experimental periodontal disease in rat. Journal of
476–483. Molecular Histology 39: 275–282.
Ricucci, D., Mannocci, F., and Pitt Ford, T.R. (2006a). A study Roed-­Petersen, B. (1969). Nasolabial cysts. British Journal of
of periapical lesions correlating the presence of a Oral Surgery 7: 84–95.
radiopaque lamina with histological findings. Oral Surgery, Roma, L.G., Lopes, C.B., Netto, J.N.S. et al. (2021). Idiopathic
Oral Medicine, Oral Pathology, Oral Radiology, and bone cavity: clinicopathologic features from a case series
Endodontics 101: 389–394. with emphasis on differential diagnosis of chronic apical
Ricucci, D., Pascon, E.A., Pitt Ford, T.R., and Langeland, periodontitis. Journal of Endodontia 47: 221–225.
K. (2006b). Epithelium and bacteria in periapical lesions. Roper-­Hall, H.T. (1938). Cysts of developmental origin in the
Oral Surgery, Oral Medicine, Oral Pathology, Oral premaxillary region, with special reference to their
Radiology, and Endodontics 101: 239–249. diagnosis. British Dental Journal 65: 405–436.
Ricucci, D., Rôças, I.N., Hernández, S., and Siqueira, J.F. Jr. Rosa, A.C.G., Teixeira, L.N., Passador-­Santos, F. et al. (2019).
(2020). ‘True’ versus ‘bay’ apical cysts: clinical, Benign odontogenic ghost cell lesions revisited and new
radiographic, histopathologic, and histobacteriologic considerations on dysplastic dentin. Clinical Oral
features. Journal of Endodontics 46: 1217–1227. Investigations 23: 4335–4343.
Ricucci, D. and Siqueira, J.F. (2008). Apical actinomycosis as Rosencrans, M. and Barak, J. (1969). Parasitic infection of the
a continuum of intraradicular and extraradicular mouth. A case report of Cysticercus cellulosae. New York
infection: case report and critical review on its State Dental Journal 35: 271–273.
involvement with treatment failure. Journal of Rubin, M.M. and Murphy, F.J. (1989). Simple bone cyst of the
Endodontia 34: 1124–1129. mandibular condyle. Journal of Oral and Maxillofacial
Ricucci, D. and Siqueira, J.F. (2010). Biofilms and apical Surgery 47: 1096–1098.
periodontitis: study of prevalence and association with Rud, J. and Pindborg, J.J. (1969). Odontogenic keratocysts: a
clinical and histopathologic findings. Journal of follow-­up study of 21 cases. Journal of Oral Surgery 27:
Endodontics 36: 1277–1288. 323–330.
Rippin, J.W. and Woolgar, J.A. (1991). The odontogenic Rudick, A. (1981). The distribution of jaw cysts accessioned
keratocyst in BCNS and non-­syndrome patients. In: in the Department of Oral Pathology, University of the
Investigative Pathology of the Odontogenic Cysts (ed. Witwatersrand, 1974–1978. MSc (Dent.) dissertation.
R.M. Browne), 212–232. Boca Raton, FL: CRC Press. University of the Witwatersrand, Johannesburg.
344 Bibliography

Rudraraju, A., Venigalla, A., Babburi, S. et al. (2019). Medicina Oral, Patología Oral y Cirugía Bucal 25:
Calretinin expression in odontogenic cysts and e364–e369.
odontogenic tumors and the possible role of calretinin in Santos, L.C., Vilas Bôas, D.S., Oliveira, G.Q. et al. (2011a).
pathogenesis of ameloblastoma. Journal of Oral Histopathological study of radicular cysts diagnosed in a
Maxillofacial Patholology 23: 349–355. Brazilian population. Brazilian Dental Journal 22: 449–454.
Rühl, G.H., Philippou, S., and Mandelartz, E. (1989). Zur Santos, P.P., Freitas, V.S., Freitas Rde, A. et al. (2011b).
Histogenese von hyalinen Bodies in odontogenen Zysten. Botryoid odontogenic cyst: a clinicopathologic study of 10
Deutsche Zeitschrift für Mund-­, Kiefer-­und Gesichts-­ cases. Annals of Diagnostic Pathology 15: 221–224.
Chirurgie 13: 145–154. Santos-­Junior, A.O., De Castro, P.L., Mateo-­Castillo, J.F., and
Rui, Z., Li-­Ying, P., Jia-­Fei, Q. et al. (2014). Smoothened gene Pinheiro, C.R. (2019). Success or failure of endodontic
alterations in keratocystic odontogenic tumors. Head and treatments: a retrospective study. Journal of Conservative
Face Medicine 10: 36. Dentistry 22: 129–132.
Rumayor, A., Carlos, R., Kirsch, H.M. et al. (2015). Ghost Saraçoğlu, U., Kurt, B., Günhan, Ö., and Güven, O. (2005).
cells in pilomatrixoma, craniopharyngioma, and calcifying MIB-­1 expression in odontogenic epithelial rests,
cystic odontogenic tumor: histological, epithelium of healthy oral mucosa and epithelium of
immunohistochemical, and ultrastructural study. Journal selected odontogenic cysts: an immunohistochemical
of Oral Pathology and Medicine 44: 284–290. study. International Journal of Oral and Maxillofacial
Rushton, M.A. (1946). Solitary bone cysts in the mandible. Surgery 34: 432–435.
British Dental Journal 81: 37–49. Saran, R.K., Rattan, V., Rajwanshi, A. et al. (1998).
Rushton, M.A. (1955). Hyaline bodies in the epithelium of Cysticercosis of the oral cavity: report of five cases and a
dental cysts. Proceedings of the Royal Society of Medicine review of literature. International Journal of Paediatric
48: 407–409. Dentistry 8: 273–278.
Rushton, V.E. and Horner, K. (1997). Calcifying odontogenic Sarode, G.S., Sarode, S.C., Tupkari, J.V. et al. (2016).
cyst–a characteristic CT finding. British Journal of Oral and Pathogenesis of Rushton bodies: a novel hypothesis.
Maxillofacial Surgery 35: 196–198. Medical Hypotheses 93: 166–169.
Said-­Al-­Naief, N., Fantasia, J.E., Sciubba, J.J. et al. (1999). Sasai, N., Toriyama, M., and Kondo, T. (2019). Hedgehog
Heterotopic oral gastrointestinal cyst: report of 2 cases and signal and genetic disorders. Frontiers in Genetics
review of the literature. Oral Surgery, Oral Medicine, Oral 10: 1103.
Pathology, Oral Radiology, and Endodontics 88: 80–86. Sathorn, C., Parashos, P., and Messer, H.H. (2005).
Sakamoto, K., Khanom, R., Hamagaki, M., and Yamaguchi, Effectiveness of single-­versus multiple-­visit endodontic
A. (2012). Ectopic production of hair keratin constitutes treatment of teeth with apical periodontitis: a systematic
Rushton’s hyaline bodies in association with review and meta-­analysis. International Endodontic
hematogenous deposits. Journal of Oral Pathology and Journal 38: 347–355.
Medicine 41: 637–641. Savage, N.W., Joseph, B.K., Monsour, P.A., and Young,
Sakamoto, K., Morita, K., Shimada, Y. et al. (2014). W.G. (1996). The glandular odontogenic jaw cyst. Pathology
Peripheral odontogenic keratocyst associated with nevoid 28: 370–372.
basal cell carcinoma syndrome: a case report. Oral Savolainen, S., Eskelin, M., Jousimies-­Somer, H., and
Surgery, Oral Medicine, Oral Pathology, Oral Radiology Ylikoski, J. (1997). Radiological findings in the maxillary
118: e19–e23. sinuses of symptomless young men. Acta Otolaryngologica
Sangüesa Nebot, C., Llorens Salvador, R., Carazo Palacios, Supplement 529: 153–157.
E. et al. (2018). Enteric duplication cysts in children: varied Scharffetter, K., BaIz-­Herrmann, C., Lagrange, W. et al. (1989).
presentations, varied imaging findings. Insights into Proliferation kinetics-­study of the growth of keratocysts.
Imaging 9: 1097–1106. Journal of Cranio-­Maxillo-­Facial Surgery 17: 226–233.
Santarosa, M. and Ashworth, A. (2004). Haploinsufficiency Schmidt, B.L. and Pogrel, M.A. (2001). The use of enucleation
for tumor suppressor genes: when you don’t need to go and liquid nitrogen cryotherapy in the management of
all the way. Biochimica et Biophysica Acta 1654: odontogenic keratocysts. Journal of Oral and Maxillofacial
105–122. Surgery 59: 720–725.
Santos, H.B., Rolim, L.S., Barros, C.C. et al. (2020). Dermoid Schneider, L.C., Mesa, M.L., and Fraenkel, D. (1988).
and epidermoid cysts of the oral cavity: a 48-­year Osteoporotic bone marrow defect: radiographic features
retrospective study with focus on clinical and and pathogenic factors. Oral Surgery, Oral Medicine, and
morphological features and review of main topics. Oral Pathology 65: 127–129.
Bibliography 345

Schneider, T., Filo, K., Locher, M.C. et al. (2014). Stafne bone Semba, I., Kitano, M., Mimura, T. et al. (1994). Glandular
cavities: systematic algorithm for diagnosis derived from odontogenic cyst: analysis of cytokeratin expression and
retrospective data over a 5-­year period. British Journal of clinicopathological features. Journal of Oral Pathology and
Oral and Maxillofacial Surgery 52: 369–374. Medicine 23: 377–382.
Schuch, L.F., de Arruda, J.A.A., Mosconi, C. et al. (2020). A Şen-­Tunç, E., Açikel, H., Sönmez, I.S. et al. (2017).
Brazilian multicentre study of 2,497 isolated cases of Eruption cysts: a series of 66 cases with clinical features.
odontogenic keratocysts. Oral Diseases 26: 711–715. Medicina Oral, Patología Oral y Cirugía Bucal 22:
Schulman, J.M. and Jordan, R.C. (2014). Oral mucus e228–e232.
extravasation phenomenon with myxoglobulosis. Journal Seppala, M., Fraser, G.J., Birjandi, A.A. et al. (2017). Sonic
of Cutaneous Pathology 41: 767–770. hedgehog signaling and development of the dentition.
Schultz, L. (1927). Cysts of the maxillae and mandible. Journal of Developmental Biology 5: 6.
Journal of the American Dental Association (1939) 14: Seward, G.R. (1962). Nasolabial cysts and their radiology.
1395–1402. Dental Practitioner 12: 154–161.
Sciubba, J.J. (2009). Neuralgia-­inducing cavitational Seward, M.H. (1973). Eruption cyst: an analysis of its clinical
osteonecrosis: a status report. Oral Diseases 15: 309–312. features. Journal of Oral Surgery 31: 31–35.
Sedano, H.O. and Gorlin, R.J. (1968). Hyaline bodies of Shade, N.L., Carpenter, W.M., and Delzer, D.D. (1987).
Rushton; some histochemical considerations concerning Gingival cyst of the adult. Case report of a bilateral
their etiology. Oral Surgery, Oral Medicine, and Oral presentation. Journal of Periodontology 58: 796–799.
Pathology 26: 198–201. Shah, A., Latoo, S., Ahmed, I., and Malik, A.H. (2009). Head
Sedano, H.O. and Pindborg, J.J. (1975). Ghost cell epithelium and neck teratomas. Journal of Maxillofacial and Oral
in odontomas. Journal of Oral Pathology 4: 27–30. Surgery 8: 60–63.
Seifert, G., Donath, K., and Jautzke, G. (1999). Unusual Shah, K.A. (2003). Myxoglobulosis in oral extravasation
choristoma of the parotidgland in a girl. A possible mucocele: an entity? Histopathology 43: 291–296.
trichoadenoma. Virchows Archives 434: 355–359. Shamaskin, R.G., Svirsky, J.A., and Kaugars, G.E. (1989).
Seifert, G., Thomsen, S.T., and Donath, K. (1981). Bilateral Intraosseous and extraosseous calcifying odontogenic cyst.
dysgenetic polycystic parotid glands: morphological Journal of Oral and Maxillofacial Surgery 47: 562–565.
analysis and differential diagnosis of a rare disease of Shamim, T., Varghese, V.I., Shameena, P.M., and Sudha,
salivary glands. Virchows Archives A 390: 273–288. S. (2008). Sebaceous differentiation in odontogenic keratocyst.
Seifi, S., Sohanian, S., Khakbaz, O. et al. (2016). Ectopic Indian Journal of Pathology and Microbiology 51: 83–84.
ciliated cyst in the mandible secondary to genioplasty and Shankland, W.E. and Bouquot, J.E. (2004). Focal osteoporotic
Lefort after two years: a case report and literature review. marrow defect: report of 100 new cases with
Iranian Journal of Otorhinolaryngology 28: 353–356. ultrasonography scans. Cranio 22: 314–319.
Sekerci, A.E., Buyuk, S.K., and Cantekin, K. (2014). Cone-­ Shanley, S., Ratcliffe, J., Hockey, A. et al. (1994). Nevoid basal
beam computed tomographic analysis of the cell carcinoma syndrome: review of 118 affected individuals.
morphological characterization of the nasopalatine canal American Journal of Medical Genetics 50: 282–290.
in a pediatric population. Surgical and Radiologic Anatomy Shapira, M. and Akrish, S. (2014). Mucoceles of the oral
36: 925–932. cavity in neonates and infants—­report of a case and
Sekine, S., Sato, S., Takata, T. et al. (2003). Beta-­catenin literature review. Pediatric Dermatology 31: e55–e58.
mutations are frequent in calcifying odontogenic cysts, but Sharif, F.N., Oliver, R., Sweet, C., and Sharif, M.O. (2015).
rare in ameloblastomas. American Journal of Pathology Interventions for the treatment of keratocystic odontogenic
163: 1707–1712. tumours. Cochrane Database of Systematic Reviews 2015
Šekoranja, D., Zupan, A., Mavčič, B. et al. (2020). Novel (11): CD008464. https://doi.org/10.1002/14651858.
ASAP1-­USP6, FAT1-­USP6, SAR1A-­USP6, and TNC-­USP6 CD008464.pub3.
fusions in primary aneurysmal bone cyst. Genes, Sharifian, M.J. and Khalili, M. (2011). Odontogenic cysts: a
Chromosomes and Cancer 59: 357–365. retrospective study of 1227 cases in an Iranian population
Selvamani, M., Devi, A.Y., Basandi, P.S., and Madhushankari, from 1987 to 2007. Journal of Oral Science 53: 361–367.
G.S. (2014). Prevalence and clinicopathological comparison Shear, M. (1960a). Primordial cysts. Journal of the Dental
of kerotocystic odontogenic tumor and orthokeratinized Association of South Africa 15: 211–217.
odontogenic cyst in South Indian sample population: a Shear, M. (1960b). Secretory epithelium in the lining of
retrospective study over 13 years. Journal of Pharmacy and dental cysts. Journal of the Dental Association of South
Bioallied Sciences 6: S127–S130. Africa 15: 117–122.
346 Bibliography

Shear, M. (1963a). The histogenesis of the dental cyst. Dental ramus. American Journal of Physical Anthropology 111:
Practitioner 13: 238–243. 425–427.
Shear, M. (1963b). Cholesterol in dental cysts. Oral Surgery, Shimada, Y., Katsube, K., Kabasawa, Y. et al. (2013).
Oral Medicine, and Oral Pathology 16: 1465–1473. Integrated genotypic analysis of hedgehog-­related genes
Shear, M. (1963c). The microscopic features of the fibrous identifies subgroups of keratocystic odontogenic tumor
walls of dental cysts. Diastema 1: 9–13. with distinct clinicopathological features. PLoS One
Shear, M. (1964). Inflammation in dental cysts. Oral Surgery, 8: e70995.
Oral Medicine, and Oral Pathology 17: 750–767. Shimoyama, T., Horie, N., Nasu, D. et al. (1999). So-­called
Shear, M. (1992). Cysts of the Oral Regions, 3e. Oxford: simple bone cyst of the jaw: a family of pseudocysts of
Wright, Butterworth-­Heinemann. diverse nature and etiology. Journal of Oral Science 41: 93–98.
Shear, M. (1994). Developmental odontogenic cysts. An Shrout, M.K., Hall, M.J., and Hildebolt, C.E. (1993).
update. Journal of Oral Pathology and Medicine Differentiation of periapical granulomas and radicular
23: 1–11. cysts by digital radiometric analysis. Oral Surgery, Oral
Shear, M. (2002a). The aggressive nature of the odontogenic Medicine, and Oral Pathology 76: 356–361.
keratocyst: is it a benign neoplasm? A review. Part 1. Shuster, A., Shlomi, B., Reiser, V., and Kaplan, I. (2012). Solid
Clinical and early experimental evidence of aggressive keratocystic odontogenic tumor—­report of a nonaggressive
behaviour. Oral Oncology 38: 219–226. case. Journal of Oral and Maxillofacial Surgery 70:
Shear, M. (2002b). The aggressive nature of the odontogenic 865–870.
keratocyst: is it a benign neoplasm? A review. Part 2. Siar, C.H. and Ng, K.H. (1988). Orthokeratinized odontogenic
Proliferation and genetic studies. Oral Oncology 38: keratocysts in Malaysians. British Journal of Oral and
323–331. Maxillofacial Surgery 16: 215–220.
Shear, M. (2002c). The aggressive nature of the odontogenic Sibda, A., Meer, S., and Altini, M. (2012). Analysis of
keratocyst: is it a benign neoplasm? A review. Part 3. demographic data obtained from pathology records. South
Immunocytochemistry of cytokeratin and other epithelial African Dental Journal 67: 376–379.
cell markers. Oral Oncology 38: 407–415. Silva, T.A., Garlet, G.P., Fukada, S.Y. et al. (2007).
Shear, M. and Pindborg, J.J. (1975). Microscopic features of Chemokines in oral inflammatory diseases: apical
the lateral periodontal cyst. Scandinavian Journal of Dental periodontitis and periodontal disease. Journal of Dental
Research 83: 103–110. Research 86: 306–319.
Shear, M. and Singh, S. (1978). Age-­standardized incidence Simiyu, B.N., Butt, F., Dimba, E.A. et al. (2013). Keratocystic
rates of ameloblastoma and dentigerous cyst on the odontogenic tumours of the jaws and associated
Witwatersrand, South Africa. Community Dentistry and pathologies: a 10-­year clinicopathologic audit in a referral
Oral Epidemiology 6: 195–199. teaching hospital in Kenya. Journal of Cranio-­Maxillo-­
Sheikh, A.B., Chin, O.Y., Fang, C.H. et al. (2016). Nasolabial Facial Surgery 41: 230–234.
cysts: a systematic review of 311 cases. Laryngoscope Simon, J.H., Enciso, R., Malfaz, J.M. et al. (2006). Differential
126: 60–66. diagnosis of large periapical lesions using cone-­beam
Shen, J., Fan, M., Chen, X. et al. (2006). Glandular computed tomography measurements and biopsy. Journal
odontogenic cyst in China: report of 12 cases and of Endodontics 32: 833–837.
immunohistochemical study. Journal of Oral Pathology Simon, J.H.S. (1980). Incidence of periapical cysts in relation
and Medicine 35: 175–182. to the root canal. Journal of Endodontics 6: 845–848.
Shetty, D.C., Rathore, A.S., Jain, A. et al. (2016). Simon, J.H.S. and Jensen, J.L. (1985). Squamous odontogenic
Orthokeratinized odontogenic cyst masquerading as tumor-­like proliferations in periapical cysts. Journal of
dentigerous cyst. International Journal of Applied and Endodontics 11: 446–448.
Basic Medical Research 6: 297–299. Şimşek-­Kaya, G., Özbudak, İ.H., and Kader, D. (2018).
Shetty, S., Angadi, P.V., and Rekha, K. (2010). Radicular cyst Coexisting sublingual dermoid cyst and heterotopic
in deciduous maxillary molars: a rarity. Head and Neck gastrointestinal cyst: case report. Journal of Clinical and
Pathology 4: 27–30. Experimental Dentistry 10: e196–e199.
Shibata, Y., Asaumi, J., Yanagi, Y. et al. (2004). Radiographic Singh, M., Rattan, K., Rani, B. et al. (2012). Mature teratoma
examination of dentigerous cysts in the transitional of the cheek. APSP Journal of Case Reports 3: 22.
dentition. Dento Maxillo Facial Radiology 33: 17–20. Siponen, M., Neville, B.W., Damm, D.D., and Allen,
Shields, E.D. (2000). Technical note: Stafne static mandibular C.M. (2011). Multifocal lateral periodontal cysts: a report
bone defect-­further expression on the buccal aspect of the of 4 cases and review of the literature. Oral Surgery, Oral
Bibliography 347

Medicine, Oral Pathology, Oral Radiology, and Endodontics Slater, L.J. (2003). Dentigerous cyst versus paradental cyst
111: 225–233. versus eruption pocket cyst. Journal of Oral and
Siriwardena, B.S., Tennakoon, T.M., and Tilakaratne, Maxillofacial Surgery 61: 149.
W.M. (2012). Relative frequency of odontogenic tumors in Slootweg, P.J. (1995). p53 protein and Ki-­67 reactivity in
Sri Lanka: analysis of 1677 cases. Pathology, Research and epithelial odontogenic lesions. An immunohistochemical
Practice 208: 225–230. study. Journal of Oral Pathology and Medicine 24:
Skálová, A., Baněčková, M., Laco, J. et al. (2021). Sclerosing 393–397.
polycystic adenoma of salivary glands: a novel neoplasm Slusarenko da Silva, Y., Stoelinga, P.J.W., and Naclério-­
characterized by PI3K-­AKT pathway alterations – new Homem, M.D.G. (2019a). The presentation of odontogenic
insights into a challenging entity. American Journal of keratocysts in the jaws with an emphasis on the tooth-­
Surgical Pathology. https://doi.org/10.1097/ bearing area: a systematic review and meta-­analysis. Oral
PAS.0000000000001807. Epub ahead of print. and Maxillofacial Surgery 23: 133–147.
Skálová, A., Gnepp, D.R., Simpson, R.H. et al. (2006). Clonal Slusarenko da Silva, Y., Stoelinga, P.J.W., and Naclério-­
nature of sclerosing polycystic adenosis of salivary glands Homem, M.D.G. (2019b). Recurrence of nonsyndromic
demonstrated by using the polymorphism of the human odontogenic keratocyst after marsupialization and delayed
androgen receptor (HUMARA) locus as a marker. enucleation vs. enucleation alone: a systematic review and
American Journal of Surgical Pathology 30: 939–944. meta-­analysis. Oral and Maxillofacial Surgery 23: 1–11.
Skaug, N. (1973). Proteins in fluid from nonkeratinizing jaw Slutzky-­Goldberg, I., Baev, V., Volkov, A. et al. (2013). Incidence
cysts. 2. Concentrations of total protein, some protein of cholesterol in periapical biopsies among adolescent and
fractions and nitrogen. Journal of Oral Pathology 2: elderly patients. Journal of Endodontia 39: 1477–1480.
280–291. Smirniotopoulos, J.G. and Chiechi, M.V. (1995). Teratomas,
Skaug, N. (1974). Proteins in fluid from nonkeratinizing jaw dermoids, and epidermoids of the head and neck.
cysts. 4. Concentrations of immunoglobulins (IgG, IgA and Radiographics 15: 1437–1455.
IgM) and some non-­immunoglobulin proteins: relevance Smith, A.J. (1991). Odontogenic cyst fluids. In: Investigative
to concepts of cyst wall permeability and clearance of Pathology of the Odontogenic Cysts (ed. R.M. Browne),
cystic proteins. Journal of Oral Pathology 3: 47–61. 123–154. Boca Raton, FL: CRC Press.
Skaug, N. (1976a). Intracystic fluid pressure in Smith, A.J. and Matthews, J.B. (1991). Odontogenic
nonkeratinizing jaw cysts. International Journal of Oral epithelium and its residues. In: Investigative Pathology of
Surgery 5: 59–65. the Odontogenic Cysts (ed. R.M. Browne), 53–85. Boca
Skaug, N. (1976b). Lipoproteins in fluid from nonkeratinizing Raton, FL: CRC Press.
jaw cysts. Scandinavian Journal of Dental Research Smith, B.C., Ellis, G.L., Slater, L.J. et al. (1996). Sclerosing
84: 98–105. polycystic adenosis of major salivary glands: a
Skaug, N. (1977). Soluble proteins in fluid from clinicopathologic analysis of nine cases. American Journal
nonkeratinizing jaw cysts in man. International Journal of of Surgical Pathology 20: 161–170.
Oral Surgery 6: 107–121. Smith, G., Matthews, J.B., Smith, A.J., and Browne,
Skaug, N. and Hofstad, T. (1972). Demonstration of R.M. (1987). Immunoglobulin-­producing cells in human
glycosaminoglycans in fluid from jaw cysts. Acta Pathologica odontogenic cysts. Journal of Oral Pathology 16: 45–48.
et Microbiologica Scandinavica Section A 80: 285–286. Smith, G., Smith, A.J., and Browne, R.M. (1984).
Skaug, N. and Hofstad, T. (1973). Proteins in fluid from Glycosaminoglycans in fluid aspirates from odontogenic
non-­keratinizing jaw cysts. 1. Separation patterns on cysts. Journal of Oral Pathology 13: 614–621.
cellulose acetate membranes and percentage distribution Smith, G., Smith, A.J., and Browne, R.M. (1988). Quantification
of the electrophoretic fractions. Journal of Oral Pathology and analysis of the glycosaminoglycans in human
2: 112–125. odontogenic cyst linings. Archives of Oral Biology 33: 623–626.
Skaug, N., Johannessen, A.C., Nilsen, R., and Matre, Smyth, A.G., Ward-­Booth, R.P., and High, A.S. (1993).
R. (1984). In-­situ characterization of cell infiltrates in Polycystic disease of the parotid glands: two familial cases.
human dental periapical granulomas. 3. Demonstration of British Journal of Oral and Maxillofacial Surgery
T lymphocytes. Journal of Oral Pathology 13: 120–127. 31: 38–40.
Slabbert, H., Shear, M., and Altini, M. (1995). Vacuolated cells Sobin, L.H. (1971). The World Health Organization’s
and mucous metaplasia in the epithelial linings of programme for the histopathological definition and
radicular and residual cysts. Journal of Oral Pathology and classification of tumours. Methods of Information in
Medicine 24: 309–312. Medicine 10: 120–122.
348 Bibliography

Soluk Tekkeşin, M.S., Mutlu, S., and Olgac, V. (2011). The Speight, P.M. and Rautava, J. (2022). Glandular odontogenic
role of RANK/RANKL/OPG signalling pathways in cyst. In: WHO Classification of Tumours Editorial Board.
osteoclastogenesis in odontogenic keratocysts, radicular Head and Neck Tumours, Section 7.1.0.7 (WHO
cysts, and ameloblastomas. Head and Neck Pathology 5: Classification of Tumours Series), 5e, vol. 9. Lyon: IARC
248–253. Press https://publications.iarc.fr/Book-­And-­Report-­Series/
Soluk Tekkeşin, M., Mutlu, S., and Olgaç, V. (2012a). Who-­Classification-­Of-­Tumours.
Expressions of bax, bcl-­2 and Ki-­67 in odontogenic Speight, P. and Soluk-­Tekkeşin, M. (2017). Odontogenic cysts
keratocysts (Keratocystic Odontogenic Tumor) in of inflammatory origin. In: WHO Classification of Head
comparison with ameloblastomas and radicular cysts. Turk and Neck Tumours, 4e (eds. A.K. El-­Naggar, J.K.C. Chan,
Patoloji Dergisi 28: 49–55. J.R. Grandis, et al.), 232–233. Lyon: IARC.
Soluk Tekkeşin, M., Olgac, V., Aksakalli, N., and Alatli, Speight, P.M. and Soluk ­Tekkeşin, M. (2022a). Radicular cyst.
C. (2012b). Odontogenic and non-­odontogenic cysts in In: WHO Classification of Tumours Editorial Board. Head
Istanbul: analysis of 5088 cases. Head and Neck 34: and Neck Tumours, Section 7.1.0.1 (WHO Classification of
852–855. Tumours Series), 5e, vol. 9. Lyon: IARC Press https://
Soluk Tekkeşin, M., Pehlivan, S., Olgac, V. et al. (2012c). publications.iarc.fr/Book-­And-­Report-­Series/
Clinical and histopathological investigation of Who-­Classification-­Of-­Tumours.
odontomas: review of the literature and presentation of Speight, P.M. and Soluk ­Tekkeşin, M. (2022b). Inflammatory
160 cases. Journal of Oral and Maxillofacial Surgery 70: collateral cysts. In: WHO Classification of Tumours
1358–1361. Editorial Board. Head and Neck Tumours, Section 7.1.0.2
Soluk Tekkeşin, M., Tuna, E.B., Olgac, V. et al. (2016). (WHO Classification of Tumours Series), 5e, vol. 9. Lyon:
Odontogenic lesions in a pediatric population: review of IARC Press https://publications.iarc.fr/Book-­And-­Report-­
the literature and presentation of 745 cases. International Series/Who-­Classification-­Of-­Tumours.
Journal of Pediatric Otorhinolaryngology 86: 196–199. Speight, P.M. and Takata, T. (2018). New tumour entities in
Song, W.C., Jo, D.I., Lee, J.Y. et al. (2009). Microanatomy of the 4th edition of the World Health Organization
the incisive canal using three-­dimensional reconstruction Classification of head and neck tumours: odontogenic and
of microCT images: an ex vivo study. Oral Surgery, Oral maxillofacial bone tumours. Virchows Archiv 472: 331–339.
Medicine, Oral Pathology, Oral Radiology, and Endodontics Sprinzl, G.M., Koebke, J., Wimmers-­Klick, J. et al. (2000).
108: 583–590. Morphology of the human thyroglossal tract: a histologic
Sorsa, T., Ylipaavalniemi, P., Suomalainen, K. et al. (1988). and macroscopic study in infants and children. Annals of
Type-­specific degradation of interstitial collagens by Otology, Rhinology, and Laryngology 109: 1135–1139.
human keratocyst capsule collagenase. Medical Science Srikant, N., Yellapurkar, S., Boaz, K. et al. (2017). Dysgenetic
Research 16: 1189–1190. polycystic disease of minor salivary gland: a rare case
Soskolne, W.A. and Shear, M. (1967). Observations on the report and review of the literature. Case Reports in
pathogenesis of primordial cysts. British Dental Journal Pathology 2017: 5279025.
123: 321–326. Stafne, E.C. (1942). Bone cavities situated near the angle of
Southam, J.C. (1974). Retention mucoceles of the oral the mandible. Journal of the American Dental Association
mucosa. Journal of Oral Pathology 3: 197–202. 29: 1969–1972.
Speight, P., Devilliers, P., Li, T.-­J. et al. (2017a). Odontogenic Standish, S.M. and Shafer, W.G. (1957). Serial histologic
keratocyst. In: WHO Classification of Head and Neck effects of rat submaxillary and sublingual gland duct and
Tumours, 4e (eds. A.K. El-­Naggar, C. JKC, J.R. Grandis, blood vessel ligation. Journal of Dental Research 36: 866–879.
et al.), 235–236. Lyon: IARC Press. Standish, S.M. and Shafer, W.G. (1959). The mucous retention
Speight, P., Fantasia, J.E., and Neville, B.W. (2017b). phenomenon. Journal of Oral Surgery 17: 15–22.
Orthokeratinised odontogenic cyst. In: WHO Classification Stathopoulos, P., Mezitis, M., Kappatos, C. et al. (2011). Cysts
of Head and Neck Tumours, 4e (eds. A.K. El-­Naggar, and tumors associated with impacted third molars: is
C. JKC, J.R. Grandis, et al.), 241. Lyon: IARC Press. prophylactic removal justified? Journal of Oral and
Speight, P.M. and Neville, B.W. (2022). Othokeratinised Maxillofacial Surgery 69: 405–408.
odontogenic cyst. In: WHO Classification of Tumours Stern, M.H., Dreizen, S., Mackler, B.F., and Levy, B.M. (1981).
Editorial Board. Head and Neck Tumours, Section 7.1.0.9 Antibody-­producing cells in human periapical granulomas
(WHO Classification of Tumours Series), 5e, vol. 9. Lyon: and cysts. Journal of Endodontics 7: 447–452.
IARC Press https://publications.iarc.fr/Book-­And-­Report-­ Stern, M.H., Dreizen, S., Mackler, B.F., and Levy, B.M. (1982).
Series/Who-­Classification-­Of-­Tumours. Isolation and characterization of inflammatory cells from
Bibliography 349

the human periapical granuloma. Journal of Dental Su, L., Weathers, D.R., and Waldron, C.A. (1997a).
Research 61: 1408–1412. Distinguishing features of focal cemento-­osseous
Stoelinga, P.J.W. (1976). Studies on the dental lamina as dysplasias and cemento-­ossifying fibromas: I. A pathologic
related to its role in the etiology of cysts and tumors. spectrum of 316 cases. Oral Surgery, Oral Medicine, Oral
Journal of Oral Pathology 5: 65–73. Pathology, Oral Radiology, and Endodontics 84: 301–309.
Stoelinga, P.J.W. (2001). Long-­term follow-­up on keratocysts Su, L., Weathers, D.R., and Waldron, C.A. (1997b).
treated according to a defined protocol. International Distinguishing features of focal cemento-­osseous dysplasia
Journal of Oral and Maxillofacial Surgery 30: 14–25. and cemento-­ossifying fibromas. II. A clinical and
Stoelinga, P.J.W. (2005). The treatment of odontogenic radiologic spectrum of 316 cases. Oral Surgery, Oral
keratocysts by excision of the overlying, attached mucosa, Medicine, Oral Pathology, Oral Radiology, and Endodontics
enucleation, and treatment of the bony defect with carnoy 84: 540–549.
solution. Journal of Oral and Maxillofacial Surgery 63: Suei, Y., Taguchi, A., Kurabayashi, T. et al. (1998). Simple
1662–1666. bone cyst: investigation on the presence of gas in the cavity
Stoelinga, P.J.W. (2019). Keratocystic odontogenic tumour using computed tomography–review of 52 cases. Oral
(KCOT) has again been renamed odontogenic keratocyst Surgery, Oral Medicine, Oral Pathology, Oral Radiology, and
(OKC). International Journal of Oral and Maxillofacial Endodontics 86: 592–594.
Surgery 48: 415–416. Suei, Y., Taguchi, A., Nagasaki, T., and Tanimoto, K. (2010).
Stoelinga, P.J.W. and Bronkhorst, F.B. (1988). The incidence, Radiographic findings and prognosis of simple bone cysts
multiple presentation and recurrence of aggressive cysts of the of the jaws. Dento Maxillo Facial Radiology 39: 65–71.
jaws. Journal of Cranio-­Maxillo-­Facial Surgery 16: 184–195. Sugar, A.W., Walker, D.M., and Bounds, G.A. (1990). Surgical
Stoelinga, P.J.W., and Slusarenko da Silva, Y. (2021). The ciliated (postoperative maxillary) cysts following mid-­face
significance of recurrent odontogenic keratocysts in bone osteotomies. British Journal of Oral and Maxillofacial
grafts. International Journal of Oral and Maxillofacial Surgery 28: 264–267.
Surgery 50: 746–749. Sugerman, P.B., Savage, N.W., and Young, W.G. (2000).
Stoelinga, P.J.W., Cohen, M., and Morgan, A.F. (1975). The Mucocele of the anterior lingual salivary glands (glands of
origin of keratocysts in the basal cell nevus syndrome. Blandin and Nuhn): report of 5 cases. Oral Surgery, Oral
Journal of Oral Surgery 33: 659–663. Medicine, Oral Pathology, Oral Radiology, and Endodontics
Stoelinga, P.J.W. and Peters, J.H. (1973). A note on the origin 90: 478–482.
of keratocysts of the jaws. International Journal of Oral Sumer, A.P., Celenk, P., Sumer, M. et al. (2010). Nasolabial
Surgery 2: 37–44. cyst: case report with CT and MRI findings. Oral Surgery,
Stojanov, I.J., Malik, U.A., and Woo, S.B. (2017). Intraoral Oral Medicine, Oral Pathology, Oral Radiology, and
salivary duct cyst: clinical and histopathologic features of Endodontics 109: e92–e94.
177 cases. Head and Neck Pathology 11: 469–476. Summers, L. (1972). Cavitation of apical cysts. Journal of
Stojanov, I.J., Schaefer, I.M., Menon, R.S. et al. (2020). Dental Research 51: 12–47.
Biallelic PTCH1 inactivation is a dominant genomic Summers, L. (1974). The incidence of epithelium in periapical
change in sporadic keratocystic odontogenic tumors. granulomas and the mechanism of cavitation in apical
American Journal of Surgical Pathology 44: 553–560. dental cysts in man. Archives of Oral Biology 19: 1177–1180.
Stoll, C., Stollenwerk, C., Riedeger, D. et al. (2005). Sun, L.S., Li, X.F., and Li, T.J. (2008). PTCH1 and SMO gene
Cytokeratin expression patterns for distinction of alterations in keratocystic odontogenic tumors. Journal of
odontogenic keratocysts from dentigerous and radicular Dental Research 87: 575–579.
cysts. Journal of Oral Pathology and Medicine 34: Sundaragiri, K.S., Saxena, S., Sankhla, B., and Bhargava,
558–564. A. (2018). Non syndromic synchronous multiple
Stoneman, D.W. and Worth, H.M. (1983). The mandibular odontogenic keratocysts in a western Indian population: a
infected buccal cyst–molar area. Dental Radiography and series of four cases. Journal of Clinical and Experimental
Photography 56: 1–14. Dentistry 10: e831–e836.
Struthers, P.J. and Shear, M. (1976). Root resorption produced Suojanen, J., Lehtonen, N., Färkkilä, E. et al. (2014).
by the enlargement of ameloblastomas and cysts of the Common matrix metalloproteinases (MMP-­8, -­9, -­25, and
jaws. International Journal of Oral Surgery 5: 128–132. -­26) cannot explain dentigerous cyst expansion. Journal of
Su, C.Y., Chien, C.Y., and Hwang, C.F. (1999). A new Clinical and Diagnostic Research 8: ZC82–ZC85.
transnasal approach to endoscopic marsupialization of the Suresh, R., Janardhanan, M., Joseph, A.P. et al. (2011). A rare
nasolabial cyst. Laryngoscope 109: 1116–1118. case of dentigerous cyst in a one year old child: the earliest
350 Bibliography

known reported occurrence. Head and Neck Pathology 5: Takeda, Y., Oikawa, Y., Furuya, I. et al. (2005). Mucous and
171–174. ciliated cell metaplasia in epithelial linings of odontogenic
Suter, V.G., Jacobs, R., Brücker, M.R. et al. (2016). Evaluation inflammatory and developmental cysts. Journal of Oral
of a possible association between a history of dentoalveolar Science 47: 77–81.
injury and the shape and size of the nasopalatine canal. Takeda, Y., Suzuki, A., and Yamamoto, H. (1990).
Clinical Oral Investigations 20: 553–561. Histopathologic study of epithelial components in the
Suter, V.G., Sendi, P., Reichart, P.A., and Bornstein, connective tissue wall of unilocular type of calcifying
M.M. (2011). The nasopalatine duct cyst: an analysis of the odontogenic cyst. Journal of Oral Pathology and Medicine
relation between clinical symptoms, cyst dimensions, and 19: 108–113.
involvement of neighboring anatomical structures using Takeda, Y. and Yamamoto, H. (2001). Salivary duct cyst: its
cone beam computed tomography. Journal of Oral and frequency in a certain Japanese population group (Tohoku
Maxillofacial Surgery 69: 2595–2603. districts), with special reference to adenomatous
Suter, V.G., Warnakulasuriya, S., Reichart, P.A., and proliferation of the epithelial lining. Journal of Oral
Bornstein, M.M. (2015). Radiographic volume analysis as a Science 43: 9–13.
novel tool to determine nasopalatine duct cyst dimensions Tal, H., Altini, M., and Lemmer, J. (1984). Multiple mucous
and its association with presenting symptoms and retention cysts of the oral mucosa. Oral Surgery, Oral
postoperative complications. Clinical Oral Investigations Medicine, and Oral Pathology 58: 692–695.
19: 1611–1618. Tamiolakis, P., Thermos, G., Tosios, K.I., and Sklavounou-­
Swanson, K.S., Kaugars, G.E., and Gunsolley, J.C. (1991). Andrikopoulou, A. (2019). Demographic and clinical
Nasopalatine duct cyst: an analysis of 334 cases. Journal of characteristics of 5294 jaw cysts: a retrospective study of 38
Oral and Maxillofacial Surgery 49: 268–271. years. Head and Neck Pathology 13: 587–596.
Syamala, D., Suresh, R., Janardhanan, M. et al. (2016). Tanaka, A., Okamoto, M., Yoshizawa, D. et al. (2007). Presence
Immunohistochemical evaluation of myofibroblasts in of ghost cells and the Wnt signaling pathway in odontomas.
odontogenic cysts and tumors: a comparative study. Journal of Oral Pathology and Medicine 36: 400–404.
Journal of Oral and Maxillofacial Pathology 20: 208–213. Tavares, D.P., Rodrigues, J.T., Dos Santos, T.C. et al. (2017).
Sykara, M., Ntovas, P., Kalogirou, E.M. et al. (2017). Oral Clinical and radiological analysis of a series of periapical
lymphoepithelial cyst: a clinicopathological study of 26 cysts and periapical granulomas diagnosed in a Brazilian
cases and review of the literature. Journal of Clinical and population. Journal of Clinical and Experimental Dentistry
Experimental Dentistry 9: e1035–e1043. 9: e129–e135.
Syyed, N., Mason, R., Thomson, E., and Downie, Tay, J.Y.Y., Bay, B.H., Yeo, J.F. et al. (2004). Identification of
J. (2019). Mandibular respiratory cysts following RANKL in osteolytic lesions of the facial skeleton. Journal
orthognathic surgery: 2 rare case reports. Clinics in of Dental Research 83: 349–353.
Surgery 4: 2400. Taylor, B.W., Erich, J.B., and Dockerty, M.B. (1966). Dermoids
Tabrizi, R., Hosseini Kordkheili, M.R., Jafarian, M., and of the head and neck. Minnesota Medicine 49: 1535–1540.
Aghdashi, F. (2019). Decompression or marsupialization; Tek, M., Metin, M., Sener, I. et al. (2013). The predominant
which conservative treatment is associated with low bacteria isolated from radicular cysts. Head & Face
recurrence rate in keratocystic odontogenic tumors? Medicine 9: 25.
A systematic review. Journal of Dentistry (Shiraz) 20: Telfer, M.R., Jones, G.M., Pell, G.M., and Eveson, J.W. (1990).
145–151. Primary bone cyst of the mandibular condyle. British
Takata, T. and Lu, Y. (2005). Ghost cell odontogenic Journal of Oral and Maxillofacial Surgery 28: 340–343.
carcinoma. In: Pathology and Genetics. Head and Neck Ten Cate, A.R. (1972). The epithelial cell rests of Malassez
Tumours. World Health Organization Classification of and the genesis of the dental cyst. Oral Surgery, Oral
Tumours (eds. L. Barnes, J.W. Eveson, P. Reichart and Medicine, and Oral Pathology 34: 950–964.
D. Sidransky), 293. Lyon: IARC Press. Teronen, O., Hietanin, J., Lindqvist, C. et al. (1996). Mast
Takata, T., Zhao, M., Nikai, H. et al. (2000). Ghost cells in cell-­derived tryptase in odontogenic cysts. Journal of Oral
calcifying odontogenic cyst express enamel-­related Pathology and Medicine 25: 376–381.
proteins. Histochemical Journal 32: 223–229. Teronen, O., Salo, T., Konttinen, Y.T. et al. (1995a).
Takeda, Y. (1985). Hyaline bodies and secondary dental Identification and characterization of gelatinases/type IV
cuticle in dentigerous cyst. Journal of Oral Pathology 14: collagenases in jaw cysts. Journal of Oral Pathology and
268–269. Medicine 24: 78–84.
Takeda, Y., Kikuchi, H., and Suzuki, A. (1985). Hyaline Teronen, O., Salo, T., Laitinen, J. et al. (1995b).
bodies in ameloblastoma: histological and ultrastructural Characterization of interstitial collagenases in jaw cyst
observations. Journal of Oral Pathology 14: 639–643. wall. European Journal of Oral Sciences 103: 141–147.
Bibliography 351

Teszler, C.B., El-­Naaj, I.A., Emodi, O. et al. (2007). Toller, P.A. (1948). Experimental investigations into factors
Dermoid cysts of the lateral floor of the mouth: a concerning the growth of cysts of the jaws. Proceedings of
comprehensive anatomo-­surgical classification of cysts the Royal Society of Medicine 41: 681–688.
of the oral floor. Journal of Oral and Maxillofacial Toller, P.A. (1964). Radioactive isotope and other
Surgery 65: 327–332. investigations in a case of haemorrhagic cyst of the
Theofilou, N.E., Lombardi, T., and Scolozzi, P. (2021). mandible. British Journal of Oral Surgery 2: 86–93.
Maxillary surgical ciliated cysts following advancement Le Toller, P.A. (1966a). Epithelial discontinuities in cysts of the
Fort I osteotomy with concomitant autogenous bone jaws. British Dental Journal 120: 74–78.
grafting: a simple coincidence or a cause-­effect Toller, P.A. (1966b). Permeability of cyst walls in vivo:
relationship? Journal of Stomatology Oral Maxillofacial investigations with radioactive tracers. Proceedings of the
Surgery https://doi.org/10.1016/j.jormas.2020.10.002. Epub Royal Society of Medicine 59: 724–729.
ahead of print. Toller, P.A. (1967). Origin and growth of cysts of the jaws.
Thoma, K.H. (1917). A histo-­pathological study of the dental Annals of the Royal College of Surgeons of England 40:
granuloma and diseased root apex. Journal of the National 306–336.
Dental Association 4: 1075–1090. Toller, P.A. (1970a). Protein substances in odontogenic cyst
Thomas, G., Pandey, M., Mathew, A. et al. (2001). Primary fluids. British Dental Journal 128: 317–322.
intraosseous carcinoma of the jaw: pooled analysis of Toller, P.A. (1970b). The osmolality of fluids from cysts of the
world literature and report of two new cases. International jaws. British Dental Journal 129: 275–278.
Journal of Oral and Maxillofacial Surgery 30: 349–355. Toller, P.A. (1971). Autoradiography of explants from
Thompson, I.O., de Waal, J., and Nortje, C.J. (1997). odontogenic cysts. British Dental Journal 131: 57–61.
Mandibular infected buccal cyst and paradental cyst: the Toller, P.A. and Holborrow, E.J. (1969). Immunoglobulins and
same or separate entities? Journal of the Dental Association immunoglobulin containing cells in cysts of the jaws.
of South Africa 52: 503–506. Lancet 2 (7613): 178–181.
Thompson, L.D.R., Herrera, H.B., and Lau, S.K. (2016). A Tolman, D.E. and Stafne, E.C. (1967). Developmental bone
clinicopathologic series of 685 thyroglossal duct remnant defects of the mandible. Oral Surgery, Oral Medicine, and
cysts. Head and Neck Pathology 10: 465–474. Oral Pathology 24: 488–490.
Thompson, L.D.R., Herrera, H.B., and Lau, S.K. (2017). Tomlinson, I.P., Roylance, R., and Houlston, R.S. (2001). Two
Thyroglossal duct cyst carcinomas: a clinicopathologic hits revisited again. Journal of Medical Genetics 38 (2):
series of 22 cases with staging recommendations. Head 81–85. https://doi.org/10.1136/jmg.38.2.81.
and Neck Pathology 11: 175–185. Tong, A.C., Ng, I.O., and Yan, B.S. (2003). Variations in
Thosaporn, W., Iamaroon, A., Pongsiriwet, S., and Ng, clinical presentations of the simple bone cyst: report of
K.H. (2004). A comparative study of epithelial cell cases. Journal of Oral and Maxillofacial Surgery 61:
proliferation between the odontogenic keratocyst, 1487–1491.
orthokeratinized odontogenic cyst, dentigerous cyst, and Tonni, G., De Felice, C., Centini, G., and Ginanneschi,
ameloblastoma. Oral Diseases 10: 22–26. C. (2010). Cervical and oral teratoma in the fetus: a
Thurnwald, G.A., Acton, C.H., and Savage, N.W. (1994). The systematic review of etiology, pathology, diagnosis,
mandibular infected buccal cyst–a reappraisal. Annals of treatment and prognosis. Archives of Gynecology and
the Royal Australasian College of Dental Surgeons 12: Obstetrics 282: 355–361.
255–263. Torabinejad, M. and Kettering, J.D. (1985). Identification and
Tiago, R.S., Maia, M.S., Nascimento, G.M. et al. (2008). relative concentration of B and T lymphocytes in human
Nasolabial cyst: diagnostic and therapeutical aspects. chronic periapical lesions. Journal of Endodontics 11:
Brazilian Journal of Otorhinolaryngology 74: 39–43. 122–125.
Timosca, G. and Gavrilită, L. (1974). Cysticercosis of the Torabinejad, M., Kutsenko, D., Machnick, T.K. et al. (2005).
maxillofacial region. Oral Surgery, Oral Medicine, and Oral Levels of evidence for the outcome of nonsurgical
Pathology 37: 390–400. endodontic treatment. Journal of Endodontics 31: 637–646.
Toida, M. (1998). So-­called calcifying odontogenic cyst: review Tortorici, S., Amodio, E., Massenti, M.F. et al. (2008).
and discussion on the terminology and classification. Prevalence and distribution of odontogenic cysts in Sicily:
Journal of Oral Pathology and Medicine 27: 49–52. 1986–2005. Journal of Oral Science 50: 15–18.
Toida, M., Tsai, C.-­S., Okumura, Y. et al. (1990). Distribution Tosios, K.I., Kakarantza-­Angelopoulou, E., and Kapranos,
of Factor XIIIa-­containing cells and collagenous N. (2000). Immunohistochemical study of bcl-­2 protein,
components in radicular cysts: histochemic and Ki-­67 antigen and p53 protein in epithelium of glandular
immunohistochemic studies. Journal of Oral Pathology odontogenic cysts and dentigerous cysts. Journal of Oral
and Medicine 19: 155–159. Pathology and Medicine 29: 139–144.
352 Bibliography

Toto, P.D., Wortel, J.P., and Joseph, G. (1982). of a case. Journal of Oral and Maxillofacial Surgery 45:
Lymphoepithelial cysts and associated immunoglobulins. 346–349.
Oral Surgery, Oral Medicine, and Oral Pathology 54: 59–65. Ustundag, E., Iseri, M., Keskin, G. et al. (2005). Cervical
Tran, P.T., Cunningham, C.J., and Baughman, R.A. (2004). bronchogenic cysts in head and neck region. Journal of
Glandular odontogenic cyst. Journal of Endodontics 30: Laryngology and Otology 119: 419–423.
182–184. Valderhaug, J. (1972). A histologic study of experimentally
Trask, G.M., Sheller, B.L., and Morton, T.H. (1985). Mandibular induced radicular cysts. International Journal of Oral
buccal infected cyst in a six-­year-­old girl: report of a case. Surgery 1: 137–147.
Journal of Dentistry for Children 52: 377–379. Valderhaug, J. (1974). A histologic study of experimentally
Trott, J.R., Chebib, F., and Galindo, Y. (1973). Factors related induced periapical inflammation in primary teeth in
to cholesterol formation in cysts and granulomas. Journal monkeys. International Journal of Oral Surgery 3:
of the Canadian Dental Association 39: 550–555. 111–113.
Trott, J.R. and Esty, C. (1972). An analysis of 105 dental cysts. Vallo, J., Suominen-­Taipale, L., Huumonen, S. et al. (2010).
Journal of the Canadian Dental Association 38: 75–78. Prevalence of mucosal abnormalities of the maxillary
Tsai, C.-­H., Weng, S.-­F., Yang, L.-­C. et al. (2004). sinus and their relationship to dental disease in
Immunohistochemical localization of tissue-­type panoramic radiography: results from the Health 2000
plasminogen activator and type 1 plasminogen activator Health Examination Survey. Oral Surgery, Oral Medicine,
inhibitor in radicular cysts. Journal of Oral Pathology and Oral Pathology, Oral Radiology, and Endodontics 109:
Medicine 33: 156–161. e80–e87.
Tsesis, I., Rosen, E., Dubinsky, L. et al. (2016). Metaplastic van Bruggen, A.P., Shear, M., du Preez, I.J. et al. (1985).
changes in the epithelium of radicular cysts: a series of Nasolabial cysts. A report of 10 cases and a review of the
711 cases. Journal of Clinical and Experimental Dentistry literature. Journal of the Dental Association of South
8: e529–e533. Africa 40: 15–19.
Tsuji, K., Wato, M., Hayashi, T. et al. (2014). The expression Van der Wal, N., Wiener, J.D., Allard, R.H.B. et al. (1987).
of cytokeratin in keratocystic odontogenic tumor, Thyroglossal cysts in patients over 30 years of age.
orthokeratinized odontogenic cyst, dentigerous cyst, International Journal of Oral and Maxillofacial Surgery 16:
radicular cyst and dermoid cyst. Medical Molecular 416–419.
Morphology 47: 156–161. van Heerden, W.F.P. and Gomes, C.C. (2022). Ghost cell
Tsuneki, M., Maruyama, S., Yamazaki, M. et al. (2013). odontogenic carcinoma. In: WHO Classification of
Inflammatory histopathogenesis of nasopalatine duct cyst: Tumours Editorial Board. Head and Neck Tumours, Section
a clinicopathological study of 41 cases. Oral Diseases 19: 7.2.2.5 (WHO Classification of Tumours Series), 5e, vol. 9.
415–424. Lyon): IARC Press https://publications.iarc.fr/Book-And-
Uchiyama, Y., Akiyama, H., Murakami, S. et al. (2012). Report-Series/Who-Classification-Of-Tumours.
Calcifying cystic odontogenic tumour: CT imaging. British van Heerden, W.F.P. and Van Zyl, A.W. (2010). Diagnosis and
Journal of Radiology 85: 548–554. management of oral lesions and conditions in the
Uddin, N., Zubair, M., Abdul-­Ghafar, J. et al. (2019). newborn. South African Family Practice 52: 489–449.
Orthokeratinized odontogenic cyst (OOC): van Heerden, W.F.P., Raubenheimer, E.J., Weir, R.G., and
clinicopathological and radiological features of a series of Kreidler, J. (1989). Giant ossifying fibroma: a
10 cases. Diagnostic Pathology 14 (28): 1–7. clinicopathologic study of 8 tumors. Journal of Oral
Ueeck, B.A., Woo, B.M., Stewart, J.C., and Assael, Pathology and Medicine 18: 506–509.
L.A. (2007). Keratinizing odontogenic cyst with verrucous van Heerden, W.F.P., Raubenheimer, E.J., and Turner,
proliferation. Journal of Oral and Maxillofacial Surgery M.L. (1992). Glandular odontogenic cyst. Head and Neck
65: 585–588. 14: 316–320.
Ueki, Y., Tiziani, V., Santanna, C. et al. (2001). Mutations in van Rensburg, L.J., Paquette, M., Morkel, J.A., and Nortjé,
the gene encoding c-­Abl-­binding protein SH3BP2 cause C.J. (2003). Correlative MRI and CT imaging of the
cherubism. Nature Genetics 28: 125–126. odontogenic keratocyst: a review of 21 cases. Oral and
Ueno, T., Takayama, R., Osada, S.I., and Saeki, H. (2018). Maxillofacial Surgery Clinics of North America 15:
Epidermoid cyst arising on the body of the tongue: case 363–382.
report and literature review. Journal of Nippon Medical Vargas, P.A., da Cruz Perez, D.E., Mata, G.M. et al. (2007).
School 85: 343–346. Fine needle aspiration cytology as an additional tool in the
Unal, T., Gomel, M., and Gunel, O. (1987). Squamous diagnosis of odontogenic keratocyst. Cytopathology 18:
odontogenic tumor-­like islands in a radicular cyst: report 361–366.
Bibliography 353

Vargas, P.A. and Gomez, R.S. (2022). Gingival cysts. In: WHO different from odontogenic cysts. Journal of Oral Pathology
Classification of Tumours Editorial Board. Head and Neck and Medicine 38: 597–604.
Tumours, Section 7.1.0.6 (WHO Classification of Tumours Vered, M., Shohat, I., Buchner, A., and Dayan, D. (2005).
Series), 5e, vol. 9. Lyon: IARC Press https://publications. Myofibroblasts in stroma of odontogenic cysts and tumors
iarc.fr/Book-­And-­Report-­Series/Who-­Classification-­Of-­ can contribute to variations in the biological behavior of
Tumours. lesions. Oral Oncology 41: 1028–1023.
Vargas, P.A. and White, S.J. (2022). Lateral periodontal cyst Vicandi, B., Jimenez-­Heffernan, J.A., Lopez-­Ferrer, P. et al.
and botryoid odontogenic cyst. In: WHO Classification of (1999). HIV-­1 (p24)-­positive multinucleated giant cells
Tumours Editorial Board. Head and Neck Tumours, 5e, in HIV-­associated lymphoepithelial lesion of the
vol. 9, Section 7.1.0.5. Lyon: IARC https://publications. parotid gland. A report of two cases. Acta Cytologica 43:
iarc.fr/Book-­And-­Report-­Series/ 247–251.
Who-­Classification-­Of-­Tumours. Vicente-­Barrero, M., Báez-­Marrero, O. et al. (2004). Cartilage
Vasconcelos, R.F., de Aguiar, M.F., Castro, W. et al. (1999a). in the walls of odontogenic keratocyst. Medicina Oral 9:
Retrospective analysis of 31 cases of nasopalatine duct 268–270.
cyst. Oral Diseases 5: 325–328. Vier, F.V. and Figueiredo, J.A. (2002). Prevalence of different
Vasconcelos, R.F., Souza, P.E., and Mesquita, R.A. (1999b). periapical lesions associated with human teeth and their
Retrospective analysis of 15 cases of nasolabial cyst. correlation with the presence and extension of apical
Quintessence International 30: 629–632. external root resorption. International Endodontics Journal
Vázquez-­Romero, M.D., Serrera-­Figallo, M.L., Alberdi-­ 35: 710–719.
Navarro, J. et al. (2017). Maxillary peripheral keratocystic Vidaković, B., Uljanić, I., Grgurević, J. et al. (2016). Botryoid
odontogenic tumor. A clinical case report. Journal of cyst, a rare type of odontogenic cyst. Acta Clinica Croatica
Clinical and Experimental Dentistry 9: e167–e171. 55: 510–514.
Vedtofte, P. and Holmstrup, P. (1989). Inflammatory Vij, H., Vij, R., Gupta, V., and Senqupta, S. (2011).
paradental cysts in the gIobulomaxillary region. Journal of Odontogenic keratocyst: a peripheral variant. Nigerian
Oral Pathology and Medicine 18: 125–127. Journal of Clinical Practice 14: 504–507.
Vedtofte, P. and Prætorius, F. (1979). Recurrence of the Vinayachandran, D. and Sankarapandian, S. (2013). Salivary
odontogenic keratocyst in relation to clinical and duct cyst: histo-­pathologic correlation. Journal of Clinical
histological features. A 20-­year follow-­up study of 72 Imaging Science 3 (Suppl. 1): 3.
patients. International Journal of Oral Surgery 8: Viveiros, S.K., Pinho, R.F.C., Custódio, M. et al. (2019). A rare
412–420. odontogenic cyst: gingival cyst of the adult. A series of
Vedtofte, P. and Praetorius, F. (1989). The inflammatory 20 new cases from a single center. Journal of Cranio-­
paradental cyst. Oral Surgery, Oral Medicine, and Oral Maxillo-­Facial Surgery 47: 647–650.
Pathology 68: 182–188. von Arx, T., Schaffner, M., and Bornstein, M.M. (2018). Patent
Velez, I., Siegel, M.A., Mintz, S.M., and Rolle, R. (2010). The nasopalatine ducts: an update of the literature and a series
relationship between idiopathic bone cavity and of new cases. Surgical and Radiologic Anatomy 40:
orthodontic tooth movement: analysis of 44 cases. Dento 165–177.
Maxillo Facial Radiology 39: 162–166. Voorsmit, R.A.C.A., Stoelinga, P.J.W., and van Haelst,
Vencio, E.F., Mota, A., Pinho, C.d.M., and Filho, U.J.G.M. (1981). The management of keratocysts. Journal
A.A.D. (2006). Odontogenic keratocyst in maxillary sinus of Maxillofacial Surgery 9: 228–236.
with invasive behaviour. Journal of Oral Pathology and Vuhahula, E., Nikai, H., Ijuhin, N. et al. (1993). Jaw cysts
Medicine 35: 249–251. with orthokeratinization: analysis of 12 cases. Journal of
Vered, M., Allon, I., Buchner, A., and Dayan, D. (2010). Is Oral Pathology and Medicine 22: 35–40.
maspin immunolocalization a tool to differentiate central Wagner, V.P., Martins, M.D., Curra, M. et al. (2015). Gingival
low-­grade mucoepidermoid carcinoma from glandular cysts of adults: retrospective analysis from two centers in
odontogenic cyst? Acta Histochemica 112: 161–168. south Brazil and a review of the literature. Journal of the
Vered, M., Buchner, A., Dayan, D. et al. (2004). Solid variant International Academy of Periodontology 17: 14–19.
of odontogenic keratocyst. Journal of Oral Pathology and Wahlgren, J., Maisi, P., Sorsa, T. et al. (2001). Expression and
Medicine 33: 125–128. induction of collagenases (MMP-­8 and -­13) in plasma cells
Vered, M., Peleg, O., Taicher, S., and Buchner, A. (2009). The associated with bone-­destructive lesions. Journal of
immunoprofile of odontogenic keratocyst (keratocystic Pathology 194: 217–224.
odontogenic tumor) that includes expression of PTCH, Wakasa, T., Kawai, N., Aiga, H., and Kishi, K. (2002).
SMO, GLI-­1 and bcl-­2 is similar to ameloblastoma but Management of florid cemento-­osseous dysplasia of the
354 Bibliography

mandible producing bone cyst: report of a case. Journal of Wertheimer, F.W., Fullmer, H.M., and Hansen, L.S. (1962). A
Oral and Maxillofacial Surgery 60: 832–835. histochemical study of hyaline bodies in odontogenic cysts
Waldron, C.A. and Koh, M.L. (1990). Central and a comparison to the human secondary dental cuticle.
mucoepidermoid carcinoma of the jaws: report of four Oral Surgery, Oral Medicine, and Oral Pathology 15: 1466.
cases with analysis of the literature and discussion of the Whitt, J.C., Dunlap, C.L., Sheets, J.L., and Thompson,
relationship to mucoepidermoid, sialodontogenic, and M.L. (2007). Keratoameloblastoma: a tumor sui generis or
glandular odontogenic cysts. Journal of Oral and a chimera? Oral Surgery, Oral Medicine, Oral Pathology,
Maxillofacial Surgery 48: 871–877. Oral Radiology, and Endodontics 104: 368–376.
Walton, R.E. (1996). The residual radicular cyst: does it exist? WHO (2022a). Head and Neck Tumours, WHO Classification
Oral Surgery, Oral Medicine, and Oral Pathology 82: 471. of Tumours Series, 5e, vol. 9. Lyon: IARC Press https://
Wang, W., Li, Y., and Tang, Y. (2015). Keratocystoma of the publications.iarc.fr/Book-­And-­Report-­Series/
parotid gland: a case report and review of previous Who-­Classification-­Of-­Tumours.
publications. British Journal of Oral and Maxillofacial WHO (2022b). Soft Tissue and Bone Tumours, WHO
Surgery 53: 655–657. Classification of Tumours Series, 5e, vol. 3. Lyon: IARC
Wang, Y.P., Chang, Y.F., Wang, J.T. et al. (2003). Press https://publications.iarc.fr/Book-­And-­Report-­Series/
Characteristics of calcifying odontogenic cyst in Who-­Classification-­Of-­Tumours.
Taiwanese. Journal of the Formosan Medical Association Wiersma, R., Hadley, G.P., Bosenberg, A.T., and Chrystal,
102: 715–721. V. (1992). Intralingual cysts of foregut origin. Journal of
Ward, J.P., Magar, V., Franks, S.J., and Landini, G. (2004). A Pediatric Surgery 27: 1404–1406.
mathematical model of the dynamics of odontogenic cyst Williams, H.K. and Williams, D.M. (1989). Persistent
growth. Analytical and Quantitative Cytology and Histology sialadenitis of the minor glands: stomatitis glandularis.
26: 39–46. British Journal of Oral and Maxillofacial Surgery 27: 212–216.
Warren, M., Xu, D., and Li, X. (2017). Gene fusions Wise, G.E., Frazier-­Bowers, S., and D’Souza, R.N. (2002).
PAFAH1B1-­USP6 and RUNX2-­USP6 in aneurysmal bone Cellular, molecular, and genetic determinants of tooth
cysts identified by next generation sequencing. Cancer eruption. Critical Reviews in Oral Biology and Medicine 13:
Genetics 212–213: 13–18. 323–335.
Warwick, J.W. (1926). Do epithelial odontomes increase in Witteveen, M.E., Flores, I.L., Karssemakers, L.H., and
size by their own tension. Proceedings of the Royal Socety of Bloemena, E. (2019). Odontogenic keratocysts located in
Medicine 19: 73–77. the buccal mucosa: a description of two cases and review
Wayman, B.E., Murata, S.M., Almeida, R.J., and Fowler, of the literature. SAGE Open Medical Case Reports 7:
C.B. (1992). A bacteriological and histological evaluation of 2050313X19849828.
58 periapical lesions. Journal of Endodontics 18: 152–155. Woldenberg, Y., Goldstein, J., and Bodner, L. (2004). Eruption
Weathers, D.R. and Waldron, C.A. (1973). Unusual cyst in the adult: a case report. International Journal of
multilocular cysts of the jaws (botryoid odontogenic cysts). Oral and Maxillofacial Surgery 33: 804–805.
Oral Surgery, Oral Medicine, and Oral Pathology 36: Wolf, J. and Hietanen, J. (1990). The mandibular infected
235–241. buccal cyst (paradental cyst). A radiographic and
Weidner, N., Geisinger, K.R., Sterling, R.T. et al. (1986). histological study. British Journal of Oral and Maxillofacial
Benign lymphoepithelial cysts of the parotid gland. A Surgery 28: 322–325.
histologic, cytologic and ultrastructural study. American Woo, S.-­B. (2016). Oral Pathology: A Comprehensive Atlas and
Journal of Clinical Pathology 85: 395–401. Text, 2e. Philadelphia, PA: Elsevier Saunders.
Wenig, B.M. (2017a). Neck. In: Non-­neoplastic Diseases of the Woo, S.-­B., Eisenbud, L., Kleiman, M., and Assael, N. (1987).
Head and Neck, AFIP Atlas of Nontumour Pathology. Series 1, Odontogenic keratocysts in the anterior maxilla: report of
Fascicle 11 (eds. B.M. Wenig, E.L.B. Childers, M.S. Richardson, two cases, one simulating a nasopalatine cyst. Oral Surgery,
et al.), 263–317. Washington, DC: ARP Press. Oral Medicine, and Oral Pathology 64: 463–465.
Wenig, B.M. (2017b). Pharynx. In: Non-­neoplastic Diseases of Wood, R.E., Nortje, C.J., Padayachee, A., and Grotepass,
the Head and Neck, AFIP Atlas of Nontumour Pathology. F. (1988). Radicular cysts of primary teeth mimicking
Series 1, Fascicle 11 (eds. B.M. Wenig, E.L.B. Childers, premolar dentigerous cysts: report of three cases. Journal
M.S. Richardson, et al.), 207–221. Washington, DC: of Dentistry for Children 55: 288–290.
ARP Press. Woolgar, J.A., Rippin, J.W., and Browne, R.M. (1987a). A
Wertheimer, F.W. (1966). A histologic comparison of apical comparative histological study of odontogenic keratocysts
cuticles, secondary cuticles and hyaline bodies. Journal of in basal cell naevus syndrome and control patients. Journal
Periodontology 37: 91–94. of Oral Pathology 16: 75–80.
Bibliography 355

Woolgar, J.A., Rippin, J.W., and Browne, R.M. (1987b). The features and immunohistochemical study of the Hedgehog
odontogenic keratocyst and its occurrence in the nevoid signaling pathway. Pathobiology 75: 171–176.
basal cell carcinoma syndrome. Oral Surgery, Oral Yamada, K., Tatemoto, Y., Okada, Y., and Mori, M. (1989).
Medicine, and Oral Pathology 64: 727–730. Immunostaining of involucrin in odontogenic epithelial
Woolgar, J.A., Rippen, J.W., and Browne, R.M. (1987c). A tumors and cysts. Oral Surgery, Oral Medicine, and Oral
comparative study of the clinical and histological features Pathology 67: 564–568.
of recurrent and non-­recurrent odontogenic keratocysts. Yamamoto, H. and Takagi, M. (1986). Clinicopathologic study
Journal of Oral Pathology 16: 124–128. of the postoperative maxillary cyst. Oral Surgery, Oral
Woolgar, J.A., Triantafyllou, A., Ferlito, A. et al. (2013). Medicine, and Oral Pathology 62: 544–548.
Intraosseous carcinoma of the jaws–a clinicopathologic Yamamoto, K., Matsusue, Y., Kurihara, M. et al. (2013). A
review. Part I: metastatic and salivary-­type carcinomas. keratocyst in the buccal mucosa with the features of
Head and Neck 35: 895–901. keratocystic odontogenic tumor. Open Dentistry Journal 7:
Worrall, S.F. (1992). Recurrent odontogenic keratocyst within 152–156.
the temporalis muscle. British Journal of Oral and Yang, X., Ow, A., Zhang, C.P. et al. (2012). Clinical analysis of
Maxillofacial Surgery 30: 59–62. 120 cases of intraoral lymphoepithelial cyst. Oral Surgery,
Wright, B.A., Wysocki, G.P., and Larder, T.C. (1983). Oral Medicine, Oral Pathology, Oral Radiology 113:
Odontogenic keratocysts presenting as periapical disease. 448–452.
Oral Surgery, Oral Medicine, and Oral Pathology 56: Ye, P., Wei, T., Gao, Y. et al. (2020). Primary intraosseous
425–429. squamous cell carcinoma arising from an odontogenic
Wright, J.M. (1979). Squamous odontogenic tumorlike keratocyst: case series and literature review. Medicina Oral,
proliferations in odontogenic cysts. Oral Surgery, Oral Patología Oral y Cirugía Bucal 26: e49–e55.
Medicine, and Oral Pathology 47: 354–358. Yeo, J.F., Rosnah, B.Z., Ti, L.S. et al. (2007).
Wright, J.M. (1981). The odontogenic keratocyst: Clinicopathological study of dentigerous cysts in
orthokeratinized variant. Oral Surgery, Oral Medicine, and Singapore and Malaysia. Malaysian Journal of Pathology
Oral Pathology 51: 609–618. 29: 41–47.
Wright, J.M. and Li, T.J. (2022). Odontogenic keratocyst. In: Yeom, H.G. and Yoon, J.H. (2020). Concomitant cemento-­
Head and Neck Tumours, Section 7.1.0.4 (WHO osseous dysplasia and aneurysmal bone cyst of the
Classification of Tumours Series), 5e, vol. 9. Lyon: IARC mandible: a rare case report with literature review. BMC
Press https://publications.iarc.fr/Book-­And-­Report-­Series/ Oral Health 20: 276.
Who-­Classification-­Of-­Tumours. Yeung, A.W.K., Tanaka, R., Khong, P.L. et al. (2018).
Wright, J.M. and Soluk ­Tekkeşin, M. (2022). Calcifying Frequency, location, and association with dental pathology
odontogenic cyst. In: Head and Neck Tumours, of mucous retention cysts in the maxillary sinus. A
Section 7.1.0.8 (WHO Classification of Tumours Series), radiographic study using cone beam computed
5e, vol. 9. Lyon: IARC Press https://publications.iarc.fr/ tomography (CBCT). Clinical Oral Investigations 22:
Book-­And-­Report-­Series/Who-­Classification-­Of-­Tumours. 1175–1183.
Wu, L., Cheng, J., Maruyama, S. et al. (2009). Yih, W.Y. and Krump, J.L. (2005). Odontogenic keratocyst in
Lymphoepithelial cyst of the parotid gland: its possible the nasopalatine duct associated with mural cartilaginous
histopathogenesis based on clinicopathologic analysis of metaplasia. Journal of Oral and Maxillofacial Surgery 63:
64 cases. Human Pathology 40: 683–692. 1382–1384.
Wysocki, G.P., Brannon, R.B., Gardner, D.G., and Sapp, Yosbimura, Y., Takada, K., Takeda, M. et al. (1970).
P. (1980). Histogenesis of the lateral periodontal cyst and Congenital dermoid cysts of the sublingual region. Report
the gingival cyst of the adult. Oral Surgery, Oral Medicine, of a case. Journal of Oral Surgery 28: 366–370.
and Oral Pathology 50: 327–334. Yoshida, M., Kumamoto, H., Ooya, K., and Mayanagi,
Xavier, G.M., Patist, A.L., Healy, C. et al. (2015). Activated H. (2001). Histopathological and immunohistochemical
WNT signaling in postnatal SOX2-­positive dental stem cells analysis of calcifying odontogenic cysts. Journal of Oral
can drive odontoma formation. Scientific Reports 5: 14479. Pathology and Medicine 30: 582–588.
Xiong, J., Gronthos, S., and Bartold, P.M. (2013). Role of the Yoshikawa, Y., Nakajima, T., Kaneshiro, S., and Sakaguchi,
epithelial cell rests of Malassez in the development, M. (1982). Effective treatment of the postoperative
maintenance and regeneration of periodontal ligament maxillary cyst by marsupialization. Journal of Oral and
tissues. Periodontology 2000 63: 217–233. Maxillofacial Surgery 40: 487–491.
Yagyuu, T., Kirita, T., Sasahira, T. et al. (2008). Recurrence of You, M.S., Kim, D.Y., and Ahn, K.M. (2017). Surgical
keratocystic odontogenic tumor: clinicopathological management of idiopathic bone cavity: case series of
356 Bibliography

consecutive 27 patients. Journal of the Korean Association Zhang, L.Z., Man, Q.W., Liu, J.Y. et al. (2018). Overexpression
of Oral and Maxillofacial Surgeons 43: 94–99. of Fra-­1, c-­Jun and c-­Fos in odontogenic keratocysts:
Yuen, H.W., Julian, C.Y., and Samuel, C.L. (2007). potential correlation with proliferative and anti-­apoptotic
Nasolabial cysts: clinical features, diagnosis, and activity. Histopathology 73: 933–942.
treatment. British Journal of Oral and Maxillofacial Zhang, M.F., Lin, S.X., Yi, C. et al. (2010). Keratocystoma of
Surgery 45: 293–297. the parotid gland:case report and immunohistochemical
Yukimori, A., Oikawa, Y., Morita, K.I. et al. (2017). Genetic investigation. Journal of Clinical Pathology 63: 758–760.
basis of calcifying cystic odontogenic tumors. PLoS One 12: Zhang, X., Wang, J., Ding, X. et al. (2014). Altered expression
e0180224. of podoplanin in keratocystic odontogenic tumours
Zadik, Y., Yitschaky, O., Neuman, T., and Nitzan, following decompression. Oncology Letters 7: 627–630.
D.W. (2011). On the self-­resolution nature of the buccal Zhao, Y.F., Jia, Y., Chen, X.M., and Zhang, W.F. (2004).
bifurcation cyst. Journal of Oral and Maxillofacial Surgery Clinical review of 580 ranulas. Oral Surgery, Oral Medicine,
69: e282–e284. Oral Pathology, Oral Radiology, and Endodontics 98:
Zecha, J.A., Mendes, R.A., Lindeboom, V.B., and van der 281–287.
Waal, I. (2010). Recurrence rate of keratocystic Zhao, Y.F., Wei, J.X., and Wang, S.P. (2002). Treatment of
odontogenic tumor after conservative surgical treatment odontogenic keratocysts: a follow-­up of 255 Chinese
without adjunctive therapies – a 35-­year single institution patients. Oral Surgery, Oral Medicine, Oral Pathology, Oral
experience. Oral Oncology 46: 740–742. Radiology, and Endodontics 94: 151–156.
Zedan, W., Robinson, P.A., Markham, A.F., and High, Zhu, L., Yang, J., and Zheng, J.W. (2014). Radiological and
A.S. (2001). Expression of the Sonic Hedgehog receptor clinical features of peripheral keratocystic odontogenic
‘PATCHED’ in basal cell carcinomas and odontogenic tumor. International Journal of Clinical and Experimental
keratocysts. Journal of Pathology 194: 473–477. Medicine 7: 300–306.
Zen, I., Soares, M., Sakuma, R. et al. (2019). Identification of Zillo Martini, M., Caroli Rocha, A., Lemos, C.A. Jr., and
oral cavity abnormalities in pre-­term and full-­term Abreu, A.F. (2010). Fibro-­osseous lesions associated with
newborns: a cross-­sectional and comparative study. simple bone cysts: three case reports and review of the
European Archives of Paediatric Dentistry 21: 581–586. literature. Minerva Stomatologica 59:
Zhang, L., Sun, Z.J., Chen, X.M., and Chen, Z. (2010). 671–676.
Immunohistochemical expression of SHH, PTC, SMO and Zuniga, J.R. (2000). Challenging the neuralgia-­inducing
GLI1 in glandular odontogenic cysts and dentigerous cysts. cavitational osteonecrosis concept. Journal of Oral and
Oral Diseases 16: 818–822. Maxillofacial Surgery 58: 1021–1028.
Zhang, L.L., Yang, R., Zhang, L. et al. (2010). Dentigerous cyst: Zurawel, R.H., Allen, C., Wechsler-­Reya, R. et al. (2000).
a retrospective clinicopathological analysis of 2082 Evidence that haploinsufficiency of Ptch leads to
dentigerous cysts in British Columbia, Canada. International medulloblastoma in mice. Genes, Chromosomes and
Journal of Oral and Maxillofacial Surgery 39: 878–882. Cancer 28: 77–81.
357

Index

a pseudocyst of maxillary sinus  254 orthokeratinised odontogenic


abscess theory, radicular cyst  31–33 radicular cyst  21–22, 24 cyst  202–203
adjunctive therapy, keratocyst retention cyst of maxillary paradental cyst  51–53
surgery  137 sinus  254 pseudocyst of maxillary sinus  254
adult gingival cyst  155–160 simple bone cyst  272–273, 276 radicular cyst  22–23
clinical features  156–157 Stafne bone cavity  281–282 retention cyst of maxillary sinus  254
clinical presentation  157 surgical ciliated cyst  263–264 simple bone cyst  272–274
diagnosis  158–160 thyroglossal duct cyst  308 Stafne bone cavity  282
differential diagnosis  159–160 ameloblastoma surgical ciliated cyst  264
histopathology  159 calcifying odontogenic cyst thyroglossal duct cyst  306
microkeratocyst  160 differentiation  191, 197, 199 anatomy, incisive canal  214–218
pathogenesis  158–159 dentigerous cyst relationship  76–77 aneurysmal bone cyst  287
radiological features  158 glandular odontogenic cyst antral cyst of the maxillary
treatment  160 differentiation  172–173 sinus  253–255
AE1/AE3 see pan‐cytokeratin keratocyst  128–129, 132 apical cyst see radicular cyst
age distribution anatomical distribution apical granuloma see periapical
botryoid odontogenic cyst  150–151 botryoid odontogenic cyst  151 granuloma
branchial cleft cyst  300 branchial cleft cyst  300 aspiration biopsy, keratocyst  129–130
calcifying odontogenic cyst  186 calcifying odontogenic
dentigerous cyst  63–65 cyst  186–187 b
dermoid cyst  292 dentigerous cyst  66–67 B lymphocytes, periapical
epidermoid cyst  292 dermoid cyst  292–293 periodontitis  28
eruption cyst  83–84 epidermoid cyst  292–293 bacterial factors, radicular cyst  26,
gingival cysts  156, 161, 163 eruption cyst  84 27, 29, 30
glandular odontogenic cyst  165–166 gingival cysts  156–157, 161, 162 basal cell nevus syndrome see naevoid
heterotopic gastrointestinal cyst  297 glandular odontogenic cyst  165–166 basal cell carcinoma syndrome
inflammatory collateral cysts  50, 52 heterotopic gastrointestinal cyst  bay cyst see pocket cyst
intraoral lymphoepithelial cyst  256 297 Bcl‐2  115
keratocyst  92–93 inflammatory collateral cysts  50, biomarkers
lateral periodontal cyst  141 52–53 dentigerous cyst  80–81
mandibular buccal bifurcation intraoral lymphoepithelial cyst  glandular odontogenic cyst 
cyst  51, 52 256 178–180
mucoceles  239–240, 252–253 keratocyst  94–95 keratocyst  130–132
nasolabial cyst  230–231 lateral periodontal cyst  142 biopsy, keratocyst  129–130
nasopalatine duct cyst  219 mandibular buccal bifurcation Blandin–Nuhn glands, mucoceles  242
orthokeratinised odontogenic cyst  cyst  50, 52–53 Bohn’s nodules  160
202 mucoceles  239–241 bone marrow, focal osteoporotic
paradental cyst  51, 52 nasopalatine duct cyst  219 defects  285–287

Shear’s Cysts of the Oral and Maxillofacial Regions, Fifth Edition. Paul M. Speight.
© 2022 John Wiley & Sons Ltd. Published 2022 by John Wiley & Sons Ltd.
358 Index

bone resorption naevoid basal cell  98–100, 109–113, epidermoid cyst  291–293
dentigerous cyst  74 115, 124–125, 204 Epstein Pearls  163
radicular cyst  23, 35–36 radicular cyst transitions  45–46 eruption cyst  83–84
Stafne bone cavity  284 Carnoy’s solution  137 focal osteoporotic bone marrow
botryoid odontogenic cyst  147, cartilage defects  285–286
150–154 keratocyst  122–123 gingival cysts  156–157, 161, 163
clinical features  150–151 nasopalatine duct cyst  229 glandular odontogenic cyst  165–166
clinical presentation  151 β‐catenin  18, 191–193 heterotopic gastrointestinal cyst 
diagnosis  152–154 cavitational osteonecrosis  286–287 296–298
differential diagnosis  154 cell proliferation inflammatory collateral cysts  49–53
pathogenesis  152 hedgehog signalling  115 intraoral lymphoepithelial cyst  256
radiological features  151–152 keratocyst  107–109, 114–115 keratocyst  89–95
recurrence  152 radicular cyst  31–33, 35 lateral periodontal cyst  141–142
treatment  154 cell rests of the dental lamina mucoceles  238–241, 252–253
branchial cleft anomalies  300 gingival cysts  28–29 nasolabial cyst  230–233
branchial cleft cyst  290, 300–305 glandular odontogenic cyst  170 nasopalatine duct cyst  218–221
bronchogenic cyst  298–300 keratocyst  107 orthokeratinised odontogenic
lateral periodontal cyst  145–146 cyst  202–204
c cell rests of Malassez pseudocyst of maxillary
calcifying cystic odontogenic tumour lateral periodontal cyst  144–145 sinus  253–254
(CCOT) see calcifying periapical periodontitis  27 radicular cyst  23–25
odontogenic cyst radicular cyst  28–30 retention cyst of maxillary
calcifying ghost cell tumours  cellular changes, radicular cyst  38–40 sinus  253–254
199–200 cemento‐osseous dysplasia  276–279 simple bone cyst  272–275
calcifying odontogenic cyst  182–200 central mucoepidermoid Stafne bone cavity  281–282
β‐catenin  191–193 carcinoma  176–178 surgical ciliated cyst  263–265
classification  182–185 central necrosis theory, radicular thyroglossal duct cyst  305–307
clinical features  185–188 cyst  31–33 clinical presentation
clinical presentation  187–188 chemokines  27–30 adult gingival cyst  157
CTNNB1 gene  191–193 cholesterol deposition botryoid odontogenic cyst  151
dentinoid formation  195, 199–200 dentigerous cyst  78 branchial cleft cyst  301
differential diagnosis  190, 199–200 radicular cyst  42–44 calcifying odontogenic
ghost cells  183–184, 191–200 ciliated cells, radicular cyst  38–40 cyst  187–188
histopathology  193–199 CK see cytokeratins dentigerous cyst  67
malignant transformation  199–200 classification  2–5 dermoid cyst  293
pathogenesis  190–193 calcifying odontogenic cyst  epidermoid cyst  293
peripheral/extraosseous 182–185 Epstein Pearls  163
lesions  188, 199 developmental cyst  289–291 eruption cyst  83–84
radiological features  188–190 developmental cyst of foregut gingival cysts  157, 161, 163
recurrence  200 origin  295–296 glandular odontogenic cyst  166
terminology  182–185 inflammatory collateral cysts  47–49 heterotopic gastrointestinal
treatment  200 keratocyst  88–89 cyst  298
Caldwell‐Luc operation  255, 262, 263, mucoceles  238 inflammatory collateral cysts 
267–269, 281 simple bone cyst  271–272 53–54
Calretinin  18 clinical features intraoral lymphoepithelial cyst  256
carcinoma adult gingival cyst  156–157 keratocyst  95–98
central mucoepidermoid  176–178 botryoid odontogenic cyst  150–151 lateral periodontal cyst  142
cystic metastatic  303–305 branchial cleft cyst  300–301 mucoceles  241–244, 253
dentigerous cyst transitions  81–82 bronchogenic cyst  299 nasolabial cyst  231–233
ghost cells  195, 199–200 calcifying odontogenic cyst  185–188 nasopalatine duct cyst  220–221
keratocyst transformation  132–133 dentigerous cyst  63–67 orthokeratinised odontogenic
mucoepidermoid  170, 172, 176–178 dermoid cyst  291–293 cyst  203–204
Index 359

pseudocyst of maxillary sinus  254 classification  3–4 immunohistochemistry  80–81


radicular cyst  23–25 diagnosis  14–19 inflammatory  75–76, 78
retention cyst of maxillary histopathology  8–9, 16–17 initiation  72–73
sinus  253–254 immunohistochemistry  17–18 keratinisation  207
simple bone cyst  274–275 molecular pathology  17, 19 malignant changes  81–82
Stafne bone cavity  282 radiology  7–8, 15–16 metaplastic changes  80
surgical ciliated cyst  264–265 cyst–tumour interface  13–14 orthokeratinised odontogenic cyst
thyroglossal duct cyst  306–307 cytokeratins (CK) relationship  206–207
collateral cyst see inflammatory dentigerous cyst  80–81 paradental cyst relationship  60–61
collateral cyst diagnostic applications, general  18 pathogenesis  11, 72–77, 207
compression, dentigerous cyst  82 glandular odontogenic cyst  178 PTCH gene  74–75, 111
computed tomography (CT) inflammatory collateral cysts  59 radiological features  67–72
calcifying odontogenic keratocyst  131 sex distribution  65–66
cyst  189, 190 radicular cyst  40 site distributions  66–67
dentigerous cyst  67–69 cytokine‐mediated remodelling  11–12 treatment  82
glandular odontogenic cyst  169 cytokines  11–12, 27–30, 35–36 dentinogenic ghost cell tumours  195,
incisive canal  214–216 cytology, keratocyst  129 199–200
inflammatory collateral cysts  56 cytotoxic/suppressor cells  27–28 dentinoid formation  195, 199–200
keratocyst  103–104 dermoid cyst  291–295
nasolabial cyst  233 d clinical features  291–293
nasopalatine duct cyst  222–223 DALT see duct‐associated clinical presentation  293
radicular cyst  26 lymphoid tissue definition  289, 291
simple bone cyst  276–278 deciduous teeth histopathology  294–295
Stafne bone cavity  283 dentigerous cyst  65, 75, 76, 84, 86 pathogenesis  294
surgical ciliated cyst  265–266 eruption cyst  84, 86 radiological features  293–294
cone beam computerised gingival cysts  156 treatment  295
tomography (CBCT) radicular cyst  22–24 developmental cysts  288–308
dentigerous cyst  69 decompression, keratocyst  137 branchial cleft  300–305
incisive canal  214–216 dental lamina bronchogenic  298–300
inflammatory collateral cysts  56 gingival cysts  28–29 classification  4–5, 289–291
keratocyst  103–104 glandular odontogenic cyst  170 dermoid  291–295
nasopalatine duct cyst  222–223 keratocyst  107, 109 epidermoid  291–295
radicular cyst  26 lateral periodontal cyst  145–146 of foregut origin  295–300
simple bone cyst  276–277 dentigerous cyst  62–82 heterotopic gastrointestinal  296–298
congenital cyst see developmental cyst age distribution  63–65 neonates, gingival  160–163
connective tissue degradation ameloblastoma relationship  76–77 sources of epithelial lining  11
dentigerous cyst  74 biomarkers  80–81 terminology  289–291
radicular cyst  35–36 bone resorption  74 thyroglossal duct cyst  305–308
cryotherapy, keratocyst  137 clinical features  63–67 see also specific forms of cyst
CT see computed tomography clinical presentation  67 diagnosis
CTNNB1 gene  14, 19, 191–193 diagnosis  67–72, 78–81 adult gingival cyst  158–160
curettage differential diagnosis  70–72 botryoid odontogenic cyst  151–154
botryoid odontogenic cyst  154 vs. dilated follicles  71–72 dentigerous cyst  67–72, 78–81
glandular odontogenic cyst  181 enamel hypoplasia  72 Epstein pearls  163
keratocyst  136 epithelial sources  72–73 eruption cyst  84
lateral periodontal cyst  150 eruption cyst  62, 83–86 glandular odontogenic cyst  166–168,
radicular cyst  46 extrafollicular  76 170–180
simple bone cyst  281 formation  73 histopathology  8–9, 16–17
cyclosporine, eruption cyst  85 frequency  63 infant gingival cyst  161–163
cystic hygroma  244, 304 growth and enlargement  73–74 inflammatory collateral cysts  54–58
cystic metastatic carcinoma  303–305 histopathology  78–80 jaw cyst  14–19
cyst of the jaw hydrostatic pressure  73 keratocyst  100–106
360 Index

diagnosis (cont’d) endotoxins  27, 29, 30 pathogenesis  85


lateral periodontal cyst  142–150 enlargement radiological features  84
median palatal cyst  223–225 dentigerous cyst  73–74 treatment  85
naevoid basal cell carcinoma keratocyst  114–117 excision
syndrome  98–100 lateral periodontal cyst  146 mucosa, keratocyst  137
nasolabial cyst  231–236 radicular cyst  26, 33–36 nasolabial cyst  236
nasopalatine duct cyst  221–223, enteric duplication cyst  296–298 experimental studies, hyaline
226–229 enucleation bodies  41–42
orthokeratinised odontogenic botryoid odontogenic cyst  154 extrafollicular dentigerous cyst  76
cyst  204–205, 207–211 calcifying odontogenic cyst  200 extraosseous odontogenic keratocyst 
radicular cyst  23–26, 36–45 glandular odontogenic cyst  181 96–97
radiological principles  7–8, 15–16 keratocyst  136
surgical ciliated cyst  264–268 lateral periodontal cyst  150 f
thyroglossal duct cyst  306–307 nasolabial cyst  236 fatty degenerative osteonecrosis in the
differential diagnosis nasopalatine duct cyst  229 jawbone (FDOJ)  286–287
adult gingival cyst  159–160 orthokeratinised odontogenic FDOJ see fatty degenerative
botryoid odontogenic cyst  154 cyst  213 osteonecrosis in the jawbone
branchial cleft cyst  303–305 surgical ciliated cyst  268–269 fine needle aspiration
calcifying odontogenic cyst  190, epidemiology cystic metastatic carcinoma 
199–200 radicular cyst  22 304–305
dentigerous cyst  70–72 see also age distribution; frequency; keratocyst  129–130
focal osteoporotic bone marrow sex distribution floor of mouth see mouth floor
defects  286 epidermal cyst  290 florid osseous dysplasia  276–279
glandular odontogenic cyst  168, epidermoid cyst  290–295 focal osteoporotic bone marrow
175–178 clinical features  291–293 defects  285–287
immunohistochemistry  17–18 clinical presentation  293 follicular cyst see dentigerous cyst
inflammatory collateral cysts  56–58 definition  289–291 foramen caecum  305
keratocyst  104–106, 125–126 histopathology  294–295 foregut
lateral periodontal cyst  143–144, pathogenesis  294 developmental cyst originating
149 radiological features  293–294 from  295–300
mandibular buccal bifurcation treatment  295 bronchogenic  298–300
cyst  57–58 epithelia heterotopic gastrointestinal  296–298
median palatal cyst  223–225 bronchogenic cyst  298–300 formation
molecular pathology  17, 19 dentigerous cyst  72 dentigerous cyst  73
mucoceles  249–250 glandular odontogenic lateral periodontal cyst  146
nasopalatine duct cyst  223–225 cyst  170–172 radicular cyst  26, 31–33
orthokeratinised odontogenic heterotopic gastrointestinal frequency  4, 5
cyst  205, 211–212 cyst  296–297 botryoid odontogenic cyst  150
paradental cyst  56–58 hyaline bodies  40–42 branchial cleft cyst  300
Stafne bone cavity  284 inflammatory collateral cysts  58 calcifying odontogenic cyst 
surgical ciliated cyst  266–267 keratocyst  107–109, 114–115 185–186
thyroglossal duct cyst  307–308 lateral periodontal cyst  144–146 dentigerous cyst  63
duct‐associated lymphoid tissue mucoceles  240–241 dermoid cyst  292
(DALT)  257 radicular cyst  31–33, 35 epidermoid cyst  292
dysgenetic disease of the parotid sources of  10–11 eruption cyst  84
glands  259–260 epithelial dysplasia  120 gingival cysts  156, 161, 163
dysplasia, epithelial  120 Epstein Pearls  162–163 glandular odontogenic cyst  165
eruption cyst  62, 83–86 heterotopic gastrointestinal
e clinical features  83–84 cyst  296
enamel clinical presentation  83–84 inflammatory collateral cysts  49–52
hypoplasia  72–73, 144, 207 diagnosis  84 intraoral lymphoepithelial cyst  256
projections  60 histopathology  85 keratocyst  89–91
Index 361

lateral periodontal cyst  141 clinical presentation  166 focal osteoporotic bone marrow
mandibular buccal bifurcation diagnosis  166–168, 170–180 defects  286
cyst  49–52 differential diagnosis  168, 175–178 gingival cysts  159, 162, 163
mucoceles  238–239, 252 histopathology  170–180 glandular odontogenic cyst 
nasolabial cyst  230 immunohistochemistry  178–180 170–180
nasopalatine duct cyst  218–219 molecular studies  178–180 inflammatory collateral cysts  61
neonatal cyst  160–163 mucoepidermoid carcinoma intraoral lymphoepithelial cyst 
orthokeratinised odontogenic similarity  170, 172, 176–178 257–258
cyst  202 pathogenesis  169–170 jaw cyst  8–9, 16–17
paradental cyst  49–52 radiological features  167–168 keratocyst  117–133
pseudocyst of maxillary sinus  254 recurrence  169, 181 lateral periodontal cyst  146–150
radicular cyst  20–24 treatment  181 mucoceles  244–250, 253
retention cyst of maxillary sinus  254 Gli1 gene  110, 170 mucous extravasation cyst  244–248
simple bone cyst  272–273 glycogen, lateral periodontal cyst  147 mucous retention cyst  248–249
Stafne bone cavity  281 goblet cells see mucous cells nasolabial cyst  234–236
surgical ciliated cyst  263–264 Gorlin–Golz syndrome see naevoid nasopalatine duct cyst  226–229
thyroglossal duct cyst  308 basal cell carcinoma syndrome orthokeratinised odontogenic
growth cyst  207–213
g dentigerous cyst  73–74 pseudocyst of maxillary sinus  255
gastric epithelium in oral cyst  296–298 keratocyst  114–117 radicular cyst  36–45
ghost cells lateral periodontal cyst  146 retention cyst of maxillary sinus  255
calcifying odontogenic cyst  radicular cyst  26, 33–36 simple bone cyst  273, 280–289
183–184, 191–200 growth factors  27–28, 30, 35 Stafne bone cavity  285
carcinoma  195, 199–200 gubernacular canal  107 superficial mucoceles  247–248
β‐catenin CTNNB1 gene  191–193 surgical ciliated cyst  268
histopathology  193–199 h thyroglossal duct cyst  307
gingival cysts  155–163 haematogenous mechanisms, hyaline HIV‐associated salivary gland
clinical features  156–157, 161, 163 bodies  41–42 disease  259
clinical presentation  157, 161, 163 haemorrhagic bone cyst see simple hobnail cells, glandular odontogenic
histopathology  159, 162, 163 bone cyst cyst  171, 173
pathogenesis  158–159, 161–163 haemosiderin  42–43, 280 HPV see human papilloma virus
treatment  160, 162, 163 hedgehog (HH) signalling human papilloma virus (HPV) 
gingival cyst of adults  155–160 pathway  12 303–305
clinical features  156–157 cell proliferation  115 hyaline (Rushton) bodies
clinical presentation  157 dentigerous cyst  74 dentigerous cyst  80
diagnosis  158–160 glandular odontogenic cyst  170 keratocyst  122
differential diagnosis  159–160 keratocyst  99–100, 112–113, 115 lateral periodontal cyst  147
histopathology  159 naevoid basal cell carcinoma nasopalatine duct cyst  229
microkeratocyst  160 syndrome  115 radicular cyst  40–42
pathogenesis  158–159 signalling pathway  99–100 hydrostatic pressure  12–13
radiological features  158 heterotopic gastrointestinal cyst  dentigerous cyst  73
treatment  160 296–298 keratocyst  115–116
gingival cyst of infants  160–163 histopathology radicular cyst  33–35
clinical features  161, 163 branchial cleft cyst  301–305
clinical presentation  161, 163 bronchogenic cyst  299 i
Epstein Pearls  162–163 calcifying odontogenic cyst  immune mechanisms, radicular
histopathology  162, 163 193–199 cyst  27–33, 35
pathogenesis  161–163 dentigerous cyst  78–80 immunohistochemistry
treatment  162, 163 dermoid cyst  294–295 dentigerous cyst  80–81
glands of Blandin–Nuhn  242 epidermoid cyst  294–295 glandular odontogenic cyst  178–180
glandular odontogenic cyst  164–181 Epstein Pearls  163 jaw cyst  17–18
clinical features  165–166 eruption cyst  85 keratocyst  110–111, 130–132
362 Index

immunosuppression, eruption cyst  85 molecular pathology  17, 19 recurrence  133–138


impacted teeth, orthokeratinised radiology  7–8, 15–16 sex distribution  93–94
odontogenic cyst  207 jaws site distribution  94–95
incisive canal  214–218 classifications  3–4 soft tissue  97–98
incisive canal cyst see nasopalatine pseudocyst  3–4, 270–287 solid  126–129
duct cyst aneurysmal bone cyst  287 terminology, history of  88–89
incisive foramen  217–218, 222 focal osteoporotic bone marrow treatment  136–138
incisive papilla  217, 220 defects  285–287 see also orthokeratinised
incisive papilla cyst  220 simple bone cyst  271–281 odontogenic cyst
infants Stafne bone cavity  281–285 Ki‐67  114–115, 179, 206
Epstein pearls  162–163 Klestadt cyst see nasolabial cyst
gingival cyst  160–162 k
inflammation KCOT (keratocystic odontogenic l
hyaline bodies  41 tumour) see keratocyst lateral periodontal cyst  141–154
keratocyst  120 keratin expression patterns  17 clinical features  141–142
nasopalatine duct cyst  229 keratocyst  122–124 clinical presentation  142
paradental cyst  58–59 keratinisation differential diagnosis  143–144, 149
periapical periodontitis  26–28 dentigerous cyst  207 histopathology  146–150
Stafne bone cavity  284–285 orthokeratinised odontogenic multicystic variant see botryoid
inflammatory collateral cysts  47–61 cyst  207–210 odontogenic cyst
age distribution  50, 52 radicular cyst  38 pathogenesis  144–146
classification  47–49 keratoameloblastoma  128–129, 132 radiological features  142–144
clinical features  49–53 keratocystoma  129, 260–261 treatment  150
diagnosis  54–58 keratocyst (odontogenic lichen planus  241–242
differential diagnosis  56–58 keratocyst)  87–139 lingual cyst
frequency  49–52 age distribution  92–93 foregut originating  297, 299
histopathology  61 aspiration biopsy  129–130 glands of Blandin–Nuhn  242
initiation  59 as a benign neoplasm  111–114 lymphoepithelial  256–258
radiological features  54–58 biomarkers  130–132 thyroglossal duct  305
site distribution  50, 52–53 biopsy  129–130 lingual mandibular bone defect see
treatment  61 classification  88–89 Stafne bone cavity
inflammatory dental cyst see clinical features  89–95 lipofuscin, radicular cyst  43–44
radicular cyst clinical presentation  95–98 lipopolysaccharide (LPS)  29,
inflammatory dentigerous cytology  129–130 30, 35–36
cyst  75–76, 78 diagnosis  100–106 loss of heterozygosity (LOH)  12–14
inflammatory paradental cyst see differential diagnosis  104–106, dentigerous cyst  74, 75, 111
paradental cyst 125–126 glandular odontogenic cyst  170
initiation epithelial sources  107–109 keratocyst  109–114
dentigerous cyst  72–73 frequency  89–91 orthokeratinised odontogenic cyst  206
inflammatory collateral cysts  59 growth and enlargement  114–117 lymphoepithelial cyst  255–259
lateral periodontal cyst  144–146 histopathology  117–133 branchial cleft  300–305
radicular cyst  26, 28–31 immunohistochemistry  110–111, intraoral  256–258
interleukins  29, 31 130–132 parotid gland  258–259
intraoral lymphoepithelial malignant transformation  132–133
cyst  256–258 naevoid basal cell carcinoma m
syndrome  98–100, 109–113, magnetic resonance imaging (MRI)
j 115, 124–125 dermoid cyst  293–294
Jacobson’s (vomeronasal) organ  217 orthokeratinised  123–124, 211–212 epidermoid cyst  293–294
jaw cyst pathogenesis  107–117, 207 keratocyst  104
diagnosis  14–19 peripheral  96–97 nasolabial cyst  233
histopathology  8–9, 16–17 PTCH gene  99–100, 108–113, 115 Stafne bone cavity  283
immunohistochemistry  17–18 radiological features  100–106 surgical ciliated cyst  265–266
Index 363

Malassez (rest cells of) see rest cells of classifications  4 multicystic lateral periodontal
Malassez sources of epithelial lining  11 cyst  145, 149–154
malignant transformation see also salivary duct cyst; salivary multiple simple bone cyst  276, 279
calcifying odontogenic cyst  gland cyst myofibroblasts, keratocyst  116–117
199–200 mitotic figures, keratocyst  118–119 myxoglobulosis  246, 247
dentigerous cyst  81–82 MMPs see matrix metalloproteinases
keratocyst  132–133 molecular pathology n
radicular cyst  45–46 glandular odontogenic cyst  naevoid basal cell carcinoma syndrome
MAML2 gene  14, 17, 19, 179–180 178–180 (NBCCS)  98–100, 109–113,
mandibular buccal bifurcation cyst jaw cyst  17, 19 115, 124–125, 204
clinical features  49–53 bronchogenic cyst  299 nasoalveolar cyst see nasolabial cyst
clinical presentation  53–54 dermoid/epidermoid cyst  292–293 nasolabial cyst  230–236
diagnosis  54 heterotopic gastrointestinal clinical features  230–233
differential diagnosis  56–58 cyst  297 clinical presentation  231–233
histopathology  61 lymphoepithelial cyst  256–258 histopathology  234–236
pathogenesis  60 mucoceles  242–244 pathogenesis  234
treatment  61 thyroglossal duct cyst  306 radiological features  233–234
mandibular infected buccal cyst see MRI see magnetic resonance imaging treatment  236
mandibular buccal mucoceles  237–253 nasolacrimal cyst  234
bifurcation cyst classification  238 nasopalatine duct cyst  214–229
marginal wisdom tooth cyst  48 clinical features  238–241, 252–253 clinical features  218–221
marsupialization clinical presentation  241–244, 253 clinical presentation  220–221
calcifying odontogenic cyst  200 differential diagnosis  249–250 diagnosis  221–223, 226–229
dentigerous cyst  82 extravasation cyst  238–241, differential diagnosis  223–225
keratocyst  136 243–248 histopathology  226–229
mandibular buccal glands of Blandin–Nuhn  242 incisive canal anatomy  214–218
bifurcation cyst  61 histopathology  244–250 median palatal cyst  223–225
nasolabial cyst  236 maxillary sinus  252–253 pathogenesis  225–226
surgical ciliated cyst  268–269 pathogenesis  244 radiological features  221–225
Maspin  18 plunging ranula  243–244 recurrence  229
matrix metalloproteinases (MMPs)  ranula  242–244, 250 treatment  229
35–36, 116, 192 retention cyst  238–241, 248–249 nasopharyngeal cyst  290, 308
maxillary sinus cyst  4, 252–255 superficial  241–242, 247–248 NBCCS see naevoid basal cell
mucoceles  252–253 terminology  238 carcinoma syndrome
pseudocyst  252–255 treatment  250–251 neck
retention cyst  252–255 mucoepidermoid carcinoma  170, developmental cyst  4–5
surgical ciliated cyst see surgical 172, 176–178 sources of epithelial lining  10–11
ciliated cyst mucosal excision, keratocyst  137 neonates
median palatal cyst  223–225 mucous cells Epstein pearls  162–163
melanin, calcifying odontogenic bronchogenic cyst  299 gingival cyst  160–162
cyst  199 dentigerous cyst  80 neoplastic changes
merging epithelial strands theory, glandular odontogenic cyst  definition  111–112
radicular cyst  31–33 171–172, 174 dentigerous cyst  76–77
metaplastic changes nasolabial cyst  235 keratocyst as  111–114
dentigerous cyst  80 nasopalatine duct cyst  225 neuralgia‐inducing cavitational
radicular cyst  38–40 radicular cyst  38–40 osteonecrosis (NICO) 
microcyst, glandular odontogenic thyroglossal duct cyst  306–307 286–287
cyst  171–174 mucous extravasation cyst  238–241, neurovascular bundles  229
microkeratocyst  107, 160 243–248 non‐odontogenic cyst
Midpalatal raphe cyst (Epstein mucous retention cyst  238–241, classification  3–4
Pearls)  162–163 248–249 sources of epithelial lining  10
minor mucous glands multicystic keratocyst  129 see also individual types. . .
364 Index

non‐surgical treatment pan‐cytokeratin (AE1/AE3)  130 Stafne bone cavity  284–285


eruption cyst  85 paradental cyst surgical ciliated cyst  267–268
inflammatory collateral cyst  61 clinical features  49–53 PCNA see proliferating cell
radicular cyst  46 clinical presentation  53–54 nuclear antigen
nutritional deficiency theory, radicular dentigerous cyst relationship  60–61 pemphigoid  241–242
cyst  31–33 diagnosis  54 periapical cyst see radicular cyst
differential diagnosis  56–58 periapical granuloma
o
histopathology  61 periapical periodontitis  26–27
OAF see osteoclast activating factor
pathogenesis  58–60 radicular cyst  28–30
odontogenic cyst
treatment  61 periapical periodontitis,
classification  2–4
parotid gland pathogenesis  27–28
frequency  4, 5
HIV‐associated salivary gland pericoronitis  7, 47–53, 57–59
immunohistochemistry  17–18
disease  259 periodic acid–Schiff (PAS) staining 
pathogenesis  10–13
keratocystoma  129, 260–261 39, 43–44, 147
PTCH gene  17, 19
lymphoepithelial cyst  258–259, peripheral (extraosseous)
sources of epithelial lining  10
300–305 odontogenic cyst
see also individual types. . .
polycystic disease  259–260 calcifying  187, 199
odontogenic keratocyst see keratocyst
sclerosing polycystic adenoma  260 keratocyst  96–97
oncogenes  12, 17, 19
PAS see periodic acid–Schiff (PAS) see also gingival cyst
’onion‐skin’ pattern  208–209
staining peripheral ostectomy  137
OPG see osteoprotogerin
pathogenesis  6–13 phases of cyst formation  10
oral epithelia, islands, keratocyst  107
botryoid odontogenic cyst  152 plunging ranula  243–244, 250
oral tonsils  255–257
calcifying odontogenic cyst  pocket cyst  24, 44–45, 59
orthokeratinised odontogenic
190–193 podoplanin  117
cyst  123–124, 201–213
dentigerous cyst  72–77, 207 polymorphonuclear leukocytes
clinical features  202–204
dermoid cyst  294 (PMNs)  27, 31–33
clinical presentation  203–204
epidermoid cyst  294 postoperative maxillary cyst see
dentigerous cyst
Epstein Pearls  163 surgical ciliated cyst
relationship  206–207
eruption cyst  85 prevalence
diagnosis  204–205, 207–212
focal osteoporotic bone marrow inflammatory collateral cysts  49–52
differential diagnosis  205, 211–212
defects  286 radicular cyst  20–23
histopathology  207–213
gingival cysts  158–159, 161–163 see also clinical features
odontogenic keratocyst relationship 
glandular odontogenic cyst  169–170 proliferating cell nuclear antigen
211–212
inflammatory collateral cysts  58–60 (PCNA)  114–115, 179
pathogenesis  205–207
intraoral lymphoepithelial cyst  prostaglandins  28, 30, 36
radiological features  204–205
256–257 pseudocyst
recurrence  213
keratocyst  107–117, 207 of the jaw  3–4, 270–287
treatment  213
lateral periodontal cyst  144–146 aneurysmal bone cyst  287
with verrucous proliferation  212–213
mandibular buccal bifurcation focal osteoporotic bone marrow
osmolality
cyst  60 defects  285–287
dentigerous cyst  73
mucoceles  244, 253 simple bone cyst  271–281
keratocyst  116
nasolabial cyst  234 Stafne bone cavity  281–285
radicular cyst  33–34
nasopalatine duct cyst  225–226 maxillary sinus  252–255
osteoclast activating factor (OAF)  29
orthokeratinised odontogenic PTCH gene  12–14, 17, 19
osteonecrosis  286–287
cyst  205–207 dentigerous cyst  74–75, 111
osteoporotic focal bone marrow
paradental cyst  58–60 glandular odontogenic cyst  170
defects  285–287
periapical periodontitis  27–28 keratocyst  99–100, 108–113, 115
osteoprotogerin (OPG)  35–36
pseudocyst of maxillary sinus  naevoid basal cell carcinoma
p 254–255 syndrome  99–100, 109–113,
p16  18 radicular cyst  26–36 115
p53  115, 179 retention cyst of maxillary sinus  orthokeratinised odontogenic
palatine papilla see incisive papilla 254–255 cyst  206
Index 365

r orthokeratinised odontogenic keratocystoma  260–261


radicular cyst  20–46 cyst  204–205 lymphoepithelial cyst  255–259
age distribution  21–22, 24 pseudocyst of maxillary sinus  254 intraoral  256–258
bacterial factors  26, 27, 29, 30 radicular cyst  25–26 of the parotids  258–259
biological factors  29–30 retention cyst of maxillary sinus  254 maxillary sinus  252–255
chemokines  27–30 simple bone cyst  273, 275–278 polycystic disease of the
cholesterol deposition  42–44 Stafne bone cavity  282–284 parotids  259–260
clinical features  23–25 surgical ciliated cyst  265–267 sclerosing polycystic adenoma  260
clinical presentation  23–25 RANKL see receptor activator of salivary gland cyst
cytokines  27–30, 35–36 nuclear factor kappa B ligand classifications  4
diagnosis  23–26, 36–45 ranula  242–244, 250 mucoceles  237–251
epithelial proliferation  31–33, 35 Rathke’s cleft cyst  308 sources of epithelial lining  11
formation  26, 31–33 receptor activator of nuclear factor satellite cyst, keratocyst  120
frequency  20–24 kappa B ligand (RANKL), sclerosing polycystic adenoma  260
growth and enlargement  26, 33–36 radicular cyst  35–36 sebaceous cyst, definition  290
histopathology  36–45 recurrence sebaceous glands, orthokeratinised
hyaline bodies  40–42 botryoid odontogenic cyst  152 odontogenic cyst  211
hydrostatic pressure  33–35 calcifying odontogenic cyst  sex distribution
immune mechanisms  27–33, 35 199–200 branchial cleft cyst  300
initiation  26, 28–31 glandular odontogenic cyst  169, 181 calcifying odontogenic cyst  186
malignant changes  45–46 keratocyst  133–138 dentigerous cyst  64–66
metaplastic changes  38–40 nasopalatine duct cyst  229 dermoid cyst  292
pathogenesis  11, 16, 26–36 orthokeratinised odontogenic epidermoid cyst  292
periapical granuloma  28–30 cyst  213 eruption cyst  84
periapical periodontitis  27–28 simple bone cyst  281 gingival cysts  156, 161, 163
pocket cyst  44–45 resection, keratocyst  137 glandular odontogenic
prostaglandins  28, 30, 36 residual radicular cyst  24–25, 44 cyst  165–166
radiological features  25–26 resorption of bone heterotopic gastrointestinal
residual  24–25, 44 dentigerous cyst  74 cyst  297
resorption of bone  23, 35–36 radicular cyst  23, 35–36 inflammatory collateral cysts  50, 52
sites  22–23 resorption of roots, dentigerous intraoral lymphoepithelial cyst 
treatment  46 cyst  69 256
radiological features respiratory epithelia, bronchogenic keratocyst  93–94
botryoid odontogenic cyst  151–152 cyst  298–300 lateral periodontal cyst  141–142
calcifying odontogenic rest cells of the dental lamina mandibular buccal bifurcation
cyst  188–190 gingival cysts  28–29 cyst  50, 52
dentigerous cyst  67–72 glandular odontogenic cyst  170 mucoceles  239–240, 252–253
dermoid cyst  293–294 keratocyst  107 nasolabial cyst  231
epidermoid cyst  293–294 lateral periodontal cyst  145–146 nasopalatine duct cyst  219
eruption cyst  84 rest cells of Malassez orthokeratinised odontogenic
focal osteoporotic bone marrow lateral periodontal cyst  144–145 cyst  202
defects  286 periapical periodontitis  27 paradental cyst  50, 52
glandular odontogenic radicular cyst  28–30 pseudocyst of maxillary sinus  254
cyst  167–168 retention cyst radicular cyst  22, 24
inflammatory collateral cysts  maxillary sinus  252–255 retention cyst of maxillary sinus 
54–58 mucoceles  238–241, 248–249 254
jaw cyst  7–8, 15–16 root resorption, dentigerous cyst  69 simple bone cyst  272–273, 276
keratocyst  100–106 Rushton bodies see hyaline bodies Stafne bone cavity  282
lateral periodontal cyst  142–144 surgical ciliated cyst  263–264
mucocele of maxillary sinus  253 s thyroglossal duct cyst  306
nasolabial cyst  233–234 salivary duct cyst  251–252 sialo‐odontogenic cyst see glandular
nasopalatine duct cyst  221–225 HIV‐associated  259 odontogenic cyst
366 Index

simple bone cyst  271–281 cell proliferation  115 simple bone cyst  281
cemento‐osseous dysplasia  276–279 dentigerous cyst  74–75 thyroglossal duct cyst  308
classification  271–272 glandular odontogenic cyst  170
clinical features  272–275 keratocyst  99–100, 112–113, 115 t
clinical presentation  274–275 naevoid basal cell carcinoma T lymphocytes, periapical
histopathology  273, 280–289 syndrome  115 periodontitis  27–28
multiple  276, 279 signalling pathway  99–100 teeth
pathogenesis  278–280 spherules  171–172 displacement
radiological features  273, 275–278 Stafne bone cavity  281–285 aneurysmal bone cyst  287
recurrence  281 clinical features  281–282 calcifying odontogenic cyst 
solitary  279 clinical presentation  282 188–189
terminology  271–272 differential diagnosis  284 dentigerous cyst  68
site distribution histopathology  285 keratocyst  91, 95–96, 100–103,
botryoid odontogenic cyst  151 pathogenesis  284–285 107
branchial cleft cyst  300–301 radiological features  282–284 nasopalatine duct cyst  220
calcifying odontogenic treatment  285 simple bone cyst  273–278
cyst  186–187 steatocystoma  290 enamel hypoplasia  72, 144, 207
dentigerous cyst  66–67 SUFU gene  99, 109–110 enamel projections  59–60
dermoid cyst  292–293 superficial mucoceles  241–242, impacted  207
epidermoid cyst  292–293 247–248 normal eruption  11
eruption cyst  84 surgical ciliated cyst  262–269 see also deciduous teeth; wisdom
gingival cysts  156–157, 161, 162 clinical features  263–265 teeth
glandular odontogenic cyst  clinical presentation  264–265 teratoid cyst, definition  289–291
165–166 differential diagnosis  266–267 teratomas, definition  290–291
heterotopic gastrointestinal cyst  297 histopathology  268 thymic cyst  308
inflammatory collateral cysts  50, 52–53 pathogenesis  267–268 thyroglossal duct cyst  305–308
intraoral lymphoepithelial cyst  256 radiological features  265–267 TNF see tumour necrosis factor
keratocyst  94–95 treatment  268–269 tongue
lateral periodontal cyst  142 surgical treatment bronchogenic cyst  299
mandibular buccal bifurcation cyst  adult gingival cyst  160 glands of Blandin–Nuhn  242
50, 52–53 botryoid odontogenic cyst  154 heterotopic gastrointestinal
mucoceles  239–241 branchial cleft cyst  305 cyst  297
nasopalatine duct cyst  219 calcifying odontogenic cyst  200 lymphoepithelial cyst  256–258
orthokeratinised odontogenic dentigerous cyst  82 thyroglossal duct cyst  306
cyst  202–203 dermoid cyst  295 tonsils  255–257, 296
paradental cyst  50, 52–53 epidermoid cyst  295 Tornwaldt cyst  308
pseudocyst of maxillary sinus  254 eruption cyst  85 transnasal marsupialization  236
radicular cyst  22–23 glandular odontogenic cyst  181 trauma
retention cyst of maxillary heterotopic gastrointestinal cyst  298 mucoceles  240
sinus  254 inflammatory collateral cysts  61 simple bone cyst  271–272, 274–279
simple bone cyst  272–274 keratocyst  136–138 treatment
Stafne bone cavity  282 lateral periodontal cyst  150 botryoid odontogenic cyst  154
surgical ciliated cyst  264 mucoceles  250–251 branchial cleft cyst  305
thyroglossal duct cyst  306 nasolabial cyst  236 bronchogenic cyst  300
Sjögren syndrome  255, 258–259 nasopalatine duct cyst  229 calcifying odontogenic cyst  200
SMO see smoothened protein orthokeratinised odontogenic dentigerous cyst  82
smoothened protein (SMO)  75, cyst  213 dermoid cyst  295
99–100, 110, 170 pseudocyst of maxillary sinus  255 epidermoid cyst  295
soft tissue keratocyst  97–98 radicular cyst  46 Epstein Pearls  163
solid odontogenic keratocyst  126–129 ranula  250 eruption cyst  85
solitary bone cyst see simple bone cyst retention cyst of maxillary gingival cysts  160, 162, 163
sonic hedgehog (SHH) protein  12 sinus  255 glandular odontogenic cyst  181
Index 367

heterotopic gastrointestinal cyst  298 simple bone cyst  281 verrucous odontogenic cyst 
inflammatory collateral cysts  61 Stafne bone cavity  285 212–213
intraoral lymphoepithelial cyst  258 surgical ciliated cyst  268–269 vomeronasal (Jacobson’s) organ  217
keratocyst  136–138 thyroglossal duct cyst  308
lateral periodontal cyst  150 tricholemmal cyst  290 w
mucoceles  250–251 tumour necrosis factor (TNF)  30, 31 whorling  171–172
nasolabial cyst  236 wisdom teeth
nasopalatine duct cyst  229 u dentigerous cyst  65–72, 82
orthokeratinised odontogenic ultrasound, nasolabial cyst  233 glandular odontogenic cyst  168
cyst  213 unicameral bone cyst see simple inflammatory collateral cysts  48–53,
pseudocyst of maxillary sinus  255 bone cyst 56, 58, 60
radicular cyst  46 keratocyst  102, 105, 107–108
ranula  250 v orthokeratinised odontogenic
retention cyst of maxillary sinus  255 vacuolated cells  39–40 cyst  203, 204
WILEY END USER LICENSE AGREEMENT
Go to www.wiley.com/go/eula to access Wiley’s ebook EULA.

You might also like