You are on page 1of 394

https://t.

me/MBS_MedicalBooksStore
The Oral-Systemic Health Connection: A Guide to Patient Care
Second Edition

https://t.me/MBS_MedicalBooksStore
The
Oral-Systemic
Health Connection SECOND EDITION

A Guide to Patient Care

Edited by
Michael Glick, dmd
Professor, Oral Diagnostic Sciences
University at Buffalo
The State University of New York
Buffalo, New York

Berlin, Barcelona, Chicago, Istanbul, London, Milan, Moscow, New Delhi,


Paris, Prague, São Paulo, Seoul, Singapore, Tokyo, Warsaw
Library of Congress Cataloging-in-Publication Data

Names: Glick, Michael, editor.


Title: The oral-systemic health connection : a guide to patient care /
edited by Michael Glick.
Other titles: Oral-systemic health connection (Glick)
Description: Second edition. | Batavia, IL : Quintessence Publishing Co, Inc., [2019] |
Includes bibliographical references and index. | 
Identifiers: LCCN 2018044816 (print) | LCCN 2018045699 (ebook) |
ISBN 9780867158083  | ISBN 9780867157888 (softcover)
Subjects: | MESH: Oral Health | Periodontal Diseases--complications |
Tooth Diseases--complications | Inflammation | Risk Factors
Classification: LCC RK61 (ebook) | LCC RK61 (print) | NLM WU 113 | DDC 617.6--dc23
LC record available at https://lccn.loc.gov/2018044816

https://t.me/MBS_MedicalBooksStore

97%
© 2019 Quintessence Publishing Co, Inc

Quintessence Publishing Co, Inc


411 N Raddant Road
Batavia, IL 60510
www.quintpub.com

5 4 3 2 1

All rights reserved. This book or any part thereof may not be reproduced, stored in a retrieval system, or
transmitted in any form or by any means, electronic, mechanical, photocopying, or otherwise, without prior
written permission of the publisher.

Editor: Zachary Kocanda


Design: Sue Zubek
Production: Kaye Clemens

Printed in the USA


C ONTENTS
Preface  vii
Contributors  viii

1 | Causation: Frameworks, Analyses, and Questions  1


Michael Glick

2 | Essential Statistical and Research Design Elements to Help Critically


Interpret the Literature  24
Barbara L. Greenberg and Michael Glick

3 | The Traveling Oral Microbiome  38


Wenche S. Borgnakke

4 | The Role of Inflammation in Oral-Systemic Interactions  86


Marcelo Freire and Thomas E. Van Dyke

5 | Obesity, Metabolic Syndrome, and Oral Health  102


Ira B. Lamster, Nadia Laniado, and Ilene Fennoy

6 | Associations Between Periodontal Disease and


Hyperglycemia/Diabetes  135
Wenche S. Borgnakke and Robert J. Genco

7 | The Cardiovascular System and Oral Infections  164


Maurizio S. Tonetti and Filippo Graziani

8 | The Association Between Oral Infections and Renal Disease  180


Karren Komitas and Effie Ioannidou

9 | The Association Between Oral Infections and Pulmonary


Disease  193
Karren Komitas and Effie Ioannidou
10 | Periodontal Infections and Adverse Pregnancy Outcomes  210
Yiorgos A. Bobetsis, Wenche S. Borgnakke, and Panos N. Papapanou

11 | Oral Infections and Cancer  231


Dominique S. Michaud

12 | Oral Manifestations of Systemic Diseases  242


Alessandro Villa and Sook-Bin Woo

13 | Oral Complications in the Immunocompromised Patient:


The Oncology Prototype  271 
Douglas E. Peterson

14 | Oral-Systemic Health: The Genomic Connection  292


John R. Shaffer and Robert J. Weyant

15 | Common Risk Factors: The Link Between Oral and


Systemic Disease  311
David M. Williams

16 | Antibiotic Prophylaxis for Patients at Risk for Infective


Endocarditis  329
Peter B. Lockhart

17 | Oral-Systemic Connection: The Salivaomics and


Exosomics Connection  342
˙
Taichiro Nonaka, Karolina Elzbieta Kaczor-Urbanowicz, and David T.W. Wong

18 | The Economic Impact of Periodontal Inflammation  357


Michael C. Alfano

Index  369
PREFA C E
A renewed interest in the oral-systemic health connection has widened the debate on
this fascinating topic, which now ranges from different plausible underlying biologic
principles to potential financial impact and effect on overall health and well-being.
This is not a new topic, but broadened implications for reimbursement models and the
recognition of oral health within the scope of other noncommunicable diseases affect
both clinical practice and health care policies.
The first written mention of the connection between infections in the oral cavity
and systemic health can be traced all the way back to cuneiform tablets from Niniveh
in Assyria circa 700 BC.1 However, since that time, anecdotes and reported cases have
given way to sophisticated research and analyses. Interpreting this research is sometimes
challenging. While distinct study designs and different statistical analyses may generate
different associations between oral infections and systemic diseases, this book provides
clarity and insight, with an ultimate goal of improving our patients’ overall health.
This second edition of The Oral-Systemic Health Connection explores a broad range
of topics that taken together enhance the reader’s appreciation of this still-emerging
association. There are several new chapters in this edition that are included based on
feedback from practitioners, scientists, and policymakers. The first chapter addresses one
of the most critical issues in the debate on the oral-systemic connection: the different
frameworks and models that are or can be used to claim causation. A new chapter on
biostatistical assessment has also been added to assist in the interpretation of the existing
literature. Two additional new chapters discuss the association between oral infections
and cancer as well as the association between oral health and renal disease. Other
chapters include updates on associations with other systemic diseases and conditions,
in addition to updates on what is known today about the role of inflammation and
the microbiome. Other important topics are addressed in new chapters on antibiotic
prophylaxis, the genomic connection, risk factors that are shared with systemic diseases
that are associated with significant morbidity and mortality, and the potential economic
impact of the oral-systemic health connection. Readers will note that there is not always
agreement among the authors about the importance and impact of the oral-systemic
health connection. No attempts have been made to reconcile these differences or reach
a consensus, and it is left to the reader to draw his or her own conclusions. Nonetheless,
similar to a much-appreciated feature in the first edition, specific answers to important
clinical considerations are included in selected chapters throughout the book.
We are inundated with data, statistics, and facts from a variety of outlets, including the
biomedical literature as well as social media and the lay press. It is therefore important
for today’s health care professionals to stay informed and remain reliable sources of
health information. This book will assist readers in understanding and appreciating the
complexity surrounding the oral-systemic health connection and, most importantly, help
to inform others about the impact of this link in order to benefit our patients.

Reference
1. Denton GB. A new interpretation of a well-known Assyrian letter. J Near Eastern Studies 1943;4:314.

vii
C ONTRIB U T OR S
Michael C. Alfano, dmd, phd Robert J. Genco, dds, phd
President, Santa Fe Group SUNY Distinguished Professor of
Professor, Dean, and Executive Vice Oral Biology and Microbiology and
President Emeritus Immunology
New York University Director, UB Microbiome Center
New York, New York University at Buffalo
The State University of New York
Yiorgos A. Bobetsis, dds, phd Buffalo, New York
Assistant Professor
Department of Periodontology Michael Glick, dmd
School of Dentistry Professor, Oral Diagnostic Sciences
National and Kapodistrian University University at Buffalo
of Athens The State University of New York
Athens, Greece Buffalo, New York

Wenche S. Borgnakke, dds, mph, phd Filippo Graziani, dds, mclindent, phd
Senior Research Associate Professor of Periodontology
Adjunct Clinical Assistant Professor Unit of Dentistry and Oral Surgery
Department of Periodontics and Oral University of Pisa
Medicine Pisa, Italy
University of Michigan School of
Dentistry Barbara L. Greenberg, msc, phd
Ann Arbor, Michigan Adjunct Professor of Dental Medicine
Touro College of Dental Medicine
Ilene Fennoy, md, mph New York Medical College
Professor of Pediatrics Valhalla, New York
Columbia University Irving Medical
Center Effie Ioannidou, dds, mds
Vagelos College of Physicians and Professor of Periodontology
Surgeons Director, Dental Clinical Research
New York, New York Center
School of Dental Medicine
Marcelo Freire, dds, phd, dmsc UConn Health
Associate Professor Farmington, Connecticut
Genomic Medicine and Infectious
Disease
J. Craig Venter Institute
La Jolla, California

viii
Karolina Elzbieta
˙ Kaczor-Urbanowicz, Dominique S. Michaud, scd
dmd, phd, msc, msl Professor of Public Health and
Visiting Assistant Project Scientist Community Medicine
Division of Oral Biology and Medicine School of Medicine
Clinical Lecturer Tufts University
Section of Orthodontics Boston, Massachusetts
UCLA School of Dentistry
Los Angeles, California Taichiro Nonaka, dds, phd
Assistant Project Scientist
Karren Komitas, dmd, phd, mdsc Division of Oral Biology and Medicine
Assistant Professor UCLA School of Dentistry
Department of Periodontics and Dental Los Angeles, California
Hygiene
School of Dentistry Panos N. Papapanou, dds, phd
University of Texas Health Science Professor of Dental Medicine
Center at Houston Chair, Section of Oral, Diagnostic, and
Houston, Texas Rehabilitation Sciences
Director, Division of Periodontics
Ira B. Lamster, dds, mmsc Columbia University College of Dental
Clinical Professor Medicine
School of Dental Medicine New York, New York
Stony Brook University
Douglas E. Peterson, dmd, phd, fds rcsed
Dean Emeritus Professor of Oral Medicine
Columbia University College of Dental School of Dental Medicine
Medicine Co-Chair, Program in Head and Neck
New York, New York Cancer and Oral Oncology
Neag Comprehensive Cancer Center
Nadia Laniado, dds, mph UConn Health
Director of Community Dentistry and Farmington, Connecticut
Population Health
Department of Dentistry/OMFS John R. Shaffer, phd
Jacobi Medical Center Assistant Professor
Albert Einstein College of Medicine Department of Human Genetics
New York, New York Graduate School of Public Health

Peter B. Lockhart, dds, fds rcsed, Assistant Professor


fds rcps
Department of Oral Biology
Research Professor School of Dental Medicine
Department of Oral Medicine University of Pittsburgh
Carolinas Medical Center Pittsburgh, Pennsylvania
Charlotte, North Carolina

ix
Maurizio S. Tonetti, dmd, phd, mmsc David M. Williams, bds, msc, phd, frcpath,
Executive Director fds rcs (engl)
European Research Group on Professor of Global Oral Health
Periodontology Bart’s and The London School of
Clinical Professor in Periodontology Medicine and Dentistry
Prince Philip Dental Hospital Queen Mary University of London
The University of Hong Kong London, England
Sai Ying Pun, Hong Kong
David T.W. Wong, dmd, dmsc
Thomas E. Van Dyke, dds, phd Felix and Mildred Yip Endowed Chair
Vice President and Senior Member of in Dentistry
Staff Associate Dean for Research
The Forsyth Institute Professor of Oral Biology
Director, UCLA Center for Oral/Head
Professor of Oral Medicine, Infection, & Neck Oncology Research
and Immunity UCLA School of Dentistry
Harvard School of Dental Medicine Los Angeles, California
Cambridge, Massachusetts
Sook-Bin Woo, dmd, mmsc
Alessandro Villa, dds, phd, mph Associate Professor
Assistant Professor Department of Oral Medicine,
Department of Oral Medicine, Infection, and Immunity
Infection, and Immunity Harvard School of Dental Medicine
Harvard School of Dental Medicine
Attending Surgeon
Associate Surgeon Division of Oral Medicine and
Division of Oral Medicine and Dentistry
Dentistry Brigham and Women’s Hospital
Brigham and Women’s Hospital Boston, Massachusetts
Boston, Massachusetts

Robert J. Weyant, ms, dmd, drph


Professor and Chair
Department of Dental Public Health
School of Dental Medicine
University of Pittsburgh
Pittsburgh, Pennsylvania

x
CHAPTER 1

Causation: Frameworks,
Analyses, and Questions
Michael Glick, dmd

Why Do We Care About analyses, and criteria can be used to claim that
periodontal disease affects systemic health.
Causation? Awareness of causal relationships will lead to
benefits such as the ability to explain and pre-
The desire to ascertain causality is one we dict outcomes that are important to patients.
encounter in everyday life. Most of us have However, data generation alone cannot answer
difficulty accepting uncertainty, and we want questions about causal relationships. Knowl-
to discern patterns and reasons to explain why edge about how the data were acquired and
things occur. This is nothing new; such quests how those data fit into causal constructs are
can be found in ancient texts on philosophy, necessary to assess and use those data to claim
science, and even religion. The word cause or causal relationships.
causal wording is often found in the biomed- One challenge is how to interpret results
ical literature, in nonscientific publications, from observational data and intervention
and in social and other media. However, it is trials. The conventional use of observational
not always clear how a claim could have been studies is to find associations that can explain
made. connections between different exposures and
My hope is that after reading this chapter, outcomes.1 Appropriately designed and per-
a reader who encounters a claim of causation formed randomized controlled trials (RCTs)
will ask what framework, analysis, or crite- have traditionally been considered one of the
rion was used to make such a claim. To even most suitable methods to establish evidence
ask such a question, a frame of reference to for causal inferences and claims. But where an
compare claims of causation will need to be RCT is impossible to carry out, observational
established. This chapter provides an overview data from cross-sectional, case control, and
of causal constructs that provide a founda- cohort studies may still be used to approximate
tion for a discourse on causal relationships. causal inferences.2
Throughout this chapter, periodontal disease RCTs were initially designed to reduce bias
is used as a proxy for oral infections, but other and have long been labeled the gold standard
chapters in this book specifically address other for evidence-based medicine, but this concept
diseases, conditions, and circumstances as a has been challenged.3,4 The limitations of RCTs
focus of discussion. In this chapter, we will spe- lie not with the randomization or the control
cifically examine how proposed frameworks, but with, for example, the use of surrogate

1
1 Causation: Frameworks, Analyses, and Questions

endpoints rather than clinical outcomes, or As discussed in this chapter, there is no right
inappropriate extrapolation, or generalization way to determine causal relationships, nor
of findings. Furthermore, RCTs do not lend are there any universally agreed upon causal
themselves to all research questions. Research frameworks, models, methods, algorithms,
questions about harm, etiology, prognosis, and criteria, or measures. The advantage of a uni-
diagnosis may be better answered by obser- versal language and framework for causation is
vational studies. Although outcomes from that it enables a dialogue about consequences
properly designed and performed RCTs of an and actions, which is needed to benefit our
appropriate size lead to an inference, such an patients.7 To quote Voltaire, “If you wish to
inference is based on a probabilistic estimate converse with me, first define your terms.”
of a clinical prediction. Even when valid, such
a prediction cannot ensure an expected out-
come. Yet RCTs still hold many advantages
over observational studies. A Brief History of the
The vast majority of research on the oral Oral Infection–Systemic
infection–systemic health relationship consists
of observational studies. Many oral diseases, Health Link
such as periodontal diseases and caries, are
complex and chronic and are probably asso- Observations of a putative link between sys-
ciated with multifactorial determinants or temic conditions and oral health were described
risk factors. The same holds true for systemic on cuneiform tablets from Assyria more than
illnesses. Except in rare circumstances, it is 2,700 years ago.8 Since that time, many prom-
difficult to determine the onset of a systemic inent figures in the history of medicine have
condition before signs and symptoms appear written about this possible association, includ-
(with the possible exception of pregnancy). ing Hippocrates (460–370 BCE), who is often
Thus, one of the hallmarks of a causal rela- portrayed as the paragon of ancient physicians
tionship—temporality (a directional sequence and allegedly claimed that it was possible to
of events where a causal event [exposure] cure arthritis by the removal of teeth, and
happens before the effect [outcome])—may Galen (131–201 CE), another dominant Greek
be practically impossible to establish. Estab- physician and the most famous and influential
lishing whether an oral infection truly occurred physician of ancient Rome.
prior to a systemic illness, with our present The term focus of infection was coined by
research armamentarium and knowledge, Willoughby Dayton Miller, a leading microbi-
may not always be possible. Furthermore, a ologist and dentist who published a series of
cause-and-effect relationship is not symmetric. articles titled “The human mouth as a focus of
For example, stopping sugar consumption may infection” in Dental Cosmos and in The Lancet
reduce the incidence of caries, but treating car- in 1891.9,10
ies lesions is not in itself a strategy for stopping Most observational reports in the medical
sugar consumption. and dental literature in the late 1800s and
Statistical analyses can infer associations early 1900s described how infected teeth
among variables and estimate measures of could influence systemic conditions, even sug-
associations under static conditions, but gesting that extraction of infected teeth could
they fall short when dealing with changing cure systemic illness. This approach to treat-
conditions. There is no universally accepted ing and curing illnesses, for example arthritis,
technique or statistical analysis to measure was later challenged by researchers. Instead
causal effects, although several methods have of describing single observations, researchers
been proposed.5,6 conducted retrospective studies that included

2
Definitions of Cause and Causation

multiple patients. In their seminal article on oral manifestations of systemic illnesses, the
the association between extracted teeth and increased susceptibility of the immune-deficient
relief of signs and symptoms of rheumatoid or immune-dysfunctional host to develop
arthritis, Cecil and Angevine famously stated adverse systemic outcomes due to oral infec-
that “Focal infection is a splendid example of tion, genetic determinants associated with
a plausible medical theory which is in danger oral health, and the association between oral
of being converted by its too enthusiastic sup- infection and infectious endocarditis are well
porters into the status of an accepted fact.”11 known and should be included in such a dis-
With better appreciation of scientific evidence, cussion (see chapters 4, 11, 12, 13, and 15).
better research design, and better diagnostic
techniques and treatments, a movement away
from the focal infection theory and against
teeth extraction was emerging.12,13
Definitions of Cause
One of the first mentions of periodontal and Causation
infection as a possible source of systemic illness
appeared in the first edition of Oral Medicine by The scientific literature is replete with words
Lester W. Burket in 1946, where he stated that implying causation: affect, cause, contribute,
“the frequent occurrence of bacteremias which determine, effect, enhance, impact, influence,
may arise from diseased periodontal tissue, lead to, produce, responsible for, result from,
might be of more importance in the causation and so on. It is important to recognize the mis-
of joint lesions.”13 In 1951, an entire issue of use of causal terminology, as authors may at
The Journal of the American Dental Associa- times employ these causal words to infer effects
tion was published as a “report, prepared under beyond study findings.
the general direction of the Council on Dental Common meanings of the word cause
Health of the American Dental Association, can be found in most dictionaries; it is more
[that] constitutes a summary and an evaluation difficult to find a common definition of
of modern scientific information about the role causation. However, we need an agreed upon
of dental foci of infection in body disease.”14 definition of cause before we can start a dis-
In the late 1980s and the early 1990s, obser- cussion about causation. According to the
vational studies from Finland highlighted an Free Dictionary, cause can be defined as “the
association between dental health (dental car- producer of an effect, result, or consequence.”
ies, periodontal disease, or both) and acute Merriam-Webster similarly defines cause as
myocardial infarction, while similar studies “something that brings about an effect or a
in the United States continued to establish a result” but elaborates as “a reason for an action
bidirectional association between periodontal or condition: motive.”22,23 Within the biomed-
health and diabetes.15–17 Apart from observa- ical field, cause is often used synonymously
tional studies, experimental animal studies with etiology. These common definitions
revealed an association between oral pathogens immediately invoke teleology—the study of
and adverse pregnancy outcomes, an associa- origin—one of the core contributors to con-
tion that was later observed in humans.18–20 fusion in what cause is intended to mean in
The discussion about the relationship between biomedical contexts.
oral disease and systemic illness was rapidly A more representational depiction of cause
shifting from an association between diseases could be described as follows: Variable A is a
to the influence of the oral microbiome (see cause of variable B if B in any way depends
chapter 3). Although a proliferation of stud- on A for its value. This rather rudimentary
ies have investigated the association between explanation can help outline different causal
periodontal disease and systemic health, 21 relationships:

3
1 Causation: Frameworks, Analyses, and Questions

• A causes B (direct causal relationship). For this relationship is a logical fallacy known
example, sugar is associated with the devel- as fallacy of affirming the consequent: We
opment of caries. Caries cannot develop know that if X is true, Y must be true. We
without the presence of sugar. Therefore, know that Y is true, so therefore X must be
sugar causes caries. In this scenario, sugar is true. If I have periodontal disease, I will have
a necessary cause (see Box 1-3). cardiovascular disease. I have cardiovascular
• A and B are consequences of a common disease, and therefore I have periodontal dis-
cause. Let’s assume smoking causes periodon- ease. Unfortunately, this fallacy is used often
tal disease and smoking causes cardiovascular by scientists and laypeople in interpreting
disease. Such an assumption may suggest a data.
common cause (smoking) but not necessar- • Both A and B cause C. Both early childhood
ily a causal relationship between periodontal caries and periodontal disease can result in
disease and cardiovascular disease. In this full-mouth extraction. As both conditions
example of considering whether A (periodon- result in the same outcome, a claim may
tal disease) is a cause of B (cardiovascular be made of an association between these
disease), smoking may be a confounder, a conditions. However, childhood caries and
third variable affecting both periodontal periodontal disease do not necessarily have
disease and cardiovascular disease and poten- a causal relationship.
tially fully explaining the putative causal • A causes C, which causes B (indirect causal
relationship between A and B. Thus, although relationship). Periodontal disease (A) is asso-
there may be an association between peri- ciated with tooth loss (C). Tooth loss (C) is
odontal disease and cardiovascular disease, associated with nutritional deficiencies (B). Is
there is, in this example, no causation. it therefore possible to claim that periodontal
• A causes B, and B causes A (bidirectional, disease (A) causes nutritional deficiencies (B)?
reciprocal, or cyclic causal relationship). • There is no connection between A and B; the
Oral conditions have been claimed to cause correlation is coincidental. For example, we
systemic inflammation, and systemic inflam- found that nearsighted individuals are more
matory conditions have been implicated in likely to have periodontal disease than indi-
the etiology of oral conditions. This recipro- viduals who are not nearsighted. Using this
cal relationship has been used to claim that example to infer cause and effect is spurious.
as diabetes will exacerbate periodontal con-
ditions and vice versa, there is a bidirectional The above examples highlight why causation
claim of causality between these conditions. is such an elusive concept. Yet it is a concept
The interrelationships between periodontitis that has been discussed and written about at
and diabetes provide an example of systemic least since the time of Aristotle (384–322 BCE).
disease predisposing to oral infection. Once Numerous theories of causation have been
the oral infection is established, it exacer- proposed, in philosophy and in biomedical disci-
bates systemic disease. In this case, it may plines, and this chapter highlights those theories
also be possible for the oral infection to pre- that could be applied to the association between
dispose to systemic disease.24 oral infection and systemic health. However, an
• B causes A (reverse causal relationship). A exhaustive historical literature review of this
cross-sectional study finds an association topic will not be undertaken. Although defining
between the presence of periodontal disease causation is important, such an endeavor often
and the presence of cardiovascular disease. digresses into philosophical and metaphysical
Did periodontal disease cause cardiovascu- dialogues. In this chapter, we will shy away from
lar disease, or did cardiovascular disease a discourse on definitions and instead highlight
cause periodontal disease? A variation of frameworks and methods that describe causal

4
Frameworks, Classifications, Criteria, and Analyses to Describe and Infer Causation

Box 1-1  |  Henle-Koch postulates26

1. The microorganism must be observed in all cases of the disease.


2. The microorganism must be isolated and grown in pure culture.
3. Microorganisms from the pure culture, when inoculated into a susceptible animal,
must reproduce the disease.
4. The microorganism must be observed in and recovered from the experimentally
diseased animal.

relationships. The intent of this chapter is not microorganism and a disease. This gold stan-
to promote a specific framework of causation dard, which was used to establish evidence for
or engage in an epistemologic discourse on a single infectious agent as a cause for a specific
causation but to provide different frameworks disease, has been challenged by the discovery
that can be used to judge claims of causation. of viruses and other microbes, the recognition
of multiple causes, and its not being adaptable
to experimentation. However, the postulates
did provide a conceptual framework for sub-
Frameworks, Classifications, sequent models of causation, including the one
Criteria, and Analyses to proposed by Sir Austin Bradford Hill.

Describe and Infer Causation


Bradford Hill criteria
Frameworks of causation are generated as an
attempt to explain connections between events. One of the most influential and revered
It is not possible in this chapter to cover all medical statisticians of his time, Sir Aus-
such proposed frameworks, but a couple of tin Bradford Hill is recognized as one of the
pertinent conceptual frameworks are high- pioneers of RCTs.27,28As a statistician and
lighted below.25 All of them can be used to an epidemiologist, Hill was concerned with
some degree when discussing the connection how the medical community interpreted and
between oral infections and systemic health. subsequently implemented results from obser-
Many recent proposed frameworks of vation and intervention trials. A method and
causation use a common terminology to delin- framework was needed to distinguish correla-
eate nuances that need to be addressed, and tions that are true signs of cause-and-effect
familiarity with this terminology will help the relationships from those that are not. In his
reader to understand the theoretical arguments president’s address to the Section on Occu-
supporting causal frameworks (see glossary). pational Medicine within the Royal Society
of Medicine in 1965, Hill asked, “What cir-
cumstances can we pass from this [respiratory
Henle-Koch postulates illness and dust in the environment] observed
association to a verdict of causation?”25 Hill
The Henle-Koch postulates (Box 1-1),26 for- put forth nine “aspects of association” (criteria,
mulated in the late 19th century, were a or causal components), based on epidemiologic
scheme to establish a causative link between a data, that might be used to assess causation

5
1 Causation: Frameworks, Analyses, and Questions

Box 1-2  |  The Bradford Hill criteria27

Strength (of an association). A small or weak association (effect size, odds ratios, relative risks,
etc) does not mean that there is not a causal effect. However, the larger or stronger the association,
the more likely that it is causal.
Consistency. Consistent findings observed by different researchers, across different study designs,
in different populations, among different locations, and with different samples strengthen the like-
lihood of a causal relationship.
Specificity. The more specific an association between a factor (a causative agent) and a specific
outcome (disease) is, the higher the probability of a causal relationship. This is similar to Henle-Koch
postulates 1 and 2.
Temporality. The exposure must precede the outcome.
Biologic gradient. If greater exposure (dose, level, duration) generally leads to greater incidence
of the effect, a stronger causal relationship is assumed.
Plausibility. A known biologic pathway may explain the pathophysiologic link between a risk factor
and the disease in question.
Coherence. The relationships should not conflict with what we know of the disease (consistency
between epidemiologic and laboratory findings). This is similar to Henle-Koch postulate 3.
Experiment. Experimental results from different experimental methods and by different investigators
are useful evidence toward a causal relationship.
Analogy. After finding a relationship between, for example, human papillomavirus (HPV) and cervical
cancer, we may more readily accept that HPV could cause another type of cancer.

(Box 1-2).29 Five of Hill’s proposed criteria of causation should be familiar with these crite-
(strength, consistency, specificity, temporality, ria as they are still frequently called on to make
and coherence) had already been used in the causal claims, yet the fallibility of the Bradford
1964 Surgeon General’s report on the dangers Hill criteria has been discussed at length.32 The
of smoking.30 Although this was not Hill’s Bradford Hill criteria have been mentioned
intent, his proposed criteria, often referred to as when discussing a causal link between peri-
the Bradford Hill criteria, have since been used odontal disease and systemic illnesses.33–35
by researchers to “prove” causal associations Using the Bradford Hill criteria may be a spe-
between numerous observational associations. cious proposition in an attempt to simplify a
Hill was very careful not to claim that his nine complex process and an unattainable goal.
criteria served as sufficient proof of causation,
instead stating that they could be used as
“evidence toward a causal relationship.” The Necessary and sufficient condition
increasing complexity of and advancements in framework
science since the publication of the Bradford
Hill criteria in 1965 have challenged his ini- In science, in medicine, and even in the legal
tial understanding of causality.31 The Bradford realm, necessary and sufficient conditions are
Hill criteria should, maybe at best, be used as used to describe a causal relationship between
a screening test to determine whether further events (Fig 1-1 and Box 1-3). A necessary con-
consideration and examination are warranted dition is a condition that is required for the
to ascertain a causal relationship. Any student occurrence of the effect (outcome or disease),

6
Frameworks, Classifications, Criteria, and Analyses to Describe and Infer Causation

Constellations of components*

I II III IV V

ID 2 ID 3 8 4 1 4
ID
1 4 1 6 1 3 5 7
5 7 6 6
Causal scenarios Outcomes
PD Possible Possible Possible Never Never
ID is a necessary
A
condition.
Oalt Possible Possible Possible Never Never

PD Always Always Always Possible Possible


ID is a sufficient
B
condition. Oalt Never Never Never Possible Possible

PD Always Always Always Never Never


ID is a necessary and
C
sufficient condition.
Oalt Never Never Never Always Always

ID is a necessary PD Possible Possible Possible Never Never


D but not a sufficient
condition. Oalt Possible Possible Possible Possible Possible

ID is a sufficient PD Always Always Always Possible Possible


E but not a necessary
condition. Oalt Never Never Never Possible Possible

ID is neither a PD Possible Possible Possible Possible Possible


F necessary nor a
sufficient condition. Oalt Possible Possible Possible Possible Possible

See Box 1-3 for more detailed explanations. *Constellations of components:


• I is only composed of one component, ID.
In this example, immune dysfunction (ID) is inves-
• II is a constellation of components (ID, 1, 2, 4, and 5).
tigated as a causative condition for the outcomes
• III is a constellation of components (ID, 1, 3, 6, and 7).
periodontal disease (PD) or an alternative or nonPD
• IV is a constellation of components (1, 3, 4, 6, and 8),
outcome (Oalt).
where in scenario F 8 is a contributory condition.
Scenarios A to F depict how a necessary and
• V is a constellation of components (1, 4, 5, 6, and 7).
sufficient condition framework changes the impact
on outcomes.

Fig 1-1  |  Necessary and sufficient conditions, and combinations of necessary and sufficient conditions.

but a necessary condition can be present with- occurrence of the outcome, but the outcome
out the occurrence of the effect. For example, can occur without the presence of a sufficient
sugar must be present for caries to develop condition. For example, exposure of a vital
(necessary condition), but sugar can be con- pulp (sufficient condition) will always produce
sumed without the accompanying development tooth pain, but tooth pain can also occur by
of caries (see Fig 1-1A). A sufficient condition other means (see Fig 1-1B).
is a condition that is always followed by the

7
1 Causation: Frameworks, Analyses, and Questions

Box 1-3  |  Necessary, sufficient, and combinations of necessary and sufficient conditions

Necessary condition. If component X (CX) is a necessary condition for outcome Y (OY), CX must always be present
for OY. But the presence of CX doesn’t always result in OY, and an alternative outcome (Oalt) may be present. However,
OY can never be present without CX.
See Fig 1-1A. If immune dysfunction (ID) is a necessary condition for periodontal disease (PD), ID must be present
for PD to be present (constellations I, II, and III). The presence of ID does not always result in PD (constellations I, II,
and III). However, PD can never be present without ID (constellations IV and V).
Another example: Mycobacterium tuberculosis (MTB) (CX) must be present for the development of tuberculosis
(TB) (OY). But MTB can be present without the development of TB. However, TB can never develop without the
presence of MTB.

Sufficient condition. If CX is a sufficient condition for OY, CX will always result in OY, but OY can also occur with
alternative conditions (Calt). However, Oalt can never be present in the presence of CX.
See Fig 1-1B. If ID is a sufficient condition for PD, PD will always be present in the presence of ID (constellations
I, II, and III), but other conditions can also cause PD (constellations IV and V). However, ID can never be present
without the presence of PD.
Another example: 38% silver diamine fluoride (CX) will always cause black staining of teeth (OY), but black staining
of teeth can also occur due to other causes.

Both necessary and sufficient. If CX is both a necessary and a sufficient condition for OY, OY cannot happen
without CX, and OY will always happen with the presence of CX. OY never happens without CX, and CX always leads
to OY.
See Fig 1-1C. If ID is both a necessary and a sufficient condition for PD, PD will never happen without ID (con-
stellations IV and V), and PD will always happen as a result of ID (constellations I, II, and III). ID always leads to PD,
and PD never happens without ID.
Another example: The presence of a third chromosome (CX) is a necessary and sufficient condition for the
development of Down syndrome (OY). There is no alternative condition (no Calt) or any alternative outcome
(no Oalt).

The concept of necessary and sufficient con- The use of the necessary and sufficient con-
ditions is sometimes invoked when describing dition framework considers each cause either
deterministic causality models, which are based a necessary condition or a sufficient condition.
on the notion that every event is caused by an This limits a causal relationship to a one-to-one
antecedent event and in accordance with the association between an observed condition or
laws of nature. Expressions of these conditions, cause and an outcome.37 A one-to-one causal
separately and in combination, are also used to model is of course highly unlikely in the bio-
describe specific conditions. medical arena, and this framework needs to be
A third type of condition, known as a con- extended to allow multiple or constellations
tributory cause, is neither a necessary nor a of conditions to explain observed outcomes.
sufficient condition by itself. A contributory A simple example that is often used when dis-
cause needs to fulfill two criteria: “(1) the attri- cussing this concept is the appearance of light
bute referred to as the cause has been shown to after flipping a light switch from off to on. If
precede the effect; (2) altering only the cause I had little (or no) knowledge of the under-
has been shown to alter the effect.”36 lying mechanism necessary for the light to
appear (presence of electricity, complete and

8
Frameworks, Classifications, Criteria, and Analyses to Describe and Infer Causation

Necessary but not sufficient. If CX is a necessary but not sufficient condition for OY, CX is always present for OY
but can also be present without OY; CX by itself does not always result in OY.
See Fig 1-1D. If ID is necessary but not sufficient for PD, ID is required for the presence of PD (constellations I, II,
and III but never IV or V) but can be present without PD (constellations I, II, and III). ID by itself does not always result in
the presence of PD but can result in PD together with other components (constellations I, II, and III).
Another example: HIV (CX) must be present to develop AIDS (OY), but HIV can be present without a person developing
AIDS; the presence of HIV by itself is not enough for the development of AIDS. An alternative constellation of conditions
(Calt) that includes HIV could have brought about AIDS, but HIV by itself could also result in an alternative outcome (Oalt).

Sufficient but not necessary. If CX is a sufficient but not necessary condition for OY, CX will always result in OY, but
CX is not necessary for OY and OY can happen without CX.
See Fig 1-1E. If ID is sufficient but not necessary for PD, ID will always result in PD (constellations I, II, and III), but
PD can happen without ID (constellations IV and V).
Another example: Full-mouth extraction (CX) will always cause edentulism (OY), but full-mouth extraction is not the
only reason for edentulism, and edentulism can happen in the absence of full-mouth extraction under alternative con-
ditions (Calt). However, there is no alternative outcome (no Oalt) (nonedentulism) when full-mouth extraction is present.

Neither necessary nor sufficient. If CX is neither a necessary nor a sufficient condition for OY, then when CX occurs
OY will sometimes happen. However, OY can also occur without CX. The condition sometimes leads to the outcome, and
sometimes the outcome can occur without the condition. There may be another unknown or alternative condition (Calt)
(contributory condition) that is neither necessary nor sufficient for OY.
See Fig 1-1F. If ID is neither necessary nor sufficient for PD, then sometimes when ID is present PD is present.
However, PD can also occur without ID. ID sometimes leads to PD, and sometimes PD can be present without ID
(constellations I, II, III, IV, and V). There may be another or alternative component (contributory condition) (component
8 in constellation IV) that is neither necessary nor sufficient for PD.
Another example: Although there is an association between low socioeconomic status (SES) and periodontal disease,
low SES (CX) on its own is not required for the development of periodontal disease (OY), and periodontal disease may
occur in the absence of low SES due to a different condition that is neither necessary nor sufficient (Calt).

fully functioning wiring, intact lightbulb, etc), I are sufficient for the effect or outcome to occur38
might be warranted in claiming that flipping the (see Fig 1-2). Each component must be present
switch caused the light to appear. More knowl- or must have occurred for the outcome to hap-
edge makes a causal claim much more complex. pen. This framework allows for different causal
Furthermore, this example also highlights that mechanisms. As highlighted in the hypothetical
a mechanistic requirement, or a biologic path- example in Fig 1-2, different combinations of
way, which was discussed by Hill, has to be single component causes provide a sufficient
considered. causal mechanism. When considering the cause
of a disease, many of the sufficient components
may be unknown, and it is therefore wise to
Sufficient-component framework include one component cause, labeled U, to
represent unknown determinants38 (see Fig
In the sufficient-component framework, suffi- 1-2). In the sufficient-component model, all
cient cause is used to indicate a complete causal the components are essential, and blocking
mechanism that includes a minimal set of com- the contribution of one prevents the onset of
ponents (conditions and events) that together the disease.

9
1 Causation: Frameworks, Analyses, and Questions

I II III
Each “pie” is composed of multiple single component
A A A causes that together represent a sufficient cause of
U B U B U G disease. I, II, and III each depict a causal mechanism.
This model depicts a multicausal framework rather
D C F E I H
than a single cause of disease.

A hypothetical example: What causes coronary The causal mechanisms for CAD are proposed to be the
artery disease (CAD)? joint presence of multiple single components:
Single component causes: I. Genetic susceptibility to develop CAD, oral
A = genetic susceptibility to develop CAD inflammation, presence of atheroma, immune
B = oral inflammation dysfunction, and unspecified events
C = presence of atheroma II. Genetic susceptibility to develop CAD, oral inflam-
D = immune dysfunction mation, presence of P gingivalis, metastasis of oral
E = p resence of a specific oral bacteria bacteria, and unspecified events
(eg, Porphyromonas gingivalis) III. Genetic susceptibility to develop CAD, metastasis
F = metastasis of oral bacteria of toxin from oral bacteria, bacteremia, presence
G = metastasis of toxin from oral bacteria of biofilm, and unspecified events
H = bacteremia A component cause (eg, genetic susceptibility to
I = presence of biofilm develop CAD [A]) that appears in all sufficient causes
U = unspecified events is a necessary cause of the outcome. An unspecified
event (U) component should always be considered.

Fig 1-2  |  Sufficient-component model of causation. (Adapted from Rothman and Greenland32 and Rothman.38)

Counterfactual framework by two events, flying via Newark or via Chi-


cago) but has to be a necessary cause (the one
When flying out of Buffalo, New York, to route that took me to San Francisco) without
attend a meeting in San Francisco, I decided which the event (attending the meeting on time)
to travel via Newark rather than through Chi- would not have occurred. As only one outcome
cago. Unfortunately, due to delays of incoming will actually occur (in this case, I missed my
flights to Newark, my onward flight from New- meeting in San Francisco), the other outcome is
ark to San Francisco was severely delayed and I not observed (I would have arrived in time for
missed my meeting in San Francisco. I lamented my meeting in San Francisco if I had traveled
that if I had taken the Chicago connection, I via Chicago) and is known as the counterfac-
would have been on time for my meeting. tual or potential outcome. Events that can be
A binary outcome event is an outcome event considered by an “if” statement are known
(attending a meeting on time in San Francisco) as counterfactuals, where the “if” portion of
that always occurs after one exposure but not the counterfactual is called the hypothetical
after the other (arriving on time for my meeting condition (or antecedent) that may lead to the
in San Francisco after traveling either through unobserved or the counterfactual (unrealized,
Newark or Chicago). Therefore, a causal effect potential, or maybe untrue) outcome.
(my travel route) of a binary event is not neces- We often use counterfactual arguments in
sarily a sufficient cause (the outcome can occur dentistry. For example, “if he had brushed his

10
Frameworks, Classifications, Criteria, and Analyses to Describe and Infer Causation

teeth, he wouldn’t have developed gingivi- even if they had been treated for periodontal
tis.” In essence, this counterfactual argument disease) on experimental data.
implies that brushing his teeth would have pre-
vented the development of gingivitis. Or stated
differently, not brushing his teeth caused the Probabilistic framework
development of gingivitis.
It would be difficult to apply a counterfactual A probabilistic cause framework explains the
cause scenario to the oral infection–systemic relationship between cause and effect using
health connection. Using the example of peri- the tools of probability theory. Contrary to
odontal disease and diabetes: If poor metabolic the deterministic cause model, the probabilis-
control was observed in individuals not treated tic cause framework provides an estimate—a
for periodontal disease and good metabolic probability rather than certainty—of an event
control was observed in individuals treated for occurring. This model is sometimes used to esti-
periodontal disease, can we claim that peri- mate the effect of dose-dependent relationships.
odontal disease affected metabolic control? For example, as was mentioned previously,
The shortcoming of this argument is that good sugar is a necessary cause for the development
metabolic control cases cannot be tested twice, of caries. But could there be a daily consump-
with and without periodontal treatment. For tion threshold for sugar where caries will
example, if we measure equal proportions of always develop (a sufficient cause)? In this theo-
poor metabolic control in the treatment and retical example, sugar consumption is not static
control groups, we cannot tell how many cases but is associated with an action (increasing the
of poor metabolic control are actually attribut- amount of sugar) and can thus be perceived in
able to the lack of periodontal treatment itself. accordance with a probabilistic causal frame-
It is quite possible that many of those patients work, as it might be possible to estimate the
with good metabolic control who underwent probability of such an event occurring.
periodontal treatment would, if left untreated, The probabilistic model may be useful to
have had good metabolic control, and, simul- estimate the probability that oral infections
taneously, many of those with good metabolic may cause a systemic illness. In other words,
control who did not receive periodontal treat- if oral infections have consistently and with a
ment would have developed poor metabolic high degree of probability been shown to be
control if treated. Thus, we cannot use the associated with a systemic outcome, we may be
argument that as lack of periodontal treatment able to claim that the oral infections probably
resulted in poor metabolic control, periodontal caused such an outcome, with a caveat that
treatment will cause good metabolic control. such a claim can only be made after random-
Although it has been argued that causal ized controlled experiments.
relationships can be elucidated from experi-
mental studies, the example above suggests
that this is not always true. One reason for this Spurious relationships
conundrum is that experimental studies cannot
always affirm or separate the effect of attribu- The presence of spurious (false or bogus) cor-
tion (what portion/percentage of cases of good relations has been recognized for more than
metabolic control could be attributed to a spe- 100 years.39 There are several tongue-in-cheek
cific exposure, such as periodontal therapy) and examples published in even very prestigious
the effect of susceptibility (what portion/per- journals. One example is the claim that choc-
centage of cases with good metabolic control olate consumption will increase cognitive
who did not undergo periodontal treatment function. The author showed that “there was a
would have developed poor metabolic control close, significant linear correlation (r = 0.791,

11
1 Causation: Frameworks, Analyses, and Questions

P < .0001) between chocolate consumption of the challenges with using the Bradford Hill
per capita and the number of Nobel laure- criteria are delineated here.
ates per 10 million persons in a total of 23
countries.”40 The author points out that “it
Strength: Strong associations are more likely
seems most likely that in a dose-dependent
to be causal than weak associations.
way, chocolate intake provides the abundant
fertile ground needed for the sprouting of Specific bacteria may be strongly associated
Nobel laureates. Obviously, these findings are with periodontal disease and may therefore
hypothesis-generating only and will have to be be considered a cause of periodontal disease.
tested in a prospective, randomized trial,” but However, an immune dysfunctional environ-
“a second hypothesis, reverse causation—that ment may also be a risk factor for periodontal
is, that enhanced cognitive performance could disease. Immune dysfunction is a confounder
stimulate countrywide chocolate consump- as it is associated with both the exposure
tion—must also be considered.” (enabling the growth of specific bacteria) and
Unfortunately, spurious correlations are often the outcome (clinical manifestations of peri-
espoused as scientifically valid cause-and-effect odontal disease). Confounders can make what
relationships both in the lay press and on social appears to be a strong association seem causal
media, where they are propagated by mech- when it is not. However, consistent findings of
anisms such as Facebook “likes.” Fallacies greater magnitudes of association would bol-
such as post hoc ergo propter hoc and cum ster claims for a causative relationship.
hoc ergo propter hoc (see glossary) can also
sometimes be viewed as spurious relationships.
Consistency: Repeated observations of an
An inability to differentiate between spurious
association are made under different circum-
relationships and valid causal relationships
stances and in different populations.
may have dire consequences, which has been
amply demonstrated by the efficacy of snake As most systemic illnesses are associated with
oil and other fraudulent claims. multiple causes, a single association, such
as periodontal disease, may be statistically
significant and consistent across different pop-
Does Periodontal Disease ulations and circumstances but still may not be
enough to explain the entire causal mechanism.
Cause Systemic Illnesses? We need to recognize that periodontal disease
by itself may be neither a necessary condition
Let’s consider periodontal disease a surrogate nor a sufficient one.
for oral infections and ask whether we can
actually claim that periodontal disease causes
Specificity: A cause leads to a single effect.
systemic conditions according to the proposed
frameworks above. It is not possible to claim that periodontal dis-
ease will only cause a single systemic illness.
Furthermore, if the Bradford Hill criteria are
Bradford Hill criteria essential to establish periodontal disease as a
cause, the specificity criterion would negate
If we apply each criterion suggested by Brad- the possibility that periodontal disease may
ford Hill, we will discover many inconsistencies be the cause of several very different systemic
that would make it difficult to use these crite- outcomes. A very influential epidemiology
ria to ascertain a causal relationship between textbook published many years ago even
periodontal disease and systemic illness. Some stated that “specificity does not confer greater

12
Does Periodontal Disease Cause Systemic Illnesses?

validity to any causal influence regarding the Experiment: Experimental evidence may iden-
exposure effect … the criterion is useless and tify a factor as a potential cause when removal
misleading.”41 of the factor or a change in its frequency will
prevent the outcome.
Temporality: The cause needs to precede the
As periodontal disease has never been shown to
effect.
be a necessary or a sufficient condition for the
It is very difficult to determine the onset of development of a systemic condition, a change
periodontal disease or most systemic illnesses in a systemic outcome may be associated with
among cohorts in observational studies. Deter- confounders or effect modifiers and not the
mining the onset of oral infections may be removal of periodontal disease (Box 1-4).
more straightforward. However, randomized controlled intervention
trials, comparing individuals with successfully
treated periodontal disease with individuals
Biologic gradient: Greater exposure leads to
with unsuccessful periodontal care, where all
greater incidence of the effect.
other potential determinants or prognostic fac-
Although we use a grading system to describe tors are kept the same between the two groups,
the level of severity of periodontal disease, may provide evidence of a causal relationship.
the factors responsible for increased severity
may not be the same factors responsible for a
Analogy: Identifying a causative factor in
potential causal relationship with a systemic
one disease may strengthen the assertion of
condition.
observing similar outcomes with the same
causative factor.
Plausibility: A biologic rationale exists for an
Knowing that periodontal disease is an
association.
inflammatory condition strengthens claims
Several plausible biologic mechanisms, for for periodontal disease as a causative factor
example inflammation, can explain a causal for systemic conditions that are known to be
relationship between periodontal disease and inflammatory in nature, but many other anal-
systemic conditions. ogies could also be proposed.

Overall, if we apply the Bradford Hill cri-


Coherence: A cause-and-effect interpretation
teria to other causal frameworks, it is unclear
of an association should not conflict with what
whether a given criterion is a necessary condi-
is known about the biology and pathogenesis
tion, a sufficient condition, or a combination
of the disease.
of necessary and sufficient conditions, or
Periodontal pathogens have been found in how it can be viewed in a counterfactual or a
atheromas in individuals with coronary artery sufficient-component causal mechanism model.
disease (CAD). They have also been shown to However, as highlighted above, several of the
induce platelet aggregation, which is a stage in Bradford Hill criteria (strength, consistency,
the pathogenesis of CAD. Similar findings have temporality, biologic gradient, and plausi-
been made in individuals with other systemic bility) can be used as norms when assessing
consequences, such as adverse pregnancy out- the impact of periodontal disease on systemic
comes. The question that needs to be asked is conditions.
this: If the periodontal pathogens were removed,
would we still have the same outcomes?

13
1 Causation: Frameworks, Analyses, and Questions

Box 1-4  |  An example to determine the difference between a confounder and an effect modifier

In a hypothetical cohort study, we measured the association between the treatment of periodontal disease (TxPD+)
versus no treatment for periodontal disease (TxPD−) (exposures) with the risk of having a myocardial infarction
(MI+) (outcome). We found that TxPD− increased the risk of MI+, with a relative risk (RR) of 2.7 (95% confidence
interval [CI]: 2.2–2.9).

Initial study
MI+ MI− Risk of MI+ RR of MI+

TxPD− a b a/(a + b) a / (a + b)


= 2.7
TxPD+ c d c/(c + d) c / (c + d)

However, other cross-sectional studies have shown that smoking is also a risk factor for MI+. We now wanted
to know if smoking was responsible for our result. So we divided our study participants into two subgroups—
smokers and nonsmokers.

Scenario 1
We found that among the subgroup of smokers TxPD− increased the risk for MI+ with an RR of 3.0 (95% CI:
2.6–3.2), but among the nonsmoking subgroup TxPD− increased the risk for MI+ with an RR of only 1.3 (95%
CI: 0.9–1.6). There must have been a modification of the causal pathway of TxPD− to MI+ by smoking, as the
exposure had a different effect among different subgroups. Smoking must have been the risk factor that modified
the effect of TxPD+ to achieve the result in our original study.

Smokers
MI+ MI− Risk of MI+ RR of MI+
TxPD − a b a/(a + b) a / (a + b)
= 3.0
TxPD + c d c/(c + d) c / (c + d)

Nonsmokers
MI+ MI− Risk of MI+ RR of MI
TxPD − a b a/(a + b) a / (a + b)
= 1.3
TxPD+ c d c/(c + d) c / (c + d)

Necessary and sufficient condition periodontal disease does not always cause a
specific outcome (systemic condition). Accord-
framework
ingly, periodontal disease is neither a necessary
Periodontal disease is not a necessary condi- nor a sufficient condition.
tion, as the systemic conditions that have been Although periodontal disease, specifically
linked to periodontal disease have been asso- the presence of periodontal pathogens, has
ciated with other causes without the presence been suggested as a contributory cause of
of periodontal disease. Periodontal disease is cardiovascular disease,42 no study has ever
not a sufficient condition, as the presence of been published that fulfills the two criteria

14
Does Periodontal Disease Cause Systemic Illnesses?

Scenario 2
We found that among the subgroup of smokers TxPD− decreased the risk for MI+ with an RR of 0.9 (95% CI:
0.7–1.1), and among the subgroup of nonsmokers TxPD− decreased the risk for MI+ with an RR of 0.7 (95% CI:
0.6–1.1). Thus, our original finding that TxPD− increased the risk for MI+ (RR of 2.7 [95% CI: 2.2–2.9]) must be
incorrect. In scenario 2, the RR for MI+ with TxPD− did not change in either subgroup of smokers or nonsmokers,
and a factor that was associated with both the exposure and the outcome but does not lie in the causative
pathway—a confounder (smoking)—must have been present.

Smokers
MI+ MI− Risk of MI+ RR of MI+
TxPD − a b a/(a + b) a / (a + b)
= 0.9
TxPD+ c d c/(c + d) c / (c + d)

Nonsmokers
MI+ MI− Risk of MI+ RR of MI+
TxPD− a b a/(a + b) a / (a + b)
= 0.7
TxPD + c d c/(c + d) c / (c + d)

Scenario 3
Among the subgroup of smokers the RR with TxPD− is 2.2 (95% CI: 2.0–2.4), while among the subgroup of
nonsmokers the RR with TxPD− is 2.7 (95% CI: 2.5–2.9). This means that the smoking is neither a confounder
nor an effect modifier.

Smokers
MI+ MI− Risk of MI+ RR of MI+
TxPD− a b a/(a + b) a / (a + b)
= 2.2
TxPD + c d c/(c + d) c / (c + d)

Nonsmokers
MI+ MI− Risk of MI+ RR of MI+
TxPD − a b a/(a + b) a / (a + b)
= 2.7
TxPD+ c d c/(c + d) c / (c + d)

If, in a subgroup analysis, which is based on a suspected confounder or a suspected effect modifier, the orig-
inal association holds up in one subgroup but not in another, the factor is an effect modifier. But if the original
association between the exposure and outcome does not hold up in both subgroups, the factor is a confounder.

15
1 Causation: Frameworks, Analyses, and Questions

(temporality and treatment effect) necessary Probabilistic framework


to make such a claim.
Numerous observational studies have found
strong measures of association, such as
Sufficient-component framework absolute risk, relative risk, odds ratio, mean dif-
ference, and standardized mean difference, that
In the sufficient-component causal mechanism can be used to claim causal inference. There is
model, each and every component is necessary. no universal agreement on what is considered
As far as we know, there are no systemic condi- a strong or weak association, as there are no
tions that cannot occur without the presence of absolute numbers that could apply to all study
periodontal disease. Thus, periodontal disease designs, populations, or circumstances. How-
cannot be a single-component cause. ever, there is little disagreement that extremely
high and consistent measures of associations
(eg, odds ratios in the range of 10 to 15 or
Counterfactual framework more) can be used to make causal inferences.
For example, the odds ratio associated with
The definition of periodontal disease and the developing lung cancer in male smokers ranges
parameters used to assess periodontal disease from a low of 9 to more than 50 depending
vary greatly.43 Thus, it would be very difficult on the number of cigarettes smoked per day.44
to assess the effect of periodontal disease on But measures of association of this magnitude
systemic outcomes using a counterfactual or even close to those found in smokers that
framework, as the effect on a systemic outcome develop lung cancer have never been seen in the
may be directly determined by how periodontal literature for the periodontal-systemic link. It
disease is defined. On the other hand, because is unclear what the lowest number to make a
periodontal pathogens may affect systemic con- causal inference might be.
ditions such as coronary heart disease (CHD), It may be possible to claim that strong
it may be possible to infer that the absence of measures of association are enough to infer
periodontal pathogens would prevent or miti- causality and that there is no need to ascertain
gate the development of such conditions. RCTs a true causative effect using existing criteria or
may be able to test this hypothesis. frameworks. This may be the case when ethical
A comprehensive causal mechanism may be concerns prohibit conducting RCTs. For exam-
considered a binary outcome event scenario ple, it would be unethical to conduct a RCT to
because a set of factors, when considered ascertain whether smoking causes lung cancer.
together, cause a binary outcome event. Thus,
omission of just one factor would change the
outcome. Because many of the systemic ill-
nesses linked to oral infections can be caused
Additional Strategies to
by a multitude of mechanisms, it would not Assess Causation
be possible to single out oral infections as a
cause using a counterfactual cause model. The The difficulties associated with conduct-
same argument was put forth when discussing ing RCTs have spurred the search for better
the shortcomings of the Bradford Hill criteria. methods to determine causal inference from
For example, if the strength of an association is observational studies. Two of these methods
weak, we cannot conclude that a strong associa- are discussed in the sections that follow.
tion might have resulted in a different outcome.

16
Additional Strategies to Assess Causation

Z (the instrumental variable [IV]) is associated


with the exposure (effect) X; X is associated Z is not associated with Unmeasured
with the outcome (disease) Y; Z is indirectly (is independent of) U confounders
associated with Y through X; U, the unknown
U
or unmeasured confounder, is associated with
both X and Y.
1. Z (the IV genotype, single nucleotide poly­
Z X Y
morphisms, or allele) must not associate with
(is independent of) U (confounders [eg, age, Genotype Obesity Periodontal
sex, smoking status, education level]). disease
2. Z must reliably associate with X (obesity).
3. Z must not associate with (independent of)
Z is not associated with (is independent of) Y
Y (periodontal disease), given X and U; or,
there is no pathway from Z to Y without going
through the exposure of interest, X.
Observational studies have found an association the same risk for developing periodontal disease (the
between obesity and periodontal disease. As most of same prognostic characteristics), where one group has
these are cross-sectional studies, a causative relationship a genetic propensity to develop obesity and the other
cannot be established. An RCT where the risk for devel- group does not. If obesity is not a causal risk factor for
oping periodontal disease is compared between two periodontal disease, we would expect similar findings of
groups, one where obesity is “assigned” to a treatment periodontal disease in individuals with a genetic predis-
group and no obesity to a control group, obviously cannot position to obesity and in individuals without a genetic
be conducted to establish a causative relationship. disposition to obesity. In other words, the pathway to
It is known that specific genotypes (Z) are associated periodontal disease in those with a genotype for obesity
with obesity (X), and these genotypes are not associated and those without did not have to pass through obesity.
with other risk factors for obesity (U) or for periodontal See Shungin et al48 for more specifics about how it was
disease (Y). It is possible to determine the presence of determined that obesity was not a causal risk factor for
periodontal disease in two groups of individuals with periodontal disease.

Fig 1-3  |  Instrumental variable method to generate a causal estimate.

Instrumental variable the outcome Y through X, yet (4) there also


exists an unknown confounder (U) that directly
Instrumental variables (IVs) can be used affects both X and Y but is not affected by
as a nonconfounded proxy and control for Z46,47 (Fig 1-3). For example, if smoking (X)
threats to internal validity in observational affects periodontal disease (Y), will encourag-
studies, such as confounding variables, mea- ing or not encouraging smoking cessation (Z,
surement error, and reverse causality. When something that will directly affect only smok-
used correctly, IVs allow for the possibility of ing and not the presence of periodontal disease)
making causal inferences from the analysis of explain the changes in the outcome (presence
(cross-sectional) observational data even in the of periodontal disease)? If Z can explain the
presence of unmeasurable confounders.45 observed effect on Y, is it possible to argue for
An instrument within the IV framework is a causal relationship even if the confounder U
the Z variable that (1) has a causal effect on cannot be measured or corrected for?
X (the actual exposure or intervention), (2) For example, a recent study used IVs to
affects the outcome (Y), and (3) only affects test the popular saying, “gain a child, lose a

17
1 Causation: Frameworks, Analyses, and Questions

tooth.” The authors exploited random natu- specific characteristics of alleles—randomiza-


ral variation in family size resulting from (1) tion, lack of effect of subsequent exposures,
the birth of multiples vs singletons, and (2) and lack of effect of subsequent exposures
the sex composition of the two firstborn chil- on the fixed genotype—can be exploited to
dren (increased likelihood of a third child if discover the effects of confounders in obser-
the two firstborn children are the same sex). vational studies.
The findings suggest that an additional birth There are several limitations of epidemio-
might indeed be detrimental to the mother’s logic studies that thwart attempts at making
oral health but not the father’s.49 causal inferences. Some of these limitations can
The IV method is similar to the counter- be mitigated with properly and appropriately
factual model of causation but differs in that conducted RCTs. The presence of confound-
Z is an actual intervention and not part of a ers and reverse causation are acknowledged as
thought experiment.47 In purely observational some of the main reasons for the discrepancy
studies (in which neither the instrument Z nor between an inference of causal relationships
the treatment X has been subject to experimen- obtained from observational data analysis and
tal manipulation), a major limitation of all IV study outcomes from RCTs. A novel type of
methods is their strong reliance on the assump- investigation, the Mendelian randomized (MR)
tion that Z is truly independent of U and Y. method, involves finding alleles that are asso-
ciated with a specific exposure and examining
the association between these genetic variants
Mendelian randomization, an and the outcome. The randomization of alleles
instrumental variable construct makes MR akin to an RCT (Fig 1-4).50
The basic premise of MR is that genotypes
As Gregor Mendel discovered during his exper- are generally not susceptible to the confound-
iments with genes for pod colors in pea plants, ing found in observational studies.51,52 The
different forms of a specific gene—alleles—were underlying concept of MR is to take advan-
associated with altered observable phenotypes. tage of situations where genotypic differences
An allele is one of a pair or series of genes produce effects similar to environmental fac-
that appears at a specific location (locus) on a tors (and vice versa) and use genetic variants
particular chromosome and controls the same as proxies for environmental exposures rather
characteristic, such as eye color. A change in than measure the exposures themselves. This
allele at a specific genetic loci is called a genetic is similar to IV analysis where the allele is the
variant and is responsible for observable phe- IV (see Fig 1-4).
notypical traits. Humans have two alleles at This methodology has already been used
each genetic position, one inherited from each to determine how confounders may affect the
parent. Mendel’s law of segregation states that association between biomarkers and clinical
alleles for a trait separate when gametes (cells outcomes, such as C-reactive protein (CRP) and
that carry half of the genetic information of incident coronary events. Observational studies
the parent from which they are inherited) are have suggested an association between the pres-
formed through meiosis. These allele pairs are ence of CRP and an elevated risk for CHD, but
then randomly united at fertilization. Thus, when the MR method (using genetic proxies for
alleles are produced randomly. Alleles are not CRP concentration) was employed it failed to
associated with subsequent behavioral, social, confirm that CRP is a risk factor for CHD.53,54
or environmental influences, and subsequent In other studies using MR, a causal relationship
behavioral, social, or environmental exposures could be confirmed between individuals who
cannot change the fixed genotypes as these are were homozygote for an allele associated with
determined at the time of conception. These higher plasma homocysteine and stroke.55

18
Conclusion

Mendelian randomization RCT

Random segregation of alleles Randomized method

Exposure: Exposure: Exposure: Exposure:


alleleEa1 alleleEa2 interventionEi1 interventionEi2

All prognostic All prognostic


characteristics are characteristics are
the same the same
between groups between groups

Compare results Compare results

Fig 1-4  |  Comparison of Mendelian randomization with RCTs. (Adapted from Smith and Ebrahim.50)

MR was performed to assess the associa- Conclusion


tion between obesity (measured by body mass
index) and periodontal disease (clinically This chapter does not provide an exhaustive list
assessed and self-reported) in a multicentered of methods and concepts used in the context of
cohort of more than 49,000 individuals.48 This causal inference. Some constructs and frame-
study could not support a causal relationship works not mentioned in this chapter include
between absolute adiposity and periodontitis. directed acyclic graphs, regression-discontinuity
Using MR to investigate a causal relation- design, difference-in-difference analysis, and
ship between oral infections and systemic propensity score matching.2,56,57
health would provide important information Furthermore, the frameworks, methods, and
to help ascertain the veracity of such an asso- criteria discussed in this chapter cannot account
ciation. However, as with other frameworks for all possible ways cause operates. Awareness
used to assess causality, MR is not a panacea to of the difficulties in establishing causality and
determine causal relationships, and challenges recognition that many different concepts are
remain to interpreting MR analyses.52 used in the scientific literature will enhance the
discussion on this important topic. However,
when a claim of causal inference is made, it is
important to know which framework, method,

19
1 Causation: Frameworks, Analyses, and Questions

or analysis was used to reach that conclusion. as a disease, that is statistically dependent;
How else can a claim be judged? that is, the presence of one alters the proba-
The benefits of establishing a true caus- bility of observing the presence of the other.
ative relationship between oral infections and causal inference  Use of a measurement of
systemic outcomes are clear—improved pre- an effect.
vention strategies, better prognostic assessment causative chain with component cause  Mul-
of disease development, better treatment strate- tiple, joint causes for the occurrence of the
gies, and overall improved patient benefits, just outcome.
to mention a few. But what would be the down- component cause  An event required for an
side of claiming a causative relationship where outcome to develop.
none exists? Resources that could be spent on confounder  A factor that is associated with
beneficial health strategies would be wasted; both the exposure and the outcome but
patient expectations, such as reducing adverse does not lie in the causative pathway. Con-
pregnancy outcomes by periodontal therapy or founders should be eliminated to prevent
preventing heart attacks and strokes, would not misrepresentation of research findings. Con-
be met; and there may be legal ramifications founders differ from effect modifiers (see
from arguing that expenditures should be made effect modifier).
to achieve an outcome that cannot be realized. confounding  More than one possible indepen-
The inability to determine causal infer- dent variable (cause) acting at the same time.
ences should not deter us from recognizing correlation  A statistical measure of an asso-
the associations that have already been estab- ciation; linear association between two
lished between oral infections and systemic continuous or ordinal variables. A correla-
health. These associations should serve as an tion is not sufficient to claim causation.
important springboard to improve interpro- counterfactual  Nonexistent or false. An out-
fessional education, practice, and research and come, after exposure to a determinant or an
to provide better and more science-based pre- agent of change, is compared in a thought
vention, interventions, and reimbursement in experiment with the same outcome but with-
a value-based health care model. But we must out a determinant or an agent of change. For
be careful not to overstate the impact of these example, suppose you examine a patient and
associations. In the end, it might be worthwhile discover she has a 6-mm pocket depth on
to take note of (but not necessarily agree with) the mesial side of the mandibular right first
Bertrand Russell’s claim that “the law of cau- molar, so you decide to insert a biodegrad-
sality, I believe, like much that passes muster able chip containing a new pharmaceutical
among philosophers, is a relic of a bygone age, agent in the pocket. After 1 month, her
surviving, like the monarchy, only because it is pocket depth is only 3 mm. Did this outcome
erroneously supposed to do no harm.” occur because of the chip? You compare the
outcome—reduced pocket depth—which
you observed, with the counterfactual out-
come, which you would have observed if
Glossary you had chosen not to insert the chip in the
pocket (a thought experiment). If you think
antecedent  The “if” portion of a counterfac- the outcome would have been different—no
tual or hypothetical condition. reduction in pocket depth—if a chip had not
antecedent event  “Going before,” or an been inserted, you will conclude that the chip
event preceding another event. caused the reduction in pocket depth.
association  A relationship between an expo- cum hoc ergo propter hoc  Latin for “with
sure or characteristic and an outcome, such this, therefore because of this.” This fallacy

20
References

is commonly used to infer a causal relation- post hoc ergo propter hoc  Latin for “after
ship when two events occurring together are this, therefore on account of this.” This
taken to have a cause-and-effect relationship. fallacy is commonly used to infer a causal
However, such a temporal relationship does relationship when an event that follows
not by itself imply a causative relationship. another is a necessary consequence of the
As an example, we can infer that wearing first event. However, such a temporal rela-
bathing suits causes consumption of ice tionship does not by itself imply a causative
cream as there is an observed association relationship. An example is that the crowing
between the presence of wearing bathing of a rooster (an antecedent event) is neces-
suits and ice cream consumption. sary for the sun to rise because sunrise is
effect modifier  An exposure that has a differ- always preceded by the crowing.
ent effect among different subgroups and lies risk factor  A factor that is acknowledged as
in the causal pathway. Effect modifiers are a determinant (brings about a change), a
not associated with the exposure but with cause, or a sufficient and component cause
the outcome. An effect modifier (a third fac- of disease.
tor) can be used to change the magnitude of sin qua non  Latin for “without which not.”
the effect. Effect modifiers differ from con- This phrase is sometimes used to imply a
founders (see confounder). necessary condition.
event  Any assignment of a value or set of val- sufficient and component cause  Multiple
ues to a variable or a set of variables; specific determinants operating sufficiently and
occurrence. jointly to form one or more causal mecha-
external validity  Refers to how well data and nisms, each of which is capable of producing
theories from one setting apply to another; or contributing to a given outcome (disease)
also known as generalizability or the exten- (see Fig 1-2).
sion of research findings and conclusions sufficient condition  If X is sufficient for Y
from a study conducted on a sample or study (sufficient condition), it means that if X is
population to the population at large. present, the outcome Y will always occur.
necessary condition  If X is necessary for Y However, the outcome may occur without
(necessary condition), it means that X must the condition. (For examples, see Fig 1-1.)
be present for Y to happen. In other words, variable  Any property or descriptor that can
a necessary condition (X) means that the take multiple values. A variable can be dis-
outcome (Y) can never happen without the crete (also known as categorical) (eg, sex) or
condition (X). However, the condition can continuous (eg, age), independent (the pre-
occur without the outcome. (For examples, dictor) or dependent (the outcome).
see Fig 1-1.)

References 5. Maldonado G, Greenland S. Estimating causal effect. Int


J Epidemiol 2002;31:422–429.
1. Glick M. Causation: A loosely founded concept in epi- 6. Hernán MA, Robins JM. Estimating causal effects from
demiology. J Am Dent Assoc 2007; 138:1532–1533. epidemiological data. J Epidemiol Community Health
2. Listl S, Jürges H, Watt RG. Causal inference from obser- 2006;60:578–586.
vational data. Community Dent Oral Epidemiol 2016; 7. Glick M. A cause célèbre: Can we agree on a common
44:409–415. definition or model for causation? J Am Dent Assoc 2017;
3. Worrall J. Causality in medicine: Getting back to the Hill 148:863–865.
top. Prev Med 2011;53:235–238. 8. Denton GB. A new interpretation of a well-known Assyr-
4. Bothwell LE, Greene JA, Podolsky SH, Jones DS. ian letter. J Near Eastern Studies 1943;4:314–315.
Assessing the gold standard—Lessons from the history 9. Miller WD. The human mouth as a focus of infection.
of RCTs. N Engl J Med 2016;374:2175–2181. Dental Cosmos 1891;33:689–713,789–804,913–919.

21
1 Causation: Frameworks, Analyses, and Questions

10. Miller WD. The human mouth as a focus of infection. 31. Fedak KM, Bernal A, Capshaw ZA, Gross S. Applying
Lancet 1891;138:340–342. the Bradford Hill criteria in the 21st century: How data
11. Cecil RL, Angevine DM. Clinical and experimental obser- integration has changed causal inference in molecular
vations on focal infection with an analysis of 200 cases of epidemiology. Emerg Themes Epidemiol 2015;12:14.
rheumatoid arthritis. Ann Intern Med 1938;12:577–584. 32. Rothman KJ, Greenland S. Causation and causal infer-
12. Reimann HA, Havens WP. Focal infection and systemic ence in epidemiology. Am J Public Health 2005;95(suppl
disease: A critical appraisal. The case against indiscrim- 1):S144–S150.
inate removal of teeth and tonsils. JAMA 1940;114:1–6. 33. Barthold PM, Mariotti A. The future of periodontal-
13. Burket LW. Oral Medicine: Diagnosis and Treatment. systemic associations: Raising the standards. Curr Oral
Philadelphia: Lippincott, 1946. Health Rep 2017;4:258–262.
14. Easlick KA. An evaluation of the effect of dental foci of 34. Linden GJ, Herzberg MC, Working group 4 of the
infection on health. J Am Dent Assoc 1951;42:615–697. joint EFP/AAP workshop. Periodontitis and systemic
15. Mattila KJ, Nieminen MS, Valtonen VV, et al. Association diseases: A record of discussions of working group 4
between dental health and acute myocardial infarction. of the Joint EFP/AAP Workshop on Periodontitis and
BMJ 1989;298:779–781. Systemic Diseases. J Clin Periodontol 2013; 40(suppl
16. Shlossman M, Knowler WC, Pettitt DJ, Genco RJ. Type 14):S20–S23.
2 diabetes mellitus and periodontal disease. J Am Dent 35. de Smit MJ, Westra J, Brouwer E, Janssen KM, Vissink
Assoc 1990;121:532–536. A, van Winkelhof AJ. Periodontitis and rheumatoid
17. Nelson RG, Shlossman M, Budding LM, et al. Periodon- arthritis: What do we know? J Periodontol 2015; 86:
tal disease and NIDDM in Pima Indians. Diabetes Care 1013–1019.
1990;13:836–840. 36. Riegelman, R. Contributory cause: Unnecessary and
18. Collins JG, Windley III HW, Arnold RR, Offenbacher S. insufficient. Postgrad Med 1979;66:177–179.
Effects of a Porphyromonas gingivalis infection on inflam- 37. Rothman KJ, Greenland S, Lash TL. Modern epidemiol-
matory mediator response and pregnancy outcome in ogy, ed 3. Philadelphia: Wolters Kluwer Health/Lippincott
hamsters. Infect Immun 1994;62:4356–4361. Williams & Wilkins, 2008.
19. Offenbacher S, Katz V, Fertik G, et al. Periodontal infec- 38. Rothman KJ. Causes. Am J Epidemiol 1976; 104:
tion as a possible risk factor for preterm low birth weight. 587–592.
J Periodontol 1996;67:1103–1113. 39. Pearson K. Mathematical contributions to the theory of
20. Offenbacher S, Jared HL, O’Reilly PG, et al. Potential evolution—On a form of spurious correlation which may
pathogenic mechanisms of periodontitis associated arise when indices are used in the measurements of
pregnancy complications. Ann Periodontol 1998; 3: organs. Proc R Soc Lond 1897;60:489–498.
233–250. 40. Messerli FH. Chocolate consumption, cognitive func-
21. Monsarrat P, Blaizot A, Kémoun P, et al. Clinical research tion, and Nobel laureates. N Engl J Med 2012; 367:
activity in periodontal medicine: A systematic mapping 1562–1564.
of trial registers. J Clin Periodontol 2016;43:390–400. 41. Rothman KJ, Greenland S. Modern Epidemiology, ed 2.
22. Cause. The Free Dictionary. https: //www.thefreedictio- Philadelphia: Lippincott Williams and Wilkins, 1998:22.
nary.com/cause. Accessed 31 December 2017. 42. Bale BF, Doneen AL, Vigerust DJ. High-risk periodontal
23. Cause. Merriam-Webster. https: //www.merriam– pathogens contribute to the pathogenesis of atheroscle-
webster.com/dictionary/cause. Accessed 31 December rosis. Postgrad Med J 2017;93:215–220.
2017. 43. Manau C, Echeverria A, Agueda A, Guerrero A, Eche-
24. Iacopino AM. Periodontitis and diabetes interrelationships: verria JJ. Periodontal disease definition may determine
Role of inflammation. Ann Periodontol 2001;6:125–137. the association between periodontitis and pregnancy
25. Parascandola M1, Weed DL. Causation in epidemiology. outcomes. J Clin Periodontol 2008;35:385–397.
J Epidemiol Community Health 2001;55:905–912. 44. Pesch B, Kendzia B, Gustavsson P, et al. Cigarette smok-
26. Koch’s postulates. Mosby’s Medical Dictionary, ed 8. ing and lung cancer: Relative risk estimates for the major
2009. https://medical-dictionary.thefreedictionary.com/ histological types from a pooled analysis of case-control
Koch%27s+postulates. Accessed 31 December 2017. studies. Int J Cancer 2012;131:1210–1219.
27. Hill AB. Principles of medical statistics. I. The aim of the 45. Martens E, Pestman W, de Boer A, Belitser SV, Klungel
statistical method. Lancet 1937;229:41–43. OH. Instrumental variables: Applications and limitations.
28. Doll R. Controlled trials: The 1948 watershed. BMJ 1998; Epidemiology 2006;17:260–267.
317:1217–1220. 46. Greenland S. An introduction to instrumental variables for
29. Hill AB. The environment and disease: Association or epidemiologists. Intern J Epidemiol 2000;29:722–729.
causation? Proc R Soc Med 1965;58:295–300. 47. Hernán MA, Robins JM. Instruments for causal infer-
30. United States Department of Health, Education and ence: An epidemiologist’s dream? Epidemiology 2006;
Welfare. Smoking and Health: Report of the Advisory 17:360–372.
Committee to the Surgeon General of the Public Health
Service. Washington, DC: United States Public Health
Service, 1964.

22
References

48. Shungin D, Cornelis MC, Divaris K, et al. Using genet- 53. Elliott P, Chambers JC, Zhang W, et al. Genetic loci asso-
ics to test the causal relationship of total adiposity and ciated with C-reactive protein levels and risk of coronary
periodontitis: Mendelian randomization analyses in the heart disease. JAMA 2009;302:37–48.
Gene-Lifestyle Interactions and Dental Endpoints (GLIDE) 54. C Reactive Protein Coronary Heart Disease Genetics Col-
Consortium. Int J Epidemiol 2015;44:638–650. laboration (CCGC), Wensley F, Gao P, et al. Association
49. Gabel F, Jürges H, Kruk KE, Listl S. Gain a child, lose between C reactive protein and coronary heart disease:
a tooth? Using natural experiments to distinguish Mendelian randomisation analysis based on individual
between fact and fiction. J Epidemiol Community Health participant data. BMJ 2011;342:d548.
2018;72:552–556. 55. Casas JP, Bautista LE, Smeeth L, Sharma P, Hingorani
50. Smith DG, Ebrahim S. What can Mendelian ran- AD. Homocysteine and stroke: Evidence on a causal
domization tell us about modifiable behavioural and link from Mendelian randomization. Lancet 2005; 365:
environmental exposures? BMJ 2005;330:1076–1079. 224–232.
51. Zheng J, Baird D, Borges MC, et al. Recent devel- 56. Dimick JB, Ryan AM. Methods for evaluating changes
opments in Mendelian randomization studies. Curr in health care policy: The difference-in-differences
Epidemiol Rep 2017;4:330–345. approach. JAMA 2014;312:2401–2402.
52. Holmes MV, Ala-Korpela M, Smith GD. Mendelian ran- 57. Moscoe E, Bor J, Bärnighausen T. Regression disconti-
domization in cardiometabolic disease: Challenges in nuity designs are underutilized in medicine, epidemiology,
evaluating causality. Nat Rev Cardiol 2017;14:577–590. and public health: A review of current and best practice.
J Clin Epidemiol 2015;68:122–133.

23
CHAPTER 2

Essential Statistical and Research


Design Elements to Help Critically
Interpret the Literature
Barbara L. Greenberg, msc, phd
Michael Glick, dmd

Clinical researchers, translational researchers, elements used to assess the literature on the
and consumers of scientific literature strive link between oral and systemic health, includ-
to inform and guide clinical care and health ing determination of the clinical importance
policy development to optimize the health and of study results.
well-being of patients and populations. An There is a misconception that if something
overwhelming amount of scientific literature is is statistically significant it is also clinically
published every day on the connection between important, and if something is not statistically
oral and systemic disease. For example, a significant it is not clinically important. This
search of the literature on the link between is not necessarily true. This chapter presents
periodontal disease, diabetes, and cardiovas- the basic concepts that are essential for under-
cular disease suggests that, on average, one new standing and determining statistical significance
article has been indexed in PubMed every day and clinical importance. The terms clinically
for the past 10 years (Fig 2-1). Unfortunately, meaningful, clinically important, and clinically
many of these articles report studies with dif- relevant are often used synonymously; in this
ferent and even dichotomous results. Yet oral chapter these terms are collectively referred to
health care professionals have a responsibility as clinically important. Clinical importance
to interpret and use this scientific information is distinct from clinical significance, which is
to inform the practice of dentistry and affect defined later in the chapter.
the overall health of patients. A discussion of statistical and clinical
The clinical literature produced is unfortu- importance first assumes that the study (1) is
nately not of equal quality, and merely being answering an important question, (2) is well
published does not ensure quality. Producers designed, (3) is well implemented, and (4) is
of scientific literature have a responsibility properly analyzed. However, the discussion
to appropriately and thoroughly report and must also include, among other things, P val-
interpret results, while consumers of scientific ues, α level, confidence intervals (CIs), minimal
literature have a responsibility to critically read clinically important differences (MCIDs), and
and understand the literature. Thorough assess- effect size. The purpose of this chapter is to
ment of the research quality of the literature facilitate the interpretation of these fundamen-
is a complex, multifaceted endeavor that in its tal elements for understanding and determining
entirety is beyond the scope of this chapter. statistical and clinical importance when con-
Instead, this chapter focuses on several critical suming scientific literature.

24
Overview of Statistical Significance

400

350

Number of publications indexed on PubMed


50% since
2008
300

250

200

150

100

50

0
49

53

58

62

66

70

74

78

82

86

90

94

98

02

06

10

14

18
19

19

19

19

19

19

19

19

19

19

19

19

19

20

20

20

20

20
50% 50%
Year

Fig 2-1  |  Periodontal disease, diabetes, and cardiovascular disease in humans: number of PubMed citations
by publication year as of March 15, 2018 (N = 6,343). For the past 10 years, one publication has been indexed
in PubMed almost every day on the links between periodontal disease and diabetes and between periodontal
disease and cardiovascular disease, with 50% of the total published in 2008 or later.

Overview of Statistical submission for publication, there is a growing


sentiment among biostatisticians that research
Significance should move away from reporting only statisti-
cal significance and P values and instead focus
The purpose of research is to make inferences on CIs and effect size to assess precision and
from a study sample that can be applied to clinical importance.1,2
the total population, whether it is to determine Most research is hypothesis driven, with
if there is an association between a risk fac- a null hypothesis (H0) that states there is no
tor or characteristic and the development of statistically significant association between
disease, to determine treatment efficacy, or to a factor and an outcome and an alternate
derive appropriate inferences about a causal hypothesis (H1) that states there is a statistically
relationship. Statistical significance testing significant association. The alternate hypothesis
is the process of reaching conclusions about is what the researcher is predicting or expect-
characteristics of a larger population using ing. The null hypothesis is what the researcher
data from a subset, or sample, of that pop- tries to disprove; statistics test the null hypoth-
ulation, and it is the fundamental approach esis of no association. Although this may sound
for making such inferences. Although statis- counterintuitive, it is similar to the approach
tical significance testing is commonly used used in the United States judicial system when
and is sometimes required for acceptance of a someone is accused of a crime. The accused is

25
2 Essential Statistical and Research Design Elements to Help Critically Interpret the Literature

presumed innocent, and the jury is asked to How Do We Determine


reject or not reject this “hypothesis.”
Based on the outcome of the statistical test, Statistical Significance?
the null hypothesis is rejected or not rejected.
When a statistical test result for a given study Hypothesis testing is based on the α level and
is significant, the null hypothesis is rejected; P value. The α level is the level of statistical sig-
when the test result is not significant, the null nificance that is set a priori by the investigator.
hypothesis is not rejected. The null hypothesis By convention, for epidemiologic observational
is not the opposite of the research hypothesis; and clinical trials, the α level is typically set
the null hypothesis is “no association.” A test at .05. However, there may be circumstances
of statistical significance answers the following where multiple null hypotheses are being tested
question: If the null hypothesis were true, what and one wants to minimize type I errors, for
is the probability that a difference (or an asso- example in genome-wide association studies
ciation) at least as big (or as strong) as the one (GWASs). In such instances, the α level may be
observed would occur, assuming that chance is set at much smaller values depending on what
operating alone? is being investigated. In the case of GWASs, the
A statistically significant result suggests that α level may be 10−8 or even smaller.4 A type I
the observed association is not likely assum- error is the incorrect rejection of a true null
ing chance alone is operating and is evidence hypothesis, also known as a false positive find-
to infer a real association between a predictor ing. A type II error is the incorrect failure to
and an outcome. Remember, we are studying reject the null hypothesis when it is false, also
a sample drawn from a larger population and known as a false negative finding (Fig 2-2).
we are interested in whether what we observe The α level can be defined as the probability
in the study sample is also happening in the of rejecting the null hypothesis when in fact
population the sample is supposed to represent. it is true or the risk of concluding there is an
Statistical significance provides a measure to association when none exists. An α level of
help us make that decision. .05 means there is a 5% probability that one
In summary, significance testing addresses will incorrectly reject the null hypothesis of no
only the yes/no question of whether there is association and conclude there is an association
a statistically significant association that is when in fact there is none. In other words, by
unlikely to be due to chance alone. Statistical predetermining an α level of .05, we accept
testing is a formal process for determining the that there is a 5% probability of reaching a
probability of obtaining the observed study false positive result. When testing the efficacy
results or an outcome more extreme given that of two drugs or treatments, the P value is used
the null hypothesis is true; it is not an indica- to determine if the difference is statistically sig-
tion of the probability of a real association. But nificant. In this instance, a P value that is not
it is important to keep in mind that study out- significant (P ≥ α) indicates that for the given
comes, even when determined to be statistically study data, the difference in the two drugs or
significant, may not confer any clinical impor- treatments is not statistically significant. That
tance or may not be scientifically meaningful.3 should not be interpreted to mean that the two
drugs or treatments are the same; the correct
interpretation is that there is insufficient evi-
dence to conclude that the drugs or treatments
are different. Studies designed specifically to
assess if two drugs or treatments are the same
often require very large sample sizes.

26
How Do We Determine Statistical Significance?

Study on the impact of periodontal treatment (TxPD) on adverse pregnancy outcomes (APO)
Hypothesis (H1) = TxPD will result in fewer APO
Hypothesis (H0) = TXPD will not affect APO

True difference
Study outcome after
providing periodontal treatment No impact
Fewer APO;
on APO;
H0 “false”
H0 “true”

Reject H0 Correct Type I error

Not reject H0 Type II error Correct

Fig 2-2  |  Example of hypothesis testing with type I and type II errors.

The P value is the probability that the study data are likely under the null hypothesis
observed result, or one that is more extreme, and there is insufficient evidence to reject the
differs from the null hypothesis (no association), null hypothesis; the study results are considered
assuming chance is operating alone and the null not statistically significant. With a significant P
hypothesis is true. The P value evaluates how value, the null hypothesis is rejected; with a P
well the study data support the null hypothe- value that is not statistically significant, the null
sis and answers the question of how likely the hypothesis is not rejected.
observed effect is in the given study sample data While statistical significance based on the P
if the null hypothesis is true. There are many value is a ubiquitous concept in the literature,
reasons for a difference between the observed there are a number of pitfalls and limitations
data and the null hypothesis if the null hypoth- of the P value, and incorrect interpretations
esis is true. As it is not known why a difference of it are very common.5 The P value is not a
may exist, the P value is the computed proba- measure of support for the alternate hypothe-
bility assuming chance was operating alone or sis that there is an association or difference; a
merely the probability that the observed out- statistically significant finding does not mean
come is due to chance.5 To determine statistical that the alternate hypothesis is accepted, nor is
significance, the calculated P value is assessed it the probability of mistakenly rejecting a true
relative to the a priori set α level. A P value null hypothesis. The P value addresses only the
less than the α level (P < α) indicates that the question of how likely the study data are given
observed study results are not likely to be due the null hypothesis.
to chance alone and suggests that the study In summary, the P value is a point estimate
data provide sufficient evidence to reject the that assesses how well the data support the null
null hypothesis; the study results are considered hypothesis of no association; it is not a measure
statistically significant. A P value greater than of support for the alternate hypothesis. The P
or equal to the α level (P ≥ α) indicates that the value does not provide information about how

27
2 Essential Statistical and Research Design Elements to Help Critically Interpret the Literature

TABLE 2-1  |  Questions answered by P value versus CI

Description Questions answered


P value A calculated probability • How well do the study sample data support the null hypothesis? Do
we reject or not reject the null hypothesis?
• Is there a statistical association? How likely are the observed results or
a more extreme result assuming chance is operating alone?

CI A calculated parameter ± a margin of errora • What is the size of the association or treatment difference?
• How precisely did the given study or trial estimate the association or
treatment effect?
• Is there a statistical association?

The margin of error determines the width of the CI.


a

well the study was designed or conducted, nor The CI is a range of values that attempts
does it provide a measure of the strength of to quantify the level of uncertainty that this
an association or difference. The P value alone range will include the desired true population
should not be the primary influence for clinical value as determined by a specific parameter.
interpretation or application of study results to Examples of a parameter include an arithmetic
clinical decision making. mean, a difference, an odds ratio, or a relative
risk, each of which will have a certain calcu-
lated value. The desired true parameter for a
How Do We Evaluate Clinical population is what we try to infer from the
result of a sample from the population. Thus,
Importance? CIs are based on sample data and give ranges
of plausible values for the true parameter. The
As noted earlier, a study outcome can be statis- CI consists of the point estimate ± a margin
tically significant but not clinically important of error (level of precision, which is depicted
or meaningful. Likewise, a study outcome that as the width of the CI) that provides a lower
is not statistically significant can be clinically and an upper confidence limit around the point
important or meaningful. The concepts used estimate. The lower and upper confidence lim-
beyond the P value include CI, MCID, and its delineate the range for the estimate, and
effect size. the difference between the lower and the upper
limit indicates the width of the CI. The width
of the CI depicts the magnitude of the effect. A
Confidence interval wide interval has more uncertainty (usually due
The CI is used to obtain information beyond to a small sample size or a high degree of mea-
the statistical significance of a result and to surement variability), but the results may still
begin to assess clinical importance. The CI be statistically significant though not always
represents the magnitude of the association clinically important. The wider the interval, the
or difference observed and the precision of less precise the estimate and the more difficult it
the estimate of the association or difference, is to make a clinical interpretation. Figures 2-3
or how much uncertainty is associated with and 2-4 show graphic representations of CIs.
the point estimate from the given study data. Typically, the CI calculated is the 95% CI.
This is in contrast to the P value, which only This means that if a study is repeated indef-
answers the yes/no question of whether the initely and a 95% CI is placed around each
observed association or treatment difference sample parameter, 95% of the intervals will
is statistically significant (Table 2-1). contain the true (population) parameter. In

28
How Do We Evaluate Clinical Importance?

Clinical assessment of marginal bone loss and gain after the use of an experimental bone
augmentation material used in six different studies

Harmful Unclear Beneficial

Almost certainly beneficial

Probably beneficial
Probably harmful

Very likely trivial


Almost certainly
harmful
Unclear
–1.0 –0.8 –0.6 –0.4 –0.2 0 0.2 0.4 0.6 0.8 1.0

Bone loss Bone gain

No change

The box is the point estimate of the CI. The CI has a lower and an upper limit, represented by the vertical lines at the end of each horizontal line.
The distance between the lower and upper limit represents the width of the CI. The determination of what is harmful, trivial, or beneficial is based
on prior clinical knowledge.

Fig 2-3  |  Example of CI and effect. (Adapted from Page.6)

other words, the 95% CI contains the true outcome from data comparing a group of
value in 95 of 100 studies performed. Thus, pregnant women who received periodontal
with the 95% CI there is a 95% probability treatment with a group of pregnant women
that an interval calculated from the sample data who did not receive periodontal treatment. If
contains the true population value; it does not the quotient of the estimated risk of developing
mean that there is a 95% chance that the true an adverse pregnancy outcome in the group
population value lies within the calculated CI of pregnant women who received periodontal
for any given study. To provide more certainty treatment divided by the estimated risk in the
that the interval contains the true population group of pregnant women who did not receive
value, the 99% CI could be calculated; how- periodontal treatment equals 1 (the risk ratio
ever, this interval is wider but less precise than equals 1), we can determine that there is no
the 95% CI. For any calculated CI (eg, 95% or difference (1, in this case, is the null value).
99%), the wider the interval is, the less precise A CI that does not contain the null value is
it is; a wider CI implies poorer precision. considered statistically significant and will cor-
For any given parameter of interest, there is a respond to a significant P value (P < .05); a CI
“no effect” or “null value,” the value indicating that contains the null value is considered not
there is no association, no effect, or no differ- statistically significant and will correspond to a
ence. For a ratio the null value is 1, and for a calculated P value that is not statistically signif-
difference the null value is 0 (see Table 2-2). icant (P ≥ .05). A CI that does contain the null
For example, a study is performed to determine value indicates that the true population param-
the risk of developing an adverse pregnancy eter could indeed be equal to the null value so

29
2 Essential Statistical and Research Design Elements to Help Critically Interpret the Literature

Marginal bone loss and gain after the use of an experimental bone
augmentation material used in five different studies (A–E)

A
Not statistically significant; not clinically important
B
Statistically significant; clinically important
C
Statistically significant; probably clinically important
D
Not statistically significant; not clinically important
E
Not statistically significant; potentially clinically important

Difference
–0.4 –0.2 0 0.2 0.4 0.6 0.8 1.0 in mm

Relevance limit (MCID)


Lines represent lower and upper limits of CI and mean differences.
Guidelines for determining level of clinical importance:
• While the “0” difference will aid in determining the statistical difference (if the CI crosses the 0, we state that there is no statistical significance),
the MCID will aid in determining clinical importance.
• Definitely not clinically important: study A—the MCID lies within the CI but is larger than the study point estimate; study D—the MCID lies outside
the upper limit of the CI and is larger than the study point estimate.
• Probably clinically important: study C—the MCID lies within the interval of the CI and is smaller than the study point estimate.
• Potentially clinically important: study E—the MCID lies within the interval of the CI and is smaller than the study point estimate, but the wide CI
also suggests potentially no clinical importance. (The difference between study C and study E is the width of the CI, and calling one “probable”
and one “potential” is a semantic difference where “probable” denotes more certainty than “potential.”)
• Definitely clinically important: study B—the MCID lies outside the lower limit of the CI and is smaller than the study point estimate.

Fig 2-4  |  Example of CI, significance, and MCID.

TABLE 2-2  |  Assessing statistical significance using CIs and P values

Null Assessing significance using Assessing significance using


Parameter value the CI the corresponding P value
Difference: mean difference, CI contains 0 = not significant P ≥ .05
standardized mean difference, 0
risk difference, rate difference CI does not contain 0 = significant P < .05

Ratio: relative risk, odds ratio, CI contains 1 = not significant P ≥ .05


1
rate ratio CI does not contain 1 = significant P < .05

that one cannot reject the null hypothesis of no Overlapping CIs do not necessarily mean there
association. Table 2-2 shows the null value for is no statistical significance.
a given parameter and guidelines for assessing In addition to the calculated CI, there are
statistical significance with the CI and P value. key clinical indicators set by the investigator or
accepted among clinicians that help determine

30
How Do We Evaluate Clinical Importance?

the clinical importance of study results. These harmful effect size would be considered harm-
include MCID and effect size. ful or potentially harmful. A CI that includes
the null value and crosses the threshold for a
beneficial and harmful effect would be con-
MCID and effect size sidered unclear. Figure 2-3 presents a graphic
The MCID is the smallest change considered representation of CI and effect size.
meaningful by a practitioner or patient and/ Similarly, if the CI contains or is less than
or that would result in a change in patient the MCID, the results would not be clinically
management. There are no standards for deter- important because the true population param-
mining the MCID other than clinical judgment eter could be less that the MCID. If the CI is
or widely accepted clinical standards for a greater than the relevance limit or MCID, the
given outcome of interest. result is considered clinically important because
The effect size is the magnitude of the desired this indicates the true population parameter is
difference between groups or the magnitude of greater than the MCID. A graphic represen-
the desired observed association that is consid- tation of the CI and the MCID is shown in
ered clinically important. There are different Fig 2-4.
measures of effect, including means, standard- As an example, a randomized controlled
ized means, relative risks, odds ratios, and trial (RCT) that assessed the effect of non-
absolute risks. The desired or expected effect surgical periodontal treatment reported a
size should be established at the outset of the statistically significant improvement in the
study, reported in the manuscript, and when metabolic control (P = .019) between indi-
appropriate (ie, in comparing results with dif- viduals with type 2 diabetes receiving therapy
ferent measurement units), standardized across compared with individuals not receiving the
the different units of measurement. The larger therapy.7 Using available data provided in the
the effect size, the easier it is to find statistical article, 95% CIs placed around the reported
significance. The determination of the desired sample means could be calculated. After 6
effect size is based on previous literature and/ months, the treatment group had a reported
or clinical knowledge. When setting the desired mean fasting plasma glucose of 150.02 mg/dL
effect size, important considerations include a (standard deviation [SD] 49.54; n = 42) com-
change that is of interest to patients or clini- pared with 167.95 mg/dL (SD 41.90; n = 48)
cians, a change in an important outcome (eg, for the control group. The calculated 95% CI
any outcome that may alter treatment deci- was 135.04 to 165.00 in the treatment group
sions), and the associated risk-benefit ratio. and 156.10 to 179.80 in the control group.
How should these results be interpreted? For
the purpose of this study question and given
Assessing clinical importance these data, the CIs are considered sufficiently
using the CI and effect size or MCID narrow. This would suggest that the data are
sufficiently precise. However, the difference
The CI indicates the magnitude and direction between the resulting CIs does not seem to be
of the effect. In addition to statistical signifi- clinically important, as neither group achieved
cance, the CI in conjunction with the a priori metabolic control (< 126 mg/dL); there is an
effect size can be used to assess whether the overlap between the two CIs, and the differ-
given study results are trivial, beneficial, or ences between the lower limits and between the
harmful. A CI that is greater than the null upper limits do not seem clinically important
value and includes or exceeds a desired positive at this level of poor metabolic control. Further-
effect size would be considered beneficial; a CI more, the authors did not mention an MCID.
that is less than the null value and includes a Thus, the clinical impact of the observed data

31
2 Essential Statistical and Research Design Elements to Help Critically Interpret the Literature

Did investigator
assign exposure?

YES NO
Experimental study Observational study
Random allocation? Comparison group?

YES NO YES NO

Randomized Nonrandomized Analytical study Descriptive study


controlled controlled
trial trial Direction?

Exposure   Outcome Exposure   Outcome Exposure   Outcome

Cohort Case- Cross-


study control sectional
study study

Cohort study: Study groups are defined by an exposure, and the exposure is tracked forward in time to an outcome.
Case-control study: Study groups are defined by an outcome, and the outcome is traced backward to an exposure.
Cross-sectional study: The exposure and the outcome are assessed in the study group at the same time (a snapshot in time).

Fig 2-5  |  Classification of study designs. (Adapted from Grimes and Schulz.10)

for this particular patient population within calculation of a CI.8 This is unfortunate, as a
this particular clinical setting is unclear, as it P value does not provide enough information
is unclear if the difference in outcomes would to inform clinical practice.
have changed any therapeutic interventions, In addition to considering an outcome clini-
had any impact on the quality of life of cally important as previously described, there is
patients, or affected potential complications also the concept of clinical significance, which
and comorbidities. encompasses the additional element of statisti-
The width of the CI indicates the precision cal significance. For a finding to be considered
of the data. There is no set range that is consid- clinically significant, it should incorporate three
ered wide; this is based on clinical knowledge features: (1) the difference or change in the
and judgment and the characteristics of the outcome observed between the experimental
chosen measurement tool, such as measure- group and the control group is of interest to
ment error. A wide CI could also be a function someone (clinician or patient); (2) this differ-
of a poorly implemented study. ence or change is observed in what is defined as
CIs are not always reported, and the data an important outcome; and (3) the difference
provided in a study do not always enable or change reached statistical significance.9

32
Study Designs

SR

Critically
Filtered
appraised
information
topics

Critically appraised
individual articles
e
enc
vid

Bia
of e

Randomized controlled trials

s
lity
Qua

Unfiltered
Cohort studies
information

Case-control studies/
Case reports/Case series

Cross-sectional epidemiologic studies/


Narrative reviews/Expert opinions

Fig 2-6  |  Quality of evidence and level of bias in different study designs. The quality of evidence increases from
the base toward the top of the pyramid as the rigor of the study design increases. The bias increases going from
the top toward the base of the pyramid. SR, systematic reviews.

Study Designs exposure, also called observational studies (Fig


2-5). As the rigor of the study design increases,
A major goal of epidemiologic research is to the potential bias decreases and the quality of
explain patterns of disease occurrence and the evidence increases (Fig 2-6).
to make causal inferences. There are several
measures to quantify associations, such as odds
ratios and relative risks, and there are different Experimental trials
study designs that can answer particular ques- RCTs, considered the gold standard for clinical
tions. The specific question being asked and epidemiologic and translational research, are
the available data, together with matters such the most rigorous study design, are typically
as ethical concerns and resources, will dictate used to assess the effectiveness of a treatment
the suitable study design. This section provides or an intervention, and provide the strongest
a brief overview of different study designs and epidemiologic evidence for making causal
highlights the appropriateness and inappropri- inferences on the relationship between an expo-
ateness of specific research designs. sure or treatment and an outcome. The terms
Applied epidemiologic and clinical research randomized trials, randomized clinical trials,
is typically divided into two major types of randomized controlled trials, and randomized
investigations: (1) studies where an exposure placebo-controlled trials are often used inter-
is being assigned, also called experimental changeably in the literature. The hallmark of
studies, and (2) studies without assigning an a randomized trial is the random allocation

33
2 Essential Statistical and Research Design Elements to Help Critically Interpret the Literature

or assignment of study participants to treat- results from an RCT may be difficult to gen-
ment, intervention, or exposure groups. The eralize (external validity) because a “normal”
main purpose of randomization is to minimize population may have many characteristics or
selection bias on the part of the investigator. other conditions that differ from those of the
In addition, randomization often increases study population. For example, it would not be
comparability of the treatment groups for possible to generalize the results from a study
variables we can measure as well as those we that looked at the success rate of immediate
are not aware of or cannot measure, thus min- versus delayed placement of dental implants
imizing the impact of potential confounders. if the exclusion criteria for the study popula-
(A confounder is a factor that is associated tion included factors that could have affected
with both the exposure and the outcome but the outcome (eg, success rate) and are com-
does not lie in the causative pathway; see page monly found in the general population, such
20.) However, to ensure that frandomization as smoking, systemic diseases, medications,
produces comparable groups, investigators or periodontal disease. Furthermore, individ-
should always assess similarity of the study uals that volunteer to participate in medical
groups at baseline for relevant known clini- research are often healthier than the general
cal and demographic characteristics and other population.
potential prognostic characteristics. In other Another issue with RCTs is a potential eth-
words, the chance of achieving the study out- ical concern, as randomization is not always
come should be the same in the intervention ethical.11 It would obviously be unethical to
(treatment, exposure) group and the noninter- assign a known harmful exposure to individuals
vention group (also referred to as the control to compare the outcome to a group that was
group or placebo group if a placebo is being not exposed to the harmful exposure. Another
used) if an intervention had not been assigned. example of an ethical concern is when indi-
Another important design element of RCTs that viduals suffering from a disease with a dire
reduces the potential for bias is blinding; in sin- outcome, such as cancer, are randomized to an
gle blinding or masking, the study participants intervention group that receives a new drug that
are unaware of what treatment group they are may potentially be beneficial and another group
assigned to; in double blinding, both the inves- is given a placebo when there is a known ther-
tigator and the study participants are unaware apy available with some degree of effectiveness.
of the group assignment. A well-designed and However, this is no longer common practice.
implemented RCT can therefore minimize Measures of association for RCTs include
selection bias, information bias (blinding of relative risks and odds ratios, with the relative
both participants and research personnel), and risk being the preferred measure. The known
confounding (the intervention group and the temporal relationship between the exposure
control group are potentially exposed to the and the outcome allows for calculation of the
same confounders). Another advantage of an incidence or risk of developing the outcome
RCT is the certainty of the temporal relation- of interest, and the relative risk is the ratio of
ship (which comes first) between an exposure the incidence of the outcome in the exposed
(in this case, treatment or intervention) and group to the incidence of the outcome in the
an outcome. nonexposed group. Relative risk is discussed
However, there are drawbacks associated further in the next section.
with RCTs. The inclusion criteria for partic- One way to assess the rigor of an RCT is
ipant selection is often restrictive, as RCTs to ascertain whether the reporting of the RCT
usually address a very specific condition among follows the Consolidation of Standards for
a very select demographic that is healthy other Reporting Trials (CONSORT) guidelines.12
than the condition of interest. Therefore, Other resources are websites where trials are

34
Study Designs

supposed to be registered prior to their initia- Because participants are free of the outcome
tion,13 such as clinicaltrials.gov, administered by (periodontal disease) at the onset of the study,
the US National Library of Medicine.14 this type of study design can determine whether
the development of the outcome is associated
with the exposure based on the difference in
Observational studies incidence (the development of the disease over
Analytical observational studies (studies that, a specific time period) between the two groups.
contrary to descriptive studies, include a Overall, cohort studies always move forward,
comparison or control group) include cohort but the data collection might not. For example,
studies, where a group of individuals free investigators can use existing dental records to
of the disease/outcome of interest with and identify individuals at risk for and not at risk
without the exposure of interest are followed for developing periodontitis according to a
prospectively forward in time; case-control defined past exposure (eg, flossing), as long as
studies, where individuals with (cases) or they can determine that at the time of expo-
without (controls) the outcome of interest are sure the individuals were free of disease. The
traced backward in time to determine possible identified individuals are then tracked forward
exposure; and cross-sectional studies, where in time to determine who developed periodon-
exposure and outcome are measured at the titis. In other words, the study uses historical
same time (see Fig 2-5). data to determine disease-free status and still
As mentioned previously, unlike with RCTs, looks forward from exposure to outcome. This
the exposure in observational studies is not is referred to as a retrospective cohort study.
assigned but observed in groups of interest. Given that individuals are disease free at
Cohort studies are prospective in nature and the study outset, cohort studies can determine
compare outcomes in groups (cohorts) of par- incidence rates in the exposed and nonexposed
ticipants with an exposure to a similar group of groups. Also, it is possible to determine tempo-
participants without an exposure (but having rality when conducting cohort studies because
the same risk for developing the outcome). In the exposure always occurs before the outcome.
short, participants in the exposed and unex- However, cohort studies may suffer from selec-
posed groups need to be similar in all aspects tion bias if baseline prognostic characteristics
except for their exposure; all prognostic charac- are not adequately accounted for with similar
teristics must be the same. In this study design, distribution in each cohort, and there is always
it is important to establish that the study par- a risk of losing study participants during the
ticipants are free of disease at the start of the progression of the study such that the final study
study and to have a clear, measurable definition sample is a biased sample. The relative risk, the
of exposure and outcome. For example, two measure of disease association in cohort stud-
groups (cohorts) of adults shown to be free of ies, is the proportion of participants who will
periodontal disease, one group that smokes cig- develop the outcome in the cohort with expo-
arettes (the exposure) and one group that does sure as a ratio of (divided by) the proportion of
not use tobacco at all, are followed forward in participants who will develop the outcome in
time for the development of periodontal disease. the cohort without the exposure. Relative risk
The two groups should be similar, with similar is the incidence of the outcome in the exposed
risk factors for the development of periodon- group relative to the incidence of the outcome in
tal disease, except for the fact that participants the nonexposed group and provides a measure
in the exposure group smoke cigarettes and of the risk of developing disease if exposed.
participants in the control group do not. The Information to pay attention to when
outcome of interest, the development of peri- interpreting cohort studies includes concerns
odontal disease, will be assessed after 2 years. with information bias, loss to follow-up, and

35
2 Essential Statistical and Research Design Elements to Help Critically Interpret the Literature

whether potential confounders were controlled always important to ascertain that the partic-
for. ipants in the control group have the same risk
In case-control studies, researchers begin of developing the outcome (same prognostic
with an observed outcome and then try to ret- characteristics) as do the cases.
rospectively determine the presence of exposure. When reading studies using a case-control
Cases used in this study design are those with design, the appropriateness of the control
the outcome of interest, and the controls are a group should be ascertained by determining
comparable group (with the same prognostic whether all potential biases that could influence
characteristics except for the presence of the the outcome were controlled for and whether
outcome of interest) based on a well-defined the controls represent the population at risk
definition of the outcome. The selection and of becoming cases. It is often difficult to make
source of cases and appropriate controls are this determination, as the needed data are not
critical in case-control studies, but it is beyond always available.
the scope of this chapter to discuss this concern A cross-sectional study assesses the presence
in more detail. In brief, cases should be rep- or absence of an exposure and the presence
resentative of all cases in the population and or absence of an outcome at a particular time
controls should be representative of all controls (prevalence of the exposure and prevalence
in the population from which they were drawn. of the outcome). Researchers may determine,
For example, cases of adults with periodontal at a particular time, the presence of children
disease are compared to adults without peri- with or without caries that consume or do not
odontal disease to determine if an exposure, consume sugar-containing beverages. As this
such as smoking cigarettes, is associated with determination represents a snapshot in time, it
the presence of periodontal disease. In this study is not possible to know if the consumption of
design, the investigator can only assess the prev- the sugar-containing beverages occurred prior
alence of disease, as there is no way to ensure to the development of caries (a temporal rela-
that the exposure happened before the disease tionship), and accordingly, if consumption of
process began. If a greater proportion of cases sugar-containing beverages is associated with
(adults with periodontal disease) have a history the development of caries. Cross-sectional
of smoking cigarettes compared to the controls, studies cannot be used to claim causative rela-
smoking cigarettes may be associated with the tionships and are typically used for prevalence
presence of periodontal disease. studies or to develop a hypothesis to be tested
As case-control studies do not have a true in more rigorously designed studies.
denominator of at-risk individuals, and because Case reports and case series are purely
the temporal relationship of exposure and out- descriptive and may, similar to other observa-
come is not clearly established, case-control tional studies, help generate hypotheses about
studies cannot use the relative risk as a measure exposure and outcomes that need to be tested
of association and instead use the odds ratio with higher-quality study designs. Descriptive
(the odds of exposure among the cases as a ratio studies are also used to monitor the health of
of [divided by] the odds of exposure among populations and assess trends over time. With-
the controls) as a measure of association. In a out comparison groups and consideration of
case-control study, the odds ratio assesses the temporality, inferences about causality are not
odds of exposure if disease is present. The odds appropriate. One concern with descriptive
ratio can be used as a measure of disease associ- studies is that sometimes the case definition
ation in a cohort study, and in that study design is not clearly specified or the study cannot be
the odds ratio assesses the odds of developing reproduced. This is true of any type of study
disease given exposure, but it is not as strong a that requires a clearly defined case definition or
measure of association as the relative risk. It is a clearly defined diagnostic outcome.

36
Further Reading

The nature of the question being asked and 4. Ioannidis JP. Preventing tooth loss with biannual dental
visits and genetic testing: Does it work? J Am Dent
the data and financial resources available will
Assoc 2015;146:141–143.
dictate the most appropriate study design. 5. Greenland S, Senn SJ, Rothman KJ, et al. Statistical
Questions about the effectiveness or harm- tests, P-values, confidence intervals, and power: A
fulness of a therapy or intervention are best guide to misinterpretations. Eur J Epidemiol 2016; 31:
337–350.
answered by RCTs. Cohort and case-control 6. Page P. Beyond statistical significance: Clinical inter-
studies are the most appropriate study designs pretation of rehabilitation research literature. Int J Sports
to begin to answer questions about etiology Phys Ther 2014;9:726–736.
and risk factors. In some instances, ethical con- 7. Mauri-Obradors E, Merlos A, Estrugo-Devesa A, Jané-
Salas E, López-López J, Viñas M. Benefits of non-
siderations will preclude RCTs being conducted surgical periodontal treatment in patients with type 2
with human volunteers (eg, studies of smoking diabetes mellitus and chronic periodontitis: A random-
and lung cancer). In these instances, data from ized controlled trial. J Clin Periodontol 2018; 45:
345–353.
cohort and case-control studies may provide
8. Teeuw WJ, Kosho MX, Poland DC, Gerdes VE, Loos
the necessary information if there is consistency BG. Periodontitis as a possible early sign of diabetes
across outcomes and strong enough measures mellitus. BMJ Open Diabetes Res Care 2017; 5:
of association.15 For questions about prognosis, e000326.
9. Brignardello-Peterson R, Carrasco-Labra A, Shah P,
cohort studies may be the most appropriate;
Azarpazhooh A. A practitioner’s guide to developing
questions about diagnosis may be answered by critical appraisal skills: What is the difference between
cross-sectional and case-control studies. clinical and statistical significance? J Am Dent Assoc
2013;144:780–786.
10. Grimes DA, Schulz KF. An overview of clinical research:
The lay of the land. Lancet 2002;359:57–61.

Conclusion 11. Nardini C. The ethics of clinical trials. Ecancermedical-


science 2014;8:387.
12. The CONSORT Statement website. http://www.consort-
statement.org/. Accessed 20 February 2018.
Given the large volume of literature on the 13. Kotsakis GA. Mandatory trial registration in oral health
oral-systemic disease connection and the research. Long overdue? J Am Dent Assoc 2017;148:
varying conclusions and quality of what is 353–356.
published, it is incumbent upon clinicians to 14. ClinicalTrials.gov website. https: //clinicaltrials.gov.
Accessed 20 February 2018.
critically interpret what they read. This chapter 15. United States Department of Health, Education and
provides a basic overview of fundamental sta- Welfare. Smoking and Health: Report of the Advisory
tistical concepts and research design elements Committee to the Surgeon General of the Public Health
Service. Public Health Service Publication No. 1103.
to facilitate interpretation and application of
Washington, DC: United States Public Health Service,
research findings into clinical decision making. 1964.

References Further Reading


1. Rothman KJ. Disengaging from statistical significance. Gordis L. Epidemiology, ed 5. Philadelphia: Elsevier Saun-
Eur J Epidemiol 2016;31:443–444. ders, 2014.
2. Poole C. Low P-values or narrow confidence intervals: Modulsky H. Intuitive Biostatistics: A Nonmathematical
Which are more durable? Epidemiology 2001; 12: Guide to Statistical Thinking, ed 4. New York: Oxford
291–294. University, 2017.
3. Berry DA. P-values are not what they are cracked up Guyatt G, Rennie M, Meade MO, Cook DJ. Users’ Guides
to be. The American Statistician Online Discussion. to the Medical Literature: A Manual for Evidence-Based
https: //amstat.tandfonline.com/doi/suppl/10.1080/00 Clinical Practice, ed 3. New York: McGraw-Hill Edu-
031305.2016.1154108/suppl_file/utas_a_1154108_ cation, 2015.
sm5342.pdf. Accessed 27 February 2018.

37
CHAPTER 3X

The Traveling
Chapter Title Oral
The Traveling
Chapter Title Oral Microbiome
Microbiome
Michael Glick,
Wenche S. Borgnakke,
dmd dds, mph, phd

Microbes are everywhere. Each time we breathe prevalence of previously unknown or not cul-
in, we inhale a million microbes or more, turable organisms.
depending on where we are. Many remain The ability to fully describe the microbiome
trapped in our nostrils, but countless continue that inhabits the human body, including the
into our bodies; fortunately, the overwhelming oral microbiome, suggests a need to reexamine
majority cause no harm. these microbes and their relationship to health
Science is now focused on identifying and disease. The use of these methods sheds
microbes and all other living organisms, thanks new light on the role of the oral microbiome
to novel technologies and computer capabilities in periodontal disease and other oral diseases
of formerly unthinkable power. Collectively, the as well as in systemic diseases and conditions.
microorganisms in the oral cavity—bacteria, This new insight will lead to novel therapeutic
archaea (one-celled, nucleus-free organisms advances that will initiate future changes in
formerly classified as archaebacteria), fungi, clinical practice. We have yet seen only the very
viruses, and protozoa—have been referred to as tip of the iceberg. The sky is the limit.
the oral microflora, oral microbiota, and more
recently, the oral microbiome. While the term
microbiome refers strictly to the total pool of
genetic material in a particular environment, “I” Am Really “We”
whereas microbiota encompasses the entirety
of the microbes, these terms are used synon- Perhaps it is time for each of us to think of
ymously to refer to all nonhuman organisms ourselves and our commensal microbes (usu-
that inhabit the human body inside and out. ally present and living in or on our bodies) as
Our understanding of the human oral micro- constituting a “we” rather than an “I.” That
biome is currently in a groundbreaking phase is, each of us can be considered a collective
of development in which science has to rethink microbiome–human body “superorganism.”
and rewrite large parts of what was hitherto The rough estimate that the number of micro-
known. High-throughput genetic-based assays, bial cells (1014) outnumbers our roughly 100
powerful and novel bioinformatics tools, and trillion human cells (1013) in the human body
superfast computers recently have made it by a ratio of 10:1 is due to only two papers
possible to comprehensively survey the human from the 1970s.1,2 However, recent estimates of
oral microbiome to identify and determine the 3.8 × 1013 (one-celled) bacteria and 3.0 × 1013

38
Unlearn the Learned

human cells put this ratio at about 1.3:1, with no differences between smokers and nonsmok-
an uncertainty of 25% and a population varia- ers in the proportions of the members of the
tion of 53% for a standard male of 70 kg/154.3 microbiomes. In contrast, sequencing of the
lb.3,4 In other words, there are almost equal 16S ribosomal RNA (rRNA) revealed that
numbers of microbial and human cells in the some genera were more abundant in nonsmok-
human superorganism. As an aside, red blood ers and others in smokers. This third method
cells constitute the overwhelming majority of also showed that participants with more severe
human cells, namely about 84%.3,4 Whereas periodontal disease, particularly smokers, had
muscle and fat cells represent the vast majority fewer varieties of microbes. Considering find-
of the weight of the superorganism, bacteria ings from previous studies as well as their own,
have a total mass of about 0.2 kg/0.44 lb.4 Bizzarro et al wrote that “the ‘pathogenic role’
Further knowledge regarding the human of few specific species is questionable. Thus,
microbiomes that inhabit various body loca- we consider now the identification of a few
tions (eg, nose, oral cavity, intestines, female targeted species for diagnostic purposes to be
reproductive tract, outer part of the urinary an out-of-date procedure.”5
system, and skin) may in the future be used in
combination with metagenomic information
to identify individuals at high risk for specific Concept 2: Could periodontal bacteria actually
diseases and thereby enable individualized be a consequence, and not the cause, of
preventive and therapeutic care, referred to as periodontal breakdown?
precision medicine, in which the oral microbi- Old knowledge. Specific periodontal pathogens
ome plays a pivotal role. Researchers are just cause periodontal tissue breakdown. Mainly
now beginning to understand some aspects of gram-negative, anaerobic bacteria cause peri-
this role. odontal disease. Clusters of microbes live in the
plaque in the periodontal sulcus. They develop
into a more mature ecosystem that eventually
morphs into the most mature, complex, patho-
Unlearn the Learned genic biofilm that causes further breakdown of
Concepts periodontal tissues.

Concept 1: “Periodontal pathogen” no more? New knowledge. Periodontal breakdown in


Old knowledge. Specific periodontal pathogens susceptible individuals creates an environ-
(bacteria and possibly other microbes living in ment suitable for particular oral microbes,
the periodontal sulcus or pocket) are especially which then flourish. Deep periodontal pockets
“virulent” and cause periodontal disease. provide an environment deprived of oxygen
that enhances habitation by certain anaerobic
New knowledge. Depending on the microbe- bacteria.
host environment interaction and the technique
used for their detection, disease severity will Bartold and Van Dyke6 questioned whether
vary, and different members of the oral micro- the so-called periodontal pathogenic bacteria
biome will be identified in varying proportions really are the cause or may be a consequence of
in subgingival plaque. the chronic inflammation known as periodon-
titis. They agreed with Löe et al7 who more
Subgingival plaque from 15 smokers and 15 than 50 years ago showed that host-parasite
nonsmokers was studied using three different interactions cause gingivitis. The oral microbi-
methods to identify the presence of bacteria.5 omes form an ecosystem that maintains health
With two of the methods, the researchers found when in equilibrium. Bartold and Van Dyke6

39
3 The Traveling Oral Microbiome

contend that the host is largely responsible for environment suitable for particular oral
the complex alterations in the local periodontal microbes, which then flourish. Deepened
environment that disturb this healthy homeosta- peri-implant pockets provide an environment
sis and facilitate the switch of commensal flora deprived of oxygen that enhances habitation
to an opportunistic pathogenic flora. Therefore, by certain anaerobic bacteria. The types of
the authors concluded that such changes are microbes and their relative abundance iden-
caused mostly by the host, not by the bacteria. tified in peri-implantitis are to a great degree
Emerging evidence supports the major role different from those found in periodontitis.11–14
of host responses modulated through genetic, Peri-implantitis is found to be prosthetically and
immunologic, and inflammatory responses; surgically triggered,15 and peri-implant predic-
stress; smoking; diet; social determinants; and tors, different from those in periodontitis, are
general health—which have been identified as identified, including female sex, malposition-
risk indicators for periodontitis8–10—as being ing, overloading, and bone reconstruction.15
the major determinants of the outcomes of Furthermore, the connection area between the
periodontitis. Therefore, the overgrowth of implant and its restoration has been shown to
periodontal microbes may well be a result of act as a reservoir for bacteria associated with
the periodontal tissue breakdown and not the peri-implant diseases.16 Also, the breakdown
cause of periodontitis. of peri-implant tissues is partially due to nor-
This concept is supported by the fact that mal inflammatory responses, either solely or
currently there is no scientific evidence that predominantly or in addition to the microbial
identifies any particular bacteria as a cause insults. Albrektsson et al on behalf of a con-
of periodontitis. Only associations have been sensus meeting in Rome in 2016 suggested that
observed; that is, deep periodontal probing the marginal tissue breakdown around dental
depths and an abundance of certain bacteria implants is mostly due to immune-osteolytic
are detected simultaneously. reactions, aided by complicating factors such
as “patient genetic disorders, patient smoking,
cement or impression material remnants in the
Concept 3: Could bacteria inhabiting the peri- peri-implant sulcus, bacterial contamination of
implant space actually be a consequence, and the implant components and technical issues
not the cause, of breakdown of peri-implant such as loose screws, mobile components or
tissues—and are they the same bacteria as fractured materials.”17
those associated with periodontitis?
Old knowledge. The microbes in the
peri-implant space in peri-implantitis are sim-
ilar to those found in periodontitis. Specific Concept 4: Dental implants may not be a
periodontal pathogens cause periodontal tissue treatment option in all patients with diabetes.
breakdown, and these bacteria are suspected Old knowledge. Implants represent an accept-
of also leading to breakdown of tissues sur- able treatment option for people with diabetes.
rounding dental implants. Clusters of microbes
live in the plaque in the periodontal sulcus. New knowledge. People with prediabetes and
They develop into a more mature ecosystem well-controlled diabetes might be at about
that eventually morphs into the most mature, the same risk for peri-implant mucositis and
complex, pathogenic biofilm that causes further peri-implantitis as those with normal blood
breakdown of peri-implant tissues. glucose levels. However, patients with poor
glycemic control are more prone to develop
New knowledge. Breakdown of peri-implant peri-implantitis.18,19 Evidence to support offer-
tissues in susceptible individuals creates an ing dental implants to patients with diabetes

40
Unlearn the Learned

often does not specify the importance or level Concept 7: Infections with bacteria are also
of glycemic control. Furthermore, studies often found inside cells.
cite implant failure or survival of relatively Old knowledge. Bacterial infections are con-
short duration instead of long-term implant tained in the intercellular spaces.
success, defined as healthy peri-implant tissues,
as an outcome.20 New knowledge. Some bacteria actually are
able to invade cells, including vascular cells
in arteries, and the white blood cells that are
Concept 5: All bodily fluids contain a supposed to kill them. Some bacteria that are
microbiome, also in health. usually part of the oral microbiome, such as
Old knowledge. Human bodily fluids and most strains of Porphyromonas gingivalis, have
surfaces, including mucosa, are sterile in health, developed sophisticated methods to not only
such as blood and nasal and pulmonary fluids. migrate between layers of cells into tissues but
also penetrate cell walls and enter host cells,
New knowledge. In health, all human bodily where they can both survive and multiply, and
fluids and mucosal surfaces harbor distinct eventually exit and infect other cells.34 Think
microbiomes that are changing constantly in “Trojan horse.” This is important because
response to environmental influences from while located intracellularly, neither the host’s
within and external to the body. Even blood21,22 immune system nor antibiotics can identify and
and breast milk,23,24 synovia (fluids in synovial annihilate these bacteria if the host has a T-cell
joints),25 the vagina,26 nonmalignant pancre- immune deficiency that allows intracellular
atic cysts,27 amniotic fluid,28 as well as mucosal bacteria to flourish.
surfaces like the ocular surface that consists of
the cornea and conjunctiva29,30 harbor bacteria,
fungi, and possibly viruses in health and may What does this new knowledge
contain microbes that travel from elsewhere in mean for the dental practitioner?
the body, including the oral cavity. The latter
was shown for bacterial communities in amni- Concepts 1 and 2 express the current beliefs
otic fluid.31,32 and synthesize sentiments that have been devel-
oping over the last few years5,6 They actually
make perfect sense. For many years, the search
Concept 6: Tobacco smoking may also directly to identify the culprit bacteria that cause peri-
affect the oral microbiome. odontal disease used methods that depended on
Old knowledge. The adverse effects of tobacco growth and observation of bacteria in a labo-
use, such as smoking cigarettes or chewing ratory. Now the members and communities of
betel nuts, are due to irritation of the mucosa the oral microbiome in health and in disease
and the toxic ingredients in the tobacco and can be explored without culturing the bacte-
its smoke. ria. They can now be detected from even small
pieces of genetic material, so it is not necessary
New knowledge. It may also be the case that to examine the entire organism, let alone live
use of tobacco products has a direct effect on ones that can be kept alive and cultured in the
the microbiota in the buccal mucosa, where laboratory. The precision with which microbes
the diversity of microbes (number of different can be identified is unprecedented and has
microbes) is less than in nonsmokers,33 hence largely necessitated relearning most of what
disturbing the normal ecosystem in health in we thought we knew about microbiology.
which homeostasis (balance) prevails. Detailed genetic analysis of bacteria, which
is increasingly available and economically

41
3 The Traveling Oral Microbiome

feasible, has demonstrated an unanticipated In this chapter, increased levels of oral


genetic diversity within species. This knowl- microbes in the oral cavity are regarded as
edge guides future prevention and treatment overgrowth of commensal microbes in envi-
protocols, which seem to point in the direction ronmental conditions conducive to their
of precision medical and dental care tailored multiplication. Therefore, the terms infection
to the individual based on personal genetic, and pathogen are not used in conjunction with
environmental, and clinical profiles.35 The organisms usually inhabiting the biofilm in
increasingly sophisticated molecular-level ana- the periodontium. Hajishengallis and Lamont
lytical tools and exploration of inflammatory described in 2016 how “pathogens” originally
responses and other host factors have shown are commensal microbes that due to changes
that factors within the host may be at least as in their environment take advantage of their
important as the potential pathogen. enhanced conditions and multiply and thrive
For now, however, these new viewpoints (“overgrow”), without any intrinsic features.37
on mechanisms will not change the need to They introduce the term keystone patho-
keep the oral cavity as healthy and free of gen (alpha-bug or bacterial driver) for such
dental plaque as possible. Nevertheless, these microbes, of which the periodontal bacterium
perspectives suggest that dental professionals P gingivalis is a prime example. In 2015, Han
must constantly be vigilant and consider the used the expression commensal-turned patho-
possible existence of systemic health conditions gen for this notion regarding another such
and diseases in need of attention, especially in bacterium, namely Fusobacterium nucleatum.31
patients with severe periodontitis.36 The term microbe refers to both live and
dead bacteria and their byproducts, as well as
archaea, viruses, and fungi, unless specifically
noted. In the human body, fungi can appear
What Is an Infection? in three forms, namely as hyphae (filaments),
pseudohyphae, and yeast. Yeast is unicellular
The term infection has various definitions. whereas the other two types are multicellular.
The main difference among definitions is that Fungal cell walls consist predominantly of chi-
some include the reaction of host tissues to the tin, a derivative of glucose.
infectious organisms and the toxins they pro-
duce while others regard merely the presence
of the infectious agent in unusual amounts or Gram-positive versus
places to be sufficient for application of the gram-negative bacteria
term, even in the absence of pronounced host
responses. The latter state is what the former Gram refers to a staining method of bacteria
definition would regard as carriage; that is, the that uses crystal violet stain. The cell walls
host acts as a carrier who does not necessarily of gram-positive bacteria take up this stain
show signs or symptoms of an infection but and therefore appear violet when observed
may do so if the host’s resistance to infection in a microscope, even after decolorization
is decreased, as during psychologic stress, for washing that includes alcohol. In contrast,
instance. Still others regard an infectious agent gram-negative bacteria are not able to retain
as one that is not normally found in the body. the purple color upon washing. In order to be
These distinctions are important for how we able to see these gram-negative bacteria in a
think about infection in conjunction with peri- microscope, a counterstain is used to color
odontal disease and the identification of oral them. For example, they will appear pink or
microbes in the rest of the body. red due to taking up eosin in the commonly
used hematoxylin and eosin stain.

42
The Human Genome

Gram-positive bacteria have a thick cell wall and guanine (G). The nucleotides A and T are
predominantly consisting of a mesh composed always bonded together as a pair, as are C and
mostly of sugars and amino acids called peptido- G, hence the expression base pair. The nucleic
glycan (polysaccharide and peptide chains), also acid RNA uses uracil (U) instead of thymine,
known as murein or mucopeptide, that provides so its base pairs are A + U and C + G.
strength and rigidity. In contrast, gram-negative
bacteria are defined by a cell envelope composed
of a thin peptidoglycan cell wall sandwiched The Human Genome Project
between a bacterial outer membrane and an The Human Genome Project was declared com-
inner cytoplasmic cell membrane. pleted with the final mapping and sequencing
Antibiotics work on gram-positive bacteria of the human genome on April 14, 2003, which
whose thick cell wall disintegrates, whereas the was 2 years ahead of the 15-year-long sched-
outer cell membrane protects gram-negative ule.41 For the first time, all 20,500 human genes
bacteria from several antibiotics, such as were cataloged after sequencing of the 3 billion
penicillin, so other antibiotics must be used. chemical base pairs that make up human DNA.
Importantly, the lipopolysaccharides in the This human genome is a “reference genome”
outer membrane of gram-negative bacteria exert from a combination of anonymous donors, but
a strong proinflammatory effect seen in some each human being has a unique gene sequence.
of the most abundant periodontitis-associated Knowledge about the human genome is piv-
bacteria, such as P gingivalis.38–40 otal to understanding how our bodies function
so that preventive and curative measures can
be devised to improve our patients’ health and
quality of life.
The Human Genome Metagenomics is the study of genetic
Genomes and DNA  material and uses sequence-based methods
that permit the genetic material from all the
The term genome was adopted in 1920 and is a microbes harvested from a biologic sample to
blend of the two words gene and chromosome. be analyzed simultaneously without the need
A genome consists of all the genetic material of for cultivating the microorganisms in a lab-
a living entity and can be considered the “Book oratory. Metagenomics provides vastly more
of Life” comprising two volumes, namely one information than the analysis of single, isolated
from each parent (https://ghr.nlm.nih.gov/ microbes. It is possible not only to determine
primer/basics/gene). This Book can be likened which organisms are present but also to deduce
to a cookbook that contains recipes for the the relative abundance of the different species.
actions of genes. A gene is a distinct sequence Furthermore, it may be possible to discover
of nucleotides (DNA or RNA) that codes for which metabolic pathways are encoded by
the production of proteins. Such protein-coding the organisms so that information can be
genes are interspersed between noncoding obtained about their functions in the body.
genes and together form a chromosome. Metagenomics is used to analyze the human
Humans possess 23 pairs of chromosomes oral microbiome in unprecedented detail.
that are all located in the nucleus of all cells,
except red blood cells that do not contain any
nucleus. Because not all genes need to be active Sequencing of the 16S rRNA gene
at all times, they turn on and off according to The 16S rRNA gene has been sequenced to
demand. DNA is made up of pairs of nucle- detect the order (sequence) in which the four
otides in various sequences. The nucleotides RNA nucleotides (A + U, C + G) appear.
are adenine (A), thymine (T), cytosine (C), The entire length of the 16S gene of RNA

43
3 The Traveling Oral Microbiome

from ribosomes has been sequenced, and this health, and disease. The human microbiome
information is stored in the publicly accessi- consists of all the microbes that inhabit the
ble Human Microbiome Project database as a inside and outside of the human body. As
reference.42 Scientists can now compare their mentioned earlier, it contains about 38 tril-
findings to the reference RNA for identification. lion (3.8 × 1013) bacteria alone,3,4 but it also
The bacterial 16S rRNA gene is the molecular contains more than 8 million protein-coding
marker most widely used to identify microbes genes, outnumbering human genes by 360:1
to study the diversity of microbial communities. (8 million:20,500).44 The human microbiome
The 16S rRNA gene is used as a reference for normally consists of beneficial, harmless, non-
sequencing for a number of reasons: pathogenic organisms that in health exist in
homeostasis in which there is a relatively stable
• It is present in almost all bacteria. equilibrium, as opposed to dysbiosis, a micro-
• It is a highly conserved gene, preserved in its bial imbalance or derangement that is often
original form through various manipulations associated with disease.
such as cell death and freezing. Because of their small size, microorganisms
• Its function has not changed over time, sug- make up only a minute fraction of the weight
gesting that random sequence changes are a of a human. Sender, Fuchs, and Milo pointed
more accurate measure of time (evolution). out4 that although initial reports suggested that
• It is sufficiently long (1,500 base pairs) for 1.5 to 4.5 lb (0.7 to 2.0 kg) of microbes live in
informatics purposes involving identification and on a 150-lb (68 kg) person, the estimated
for comparisons to detect similarities and mass of the bacteria may actually be only 0.2
differences between genetic samples. kg (200 g, about 0.5 lb), representing about
0.3% of the overall body weight.4
Importantly, as in metagenomics, the The microbiome is necessary for human
microbes do not have to be whole or alive for survival. Its components digest food, synthe-
this method to be used. The DNA can stem from size essential nutrients and vitamins, educate
dead organisms or from only pieces thereof. and maintain the innate (inborn) and adaptive
Consequently, even bits of the cell walls (endo- (acquired) immune system while maintaining
toxins) are sufficient for determining the identity self-tolerance to avoid autoimmunity, regulate
of a certain microbe. A brief overview of this immune homeostasis, fight off infections, and
so-called next-generation workflow for health maintain homeostasis and healthy equilibrium.
care professionals is provided by Kilian et al.43

The Human Microbiome Project


The Human Microbiome Project was launched
The Human Microbiome by the US National Institutes of Health to
characterize the human microbiome and
A microbiome is the totality of microorganisms subsequently analyze its role in health and
(microbes), their genetic elements (genomes), disease.45 The Human Microbiome Project
and environmental interactions in a particular is the largest-ever 5-year project aimed at
environment. The term was coined in 2001, identifying and cataloging the human micro-
which indicates the novelty of this concept. biome. Researchers sampled 300 healthy 18- to
Some consider the microbiome a “newly dis- 40-year-old individuals from two US cities.
covered organ,” because its existence was not Even the presence of minor “gum disease” was
generally recognized until the late 1990s. enough to exclude a subject from the study.46
Researchers are only beginning to under- The study was focused on identifying the
stand its vast impact on human existence, microbes residing in five body areas (Fig 3-1).

44
The Human Oral Microbiome

Fig 3-1  |  Body sites for sampling microbiomes for the


Human Microbiome Project.
Nasal
Oral

Skin

Gastrointestinal

Urogenital

Researchers sampled 15 body sites in each man represent transient colonization, but some may
and 18 sites in each woman.47 turn out to inhabit the mouth on a regular
Nine of the 15 or 18 sites sampled were basis. Knowing which microbes live in the oral
located in the oral cavity, which is a testament cavity is important for targeting measures to
to the immensely important role the oral micro- improve or attain oral health.
biome plays in human health and disease.

The Human Oral Microbiome


The Human Oral Microbiome Database
The human oral microbiome is the most
Currently, about 700 different bacterial species studied human microflora. The Human Oral
have been identified in the human oral cavity, Microbiome Database was created to provide
but estimates suggest that the actual number the scientific community with a comprehensive
may be as many as 1,200.48 The oral cavity database of the oral bacterial species.42 It is
differs from all other human microbial habitats basically a ledger that provides information on
by the simultaneous presence of two types of the genomes of oral bacteria.
surface for microbial colonization: shedding An example follows of the classification hier-
surfaces (mucosa) and solid surfaces (teeth and archy of subgroups, namely for P gingivalis,
different fixed and removable oral appliances). formerly known as Bacteroides gingivalis. As
This intrinsic property of the oral cavity pro- displayed in Box 3-1 and Fig 3-2, the genomes
vides immense possibilities for a diverse range of a total of 19 different strains or subspecies
of microbiota. Saliva contains around 100 mil- have been identified.50 This was the result of
lion bacteria and archaea per milliliter, by far testing 27 strains that turned out to be clus-
the two most common types of microbe in the tered into two groups of shared proteins.50
mouth.49 Such development of strains or subspecies
Currently, only about half of the microor- demonstrates the capability of P gingivalis to
ganisms detected in the mouth are officially adapt to new environments.
named, and only about three-quarters of those The same order of classification hierarchy for
are indisputably oral commensals. The rest may Streptococcus mutans is Bacteria, Firmicutes,

45
3 The Traveling Oral Microbiome

Box 3-1  |  Tree of life classification of P gingivalis

Hierarchy Bacterium
Domain Bacteria
Phylum Bacteroidetes
Class Bacteroides
Order Bacteroidales
Family Porphyromonadaceae
Genus Porphyromonas
Species gingivalis
# Strains >19

Fig 3-2  |  P gingivalis, a keystone periodontal bacte-


rium and community activist.

Bacilli, Lactobacillales, Streptococcaceae, as shown for the three individuals in Fig 3-3.51
Streptococcus, and mutans. Moreover, the distribution and abundance of
bacteria at the phylum level in the different
oral sites sampled varied substantially among
Does a core oral microbiome exist? three individuals51 (Fig 3-4). Interestingly, bac-
teria formerly regarded as causing periodontitis
Bacteria were harbored in the three subjects as well as in
Researchers have wondered about the existence multiple participants in a larger study52 but typ-
of a core set of oral microorganisms that are ically only as minor components of the plaque.
common to most humans. Only with recently Distinguishing oral microbiomes in health
developed novel techniques that are still being and in periodontitis is difficult because up to
refined is it possible to begin to look for pos- 95% of microbe genera are present in both
sible answers to this question. When three health and disease, and periodontal prob-
healthy, unrelated individuals were studied, ing measures cannot attest to whether a site
samples originating from shedding surfaces is in the process of breaking down or is on
(mucosa of the tongue, cheek, and palate) the mend. Therefore, a group of researchers
were clearly distinguishable from samples that calculated the ratio of bacterial dysbiosis as
were obtained from solid surfaces (teeth).51 The an expression of the periodontal breakdown
three people had only one-quarter (26%) of activity.53 Such dysbiosis ratios turned out to
the 6,315 unique bacterial gene sequences in be correlated with the severity of periodontitis
common, but the abundance of the families of assessed by pocket depth and might hence be
bacteria to which these belonged were almost useful for both risk prediction and monitoring
identical (99.8%) in the three persons. This of healing pockets.
supports the concept of a core microbiome in A study of 66 healthy Han Chinese subjects
health, but only at the bacterial family level. spanning in age from neonatal (3 to 4 days) to
However, the proportions of even the com- elderly (62 to 76 years) clearly demonstrated
mon microbes vary widely among individuals, that the oral cavity is a highly diversified space

46
The Human Oral Microbiome

20

S1
S2
S3
15

10

0
Streptococcus; 803
Streptococcus; 165
Corynebacterium; 280
Neisseria; 637
Rothia; 513
Streptococcus; 230
Veilfonellaceae; 110
Actinomyces; 419
Granulicatella; 335
Streptococcus; 262
Porphyromonas; 475
Firmicutes; 592
Streptococcus; 452
Corynebacterium; 145
Actinomyces; 374
Fusobacterium; 202
Rothia; 64
Corynebacterium; 610
Haemophilus; 697
Prevotella; 181
Neisseria; 591
Actinomyces; 752
Streptococcus; 669
Fusobacterium; 758
Gampytobacter; 316
Neisseriaceae; 683
Streptococcus; 679
Actinomyces; 382
Neisseriaceae; 508
Rothia; 721
Haemophilus; 210
Streptococcus; 676
Fusobacterium; 608
Streptococcus; 109
Haemophilus; 5
Actinomyces; 500
Leptotrichia; 9
TM7; 40
Neisseriaceae; 42
Capnocytophaga; 517
Haemophilus; 16
Fusobacterium; 375
Granulicatella ; 23
Capnocytophaga; 345
Derxia; 321
Acidovorax; 797
Haemophilus; 182
Actinomyces; 578
Capnocytophaga; 18
Moryella; 350
Cardiobacterium; 192
Fig 3-3  |  Shared abundant phylotypes in three oral microbiomes (S1, S2, and S3) and their relative abundance.
Only abundant phylotypes that contributed to at least 0.1% of the individual microbiome are shown. The most
abundant phylotypes (0.5% or more of the microbiome) are grouped separately in the left panel. Phylotypes were
defined as operational taxonomic units (OTUs) clustering sequences at a 3% genetic difference. The highest
taxon (in most cases, genus) at which the OTU was identified is shown together with the cluster identification
number. Different colors indicate three different microbiomes. (Reprinted from Zaura et al51 with permission.)

100%

80%
Actinobacteria

Bacteroidetes
60%
Firmicutes

Fusobacteria

Proteobacteria 40%

Spirochaetes

TM7 20%
Unclassified
bacteria

0%
B_incisor
L_incisor
Appr_incisor
B_molar
L_molar
Appr_molar
Palate
Cheek
Tongue
Saliva
Average

B_incisor
L_incisor
Appr_incisor
B_molar
L_molar
Appr_molar
Palate
Cheek
Tongue
Saliva
Average

B_incisor
L_incisor
Appr_incisor
B_molar
L_molar
Appr_molar
Palate
Cheek
Tongue
Saliva
Average

S1 S2 S3

Fig 3-4  |  Average and site-specific relative distribution of bacterial phyla in three individuals (S1, S2, and S3).
Unclassified bacteria were reads without a recognizable match in the full 16S rRNA reference database. B, buccal;
L, lingual; Appr, approximal surface of either an incisor or a molar. (Reprinted from Zaura et al51 with permission.)

47
3 The Traveling Oral Microbiome

Fig 3-5  |  Venn diagram of shared and differ-


ent OTUs of the oral bacterial communities. A
large number of OTUs could be detected only
in saliva (11,171), supragingival dental plaque
(13,333), and buccal mucosa (6,302), whereas
Saliva only 3,619 OTUs were shared among all three
11,171 niches. (Reprinted from Xu et al54 with permis-
sion.)

3,193 3,427
3,619
Supragingival Buccal
dental plaque 806 mucosa
13,333 6,302

with highly different microbiomes according that the overall function of the complex oral
to dentition stage (age) and location within the microbiome is more important than any of its
mouth and that any core oral microbiome must individual components in understanding oral
be specific for the respective age/dentition status health and disease, which could serve as the
and oral niche/location/medium (dental plaque, basis for potentially exploring novel therapeu-
saliva, and buccal swabs).54 Figure 3-5 illus- tic targets.56
trates the small subset of microbes measured as
operational taxonomic units (OTUs) that are
common for all three as well as pairwise. Fungi
Several microorganisms that are common In 2010, a group of researchers characterized
elsewhere in the body inhabit or are frequent for the first time the “basal” fungal microbi-
guests in the oral cavity. For example, Staphylo- ome (mycobiome) of the oral cavity in healthy
coccus aureus is frequently detected, although individuals. Figure 3-6 shows the fungus genera
it is not a bacterium normally considered a reg- present in each of the 20 subjects examined,
ular member of the oral microbiome. In people illustrating the great diversity and individual
with cleft lip and palate, transmission to the differences in the composition of their myco-
mouth can easily occur from the nasal micro- biomes.58 The number of different genera of
biota. In a study of young children with cleft lip fungi among the healthy subjects ranged from
and palate, those with fistulas had significantly 3 to 16. While the commensal oral fungal com-
more S aureus in the oral cavity than did those munity plays an important, even critical, role
without fistulas, and those with larger fistulas in health and disease in humans, nonetheless
had greater counts of S aureus than those with it has largely been ignored.59
smaller fistulas.55
As in the total human microbiome, the
abundance and diversity of the oral micro- Saliva
biome presents homeostasis in health but It is estimated that human saliva contains
dysbiosis in disease.56,57 Therefore, it is likely 108 to 109 bacteria per milliliter.60 However,

48
The Human Oral Microbiome

Zygosaccharomyces Wallemia
100– Verticillium Trichaptum
Tremel/ales Torulaspora
– Subulispora Stemphy/ium
Sebacina Rhizopogon
90– Pyrenophora Pueraria
Pongo
– Phaeosphaeria
Peziza Orpinomyces
80– Ophiostoma Nectria
Macrophomina Lecania
– Lasiodiplodia lssatchenkia
Guignardia Gigaspora
70– Flammulina Filobasidiales
– Epicoccum
Frequency (%)

Davidiella
Cystofilobasidium Corynespora
60– Conoplea Clavispora
Claviceps Chaetomium
– Bagnisiella Avena
50– Amanfta Acremonium
Campylobacter Xylariales
– Trichosporon Sordariomycete
Schizosaccharomyces Pleosporaceae
40– Pichia Phoma
Paecilomyces Ochroconis

Nigrospora Lewia
30– Hypocreales Hormonema
Emericella Atractylodes
– Ascomycete Trichocomaceae
Stachybotrys Schizophyllum
20– Penicillium Dothideomycete
– Gibberella Cryptococcus
Glomus Eurotium
10– Saccharomyces Fusarium
Aspergillus Alternaria
– Teratosphaeria Dothioraceae
Saccharomycetales Aureobasidium
0– Cladosporium Candida
Non-culturable
A1 A2 A3 B1 B2 B3 C1 C2 C3 D1 D2 E1 E2 E3 F1 F2 G1 G2 G3 H2
Sample
Fig 3-6  |  Overall distribution of fungi in oral rinse samples obtained from 20 healthy individuals. Δ, sample con-
taining 16 fungal genera; *, sample containing 3 fungal genera. (Reprinted from Ghannoum et al58 with permission.)

the bacterial composition of saliva samples stimulated saliva by chewing for 1 minute on
is representative of neither supragingival nor paraffin, for instance.
subgingival plaque nor or of any other oral After studying a large extended Ashkenazi
biofilm. Furthermore, the different methods Jewish family whose members lived in dif-
used to sample the saliva—including collecting ferent locations, Shaw et al63 concluded that
saliva upon passive drooling or after stimulat- the human salivary microbiome is more a
ing salivary production by chewing paraffin, result of environmental factors like shared
active swishing with a liquid (mouthrinse or households rather than due to genetics. Such
mouthwash), insertion of paper points, or a household-shaped salivary microbiome can
buccal or tongue swabs—may affect the rep- persist for several years after leaving the ini-
resentativeness of the oral microbiome. tial environment. Time will tell whether a core
However, it was demonstrated that no or very universal human salivary microbiome in health
little variation occurs in the microbial profiles will emerge.64
over a 24-hour period and after 1 week in stim-
ulated saliva, which means the diurnal timing
is not crucial when obtaining salivary samples Distribution of oral microbes
for analysis.61 Furthermore, the same research-
ers showed that bacterial profiles in stimulated Intraoral diversity
saliva are sufficiently similar to unstimulated Within the oral cavity, groups of microbial spe-
saliva and hence can be used for analysis.62 This cies arrange themselves into surface-localized
is important because collection of unstimulated communities that vary considerably in compo-
saliva samples from passive drooling at rest sition according to their exact location. Even
takes much longer than collecting samples of adjacent oral sites “only millimeters or less”

49
3 The Traveling Oral Microbiome

Second
layer

Basal
layer

Tooth side
Fig 3-7  |  Localization of the most abundant species in supragingival biofilms. Streptococcus species (yellow)
form a thin band on top of the biofilm, almost engulf the biofilm, or present as small cells scattered through the top
layer of the biofilm. (B) Cells from the CFB cluster of bacteria in the top layer of the biofilm, without defined struc-
ture. (C) Lactobacillus species (red) forming long strings through the top layer. (D) Actinomyces species (yellow)
plaque attached to the tooth. (E) Actinomyces species (green) and cocci forming initial plaque. (F) Multispecies
initial plaque composed of Streptococcus species (yellow), yeast cells (green), and unidentified bacteria (red). (G)
Streptococcus species (green) and Lactobacillus species (red) forming initial plaque. Black holes might be chan-
nels through the biofilm. Panels A, B, C, E, and F are double-stained with probe EUB338 labeled with FITC or Cy3.
Scale bars are 10 μm. CFB cluster, Cytophaga-Flavobacterium-Bacteroides cluster, including Parvimonas micra, P
gingivalis, Prevotella intermedia, Porphyromonas endodontalis, and Prevotella nigrescens. (Reprinted from Zijnge
et al65 with permission.)

apart exhibited “strikingly different” microbial There are considerably more differences
community compositions.65 The dorsum of the between the microbiomes in supragingival and
tongue specifically can harbor several bacte- subgingival dental plaque for various reasons,
rial species detected in ventilator-associated such as the availability of oxygen. Until rather
pneumonia. recently, our knowledge regarding the bacte-
rial composition of plaque was largely based
Tooth surfaces and periodontal pockets. There on studies conducted in the 1970s that used
are considerable differences in bacterial com- culture and electron microscopic techniques.67
position among teeth at different intraoral Zijnge et al used the novel Fluorescence In
locations and even among different surfaces Situ Hybridization (FISH) technique for the
of the same tooth.66 Pronounced differences first time to provide visualization of the most
were observed in incisors and canines, where abundant bacterial phyla and species as well
Streptococcus species were found on 40% to as the composition and structure of the supra-
70% of the vestibular surfaces but were almost and subgingival biofilm from extracted human
absent on the lingual surfaces. teeth.65 Figures 3-7 to 3-10 visualize the most

50
The Human Oral Microbiome

Fig 3-8  |  Localization of the most abundant species in subgingival biofilms. (A) Overview of the subgingival
biofilm with Actinomyces species (green), bacteria (red), and eukaryotic cells (large green cells on top). (B) Spiro-
chaetes (yellow) outside the biofilm. (C) Detail of Synergistes (yellow) in the top layer in close proximity to eukary-
otic cells (green). (D) CFB cluster (yellow) in the top and intermediate layer. (E) F nucleatum in the intermediate
layer. (F) Tannerella species (yellow) in the intermediate layer. Each panel is double-stained with probe EUB338
labeled with FITC or Cy3. The yellow color results from the simultaneous staining with FITC and Cy3-labeled
probes. Scale bars are 10 μm. CFB cluster, Cytophaga-Flavobacterium-Bacteroides cluster, including P micra,
P gingivalis, P intermedia, P endodontalis, and P nigrescens. (Reprinted from Zijnge et al65 with permission.)

abundant species in supragingival (see Fig in the following pages, beginning with a sche-
3-7) and subgingival biofilm (see Fig 3-8), the matic representation (Fig 3-11).68
bacteria most abundant in periodontitis (see Figures 3-12 to 3-1468 show bacteria made
Fig 3-9), and various aggregations of bacteria visible by 10 fluorescent probes by the com-
located within the biofilm (see Fig 3-10).65 plex Combinatorial Labeling and Spectral
Combining the FISH technique with confocal Imaging Fluorescence In Situ Hybridization
scanning fluorescent microscopy and histology (CLASI-FISH) technique developed by the
shed new light on these features.67 Mark Welch Valm and Welch team,69 a major scientific
et al introduced the notion of “biogeography” breakthrough that for the first time enabled
to describe the complex multispecies microbi- visualization of the biogeography of 15 differ-
omes like the human oral microbiome at the ent taxa simultaneously to further explore the
micron scale, suggesting interpretation of the various components of a microbiome as well
so-called “hedgehog,” “corncob,” and “cauli- as its architectural structure.70
flower” structures that are briefly illustrated

51
3 The Traveling Oral Microbiome

Fig 3-9  |  Localization of species associated with periodontitis. (A) Overview of the subgingival biofilm with
CFB-cluster species (red) and Prevotella species (yellow). Because Prevotella species are part of the CFB cluster
of bacteria, cells appear in yellow. (B) Top of the biofilm with a microcolony of P micra (yellow). (C) Microcolonies
of P gingivalis (yellow) in the top layer. (D) Microcolonies of P endodontalis (yellow) in the top layer. (E) Microcol-
onies of P intermedia in the top layer. Panels B, C, D, and E are double-stained with probe EUB338 labeled with
FITC or Cy3. Scale bars are 10 μm. CFB cluster, Cytophaga-Flavobacterium-Bacteroides cluster, including P mi­
cra, P gingivalis, P intermedia, P endodontalis, and P nigrescens. (Reprinted from Zijnge et al65 with permission.)

Fig 3-10  |  Bacterial aggregates in oral


plaque. (A) Transversal view of a test-tube
brush found in subgingival plaque com-
posed of filamentous cells from the CFB
cluster. (B) Tannerella species (yellow) in a
test-tube brush. (C) Longitudinal view of
a test-tube brush with Lactobacillus spe-
cies (red rods) as central structures. F nu­
cleatum (green) and CFB-cluster filaments
radiating from the central structures. (D)
Longitudinal and transversal view of a
test tube brush stained with the eubac-
terial probe. (E) Transversal view of the
test-tube brush in panel D, composed of
Synergistetes group A species. (F) Trans-
versal view of Streptococcus species
(green) aggregation around a central cell
(not stained) in supragingival plaque. (G)
Transversal view of supragingival plaque
with Streptococcus species (green coc-
ci) and C albicans (green hyphae) in the
top layer of the biofilm and forming corn
cob structures growing outwards. Bars
are 10 μm. CFB cluster, Cytophaga-
Flavobacterium-Bacteroides cluster, in-
cluding P micra, P gingivalis, P intermedia,
P endodontalis, and P nigrescens. (Re-
printed from Zijnge et al65 with permission.)

52
The Human Oral Microbiome

Fig 3-11  |  Summary hypothesis for inter-


pretation of hedgehog structures. Coryne­
bacterium filaments bind to an existing biofilm
02, saliva, sugars
containing Streptococcus and Actinomyces.
At the distal tips of the Corynebacterium fila-
ments, corncob structures form in which the CO2, lactate,
filaments are surrounded by cocci, including acetate, H2O2
Streptococcus and Porphyromonas, in direct
contact with the Corynebacterium filament Anoxic
Tooth
as well as Haemophilus/Aggregatibacter in
contact with Streptococcus. Clusters of Neis­
seriaceae also occupy the periphery of the
hedgehog. The Streptococcus cells create
a microenvironment rich in CO2, lactate, and
acetate, containing peroxide, and low in ox-
ygen. Elongated filaments of Fusobacterium Base Annulus Perimeter
and Leptotrichia proliferate in this low-oxygen,
high-CO2 environment in an annulus just Crevicular fluid
proximal to the corncob-containing peripher-
al shell of the hedgehog. The CO2-requiring Corynebacterium Porphyromonas Fusobacterium Other
Capnocytophaga also proliferates abundantly Streptococcus Neisseriaceae Leptotrichia
Haemophilus/ Capnocytophaga Actinomyces
in and around this annulus. The base of the Aggregatibacter
hedgehog is dominated by Corynebacterium
filaments and thinly populated by additional
rods, filaments, and/or cocci. (Reprinted from
Mark Welch et al68 with permission.)

Fig 3-12  |  Corncob structures formed by Cory­ Fig 3-13  |  A cauliflower structure in plaque com-
nebacterium and cocci in plaque. Corynebacterium posed of Lautropia, Streptococcus, Haemophilus/
cells (magenta) are visible as long filaments, with coc- Aggregatibacter, and Veillonella. Scattered cells of
ci (green) bound to the tips of the filaments. Partially Prevotella, Rothia, and Capnocytophaga are also
disrupted plaque was hybridized with a probe for Co­ visible. (Reprinted from Mark Welch et al68 with per-
rynebacterium and a universal bacterial probe. Scale mission.)
bar is 20 μm. (Reprinted from Mark Welch et al68 with
permission.)

53
3 The Traveling Oral Microbiome

10 um
a b

Fig 3-14  |  Complex corncob structures in supra­


gingival plaque. (a and b) Clusters of corncobs at
the perimeter of hedgehog structures. (a) Whole
mount of plaque hybridized with probes for Coryne­
bacterium, Fusobacterium, Streptococcus, Porphyro­
monas, and Haemophilus/Aggregatibacter. (b) Meth-
Corynebacterium Fusobacterium acrylate-embedded section hybridized with probes for
Streptococcus Leptotrichia Corynebacterium, Streptococcus, Porphyromonas,
Porphyromonas Capnocytophaga
and Haemophilus/Aggregatibacter. (c) Gallery of repre-
Haemophilus/Aggregatibacter Neisseriaceae
c sentative images showing types of corncobs frequently
observed. Scale bar in c is 5 μm. (Reprinted from Mark
Welch et al68 with permission.)

Periapical periodontitis. Biofilm adhering to not caused by any specific bacterium; rather
gutta-percha from a purulent infection around it is associated with a heterogeneous group of
the apex of an endodontically treated tooth organisms consisting of at least fungi, viruses,
was compared to biofilm from similarly treated and bacteria, including several currently uncul-
teeth with apparently healthy periapical tissues, tured bacteria.
using the FISH technique.71 Whereas the lat- Microbes from both primary and posttreat-
ter contained few to no bacteria, the former ment apical periodontitis have easy access
revealed dense bacterial aggregates of varying into the neighboring tissues and blood vessels,
composition (Fig 3-15). which may be important because of their high
Studying bacterial diversity in 10 root canals prevalence. A 2017 review found a worldwide
with acute apical abscesses demonstrated that prevalence of 7% to 86% for primary apical
a multitude of different bacteria were involved, periodontitis and 10% to 62% for apical peri-
but none were present in all specimens, indicat- odontitis after endodontic treatment.74
ing great differences between the participants.72
The bacteria most frequently encountered were Dental implants. Compared to patients with
anaerobic gram-negative bacteria of the phylum healthy peri-implant tissues, P gingivalis,
Firmicutes, followed by Bacteroidetes. Another Prevotella intermedia, Tannerella forsythia, and
study found Candida to be more frequently F nucleatum were found to be more abundant
isolated from people with hyperglycemia/ in patients with peri-implantitis, whereas no
diabetes than in normoglycemic patients with important roles seemed to be played by Aggre-
endodontically treated teeth.73 In conclusion, gatibacter actinomycetemcomitans (previously
endodontics-related microbial overgrowth is called Actinobacillus actinomycetemcomitans)

54
The Human Oral Microbiome

Fig 3-15  |  FISH staining of biofilm fractions detected


in the transport media of the retrieved gutta-percha
fractions, as detected by epifluorescence microscopy.
A selection of the taxa is presented in the correspond-
ing subfigures. The FISH probes used for the staining
are provided in parentheses. Scale bars are 10 μm. (Re-
printed from Zehnder et al71 with permission.)

A: Prevotella sp. et rel. (PRV392-Cy3) B: Fusobacteria sp. (FUS664-Cy3)


C: Treponemes Cluster 1 (TrepG1-679-Cy3) D: Synergistes Cluster A (SYN-A1409-FAM)

E: Selenomonas sp. et rel. (SEL1150-FAM) F: Parvimonas micra (Pmic 740-Cy3)


G: Aggregatibacter actinomycetemcomitans (Aact639-Cy3)
H: Actinomyces naeslundii, A. oris et rel. (L-Act476-2-Cy3)
I: Campylobacter rectus et rel. (CAMP655-Cy3)

J: Porphyromonas gingivalis (L-Pgin1005-2-Cy3) L: Prevotella nigrescens (Pnig657-Cy3)


K: Tannerella forsythia Cluster 1 (Tfor127-Cy3) M: Prevotella denticola (Pdent654-FAM)

and Staphylococcus species.11 The first system- Helicobacter pylori was found to be cor-
atic review and meta-analysis, based on 12 related with the level of hyperglycemia assessed
studies exploring the effect of hyperglycemia by glycated hemoglobin (HbA1c), which reflects
on peri-implant tissue health, concluded that long-term blood sugar levels, in a dose-response
whereas there was no significant difference manner.75 It can be debated whether H pylori
regarding peri-mucositis, peri-implantitis was is a commensal or a frequently encountered
significantly more frequently present in peo- transient member of the oral microbiome.75
ple with diabetes.18 Meta-analysis of data from
seven sufficiently similar studies calculated that Orthodontic appliances. Based on 13 stud-
people with diabetes had almost a 50% greater ies, a 2017 systematic review concluded that
risk (RR = 1.46; 95% CI: 1.21–1.77) and 90% placement of orthodontic brackets leads to an
greater odds (OR = 1.89; 95% CI: 1.31–2.46) increase in the abundance of certain bacteria in
for peri-implantitis than normoglycemic the dental plaque.76 However, several months
implant recipients. Meta-analysis of the three after the removal of the orthodontic appliances,
studies that included only nonsmokers (247 the bacterial abundance seemed to return to
hyperglycemic and 74 normoglycemic partici- pretreatment levels. Nonetheless, a prospec-
pants with a total of 706 implants) calculated tive cohort study monitoring four periodontal
a three-fold greater risk for peri-implantitis bacteria concluded that local factors related to
among those with hyperglycemia (RR = 3.39, the orthodontic appliances influenced the alter-
95% CI: 1.06–10.81).18 The latter results cir- ations in the subgingival plaque that resulted in
cumvent the confounding effect of smoking. increases in inflammation and gingival bleed-
ing.77 Also, both fixed and removable space

55
3 The Traveling Oral Microbiome

retainers are shown to cause an increase in the bacteria usually associated with periodontitis—
abundance of both bacteria and yeast (mostly to be strongly associated with periodontitis.84
Candida) colonization.78 Conversely, a study The finding that the prevalence of the oppor-
among women free of periodontitis receiving tunistic bacteria E coli, E faecalis, S aureus, and
clear aligners showed decreased diversity in P aeruginosa is correlated with the presence
the subgingival microbiome with changes in and severity of periodontal disease could be
the bacterial communities but no development regarded as confirmation that host factors are
of periodontitis after the first 3 months upon important in invasion by common inhabitants
aligner placement.79 of other body sites that multiply in the mouth
under certain favorable conditions. This dis-
Removable dentures. Removable dentures can covery further supports the concepts that, in
function as a reservoir and lead to an increase periodontitis, (1) bacteria (and viruses) interact
in the microbial load consisting of both bac- with microbes that do not normally inhabit the
teria and fungi.80 Biofilm from such dentures mouth and (2) groups of several microbes, not
is shown to contain a high number of oral only one bacterium, are found in periodontitis.
bacteria that are usually associated with oral
diseases and are resistant to several antibiotics
and other antimicrobial drugs.81 The micro- Intraindividual and interpersonal diversity
biome of denture wearers is demonstrated to Saliva. The composition of the salivary micro-
differ from that of individuals with only nat- biome varies both within and among persons
ural teeth in both diversity and composition and over time. Figure 3-16 presents the relative
and is influenced by the presence or absence proportions of the most frequently identified
of natural teeth as well as the location of the bacterial phyla for each of five individuals at
denture.82 It was also illustrated that the devel- different time points. The salivary microbial
opment of denture stomatitis is much more community appeared to be stable over at least
complex than a simple overgrowth of C albi- 583 to 7 days.61 However, samples taken up to
cans because bacteria also play a role. 29 days after baseline showed that the compo-
sitions of microbiome samples taken at closer
Oral epithelial cells: Hostels for traveling time intervals were not necessarily more similar
microbes from elsewhere. A variety of visit- than those obtained over longer intervals in
ing (transient) bacterial species are harbored any individual. The saliva samples were dom-
in oral epithelial cells, which together could inated by the seven major groups (phyla), but
be considered a reservoir for these bacteria. only five groups were found in all participants
In one study, Enterococcus faecalis was the at all times (see Fig 3-16). The remaining two
most prevalent visiting species found in oral were also found in all participants but not at
epithelial cells (20.6%) but was identified only every time point.
in people with deep periodontal pockets.83 No Stimulated saliva samples from 30 partici-
correlation was found with age, sex, bleeding pants (10 with periodontitis, 10 with caries,
on probing (BOP), or supragingival biofilm. and 10 orally healthy) were analyzed with a
Half of those with great attachment loss also novel technique called HOMINGS (Human
harbored Pseudomonas aeruginosa. Oral Microbe Identification using Next Gen-
Another study found that subgingival plaque eration Sequencing) and did indeed contain
samples contained DNA from these two bacterial profiles that differed among the
bacteria as well as from two more non-oral three groups as seen in Fig 3-17.85 These pro-
pathogenic bacteria (Escherichia coli and S files were further confirmed by metagenomic
aureus). DNA from these four bacteria was and metatranscriptomic analysis that support
found—alone or together with DNA from a hypothesized genetic expression.86

56
The Human Oral Microbiome

100–

80–


Unclassified bacteria
60– Spirochaetes
Fusobacteria
– TM7
Bacteroidetes
40–
Actinobacteria
– Proteobacteria
Firmicutes
20–

0–
29

15

15

1
4
1
4

5
4–

5–
5–
4–
4–
1
5

1
7

1
5

l
5–

Al
1–

2–

3–
1–
1–

2–
2–

3–
3–

Fig 3-16  |  Relative abundance of predominant phyla across 15 salivary microbiomes, namely three from each
of five individuals on three different days. Sample numbers include subject ID and the follow-up day after the first
sampling time point (day 1). The days are indicated by the number after the hyphen (-) on the horizontal axis. For
instance, the saliva samples from the leftmost participant were taken on days 1, 5, and 29. Bacterial phyla are
indicated by color. The rightmost column, designated as All, displays the average phyla proportions when the
individual samples were pooled. Both Streptococcus and Staphylococcus species belong to the order Bacilli,
which are part of the phylum Firmicutes (dark blue). (Reprinted from Lazarevic et al64 with permission.)

A periodontal intervention study reported Geographic diversity. Several research teams


a strong correlation between reduction in have explored whether various populations
abundance of bacteria in the subgingival in different countries and of different racial
and salivary microbiomes.87 Furthermore, backgrounds indeed share a core oral micro-
the effect of smoking is sufficiently strong biome despite their individual variation. There
to enable distinction between the salivary is scant evidence that some evolutionary lin-
microbiomes in smokers and in nonsmokers eages of bacteria have adapted to particular
receiving single-tooth implants in the posterior ethnic groups.89 With the limited informa-
mandible.88 Smokers harbor oral microbiomes tion available, few of these differences can be
containing lower richness and diversity than directly related to differences in prevalence
nonsmokers but with certain bacteria occurring of periodontal disease. Asian populations are
more frequently. Specifically, the abundance regularly colonized with A actinomycetemcom-
of P gingivalis was significantly greater in itans serotype C with questionable pathogenic
smokers, which was related to greater sever- potential. On the contrary, the JP2 clone of A
ity of peri-implant marginal bone loss during actinomycetemcomitans causes significantly
the 3-month bone healing period following higher prevalence of aggressive periodontitis in
implant placement.88 Consequently, smoking adolescents whose descent can be traced back
might directly affect the microbiomes as man- to the Mediterranean region and western parts
ifested in both dental plaque and saliva. of Africa.89

57
3 The Traveling Oral Microbiome

45
40 Prevotella veroralis Veillonella atypica
Campylobacter concisus Porphyromonas sp oral taxon
35 Streptococcus sanguinis Prevotella pallens
Gemella sanguinis Veillonella atypica
30
Megasphaera micronuciformis Prevotella salivae
25 Oribacterium sinus Prevotella histicola
Neisseria pharyngis Fusobacterium periodonticum
20 Veillonella rogosae Rothia mucilaginosa
15 Neisseria flavescens Haemophilus parainfluenzae
Leptotrichia sp oral taxon 417 Prevotella melaninogenica
10
5
0
a Periodontitis Oral health Dental caries

25 Veillonella genus probe 1


Actinomyces genus probe 4

20 Streptococcus genus probe 1


Leptotrichia genus probe 1
Prevotella genus probe 1
15
Fusobacterium genus probe 4
Granulicatella genus probe
10 Veillonella genus probe 2
Neisseria genus probe 2

5 Streptococcus genus probe 4

0
b Periodontitis Oral health Dental caries

Fig 3-17  |  Relative abundance of predominant species-level and genus-level probe targets in saliva. (a) Relative
abundance of the 20 most predominant species-level probe targets in each group. (b) Relative abundance of the
10 most predominant genus-level probe targets in each group. (Reprinted from Belstrøm et al85 with permission.)

In Korean adults with advanced periodonti- subgroups (genera) with non-Amerindians


tis, all diseased sites harbored Fusobacterium from previous studies and had a lower rich-
species, while P gingivalis, Treponema spe- ness of genera (51 versus 177 genera reported
cies, and T forsythia were detected in more in non-Amerindians). In addition, their micro-
than 96% of the sites.90 Even in periodontally biota included some bacteria common in soil.
healthy subjects, Fusobacterium species were In another study, saliva samples were
present in the highest proportion (58%) of obtained from 10 individuals from each of 12
sites, while Treponema species, P gingivalis, worldwide locations.92 Overall, approximately
and T forsythia were each detected in about 13.5% of the total variance in the composition
one-fifth of healthy sites. of genera was due to differences among indi-
The oral mucosa microbiota in cheek swabs viduals. Investigation of some environmental
from six Amazon Amerindians of Guahibo variables revealed a significant association
ethnicity living in Venezuela was highly dom- between the genetic distances among loca-
inated by four bacterial groups (phyla).91 tions and the distance of each location from
These Amerindians shared only 23% of the the equator.

58
The Human Oral Microbiome

There is high diversity in the salivary micro- their categorical interactions are exemplified in
biome within and among individuals, indicative Panel B, showing strong and weak cooperation,
of an enormous geographic diversity in the exploitation, and competition.96
human salivary microbiome. Microbial com- Figure 3-19 illustrates four ways in which
munity diversity in the Echuya Batwa, a former two oral bacteria that coexist in a symbiotic
hunter-gatherer group from Uganda, is signifi- relationship within a biofilm can cooperate,
cantly higher than that in agricultural groups resulting in one species enhancing the virulence
from Sierra Leone and the Democratic Republic of the other.97
of the Congo.93 Forty microbial genera previ- In contrast to microorganisms growing in
ously not described in the human oral cavity a planktonic (freely floating) state, the inhab-
were identified in the Batwa. Their distinctive itants of communities within this densely
composition of the salivary microbiome may structured biofilm are effectively protected
have been influenced by recent changes in life- from host defense mechanisms and from ther-
style and diet. apeutic agents, including antimicrobial agents.
Knowledge regarding geographic variations For example, enterococci were harvested by
in the oral microbiome and their relationship to inserting paper points into the periodontal
periodontal disease and other oral conditions pocket of patients with periodontitis and
could be clinically important for considering showed a strong association with biofilm
precision periodontal treatment in the future. formation.98 These bacteria turned out to
be resistant to vancomycin, erythromycin,
ciprofloxacin, teicoplanin, amoxicillin, and
The dynamic oral microbiome gentamicin and confirmed the role of the sub-
The teeth are unique in that they do not con- gingival dental plaque as a reservoir for such
stantly renew their outer layer, shedding the antibiotic-resistant bacteria.
old, as other human tissues do. Therefore, Like human microbiomes in other body loca-
they allow a subgingival biofilm, consisting of tions, such a microcosm includes organisms of
a highly variable and complex community of varying types and proportions, depending on
microbes, to colonize, organize, and develop for whether its environment is in homeostasis.
extended periods. Bacteria within dental plaque Once the milieu is altered, this delicate balance
biofilms are in a dynamic state and interact with is interrupted to the benefit of certain com-
each other, embedded within a self-produced mensal microbes, which subsequently exhibit
matrix of extracellular polymeric substance opportunistic overgrowth to the detriment of
that consists of polysaccharides, lipids, proteins, other members of this biofilm community;
and nucleic acids. In the subgingival plaque the latter organisms then diminish in amount,
matrix, there is also abundant extracellular possibly even to rates below detection. The
DNA (eDNA) located outside the microbes but oral microbiome formation and the virulence
emanating from them. In addition to carriage of of its various components are influenced by
DNA, this eDNA may play an important role environmental factors such as acidity (pH),
in early biofilm formation and possibly in hori- temperature, cytokines, hormones, and oxida-
zontal transfer of DNA between oral bacteria,94 tive stress.99
as well as in protection against desiccation or Babies are exposed to a multitude of micro-
radiation and avoiding overcrowding.95 organisms both in utero and upon delivery. The
Various organizational patterns of cohabita- oral microbiome of a newborn infant changes
tion by different microbial species living in the quickly and is influenced by many factors,
same biofilm (dental plaque) that could contain among them the feeding mode.100 This means
bacteria, fungi, and possibly viruses are illus- that the abundance and composition of the
trated schematically in Fig 3-18 (Panel A), and bacterial oral microbiome in newborns differ

59
3 The Traveling Oral Microbiome

Fig 3-18  |  Interspecies bacterial interac-


tions in multispecies biofilm spatial organi-
zation. (a) Spatial organization of bacteria in
1 2
biofilms: (1) intermixinga; (2) layered struc-
ture without patchy patterninga; (3, 5b) in-
terspecific segregationa; (4) layered struc-
Strong cooperation
ture with patchy patterninga,c; (6) patchy
patterning structureb,c; increased biomass
3 of one or all member species in mixed spe-
ciesb; decreased biomass of one or all mem-
ber species in mixed species; one species
Weak cooperation
is dom­inanta,c; (b) Interspecies interactions:
cooperation, exploitation and competition.
4
Cooperation and exploitation lead to in-
creased biomass of one or all member spe-
cies in mixed-species biofilms compared to
Exploitation
single-species biofilms (a1 to a4), whereas
competition results in decreased biomass
5 6
of all member species in mixed-species bio-
films compared to single-species biofilms (a5
and a6). Arrows and vertical bars represent
Competition
growth facilitation and inhibition, respectively.
(Reprinted from Liu et al96 with permission.)
a b

Fig 3-19  |  Ways that a microbe can pro-


Symbiosis
mote the virulence of another in a mixed bio-
film: (A) subvert host defenses by limiting/
A. Subverting immunity killing by phagocytosis or complement
activity; (B) provide novel carbon sources
or nutrients; (C) inhibit the growth of com-
B. Providing nutrients peting microbes, allowing overgrowth; (D)
provide novel terminal electron acceptors,
allowing more efficient energy production.
C. Inhibiting competing microbes (Reprinted from Selleck and Gilmore97 with
permission.)

D. Enabling aerobic respiration

between breastfed and formula-fed babies.100 In the former.101 The organophosphate insecticide
addition, changes in the microbiome compo- azinphos-methyl was detected in the blood
sition within both groups were observed over of 30 of the farmworkers, and 27 of them
time, especially for Streptococcus species whose showed a significantly altered composition of
abundance increased markedly between ages 4 the buccal microbiome, including a decrease
through 8 weeks. in the abundance of the genus Streptococcus
A longitudinal study comparing buccal and with extinction suggested of entire taxa
swabs from 65 adult farmworkers to 52 adults in some individuals. Any long-term health
who did not work on farms found significant consequences of such pesticide exposure and
seasonal changes in the oral microbiome rep- subsequent changes in the buccal microbiome
resented by seven commensal bacterial taxa in are currently unexplored.101

60
The Human Oral Microbiome

100% –
Other
90% – TM7
80% – Gammaproteobacteria
Relative abundance of reads

70% – Betaproteobacteria
60% – Coriobacteria
50% – Actinobacteria
40% – Erysipelotrichi
30% – Clostridia
20% – Bacilli
10% – Flavobacteria
0% – Bacteroidia
Fusobacteria

Low Candida load High

Fig 3-20  |  Relative abundance of bacterial taxa in saliva samples at the class level. The sample order corre-
sponds to the increase in C albicans load in the samples. The Candida load was measured as the proportion
of internal transcribed gene (Candida) over 16S gene (bacteria) abundance by quantitative polymerase chain
reaction. The increase in Candida load was statistically significantly, positively correlated with reads classified as
Bacilli (includes streptococci), while the Candida load correlated negatively with the classes Bacteroidia, Flavo-
bacteria, and Fusobacteria. (Reprinted from Kraneveld et al102 with permission.)

Numerous short- and long-lasting changes salivary microbiome decreased significantly,


in a human’s life cause more or less divergence with increasing load of the yeast C albicans.
from “normal” status in the oral cavity, buccal Not only were there fewer kinds of bacteria
mucosa, gingival and periodontal tissues, gin- present in persons with more C albicans, but
gival crevicular fluid (GCF), and saliva. Many their salivary bacteria composition also shifted
of their effects on the subgingival microcom- markedly to being dominated by sugar-loving
munity are unknown, but emerging research and acid-producing bacteria, namely strepto-
reports some known effects. cocci (Fig 3-20). Such a synergistic relationship
between C albicans and streptococci has been
hypothesized to be an important mechanism
Modifiable changes for maintaining good oral health.103
Because the healthy state is so delicate, it can be This leads to the question of whether con-
questioned whether ardent use of bacteriostatic trolling the oral environment to become less
mouthrinse in the absence of periodontitis or acidic may be a potential preventive measure
gingivitis may actually do some harm and dis- for overgrowth of Candida as well as for pre-
turb the balance, especially due to the common vention of caries, especially on the roots.
alcohol content that also is a desiccant. One way of changing the abundance of bac-
To enable researchers to explore the inter- teria and the composition of the supra- and
actions between fungi and bacteria, 82 older subgingival87 microbiomes, at least tempo-
Dutch community dwellers delivered unstim- rarily, is by periodontal therapy that leads to
ulated saliva samples (passive drooling into a decrease in the abundance of certain peri-
a sterile cup) first thing on awakening in the odontal bacteria, especially those most closely
morning.102 The bacterial diversity of the associated with periodontitis.104,105 Home oral

61
3 The Traveling Oral Microbiome

hygiene may also alter the oral microbiome, output and hence a decrease in the abundance
such as brushing with a toothpaste that con- of the microbial defense substances contained
tains enzymes and proteins that enhance the in saliva.113 An example of such a substance
natural defense mechanisms of saliva and shifts is the family of histatins that contains natural
the ecologic balance of the oral bacterial micro- defense properties against oral microbes such
biome community toward health.106 as bacteria and fungi and, when present in
Another way is to decrease or cease tobacco diminished concentration, may predispose to
smoking107 and betel nut chewing.108 Even candidal overgrowth.114
subtle changes to some aspects of cigarette Systemic illness also causes changes in the
smoking, for instance degree of mentholation, oral microbiome, especially ailments that affect
show temporal changes in the oral microbi- the immune system, such as psychologic stress,
ome after 14 days.109 Al-Hebshi et al examined HIV, or the use of immune-suppressive medi-
491 species-level taxa belonging to 178 genera cation in conjunction with transplants.115 Even
and 11 phyla in the bacteriome of smokeless the progression of gingivitis to periodontitis
tobacco users.110 Richness and diversity of leads to interspecies interactions resulting in
the species were greatest in users of Swedish microbiome transitions.116
snus and lowest in users of Yemeni shammah; A definitive way to change the oral micro-
the effects of smokeless tobacco on the oral biome is to become pregnant and give birth or
microbiome were found to be qualitative and otherwise terminate the pregnancy. A Japanese
quantitative as well as functional. A study of study following 132 healthy pregnant women
oral microbes in oral wash from 1,204 US and 51 similar but nonpregnant women exam-
adults confirmed that the compositions of the ined seven microbial species three times from
microbiomes in current and noncurrent (former weeks 7 through 39 of pregnancy and found
and never) smokers were different, with those the total abundance to be greater in pregnancy.
in smokers harboring less diversity.33 Further- The incident proliferation of P gingivalis and A
more, the microbiomes depleted by smoking actinomycetemcomitans was especially signif-
lead to shifts in functionality that potentially icant, whereas P intermedia and F nucleatum
could impact smoking-related diseases. While a did not change over time.117 Another Japanese
lower diversity in the buccal mucosal microbi- study comparing similar pregnant (n = 30)
ome was found in smokers when comparing 23 and nonpregnant women (n = 30) found no
current smokers with 20 never-smokers, there difference in salivary flow, but the prevalence
also were oral sites whose microbiome com- of oral yeast (likely mostly Candida species) in
position did not differ by smoking status.111 pregnant women was double that of nonpreg-
A third way to change the abundance of bac- nant women.118 Moreover, during the 6-month
teria is to decrease the level of hyperglycemia, study, the abundance of yeast increased sta-
as high levels of blood glucose enhance the tistically significantly in the pregnant women
growth of certain oral bacteria, even to such a from the first to the third trimester but not
degree that the microbiomes actually exhibit in the nonpregnant women, likely due to the
distinct clustering by various blood glucose lev- measured greater salivary acidity in pregnancy.
els.112 Importantly, hyperglycemia and smoking
affect the oral microbiome in different ways,
and when both occur their synergistic effect
on the microbiome is greater than the sum of Traveling Oral Microbes
their respective effects.112 Hyposalivation can
be a consequence of long-term hyperglyce- The effects of oral microbes on the rest of
mia that damages the nerves of the salivary the body may be categorized as indirect and
glands, which leads to decreased salivary direct. The indirect effects consist of the body’s

62
Traveling Oral Microbes

general cascade of inflammatory reactions to is described. This is considered intrapersonal


those microbes and bacterial endotoxins. The travel. In the next section, interpersonal travel
direct effect is observed as the consequences of between humans is addressed.
specific oral organisms traveling to and colo-
nizing body locations outside the oral cavity.
Therefore, the particular microbes need to be Intrapersonal
identified via detection of the microorganism By air: Aspiration. Pulmonary aspiration refers
itself or specific antibodies to it. The rest of to the entry of material from the oropharynx
this chapter focuses on the direct effect of oral (upper respiratory tract) or gastrointestinal
microbes venturing outside the oral cavity and tract into the lungs via the larynx and lower
emphasizes the logistics of their travels. respiratory tract, which is the part of the respi-
ratory system from the trachea to the lungs.
When food or drink is aspirated, people use
Travel destinations the expression that the particle or liquid “went
In addition to the oral cavity and connected down the wrong (wind) pipe.”
spaces, oral microbes have been identified in
the following body sites: By cell invasion: Attachment, entry/internal-
ization, trafficking, persistence, and exit. P
• Oropharynx gingivalis is able to transfer from one cell to
• Pathologic heart valves another, even when the cells are of different
• Heart valve prostheses types and when the cells are not in cell-to-cell
• Surfaces of intracardiac devices (pacemakers contact, although that speeds up the transit.34
and implantable cardioverter-defibrillators) Usually, autophagy is intended to and succeeds
• Atherosclerotic plaque in the cardiovascular in enclosing and devouring its target, such as
system (coronary arteries, carotid arteries, and a microbe that can cause harm. However, Fig
coronary thrombi in myocardial infarction) 3-21 illustrates conceptually how P gingivalis
• Brain (atherosclerotic thrombi in ischemic makes use of the autophagic pathway in the
stroke, brain tissue, and meninges) case of human coronary artery endothelial cells
• Respiratory tract (lungs) (HCAEC) by the following:
• Liver
• Gastrointestinal system • Adhesion: attaching to the outside of the cell
• Pancreas • Internalization: entering into the cell
• Joints (synovial fluid) • Trafficking: moving inside the attacking ves-
• Vertebrae icle (autophagosome) inside the cell
• Vertebral disc • Persistence: staying alive and multiply-
• Mother-infant unit in pregnancy (fetal brain ing inside the autophagosome, to which P
tissue, amnion, chorion, amniotic fluid, gingivalis signals not to cause the final dis-
umbilical cord, and placenta) integration of its contents
• Breast milk • Exit: leaving the autophagosome still viable
• Vagina, cervix and in great numbers able to infect other
• Voice prostheses cells34,119

An important feature of the virulence of P


Travel options gingivalis is that this bacterium can not only
Dissemination of oral microbes occurs in var- enter and stay viable inside cells but even
ious ways. First, travel within a human body multiply within cardiovascular cells and ath-
via different avenues and by various vehicles erosclerotic plaques in humans and eventually

63
3 The Traveling Oral Microbiome

Fig 3-21  |  Proposed entry, trafficking, and


persistence of strains of P gingivalis that
Adhesion (0–15 m)
utilize the autophagic pathway in human
carotid artery endothelial cells. m, minutes.
Signaling
(Reprinted from Olsen and Progulske-Fox34
Rough with permission.)
Internalization endoplasmic
(15–16 m) reticulum
Early phagosome
(Rab5)

Late phagosome
(Rap1)
Early autophagosome
(Atg7 and BiP)

Trafficking (60–240 m)

Late autophagosome
(BiP and Lamp1)

Phagolysosome Replicating vacuole (2)


(Lamp1 and cathepsin L)

Autolysosome Persistence
(Lamp1 and cathepsin L) (> 240 m)

exit.34 This technique is often referred to as Figure 3-22 illustrates schematically two pro-
a “Trojan horse” mechanism. There are dif- posed mechanisms for microbial involvement
ferences in virulence between the different P in atherogenesis, namely bacteremia-mediated
gingivalis strains, among which the invasion delivery to the left and phagocyte-mediated
and exit mechanisms vary and also depend delivery to the right.119 Note the release of
on the type of host cell.34 P gingivalis is bacteria upon the death of the host cell, illus-
also able to adhere to and eventually invade trated to the right in Fig 3-22. This idea is not
human fibroblasts, the most abundant cell in just speculation, as shown in a human study
the periodontium, which induces expression in which DNA from periodontal bacteria
of proinflammatory mediators that contribute was found in 78% of carotid atherosclerotic
to the breakdown of periodontal tissues and plaques with internal hemorrhage.121 Older
chronic inflammation.120 Other oral plaque Japanese men with hypertension had highly
bacteria, such as S mutans, are also able to use elevated levels of the periodontal bacteria A
this pathway with HCAEC, and still others can actinomycetemcomitans and P intermedia in
stimulate P gingivalis to use this mechanism, the subgingival plaque compared to normo-
such as F nucleatum and Filifactor alocis.34 A tensive men, whereas there was no difference
2018 review also describes emerging research in women.122 This could be interpreted to be
of the influence of P gingivalis on adult den- a clinical manifestation of periodontal bacte-
tal stem cells isolated from dental pupal and ria–associated atherosclerosis, which would
periodontal ligament cells as well as exfoliated be in line with a 2016 systematic review
primary teeth.120 and meta-analysis based on 12 studies that

64
Traveling Oral Microbes

Bacterial component of atheromas


Blood flow
Bacteremia MCP-1 Vascular lumen
Blood MN

Apoptotic EC
Leukocyte
Intima
transmigration
Spreading (EC layer)
infection

Growth Spreading
SMC factors infection
proliferation
Media
Tissue MΦ (SMCs)

Fig 3-22  |  Conceptual model of two avenues for systemic dissemination and delivery to the site of inflamma-
tion of the microbial component of atherogenesis. Bacteremia-delivered bacteria invade the endothelial layer
and further spread into deeper tissue (left). Activation of infected endothelia are represented with the release of
proinflammatory chemokines (such as MCP-1) in the lumen, activating blood monocytes (MN) and macrophages
(MΦ) and promoting their adhesion and diapedesis. Phagocyte-delivered viable bacteria internalized in transmi-
grating leukocytes (center) that can switch them into an uncultivable state (red into green), while their internal-
ization by phagocytes can reactivate them (from green to red). Atheromas can grow due to smooth muscle cell
proliferation mediated by macrophage-secreted growth factors. Bacteria are also released upon host cell death
(right). EC, endothelial cell releasing intracellular bacteria; MΦ, macrophage with internalized bacteria; MCP-1,
monocyte chemotactic protein (a proinflammatory chemokine). (Reprinted from Reyes et al119 with permission.)

calculated an increased odds of 50% (OR: 1.50; cells, and its different strains/subspecies pos-
95% CI: 1.27–1.78) for those with moderate/ sess different levels of invasive capabilities
severe periodontitis to suffer from hyperten- that again are associated with the severity of
sion.123 Another study demonstrated for the periodontitis.126
first time in 2017 that intensive periodontal
therapy leads to a significant decrease in blood By direct transfer: Genetic material. In the
pressure and endothelial microparticles up to 6 human superorganism, the bacteria are in
months posttreatment, without the aid of any frequent, intimate contact with human cells.
antihypertensive medication.124 Bacterial DNA, including DNA from bacteria
Intriguingly, not only can periodontal bacte- that are often part of the oral microbiome, inte-
ria invade gingival cells in periodontitis; they grates into the human somatic genome. This is
can even form a complex biofilm inside the gin- often referred to as lateral gene transfer. Figure
giva that seems to differ in composition from 3-23 shows the integration of genetic material
that in the subgingival plaque from which they from the human papillomavirus (HPV) into
originated.125 The virulence of the keystone P human chromosome 8.127 Note the connection
gingivalis depends partly on its ability to invade from the eighth chromosome to the HPV. The

65
3 The Traveling Oral Microbiome

Fig 3-23  |  Identification of HPV integration


into the HeLa genome. The integration of hu-
man papillomavirus genome HPV NC_001357
(red) into chromosome 8 in the HeLa cell ge-
nome represented by human genome version
19 (blue) is supported by read pairs with one
read mapping to HPV and the other mapping to
the human genome (purple lines). The log-
transformed coverage of the reads supporting
integration (purple histogram: axis minimum
= 0, axis maximum = 4) is consistent with the
known integration of the HPV E6 and E7 genes
shown in yellow on the HPV genome. The log-
transformed coverage of the viral mate pairs is
also shown (red histogram). bp, base pair. (Re-
printed from Riley et al127 with permission.)

connections from chromosome 8 to the viral significantly increased the risk for bacteremia
oncogenes E6 and E7 expressions are also indi- after tooth brushing two- to three-fold.129
cated.127 Such transfer occurs more frequently Blood tests 30 seconds and 10 minutes,
in cancer and is thought to promote or possibly respectively, after full-mouth flossing and scal-
contribute to oncogenesis.127 ing and root planing (SRP) in one quadrant
in patients with chronic periodontitis demon-
By blood: Bacteremia. Whenever the integrity strated the development of total bacteremia in
of the oral cavity is compromised, it presents an 30% of the former versus 43.3% of the latter,
opportunity for bacteremia, which is the pres- a nonstatistical difference.130 Furthermore, the
ence of bacteria in the blood. Oral bacteria slip incidence of bacteremia with viridans strep-
into the bloodstream during most professional tococci that include several species and are
dental procedures. However, such bacteremias often associated with infective endocarditis
are short lived and transient, and the highest was 26.7% after both activities, involving
intensity is limited to the first 30 minutes after 11.4% of the total bacteremia in flossers ver-
a triggering episode.128 This is the conclusion of sus 7.6% of those undergoing SRP. Whereas
a review of 50 pertinent reports. Yet some indi- gingival inflammation was correlated with the
viduals are not able to clear the bacteria before incidence of both total bacteremia and viridans
they lodge themselves in new garages on their streptococci, there was no difference between
journey, such as arterial walls and organs. The the two groups in either of the two types of
current consensus is that bacteremia involving bacteremia.130
oral bacteria is caused by everyday activities to Similarly, a systematic review of nine studies
a much greater extent than occasional proce- found 49.4% of patients receiving periodon-
dures by dental professionals. For instance, a tal therapy to develop bacteremia, with the
systematic review and meta-analysis based on species Streptococcus and Actinomyces being
12 reports concluded that the accumulation most abundant and Streptococcus viridans,
of dental plaque and gingival inflammation A actinomycetemcomitans, P gingivalis, and

66
Traveling Oral Microbes

Parvimonas micra (formerly Peptostreptococ- By bloody travel vehicles. Various mechanisms


cus micros and Micromonas micros) occurring for the systemic dissemination of oral microbes
most frequently.131 However, no conclusions via the circulating blood have been suggested.
could be drawn regarding the magnitude or These include flowing freely in the bloodstream;
duration of the bacteremia. catching a ride with a “raft” of lipoproteins,
sometimes along with medium-sized choles-
By blood: Fungemia. Fungemia is the term terol molecules; adhering to red blood cells;
used to describe the situation in which fungi and hiding intracellularly. The last mechanism
are present in the blood. The yeast C albicans is an especially effective mode of transport
has been implicated in some cases of infective that can be likened to the Trojan horse model.
endocarditis. However, the C albicans organ- The microbes are engulfed or cloaked in a
isms that invade the bloodstream do not seem phagocytosis-like fashion, thereby not only
to originate in the oral cavity.132 A Brazilian being transported but also evading discovery
team simultaneously examined the palatal and attacks by the host defense system (circu-
mucosa, the palatal surface of the maxillary lating phagocytes) and antibiotics. P gingivalis,
denture, and the blood of patients with denture the “atherogenic bacterium,” especially uses
stomatitis for the presence of C albicans.133 The this method. Bacteria internalized in white
researchers found a strong relationship between blood cells (phagocytes) can persist in the cir-
denture stomatitis and Candida species from culation for extended periods.
the palatal mucosa and maxillary denture.
Despite abundant amounts of the yeast in these By nerves: Following nerve pathways. Viruses
two locations, however, use of a novel DNA and bacteria, as well as fungi, seem to be able
identification method detected no C albicans to travel along nerve corridors once trans-
in the blood. Conversely, fungemia caused ported to the nerve tissue via bacteremia.
by Candida (C albicans and C parapsilosis),
named candidemia, as well as invasive candidi- Viruses. Herpes simplex virus (HSV) lies dor-
asis are serious and may cause life-threatening mant in the nervous system throughout the
complications in neonates.134,135 life course of the host,136 and the virus seems
to travel along neural networks. Emerging
By bloody entryways and access roads. Routes evidence explores the role of infection and
of possible entry of oral microorganisms into inflammation in the neuropathology of Alz-
the systemic circulation include the following: heimer’s disease, especially in its late-onset
form, and neurotropic viruses from the Her-
• Root canal: periapical and lateral lesions pesviridae family, especially cytomegalovirus
into the alveolar and adjacent blood vessels, (CMV) and human herpesvirus (HHV)-1 and
including those in the maxillary sinus -2.137 A first study of its kind to explore the
• Periodontium: into capillaries and other potential role of HSV and CMV during a
small blood vessels in junctional epithelium lifetime as opposed to focusing on the older
and gingival connective tissues or into the age groups, analyses of data from the third
alveolar blood vessels National Health and Nutrition Examination
• Mucosal lesions with injury to capillaries in survey (NHANES) regarding seropositivity to
the oral cavity: injuries caused by mastica- HSV-1and CMV found that such seropositiv-
tion of hard food items or biting of tongue ity was associated with cognitive impairment
or cheek; denture-related ulcers; candidiasis; at all ages over 6 years.136 HSV-1 seropositivity
and pierced tissue (with jewelry): tongue, lip, was associated with lower reading and spa-
and maybe face via saliva tial reasoning test scores in children ages 6 to
16 years and was linked to impaired coding

67
3 The Traveling Oral Microbiome

speed. CMV seropositivity was associated commonly encountered in the oral cavity
with impaired learning and recall in those with or without oral diseases is potentially
aged 20 to 59 years, and HSV-1 seropositiv- important due to the aging of the world’s pop-
ity was associated with immediate memory ulation and to the fact that Alzheimer’s disease
impairment in the oldest group (> 60 years).136 accounts for 60% to 80% of dementia cases.142
In addition to HSV and CMV, hepatitis C Any infection and its subsequent sys-
virus may also be associated with Alzheimer’s temic inflammation can lead to formation
disease.137 of amyloid-beta (Aß or Abeta) that consists
of peptides of 36 to 43 amino acids. Aß is
Bacteria. Bacterial infection and its subse- deposited as senile plaques, a hallmark of Alz-
quent inflammatory responses are increasingly heimer’s disease together with synaptic loss
accepted as potential risk factors for Alzheimer’s and neurofibrillary tangles consisting of hyper-
disease, as shown in cadavers.138 It seems that phosphorylated tau protein. Aß is a protective
periodontitis is increasingly recognized as a risk substance against oxidative stress and has anti-
factor contributing to Alzheimer’s disease.139 microbial properties.37 Neuroinflammation is
Some of the bacteria often found in periodontitis, suggested to contribute to aggravation and
for example P gingivalis and some spirochetes, possibly to initiation of Alzheimer’s disease.143
have developed sophisticated survival mecha- Emerging evidence explores the role of infec-
nisms to avoid detection by the immune system tion and inflammation in Alzheimer’s disease,
and therefore may reach the brain tissue by especially its late-onset form, and neurotropic
traveling along the nerves leading thereto, viruses from the family Herpesviridae, espe-
in addition to the dissemination by blood. In cially HHV-1, HHV-2, and CMV, are suggested
especially susceptible individuals, they can acti- to be associated within Alzheimer’s disease neu-
vate and perpetuate a series of inflammatory ropathology.137 However, the degree to which
reactions, aided by the chronicity of periodon- these viruses actually originate in the oral
titis and its responding incessant inflammatory cavity and contribute to general inflammatory
reactions.139,140 A study attempted to shed light cascades or actually migrate to the brain needs
on this association by creating and relating two further exploration.
scales, namely the “Cognitive and periodontal P gingivalis infection has been reported to
impairment state,” including measures of the cause sleep pattern disturbances by changing
mental state and BOP, and the “Inflammatory molecular clock activity in glial cells register-
state” that measured levels of cytokines.141 ing light and darkness, and this in turn can
Whether any causal links exist between the two affect the clearance via the lymphatic system
scales or whether this association simply rep- of potentially disease-associated substances,
resents co-occurrence, as inflammation is part such as Aß.144 This possibility has led authors
of the pathology of both BOP and Alzheimer’s to wonder whether improved management of
disease, needs further exploration. commensal periodontal bacteria may delay or
prevent the onset or progression or Alzheimer’s
Fungi. The same travel mode along nerve cor- disease.144 This idea is intriguing as sleep dis-
ridors is suggested to also involve fungi such as turbances and circadian rhythm disruption are
C albicans, which is suspected to be involved also risk factors for periodontitis,9 but they
in late-onset Alzheimer’s disease.140 may turn out to simply be comorbidities with-
out any causal links.
A large population study reported that
Associated conditions people with periodontitis or gingivitis had an
Alzheimer’s disease. The potential involve- adjusted hazard ratio (HR) for having Alzhei-
ment of viruses, bacteria, and fungi that are mer’s disease of more than twice that of those

68
Traveling Oral Microbes

periodontally healthy (aHR: 2.54; 95% CI: of suffering from primary open angle glaucoma
1.30–3.35).145 In a 5-year study, those with (POAG) than the first quartile (OR: 3.25; 95%
levels of serum antibodies to periodontal CI: 1.20–10.55). This study also included an
bacteria associated with moderate or severe animal model as a proof of concept in which
periodontitis were at twice the risk for devel- mice with glaucoma were injected with bac-
oping incident Alzheimer’s disease as those terial material (lipopolysaccharide), which
without.146 In 2015, periodontitis was shown caused deterioration in the retinal and optical
for the first time in humans to be associated nerves.150
with the load of Aß in the brain.147 A study among 21 patients with idiopathic
As with any other evidence linking oral uveitis, an infection of the interior of the eye,
microbes/periodontitis and general health, and 22 healthy people assessed 11 bacterial
there exists a possibility that periodontitis species in oral plaque samples and inflamma-
merely co-occurs with systemic diseases and tory markers in the GCF, serum, and saliva.151
conditions. Common inflammatory-based The abundance of five of the bacterial species,
chronic diseases, especially in susceptible indi- including A actinomycetemcomitans and F
viduals, generally increase and become more nucleatum, was greater in the patients than in
severe with age when the immune responses their healthy counterparts and were accom-
deteriorate. As Bartold reminds us, while at panied by altered inflammatory biomarkers
the same time pointing out that the only solid observed both locally and systemically.
evidence for the oral-systemic health connec-
tion is between periodontitis and diabetes/
hyperglycemia, “The increasing number of Interpersonal
periodontal-systemic associations corrupts By direct transfer: Direct contact between
the ability of dentists to distinguish which of humans. Speculation abounds regarding
the associations are spurious and which are whether mode of delivery would lead to dif-
valid.”148 That is, any biologic, causal relation- ferences in the composition of the neonatal oral
ship may not exist despite evidence for strong microbiome. A study of maternal and newborn
associations. Reporting on genetic findings, microbiomes found only minor differences in
Carter et al also noted the importance of the the oral gingival microbiome immediately
simultaneous occurrence of several chronic after birth in infants delivered vaginally and
inflammation- and age-related diseases: by cesarean section. At the age of 6 weeks,
“genes related to cognitive disorders, Alzhei- the microbiomes at various body sites of the
mer’s disease, and dementia, and its co-morbid baby did not show any correlation with deliv-
conditions type 2 diabetes, obesity, and cardio- ery method.152 On the contrary, a 2017 review
vascular disease.”149 reported such a difference in the first 6 months
of a baby’s life, while noting that development
Diseases of the eye: Glaucoma and uveitis. of accurate, DNA-based methods for fungus
The oral microbiome has been associated identification are still lacking.153 This review
with the neurodegenerative disease glaucoma concluded that the most frequently studied
that affects the retina and optic nerve, as illus- fungus in vertical transmission from mother
trated by the significantly greater load of oral to child is C albicans.153
bacteria in mouthwash from people with (n
= 58) glaucoma compared to glaucoma-free By indirect transfer: Via saliva. Oral microbes
but otherwise similar individuals (n = 45).150 are transmitted via kissing, sharing of utensils,
Exploring this association in the opposite direc- parents licking or cleaning a pacifier or pre-
tion, those with the highest quartile of bacterial chewing food for their babies, oral sex, biting,
loads had more than three times greater chance and so on.

69
3 The Traveling Oral Microbiome

Cariogenic bacteria are transmitted from bite mark has been abandoned, and instead
parents or other caregivers to the child, whose identification via the culture-independent DNA
oral cavity is sterile at birth. There is limited analysis is generally accepted as hard evidence
contemporary evidence for parent-to-child in court. A research team found that strepto-
transmission of bacteria usually associated coccal DNA from bite mark samples aligned
with periodontal disease. A study genetically significantly more closely with profiles gen-
identifying A actinomycetemcomitans and P erated from the teeth responsible for the bite
gingivalis in subgingival plaque in families with than with those from other teeth in the same
periodontitis revealed interspousal transmis- individual. Hence, streptococcal DNA can be
sion of A actinomycetemcomitans in 4 of 11 used directly from bite marks.161
married couples (36%) and of P gingivalis in 2 Another team examined three genomic
of 10 married couples (20%).154 Parent-to-child regions of streptococcal DNA to discriminate
transmission of A actinomycetemcomitans was between swabs from bite marks and teeth from
seen in 6 of 19 families (32%), whereas P gingi- 16 participants.162 Using three different tech-
valis was not transmitted from parent to child niques, the researchers found probabilities of
in any of the study families. 92%, 99%, and 100%, respectively, for correct
When salivary DNA in edentulous infants identification of the tooth whose plaque was
and their mothers was compared, it became found in the bite mark.
clear that even the young, predental baby Bacterial DNA is preserved for a longer time
hosted a wide variety of bacteria.155 A total of in bite marks than in, for instance, decompos-
397 bacterial genera were identified. Only 28 ing human cells in a corpse and may therefore
were different between the adult and the child, be useful if collected after longer time spans
27 of them seen in the adults. Streptococcus after the bite occurred. Bacterial DNA is more
was the predominant genus in infant saliva, convenient in such cases because it could be
significantly more abundant than in the moth- difficult to distinguish human DNA between
ers (62.2% in infants versus 20.4% in adults). the biter and the bitten. Consequently, oral bac-
This study demonstrated that a diverse bac- terial DNA may be used in forensic dentistry
terial community exists in the oral cavity of to provide documenting information on the
infants even before tooth eruption. It remains identification of the biter.
to be seen which environmental factors affect
the further colonization and subsequent risks Pacifiers. Many parents clean their infant’s pac-
for oral and gastrointestinal disease. ifier with their own saliva by sucking it. In so
Growing evidence supports an oral origin doing, parents transfer their oral microbes to
of bacteria that cause chorioamnionitis, an the child.163 Based on a study of 184 infants,
inflammation of the fetal membranes (amnion pediatricians concluded that vaginal delivery
and chorion), as well as the placenta156,157 and and parents’ cleaning of pacifiers with their
umbilical cord.158 While bacteremia is the main saliva generated independent and additive, sig-
suspected travel mode,159,160 receptive oral sex nificant protective effects against development
from a partner with periodontal disease can of asthma (88% decrease) as well as eczema
also be the cause.159 Interestingly, a compre- and sensitization (63% decrease for each) at
hensive review concluded in 2017 that “The 18 months old. The authors speculated that
composition of the placental microbiome is the child’s immune system is stimulated by
distinct from that of the vagina and has been microbes in the mother’s birth canal and the
reported to resemble the oral microbiome.”160 parents’ saliva.
Bite marks in humans by humans are mostly However, various professional dental organi-
seen in violent crimes, especially in rape attacks zations have strongly opposed recommending
and child abuse. The use of morphology of the this practice because of the risk of transfer of

70
Traveling Oral Microbes

oral organisms that contribute to the develop- recovered a nearly complete genome represent-
ment of caries and periodontal disease. ing a new strain of a periodontal bacterium,
namely P gingivalis JCVI SC001.165
By indirect transfer: Layover in dental labora-
tory or hospital equipment. Removable dentures
are often in need of adjustment and repair due
to resorption of the edentulous alveolar ridges Travelers’ qualifications, methods,
after tooth extraction. Bacteria that inhabit the and frequencies
oral microbiome and can turn into opportunis-
tic pathogens in a favorable milieu have been Oral bacteria easily enter the bloodstream
found in pumice samples from dental labora- when the periodontium is inflamed due to
tories. Additionally, studies have shown that infection. The ease with which this occurs
the pumicing procedure produces splatter and seems to depend on the severity and extent of
aerosols, through which the bacteria can spread. the periodontal infection and inflammation and
is greater in people with poor oral hygiene.
By indirect transfer: Contaminated gloves and That is, the worse the periodontal disease, the
other protective gear. A
 study of health care more easily will bacteria penetrate the blood
personnel at four hospitals and one medical vessel wall and enter the bloodstream. Once in
center used fluorescent lotion and disclosed the blood, the microbes from the periodontal
that contamination of the skin and clothing of biofilm and their byproducts—such as live or
health care personnel occurs frequently during dead microbes and pieces of bacterial cell walls
removal of contaminated personal protective (lipopolysaccharides)—will travel to various
equipment (PPE), such as gloves and gowns, destinations in the body. Lipopolysaccharides
on average in 80% of the cases with incorrect are large molecules that are known also as
PPE removal and even in one-third of the cases endotoxins. They are a major constituent of
following correct PPE removal.164 It should be the outer cell membrane of gram-negative bac-
mentioned that 1 and 3 months after educa- teria, for instance those often associated with
tional intervention, the contamination rate periodontal destruction.
dropped to a stable 12%. Endotoxins can elicit strong immune
responses. Humans are much more sensitive
By air: Sneeze propelled. Although periodontal to them than are other mammals, such as mice.
bacteria are reported to be part of atmospheric A dose of 1 µg/kg induces shock in humans,
air high above the ocean, it is not known but mice will tolerate a dose up to 1,000 times
whether such high flyers actually can infect higher.166 This is a major reason why findings
a new host on landing and multiply in the from research conducted in mice with exper-
adopted environment. Moreover, the literature imental periodontal disease should be viewed
currently seems devoid of confirmed coloniza- with caution, as the results may not transfer
tion through human-to-human transmission directly to humans.
of oral microbes after sneezing or coughing.
However, such transmission may be possible.
Travel qualifications
By water: Sink drain. A first report of its kind Through the use of powerful novel techniques,
was published in April 2013 where DNA from an unexpected genetic diversity within the
P gingivalis was identified in the complex bacterial species has recently come to light. In
biofilm in the drain of a sink in a hospital bath- many cases, the evolution of specific species
room. Applying a novel technique of generating and their subspecies is disproportionately asso-
genomic DNA from single cells, the researchers ciated with infection or overgrowth.89 Only

71
3 The Traveling Oral Microbiome

a limited number of species and strains are • The combined abundances of Veillonella
detected in bacteremias so far. It may be that and Streptococcus species in atherosclerotic
certain microbial attributes play a role in the plaques correlated with their abundance in
ability of specific bacterial species to penetrate the oral cavity.
and survive in the bloodstream, invade distant • DNA from several additional bacteria was
tissues and organs, and ultimately multiply common to the atherosclerotic plaque and
there. Reportedly, some species exhibit innate oral or gut samples within the same individual.
attributes that enhance such activities, includ-
ing the ability to do the following: In the year 2000, Haraszthy et al identified
DNA from periodontal bacteria in 80% of
• Adhere to multiple different surfaces 50 atheromatous plaque specimens obtained
(microbes, tissues, immune cells, and sali- from patients undergoing carotid endarterec-
vary molecules) tomy,169 and several studies have confirmed
• Decrease the phagocytic activity of cells that these findings in specimens in, for example,
would engulf them coronary bypass graft surgery.170 A 2016 sys-
• Penetrate between cells in tissue and into tematic review included 63 studies with a total
host cells of 1,791 patients and reported the presence of
• Initiate, maintain, and ensure long-term 23 oral commensal bacteria within atheroscle-
survival of an intracellular localization for rotic plaques in patients undergoing carotid
transportation, defense, and multiplication endarterectomy, catheter-based atherectomy,
(Trojan horse model) or similar procedures.171 A study of specimens
from atherosclerotic plaque from the carotid
Because only certain subspecies (strains) of arteries found DNA from periodontal bacteria
bacteria seem to travel systemically, it is nec- to not only be present in between 24% and
essary to identify exactly which strains cause 68% of the samples but also to be associated
harm in order to target those organisms in with intraplaque hemorrhage, which is a risk
future therapy and prevention.167 factor for ischemic stroke.121
Overall, these findings strongly support
the hypothesis that the oral cavity can be a
Travel share: The mouth, the gut, and source for atherosclerotic plaque–associated
atherosclerosis bacteria. These bacteria may contribute to
A landmark study was designed to test the further inflammation and eventual destabili-
hypothesis that bacteria from the mouth and/or zation and rupture of the plaque, ultimately
the gut could end up in atherosclerotic plaque causing a heart attack or ischemic stroke. For
and thus contribute to the development of car- example, bacterial DNA from various bacterial
diovascular disease.168 Using 16S rRNA genes oral species and Chlamydia pneumoniae was
to survey and compare the bacterial diversity identified in thrombus aspirates and arterial
of oral, gut, and atherosclerotic plaque samples blood from patients with ST-segment-elevation
from 15 individuals with atherosclerosis and myocardial infarction undergoing primary
15 healthy controls, Koren et al168 found the percutaneous coronary intervention (n = 101;
following: 76% male; mean age: 63.3 years).172 Interest-
ingly, the concentration of bacterial DNA was
• Bacterial DNA was present in atherosclerotic 16 times greater in the thrombi than in the
plaque. blood samples. About four of five (78.2%) of
• Veillonella and Streptococcus species were the thrombi contained DNA from oral viridans
detected in the majority of the atheroscle- streptococci mostly associated with endodontic
rotic plaque samples. infection, and DNA from periodontal bacteria

72
Traveling Oral Microbes

was identified in one-third (34.7%) of the 90% will resolve within 2 years, except in
thrombi. Importantly, the 30 participants with some immunocompromised individuals, such
panoramic radiographs had significant dental as those infected with HIV. For example, an
restorations and pathology, including end- Italian study among 305 HIV-positive men
odontic treatment in two-thirds (66.7%) and found 265 (86.9%) to have HPV detected in
periapical abscesses in almost half (46.6%), of either oral rinse or anal swabs.177
which one-third occurred in conjunction with Importantly, HPV does not multiply in saliva
endodontically treated teeth.172 In the thrombi, or blood. It establishes productive infections
the odds of finding streptococci were 13.2 times only in keratinocytes of the skin or mucous
greater in those with periapical abscesses than membranes. More than 40 HPV types can
in those with healthy periapical tissues (OR: infect the mouth and throat as well as the gen-
13.2; 95% CI: 2.11–82.5). In patients under- ital areas of both males and females. HPV is
going extracorporeal circulation auxiliary to not disseminated via bacteremia but is easily
open-heart surgeries, those with periapical transferred by direct contact, most often during
abscesses were more likely to develop infec- vaginal and anal sex; it may also be passed on
tious complications, such as septicemia, than during oral sex. HPV may also be transmitted
those with healthy periapical tissues.173 via kissing. Whether the exact means of trans-
mission is direct contact or via saliva remains
to be determined. There is as yet no evidence
Frequent traveler: Human papillomavirus that saliva acts as an HPV transmission vehicle
Unquestionably, the most important microbe for oral or genital contamination. However,
transmitted easily by direct contact between given the presence of shed epithelial cells in
people is HPV. HPV might not be thought of oral fluids, this route is quite possible.
as part of the normal human oral microbiome, In addition to causing cancer, HPV infections
and no significant association was identified can cause other health problems, including
between the presence of periodontitis and HPV 100% of genital warts; recurrent respiratory
in oral rinse specimens by adjusted analyses of papillomatosis, a rare condition in which warts
the NHANES 2009–2012 data.174 However, grow in the throat; and juvenile-onset recur-
a pilot study demonstrated the potential for rent respiratory papillomatosis seen in babies
periodontal pockets to be reservoirs for HPV infected by their mothers during delivery. The
by scraping such pockets and identifying mes- types of HPV that can cause genital warts are
senger RNA from HPV in study participants not the same as the types of HPV that can
with chronic generalized periodontitis in 50% cause cancers. Warts can appear from weeks
of the eight participants who showed no signs to months after HPV is contracted,178 and the
of HPV-related diseases.175 patient might be unaware of carrying the virus
As HPV is the most common sexually until symptoms occur.
transmitted virus, the US Centers for Disease
Control and Prevention (CDC) estimates that World traveler: Prevalence of HPV. There are
up to 80% of the US population will harbor more than 100 different kinds of HPV, but not
HPV at some time during their lifetime.176 Most all of them cause health problems. According
affected persons do not realize they are carriers to the World Health Organization (WHO), the
and are passing the virus on to a sex partner, two most prevalent HPV types are HPV-16
and the vast majority of cases go unnoticed. (3.2%) and HPV-18 (1.4%).179 The global
No HPV test for men has been approved by prevalence of infection with HPV in women
the CDC.176 The majority of HPV infections without cervical abnormalities is 11% to 12%;
(many would regard this as carriage, not infec- rates are higher in sub-Saharan Africa (24%),
tion) are undetectable after 1 year, and about Eastern Europe (21%), and Latin America

73
3 The Traveling Oral Microbiome

(16%). Particularly high HPV prevalence is was 4.8% (95% CI: 3.2–7.3%).185 The overall
seen in Eastern Africa and the Caribbean, oral HPV clearance was reported to be 0% to
where rates exceed 30%. In general, higher 80% between studies, and the median time to
rates are observed in less developed regions clearance varied from 6.5 to 18 months.
of the world.179 For instance, 75.3% of the In the United States, almost 80 million indi-
population in a study in India was infected by viduals are currently infected with HPV, which
HPV.180 amounts to almost a full quarter of the entire
HPV was detected in 4.0% of oral rinse spec- US population of 327 million of all ages. About
imens obtained from 1,688 healthy men, aged 14 million people will contract the infection
18 to 74 years (median 31 years), from the each year, and about 360,000 men and women
United States, Mexico, and Brazil. The HPV will develop genital warts.176 Around 7% of the
types were similar in the three countries, except US adult population, namely 10.1% of males
that HPV-55 was seen more often in Mexico.181 and 3.6% of females, have one or more of 37
The strongest predictor for the presence of oral types of oral HPV, detected in purified DNA
HPV was smoking. However, the prevalence of from oral exfoliated cells obtained from oral
oral HPV in these men was only one-tenth that rinse.186,187 Publication in 2017 of analyses of
of infection at genital sites (4% versus approx- data from 2011 to 2014 reported that the prev-
imately 40%). The reason for this discrepancy alence of any oral HPV was 7.3% among US
is not clear, but the authors speculated that the adults aged 18 to 69 years, with 11.5% of men
mouth may be more resistant to infection than and 3.3% of women.188
the anogenital area. Over 12 months, 4.4% of
these men acquired new oral HPV. However, Vaccination and testing for HPV. The CDC
most were cleared within 1 year, after a median now recommends two (down from three) doses
infection period of less than 7 months.182 of HPV vaccine for all girls and boys aged 11
A large population study in China came to or 12 years, as well as for older unvaccinated
a similar conclusion upon following 3,289 individuals, with the second dose given 6 to 12
healthy, rural men and women aged 25 to 69 months after the first.189 In addition to prevent-
years for a mean of 18.3 months with oral ing genital HPV infection, it is shown that HPV
swab specimens,183 namely that the majority vaccination induces neutralizing antibodies in
of these new oral HPV infections were cleared oral fluids in both humans190 and mice; the
within 1 year, with 89.7% cleared after only 3 latter were subjected to different vaccination
months. Recent oral sex was associated with modes that all completely prevented oral infec-
greater incidence of mucosal HPV infection tion with HPV-16.191
with an adjusted hazard ratio of more than As an aside, a study conducted among 10%
10-fold (aHR: 10.13; 95% CI: 2.14–48.06). of Danish dentists showed that they had no
A much greater prevalence of oral (measured increased risk of being infected with HPV.192
in saliva) HPV infections (24%) was reported Dental practitioners are in an eminent posi-
among 100 women aged 22 to 52 years with tion to be alert to any signs of epithelial
known HPV genital infections compared to 8% changes in the oral mucosa. As partners of
in 25 women aged 20 to 49 years without such the patient-centered health care team, den-
genital lesions.184 tal professionals can also contribute to their
Based on nine reports involving 3,762 patients’ general health care by educating and
individuals, a systematic review and meta- encouraging them to obtain HPV vaccination
regression concluded that worldwide and as part of overall health. However, it seems
among people without HIV or cancer, 7.5% dentists may need to increase their literacy on
had oral infections with any type of HPV, and this topic193 and overcome some perceived bar-
the annual cumulative incidence (new cases) riers to patient education.194

74
Traveling Oral Microbes

Intermittent traveler: Herpes simplex virus Many dental professionals refrain from
Several types of viruses inhabit the oral cavity treating a patient during the active phase of an
and are more often associated with periodontal outbreak. This is due to concern about poten-
pockets than gingivitis. The role of these viruses tial viral transmission either via direct contact
as causative agents for periodontitis has been between dental personnel and a patient’s cold
questioned. Nevertheless, HSV merits mention sores or via a virus-containing aerosol created
as a frequent, although intermittently dormant, by air blown into the mouth and bounced back
member of the oral microbiome. over the lips of an infected patient.
After infecting an individual, HSV takes up
permanent residence in the host. HSV spreads
easily between humans, mostly via direct con- Lifelong lurking traveler: Epstein-Barr virus
tact such as kissing and caressing but also The Epstein-Barr virus (EBV), also known as
through shared utensils and towels, lip balm, human herpesvirus 4, is a common member
and razors as well as via sexual contact. The of the human herpesvirus family and as such
carrier from whom the virus is transferred lingers in the human body for life.196 Ironically,
needs not have signs or symptoms of the infec- the EBV infects not only epithelial cells but
tion, such as cold sores. Such transmission is also the B cells of the immune system that are
termed asymptomatic viral shedding. supposed to combat this virus. It stays dormant
HSV-1 is usually acquired in childhood, in the B cells until a triggering event causes it
whereas type 2 (HSV-2) typically spreads to proliferate. Worldwide, more than 90% of
through sexual contact. However, HSV-1 can adults are EBV seropositive.
also be transmitted via oral sex and cause EBV is usually acquired in early childhood or
genital cold sores. In 2015–2016, the over- adolescence. Children are susceptible as soon
all prevalence of HSV-1 in the US population as the antibodies inherited from their mother
aged 14 to 49 years was about half (48.1%), wear off, and about half of 5-year-olds in the
with significantly more females (50.9%) than United States have been exposed to the virus. If
males (45.2%) infected.195 HSV-1 is seen in 6 infection instead occurs during adolescence or
in 10 of the age group 40 to 49 years. More during the teen years, infectious mononucleo-
than one-tenth (12.1%) of that same popu- sis will manifest itself in one-third to one-half
lation is infected with HSV-2, ranging from of the cases in the United States. Because the
0.8% of the youngest (14 to 19 years) to 1 virus is transferred via saliva (and genital secre-
in 5 (21.2%) of the 40- to 49-year-olds.195 tions), infectious mononucleosis is known as
While whites have a prevalence of 8.1%, 1 in the kissing disease (or mono). EBV has also
3 (34.6%) non-Hispanic blacks have HSV-2 been identified in oral diseases such as oral
infection. lichen planus and oral hairy leukoplakia.
About 50 million US adults experience
between one and three symptomatic episodes
every year. After the first outbreak of a her- Reverse traveler: Helicobacter pylori bacterium
petic lesion, the virus moves from the skin to Although not discovered until 1984,197 H pylori
the central nervous system. There it will stay is the bacterium responsible for stomach and
dormant until a triggering event, such as stress, duodenal ulcers. H pylori is one of the most
illness, fever, sun exposure, menstrual period, common pathogenic infective agents; about half
or surgery, interrupts the host’s homeostasis the world’s population is affected. Its preva-
and decreases resistance. The outbreaks may lence correlates closely with socioeconomic and
occur in the buccal and gingival mucosa or on hygienic conditions. Developing countries have
the tonsils, as well as on the lips. a prevalence of 80% to 90%, whereas that of
developed countries is usually less than 40%.198

75
3 The Traveling Oral Microbiome

After pooling results from different stud- along with the systemic multi-antibiotic treat-
ies, a meta-analysis concluded that there is a ment of H pylori. Close collaboration between
close relation between infection with H pylori the patient’s dental professionals and gastro-
in the oral cavity and in the stomach; 45.0% enterologist or other medical care provider is
of gastrically infected individuals and 23.9% pivotal.
of individuals without stomach infection have Because a large proportion of adults harbor
oral H pylori.199 Initial infection with H pylori H pylori in the oral cavity, dental treatment
usually occurs in childhood and has been could result in frequent exposure of the den-
observed in live, cultivable form in primary tal care providers via aerosols. Some studies
endodontic infections.200 have found dental personnel to be at greater
Even in the absence of gastritis, H pylori is risk, whereas others have not. Polish dentists
frequently detected in the oral cavity, especially who had practiced longer than 15 years had a
in people with periodontitis; in dental plaque much elevated rate of infection in their gingi-
in up to 88% of people; in benign laryngeal val sulci, and male dentists were much more
lesions; on the surface of oral cancer lesions; often infected than female dentists.202 More-
and in salivary gland secretions. Additionally, over, a serologic study at a dental school in
the same type of H pylori was identified in the Japan showed that clinical dental professionals
mouth (saliva and plaque) and in the stom- were at 2.7 times greater risk for contracting H
ach, and in one study none of the participants pylori infection over a 6-year period than were
had oral H pylori infection without gastric nonclinical employee controls.203
infection.198
There is no consensus whether H pylori
should be regarded as a transient or a resident Fatal dead ends, road blocks, and tourist traps:
member of the oral biofilm. The mechanism Antibiotics
by which H pylori reaches the oral cavity is Usually the white blood cells, of which 40%
unknown. It is possible that occasional reflux to 70% are neutrophils, a type of granulocyte
or vomit from the gastric (juice) reservoir and phagocyte, defend our bodies by attack-
allows colonization of the oral cavity. It is also ing, ingesting, and poisoning with enzymes,
possible that the reverse is true. In addition, thereby killing dangerous, infectious microbes.
oral presence of H pylori could even present a However, the 2004 discovery of such defense
mode of interpersonal transmission, including systems gone awry pertains to “neutrophil
transmission from mother to child.198 extracellular traps” (NETs) released from
Pooling of findings from 26 studies showed hyperactive or overly reactive neutrophils that
that aggressive systemic antibiotic therapy might conduct their entrapment and microbial
resolved the presence of H pylori in the stom- killing to the detriment of the host as it can
ach in about 90% of individuals but in the result in destruction of tissues, such as the peri-
mouth in only 6%.199 That is, 94% still har- odontium, assisted by enzymes released by P
bored the bacterium in the mouth (tongue gingivalis.204
dorsum, supragingival and subgingival plaque, Antibiotics are used against bacteria, but in
and the saliva) after the intensive antibiotics the interest of limiting further development
course. The oral cavity may act as a perma- of resistant bacteria, myriad other substances
nent or transient reservoir or sanctuary for H with antimicrobial properties are currently
pylori and may be the source of reinfection being investigated for their potential role in
of the stomach, as confirmed by a systematic controlling the dissemination of oral bacteria
review.201 It has been suggested that profes- to the rest of the body as well as for prevent-
sional removal of plaque and calculus as well ing further breakdown of periodontal tissues.
as home oral hygiene procedures be performed The following examples have a focused range

76
Future Travels

A broadly distributed ‘supercore’ pathway: coenzyme A biosynthesis (COA-PWY) Major contributors

Bm
An

Sp
Td

Pf
–1

S
Streptococcus
–2 Lactobacillus
Bacteroides

28 pathways
–3 Haemophilus
Corynebacterium
–4 Propionibacterium
Prevotella
–5 Other
a –6 Unclassified
6 body sites
A human microbiome-enriched pathway: adenosylcobalamin salvage from cobinamide (PWY-6269)
Bm
An

Sp
Td

Pf
–1
S
Bacteroides
Porphyromonas
–2 Parabacteroides
17 pathways

–3 Streptococcus
Prevotella
–4 Escherichia
Klebsiella
–5
b –6
Other
Unclassified

A body area (oral)-enriched pathway: nitrate reduction (DENITRIFICATION-PWY)


Bm
An

Sp
Td

Pf
S

–1 Neisseria
Actinomyces
–2 Lautropia
21 pathways

–3 Capnocytophaga
Kingella
–4 Cardiobacterium
Pseudomonas
–5
c –6
Other
Unclassified
log10(rel. abund.)
–6 –5 –4 –3 –2 Anterior nares Buccal mucosa Supragingival plaque Tongue dorsum Stool Posterior fornix
Skin | n = 157 Oral | n = 183 Oral | n = 191 Oral | n = 210 Gut | n = 249 Vaginal | n = 97

Fig 3-24  |  Core and distinguishing functions of human body site microbiomes: (a) 28 metabolic pathways were
core at all 6 major body sites (“supercore” pathways); (b) 2 supercore pathways and 17 additional pathways
were core in multiple body areas and enriched among human-associated taxa (“human microbiome–enriched”
pathways); (c) 21 pathways were considerably more abundant at 1 body site than at sites from all other body
areas (“body site–enriched” pathways). Heat map values reflect the first quartile of relative abundance. In path-
way bar plots, total (community) abundance is log-scaled, and the contributions of the top seven genera are
proportionally scaled within the total. The six major body sites: An, anterior nares; Bm, buccal mucosa; Sp, su-
pragingival plaque; Td, tongue dorsum; S, stool; Pf, posterior fornix. Major contributors: “Other” encompasses
contributions from additional, known genera; “unclassified” encompasses contributions of unknown taxonomy.
(Reprinted from Lloyd-Price207 et al with permission.)

of action, namely wild blueberry polyphe- individual microbial communities can be iden-
nols that target F nucleatum205 and highbush tified. For example, in 2017 the second wave
blueberry proanthocyanidins targeting A of the National Institutes of Health Human
actinomycetemcomitans.206 Microbiome Project provided 1,631 new
metagenomes from 265 healthy individuals,
bringing the project total to 2,355 genomes
and hence contributing twice the number of
Future Travels hitherto known genomes (Fig 3-24).207 Note
the important, prominent role of the 9 oral
The oral microbiome in humans is extraordi- microbiome sites of the total of 15 in males
narily complex. This community of microbes, and 18 in females.
along with that of the gut, contains the great- By the time of this study, several of the 265
est diversity and abundance of all human healthy adults had provided data from up to
microbiomes. Now that more rapid, reli- three time points, allowing temporal variation
able, and affordable molecular techniques to be observed and classified into rapidly vari-
are available—and many more are in speedy able, moderately variable, and stable subsets.207
development or refinement—the genomes of This is a major step toward understanding the

77
3 The Traveling Oral Microbiome

function and dynamics of microbiomes at a 4. Sender R, Fuchs S, Milo R. Revised estimates for the
number of human and bacteria cells in the body. PLoS
personal level on the path to personalized or
Biol 2016;14:e1002533.
precision medicine, which has been dubbed 5. Bizzarro S, Loos BG, Laine ML, et al. Subgingival micro-
“the way forward.”208 biome in smokers and non-smokers in periodontitis: An
In addition to studying individual micro- exploratory study using traditional targeted techniques
and a next-generation sequencing. J Clin Periodontol
bial communities, researchers must study the 2013;40:483–492.
interaction, organization, and communication 6. Bartold PM, Van Dyke TE. Periodontitis: A host-mediated
(eg, cell-to-cell signaling) of these commu- disruption of microbial homeostasis. Unlearning learned
nities. There are still many microbes in the concepts. Periodontol 2000 2013;62:203–217.
7. Löe H, Theilade E, Jensen SB. Experimental gingivitis
human mouth and the rest of the body whose in man. J Periodontol 1965;36:177–187.
identity and function are largely or com- 8. Borgnakke WS. “Non-modifiable” risk factors for peri-
pletely unknown. These longtime or transient odontitis and diabetes. Curr Oral Health Rep 2016;3:
270–281.
members of the human microbiomes and the
9. Borgnakke WS. Modifiable risk factors for periodontitis
interactions between them must be studied over and diabetes. Curr Oral Health Rep 2016;3:254–269.
time and under myriad internal conditions and 10. Genco RJ, Borgnakke WS. Risk factors for periodontal
external influences within the host and from disease. Periodontol 2000 2013;62:59–94.
11. de Waal YC, Eijsbouts HV, Winkel EG, van Winkelhoff
the greater surrounding environment to learn
AJ. Microbial characteristics of peri-implantitis: A
about their function, flexibility, and reactions case-control study. J Periodontol 2017;88:209–217.
to conditions and stimuli such as diseases and 12. Eick S, Ramseier CA, Rothenberger K, Brägger U, Buser
antibiotics present in the host. Not until these D, Salvi GE. Microbiota at teeth and implants in partially
edentulous patients; A 10-year retrospective study. Clin
and other mechanisms are better understood
Oral Implants Res 2016;27:218–225.
will it be possible to successfully manipulate 13. Lafaurie GI, Sabogal MA, Castillo DM, et al. Microbiome
individual human microbiomes in the quest for and microbial biofilm profiles of peri-implantitis: A
improved oral health. systematic review. J Periodontol 2017;88:1066–1089.
14. Rakic M, Grusovin MG, Canullo L. The microbiologic
Exciting and exotic travels continue to clear profile associated with peri-implantitis in humans: A
new paths on which to walk—or run—wher- systematic review. Int J Oral Maxillofac Implants 2016;
ever they lead into the unexplored lands of 31:359–368.
the microbes inhabiting the human body. This 15. Canullo L, Tallarico M, Radovanovic S, Delibasic B,
Covani U, Rakic M. Distinguishing predictive profiles for
journey will be bursting with new discoveries patient-based risk assessment and diagnostics of
that help improve the future understanding plaque induced, surgically and prosthetically triggered
of interactions among the host genome, epig- peri-implantitis. Clin Oral Implants Res 2016; 27:
1243–1250.
enome, and oral microbiome. Medical and
16. Canullo L, Penarrocha-Oltra D, Covani U, Rossetti PH.
dental professionals have the duty and priv- Microbiologic and clinical findings of implants in healthy
ilege to skillfully apply these discoveries in a condition and with peri-implantitis. Int J Oral Maxillofac
personalized manner to promote the health, Implants 2015;30:834–842.
17. Albrektsson T, Canullo L, Cochran D, De Bruyn H.
including oral health, of each individual human
“Peri-implantitis”: A complication of a foreign body or a
being.209,210 man-made “disease.” Facts and fiction. Clin Implant
Dent Relat Res 2016;18:840–849.
18. Monje A, Catena A, Borgnakke WS. Association
between diabetes mellitus/hyperglycaemia and

References peri-implant diseases: Systematic review and


meta-analysis. J Clin Periodontol 2017;44:636–648.
1. Savage DC. Microbial ecology of the gastrointestinal 19. Naujokat H, Kunzendorf B, Wiltfang J. Dental implants
tract. Annu Rev Microbiol 1977;31:107–133. and diabetes mellitus—A systematic review. Int J Implant
2. Luckey TD. Introduction to intestinal microecology. Am Dent 2016;2:5.
J Clin Nutr 1972; 25: 1292–1294. 20. Misch CE, Perel ML, Wang HL, et al. Implant success,
3. Sender R, Fuchs S, Milo R. Are we really vastly outnum- survival, and failure: The International Congress of Oral
bered? Revisiting the ratio of bacterial to host cells in Implantologists (ICOI) Pisa Consensus Conference.
humans. Cell 2016;164:337–340. Implant Dent 2008;17:5–15.

78
References

21. Potgieter M, Bester J, Kell DB, et al. The dormant blood 38. Ohno T, Yamamoto G, Hayashi JI, et al. Angiopoietin-like
microbiome in chronic, inflammatory diseases. FEMS protein 2 regulates Porphyromonas gingivalis
Microbiol Rev 2015;39:567–591. lipopolysaccharide-induced inflammatory response in
22. Damgaard C, Magnussen K, Enevold C, et al. Viable human gingival epithelial cells. PLoS One 2017; 12:
bacteria associated with red blood cells and plasma in e0184825.
freshly drawn blood donations. PLoS One 2015; 10: 39. Fitzsimmons TR, Ge S, Bartold PM. Compromised
e0120826. inflammatory cytokine response to P. gingivalis lps by
23. Williams JE, Carrothers JM, Lackey KA, et al. Human fibroblasts from inflamed human gingiva. Clin Oral Inves-
milk microbial community structure is relatively stable tig 2018;22:919–927.
and related to variations in macronutrient and micronu- 40. Waller T, Kesper L, Hirschfeld J, et al. Porphyromonas
trient intakes in healthy lactating women. J Nutr 2017; gingivalis outer membrane vesicles induce selective
147:1739–1748. tumor necrosis factor tolerance in a toll-like receptor
24. Pannaraj PS, Li F, Cerini C, et al. Association between 4- and mtor-dependent manner. Infect Immun 2016;
breast milk bacterial communities and establishment 84:1194–1204.
and development of the infant gut microbiome. JAMA 41. Collins FS, Morgan M, Patrinos A. The Human Genome
Pediatr 2017;171:647–654. Project: Lessons from large-scale biology. Science
25. Reichert S, Haffner M, Keysser G, et al. Detection of 2003;300:286–290.
oral bacterial DNA in synovial fluid. J Clin Periodontol 42. Chen T, Yu WH, Izard J, Baranova OV, Lakshmanan A,
2013;40:591–598. Dewhirst FE. The Human Oral Microbiome database: A
26. Haque MM, Merchant M, Kumar PN, Dutta A, Mande web accessible resource for investigating oral microbe
SS. First-trimester vaginal microbiome diversity: A taxonomic and genomic information. Database (Oxford)
potential indicator of preterm delivery risk. Sci Rep 2017; 2010;2010:baq013.
7:16145. 43. Kilian M, Chapple IL, Hannig M, et al. The oral microbi-
27. Li S, Fuhler GM, Bn N, et al. Pancreatic cyst fluid harbors ome—An update for oral healthcare professionals. Br
a unique microbiome. Microbiome 2017;5:147. Dent J 2016;221:657–666.
28. Perez-Muñoz ME, Arrieta MC, Ramer-Tait AE, Walter J. 44. Clamp M, Fry B, Kamal M, et al. Distinguishing
A critical assessment of the “sterile womb” and “in utero protein-coding and noncoding genes in the human
colonization” hypotheses: Implications for research on genome. Proc Natl Acad Sci U S A 2007; 104:
the pioneer infant microbiome. Microbiome 2017;5:48. 19428–19433.
29. Shin H, Price K, Albert L, Dodick J, Park L, 45. Proctor LM. The Human Microbiome Project in 2011
Dominguez-Bello MG. Changes in the eye microbiota and beyond. Cell Host Microbe 2011;10:287–291.
associated with contact lens wearing. MBio 2016; 7: 46. Aagaard K, Petrosino J, Keitel W, et al. The Human
e00198. Microbiome Project strategy for comprehensive
30. Lu LJ, Liu J. Human microbiota and ophthalmic disease. sampling of the human microbiome and why it matters.
Yale J Biol Med 2016;89:325–330. FASEB J 2013;27:1012–1022.
31. Han YW. Fusobacterium nucleatum: A commensal- 47. Human Microbiome Project Consortium. Structure,
turned pathogen. Curr Opin Microbiol 2015; 23: function and diversity of the healthy human microbiome.
141–147. Nature 2012;486:207–214.
32. Reyes L, Phillips P, Wolfe B, et al. Porphyromonas gingi­ 48. Dewhirst FE, Chen T, Izard J, et al. The human oral
valis and adverse pregnancy outcome. J Oral Microbiol microbiome. J Bacteriol 2010;192:5002–5017.
2017;10:1374153. 49. Wade WG. Characterisation of the human oral micro-
33. Wu J, Peters BA, Dominianni C, et al. Cigarette smoking biome. J Oral Biosci 2013;55:143–148.
and the oral microbiome in a large study of American 50. Chen T, Siddiqui H, Olsen I. In silico comparison of 19
adults. ISME J 2016;10:2435–2446. Porphyromonas gingivalis strains in genomics, phylo-
34. Olsen I, Progulske-Fox A. Invasion of Porphyromonas genetics, phylogenomics and functional genomics. Front
gingivalis strains into vascular cells and tissue. J Oral Cell Infect Microbiol 2017;7:28.
Microbiol 2015;7:28788. 51. Zaura E, Keijser BJ, Huse SM, Crielaard W. Defining the
35. Garcia I, Kuska R, Somerman MJ. Expanding the foun- healthy “core microbiome” of oral microbial communi-
dation for personalized medicine: Implications and ties. BMC Microbiol 2009;9:259.
challenges for dentistry. J Dent Res 2013; 92(7 suppl): 52. Paster BJ, Boches SK, Galvin JL, et al. Bacterial diversity
S3–S10. in human subgingival plaque. J Bacteriol 2001; 183:
36. Borgnakke WS, Glick M, Genco RJ. Periodontitis: The 3770–3783.
canary in the coal mine. J Am Dent Assoc 2013;144: 53. Meuric V, Le Gall-David S, Boyer E, et al. Signature of
764–766. microbial dysbiosis in periodontitis. Appl Environ Micro-
37. Hajishengallis G, Lamont RJ. Dancing with the stars: biol 2017;83:e00462-17.
How choreographed bacterial interactions dictate noso- 54. Xu X, He J, Xue J, et al. Oral cavity contains distinct
symbiocity and give rise to keystone pathogens, niches with dynamic microbial communities. Environ
accessory pathogens, and pathobionts. Trends Micro- Microbiol 2015;17:699–710.
biol 2016;24:477–489.

79
3 The Traveling Oral Microbiome

55. Tuna EB, Topcuoglu N, Ilhan B, Gençay K, Kulekçi G. 72. Nobrega LM, Montagner F, Ribeiro AC, Mayer MA,
Staphylococcus aureus transmission through oronasal Gomes BP. Bacterial diversity of symptomatic primary
fistula in children with cleft lip and palate. Cleft Palate endodontic infection by clonal analysis. Braz Oral Res
Craniofac J 2008;45:477–480. 2016;30:e103.
56. Solbiati J, Frias-Lopez J. Metatranscriptome of the oral 73. Gomes CC, Guimaraes LS, Pinto LCC, et al. Investiga-
microbiome in health and disease. J Dent Res tions of the prevalence and virulence of Candida albicans
2018;97:492–500. in periodontal and endodontic lesions in diabetic and
57. Marsh PD. In sickness and in health—What does the normoglycemic patients. J Appl Oral Sci 2017; 25:
oral microbiome mean to us? An ecological perspective. 274–281.
Adv Dent Res 2018;29:60–65. 74. Persoon IF, Özok AR. Definitions and epidemiology of
58. Ghannoum MA, Jurevic RJ, Mukherjee PK, et al. Char- endodontic infections. Curr Oral Health Rep 2017; 4:
acterization of the oral fungal microbiome (mycobiome) 278–285.
in healthy individuals. PLoS Pathog 2010;6:e1000713. 75. Bajaj S, Rekwal L, Misra SP, Misra V, Yadav RK, Srivas-
59. Ghannoum M. The mycobiome: The largely overlooked tava A. Association of Helicobacter pylori infection with
resident fungal community plays a critical role in human type 2 diabetes. Indian J Endocrinol Metab 2014; 18:
health and disease. The Scientist. http: //www. 694–699.
the-scientist.com/?articles.view/articleno/45153/title/ 76. Guo R, Lin Y, Zheng Y, Li W. The microbial changes in
the-mycobiome/. Published 1 February 2016. Accessed subgingival plaques of orthodontic patients: A system-
25 March 2018. atic review and meta-analysis of clinical trials. BMC Oral
60. Larsen T, Fiehn NE. Dental biofilm infections—An Health 2017;17:90.
update. APMIS 2017;125:376–384. 77. Yáñez-Vico RM, Iglesias-Linares A, Ballesta-Mudarra S,
61. Belstrøm D, Holmstrup P, Bardow A, Kokaras A, Fiehn Ortiz-Ariza E, Solano-Reina E, Perea EJ. Short-term
NE, Paster BJ. Temporal stability of the salivary micro- effect of removal of fixed orthodontic appliances on
biota in oral health. PLoS One 2016;11:e0147472. gingival health and subgingival microbiota: A prospective
62. Belstrøm D, Holmstrup P, Bardow A, Kokaras A, Fiehn cohort study. Acta Odontol Scand 2015;73:496–502.
NE, Paster BJ. Comparative analysis of bacterial profiles 78. Arikan V, Kizilci E, Ozalp N, Ozcelik B. Effects of fixed
in unstimulated and stimulated saliva samples. J Oral and removable space maintainers on plaque accumu-
Microbiol 2016;8:30112. lation, periodontal health, candidal and Enterococcus
63. Shaw L, Ribeiro ALR, Levine AP, et al. The human sali- faecalis carriage. Med Princ Pract 2015;24:311–317.
vary microbiome is shaped by shared environment rather 79. Guo R, Zheng Y, Liu H, Li X, Jia L, Li W. Profiling of
than genetics: Evidence from a large family of closely subgingival plaque biofilm microbiota in female adult
related individuals. MBio 2017;8:e01237-17. patients with clear aligners: A three-month prospective
64. Lazarevic V, Whiteson K, Hernandez D, François P, study. Peer J 2018;6:e4207.
Shrenzel J. Study of inter- and intra-individual variations 80. Marchi-Alves LM, Freitas D, de Andrade D, et al. Char-
in the salivary microbiota. BMC Genomics 2010; 11: acterization of oral microbiota in removable dental
523. prosthesis users: Influence of arterial hypertension.
65. Zijnge V, van Leeuwen MB, Degener JE, et al. Oral Biomed Res Int 2017;3838640.
biofilm architecture on natural teeth. PLoS One 2010; 81. Silva SS, Ribeiro MO, Gomes FI, et al. Occurrence and
5:e9321. antimicrobial susceptibility of enteric rods and Pseudo-
66. Simón-Soro A, Tomás I, Cabrera-Rubio R, Catalan MD, monads isolated from the dental prostheses biofilm. J
Nyvad B, Mira A. Microbial geography of the oral cavity. Appl Oral Sci 2016;24:462–471.
J Dent Res 2013;92:616–621. 82. O’Donnell LE, Robertson D, Nile CJ, et al. The oral
67. Zijnge V, Ammann T, Thurnheer T, Gmür R. Subgingival microbiome of denture wearers is influenced by levels
biofilm structure. Front Oral Biol 2012;15:1–16. of natural dentition. PLoS One 2015;10:e0137717.
68. Mark Welch JL, Rossetti BJ, Rieken CW, Dewhirst FE, 83. Colombo AV, Barbosa GM, Higashi D, di Micheli G,
Borisy GG. Biogeography of a human oral microbiome Rodrigues PH, Simionato MR. Quantitative detection of
at the micron scale. Proc Natl Acad Sci U S A 2016; Staphylococcus aureus, Enterococcus faecalis and
113:E791–E800. Pseudomonas aeruginosa in human oral epithelial cells
69. Chen CY, Tsai YF, Chang WY, Yang SC, Hwang TL. from subjects with periodontitis and periodontal health.
Marine natural product inhibitors of neutrophil-associated J Med Microbiol 2013;62:1592–1600.
inflammation. Mar Drugs 2016;14. 84. da Silva-Boghossian CM, do Souto RM, Luiz RR,
70. Olsen I. Organization of supragingival plaque at the Colombo AP. Association of red complex, A. actinomy­
micron scale. J Oral Microbiol 2018;10:1438722. cetemcomitans and non-oral bacteria with periodontal
71. Zehnder M, Rechenberg DK, Thurnheer T, Lüthi-Schaller diseases. Arch Oral Biol 2011;56:899–906.
H, Belibasakis GN. Fishing for gutta-percha-adhered 85. Belstrøm D, Paster BJ, Fiehn NE, Bardow A, Holmstrup
biofilms in purulent post-treatment apical periodontitis. P. Salivary bacterial fingerprints of established oral
Mol Oral Microbiol 2017;32:226–235. disease revealed by the Human Oral Microbe Identifi-
cation using Next Generation Sequencing (HOMINGS)
technique. J Oral Microbiol 2016;8:30170.

80
References

86. Belstrøm D, Constancias F, Liu Y, et al. Metagenomic 102. Kraneveld EA, Buijs MJ, Bonder MJ, et al. The relation
and metatranscriptomic analysis of saliva reveals between oral Candida load and bacterial microbiome
disease-associated microbiota in patients with peri- profiles in Dutch older adults. PLoS One 2012; 7:
odontitis and dental caries. NPJ Biofilms Microbiomes e42770.
2017;3:23. 103. Diaz PI, Strausbaugh LD, Dongari-Bagtzoglou A.
87. Kageyama S, Takeshita T, Asakawa M, et al. Relative Fungal-bacterial interactions and their relevance to oral
abundance of total subgingival plaque-specific bacteria health: Linking the clinic and the bench. Front Cell Infect
in salivary microbiota reflects the overall periodontal Microbiol 2014;4:101.
condition in patients with periodontitis. PLoS One 2017; 104. Mombelli A, Almaghlouth A, Cionca N, Cancela J, Cour-
12:e0174782. voisier DS, Giannopoulou C. Microbiologic response to
88. Duan X, Wu T, Xu X, et al. Smoking may lead to marginal periodontal therapy and multivariable prediction of clin-
bone loss around non-submerged implants during bone ical outcome. J Periodontol 2017;88:1253–1262.
healing by altering salivary microbiome: A prospective 105. Martins SHL, Novaes Jr AB, Taba Jr M, et al. Effect of
study. J Periodontol 2017;88:1297–1308. surgical periodontal treatment associated to antimicro-
89. Rylev M, Kilian M. Prevalence and distribution of prin- bial photodynamic therapy on chronic periodontitis: A
cipal periodontal pathogens worldwide. J Clin randomized controlled clinical trial. J Clin Periodontol
Periodontol 2008;35:346–361. 2017;44:717–728.
90. Choi BK, Park SH, Yoo YJ, et al. Detection of major 106. Adams SE, Arnold D, Murphy B, et al. A randomised
putative periodontopathogens in Korean advanced adult clinical study to determine the effect of a toothpaste
periodontitis patients using a nucleic acid-based containing enzymes and proteins on plaque oral micro-
approach. J Periodontol 2000;71:1387–1394. biome ecology. Sci Rep 2017;7:43344.
91. Contreras M, Costello EK, Hidalgo G, et al. The bacterial 107. Ebersole JL, Steffen MJ, Thomas MV, Al-Sabbagh.
microbiota in the oral mucosa of rural Amerindians. Smoking-related cotinine levels and host responses in
Microbiology 2010;156:3282–3287. chronic periodontitis. J Periodontal Res 2014; 49:
92. Nasidze I, Li J, Quinque D, Tang K, Stoneking M. Global 642–651.
diversity in the human salivary microbiome. Genome 108. Hernandez BY, Zhu X, Goodman MT, et al. Betel nut
Res 2009;19:636–643. chewing, oral premalignant lesions, and the oral micro-
93. Nasidze I, Li J, Schroeder R, Creasy JL, Li M, Stoneking biome. PLoS One 2017;12:e0172196.
M. High diversity of the saliva microbiome in Batwa 109. Chopyk J, Chattopadhyay S, Kulkarni P, et al. Temporal
pygmies. PLoS One 2011;6:e23352. variations in cigarette tobacco bacterial community
94. Rostami N, Shields RC, Yassin SA, et al. A critical role composition and tobacco-specific nitrosamine content
for extracellular DNA in dental plaque formation. J Dent are influenced by brand and storage conditions. Front
Res 2017;96:208–216. Microbiol 2017;8:358.
95. Jakubovics NS, Burgess JG. Extracellular DNA in oral 110. Al-Hebshi NN, Alharbi FA, Mahri M, Chen T. Differences
microbial biofilms. Microbes Infect 2015;17:531–537. in the bacteriome of smokeless tobacco products with
96. Liu W, Røder HL, Madsen JS, Bjarnsholt T, Sørensen different oral carcinogenicity: Compositional and
SJ, Burmølle M. Interspecific bacterial interactions are predicted functional analysis. Genes (Basel) 2017; 8:
reflected in multispecies biofilm spatial organization. 106.
Front Microbiol 2016;7:1366. 111. Yu G, Phillips S, Gail MH, et al. The effect of cigarette
97. Selleck EM, Gilmore MS. Oxygen as a virulence deter- smoking on the oral and nasal microbiota. Microbiome
minant in polymicrobial infections. MBio 2016; 2017;5:3.
7:e01249-16. 112. Ganesan SM, Joshi V, Fellows M, et al. A tale of two
98. Bhardwaj SB, Mehta M, Sood S, Sharma J. Biofilm risks: Smoking, diabetes and the subgingival microbi-
formation by drug resistant Enterococci isolates ome. ISME J 2017;11:2075–2089.
obtained from chronic periodontitis patients. J Clin Diagn 113. Borgnakke WS, Anderson PF, Shannon C, Jivanescu
Res 2017;11:DC01–DC03. A. Is there a relationship between oral health and diabetic
99. Pöllänen MT, Paino A, Ihalin R. Environmental stimuli neuropathy? Curr Diab Rep 2015;15:93.
shape biofilm formation and the virulence of periodontal 114. Trindade F, Amado F, Oliveira-Silva RP, et al. Toward the
pathogens. Int J Mol Sci 2013;14:17221–17237. definition of a peptidome signature and protease profile
100. Al-Shehri SS, Sweeney EL, Cowley DM, et al. Deep in chronic periodontitis. Proteomics Clin Appl 2015;9:
sequencing of the 16S ribosomal RNA of the neonatal 917–927.
oral microbiome: A comparison of breast-fed and 115. Diaz PI, Hong BY, Frias-Lopez J, et al. Transplantation-
formula-fed infants. Sci Rep 2016;6:38309. associated long-term immunosuppression promotes
101. Stanaway IB, Wallace JC, Shojaie A, et al. Human oral oral colonization by potentially opportunistic pathogens
buccal microbiomes are associated with farmworker without impacting other members of the salivary bacte-
status and azinphos-methyl agricultural pesticide expo- riome. Clin Vaccine Immunol 2013;20:920–930.
sure. Appl Environ Microbiol 2017;83.

81
3 The Traveling Oral Microbiome

116. Diaz PI, Hoare A, Hong BY. Subgingival microbiome 131. Horliana AC, Chambrone L, Foz AM, et al. Dissemination
shifts and community dynamics in periodontal diseases. of periodontal pathogens in the bloodstream after peri-
J Calif Dent Assoc 2016;44:421–435. odontal procedures: A systematic review. PLoS One
117. Fujiwara N, Tsuruda K, Iwamoto Y, et al. Significant 2014;9:e98271.
increase of oral bacteria in the early pregnancy period 132. Meurman JH, Siikala E, Richardson M, Rautemaa R.
in Japanese women. J Investig Clin Dent 2017; 8: Non-Candida albicans Candida yeasts of the oral cavity.
e12189. In: Mendez-Vilas A (ed). Communicating Current
118. Rio R, Simões-Silva L, Garro S, Silva MJ, Azevedo Á, Research and Educational Topics and Trends in Applied
Sampaio-Maia B. Oral yeast colonization throughout Microbiology. Badajoz, Spain: Formatex, 2007;719–731.
pregnancy. Med Oral Patol Oral Cir Bucal 2017; 22: 133. de Oliveira CE, Gasparoto TH, Dionisio TJ, et al. Candida
e144–e148. albicans and denture stomatitis: Evaluation of its pres-
119. Reyes L, Herrera D, Kozarov E, Roldá S, Progulske-Fox ence in the lesion, prosthesis, and blood. Int J
A. Periodontal bacterial invasion and infection: Contri- Prosthodont 2010;23:158–159.
bution to atherosclerotic pathology. J Clin Periodontol 134. Harrington R, Kindermann SL, Hou Q, Taylor RJ, Azie
2013;40(suppl 14):S30–S50. N, Horn DL. Candidemia and invasive candidiasis
120. Kriebel K, Hieke C, Müller-Hilke B, Nakata M, Kreike- among hospitalized neonates and pediatric patients.
meyer B. Oral biofilms from symbiotic to pathogenic Curr Med Res Opin 2017;33:1803–1812.
interactions and associated disease—Connection of 135. Calley JL, Warris A. Recognition and diagnosis of inva-
periodontitis and rheumatic arthritis by peptidylarginine sive fungal infections in neonates. J Infect 2017;74(suppl
deiminase. Front Microbiol 2018;9:53. 1):S108–S113.
121. Brun A, Rangé H, Prouvost B, et al. Intraplaque hemor- 136. Tarter KD, Simanek AM, Dowd JB, Aiello AE. Persistent
rhage, a potential consequence of periodontal bacteria viral pathogens and cognitive impairment across the life
gathering in human carotid atherothrombosis. Bull course in the Third National Health and Nutrition Exam-
Group Int Rech Sci Stomatol Odontol 2016;53:e11. ination Survey. J Infect Dis 2014;209:837–844.
122. Aoyama N, Suzuki JI, Kumagai H, et al. Specific peri- 137. Sochocka M, Zwolinska K, Leszek J. The infectious
odontopathic bacterial infection affects hypertension in etiology of Alzheimer’s disease. Curr Neuropharmacol
male cardiovascular disease patients. Heart Vessels 2017;15:996–1009.
2018;33:198–204. 138. Emery DC, Shoemark DK, Batstone TE, et al. 16S rRNA
123. Martin-Cabezas R, Seelam N, Petit C, et al. Association next generation sequencing analysis shows bacteria in
between periodontitis and arterial hypertension: A Alzheimer’s post-mortem brain. Front Aging Neurosci
systematic review and meta-analysis. Am Heart J 2016; 2017;9:195.
180:98–112. 139. Pritchard AB, Crean S, Olsen I, Singhrao SK. Periodon-
124. Zhou QB, Xia WH, Ren J, et al. Effect of intensive peri- titis, microbiomes and their role in Alzheimer’s disease.
odontal therapy on blood pressure and endothelial Front Aging Neurosci 2017;9:336.
microparticles in patients with prehypertension and 140. Olsen I, Singhrao SK. Can oral infection be a risk factor
periodontitis: A randomized controlled trial. J Periodon- for Alzheimer’s disease? J Oral Microbiol 2015;7:29143.
tol 2017;88:711–722. 141. Sochocka M, Sobczyński M, Sender-Janeczek A, et al.
125. Baek K, Ji S, Choi Y. Complex intratissue microbiota Association between periodontal health status and
forms biofilms in periodontal lesions. J Dent Res 2018; cognitive abilities; The role of cytokine profile and
97:192–200. systemic inflammation. Curr Alzheimer Res 2017; 14:
126. Baek KJ, Ji S, Kim YC, Choi Y. Association of the inva- 978–990.
sion ability of Porphyromonas gingivalis with the severity 142. Alzheimer’s Association. 2016 Alzheimer’s disease facts
of periodontitis. Virulence 2015;6:274–281. and figures. Alzheimers Dement 2016;12:459–509.
127. Riley DR, Sieber KB, Robinson KM, et al. Bacteria-human 143. Teixeira FB, Saito MT, Matheus FC, et al. Periodontitis
somatic cell lateral gene transfer is enriched in cancer and Alzheimer’s disease: A possible comorbidity
samples. PLoS Comput Biol 2013;9:e1003107. between oral chronic inflammatory condition and
128. Parahitiyawa NB, Jin LJ, Leung WK, Yam WC, Sama- neuroinflammation. Front Aging Neurosci 2017;9:327.
ranayake LP. Microbiology of odontogenic bacteremia: 144. Harding A, Robinson S, Crean S, Singhrao SK. Can
Beyond endocarditis. Clin Microbiol Rev 2009;22:46–64 better management of periodontal disease delay the
[erratum 2009;22:386]. onset and progression of Alzheimer’s disease? J Alzhei-
129. Tomás I, Diz P, Tobías A, Scully C, Donos N. Periodontal mers Dis 2017;58:337–348.
health status and bacteraemia from daily oral activities: 145. Tzeng NS, Chung CH, Yeh CB, et al. Are chronic peri-
Systematic review/meta-analysis. J Clin Periodontol odontitis and gingivitis associated with dementia? A
2012;39:213–228. nationwide, retrospective, matched-cohort study in
130. Zhang W, Daly CG, Mitchell D, Curtis B. Incidence and Taiwan. Neuroepidemiology 2016;47:82–93.
magnitude of bacteraemia caused by flossing and by
scaling and root planing. J Clin Periodontol 2013; 40:
41–52.

82
References

146. Noble JM, Scarmeas N, Celenti RS, et al. Serum IgG 161. Hsu L, Power D, Upritchard J, et al. Amplification of oral
antibody levels to periodontal microbiota are associated streptococcal DNA from human incisors and bite marks.
with incident Alzheimer disease. PLoS One 2014; 9: Curr Microbiol 2012;65:207–211.
e114959. 162. Kennedy DM, Stanton JA, Garcia JA, et al. Microbial
147. Kamer AR, Pirraglia E, Tsui W, et al. Periodontal disease analysis of bite marks by sequence comparison of strep-
associates with higher brain amyloid load in normal tococcal DNA. PLoS One 2012;7:e51757.
elderly. Neurobiol Aging 2015;36:627–633. 163. Hesselmar B, Sjöberg F, Saalman R, Aberg N, Adleberth
148. Bartold PM, Mariotti A. The future of periodontal-systemic I, Wold AE. Pacifier cleaning practices and risk of allergy
associations: Raising the standards. Curr Oral Health development. Pediatrics 2013;131:e1829–e1837.
Rep 2017;4:258–262. 164. Tomas ME, Kundrapu S, Thota P, et al. Contamination
149. Carter CJ, France J, Crean S, Singhrao SK. The Porphy­ of health care personnel during removal of personal
romonas gingivalis/host interactome shows enrichment protective equipment. JAMA Intern Med 2015; 175:
in GWASdb genes related to Alzheimer’s disease, diabe- 1904–1910.
tes and cardiovascular diseases. Front Aging Neurosci 165. McLean JS, Lombardo MJ, Ziegler MG, et al. Genome
2017;9:408. of the pathogen Porphyromonas gingivalis recovered
150. Astafurov K, Elhawy E, Ren L, et al. Oral microbiome from a biofilm in a hospital sink using a high-throughput
link to neurodegeneration in glaucoma. PLoS One 2014; single-cell genomics platform. Genome Res 2013;23:
9:e104416. 867–877.
151. Akcali A, Güven Yilmaz S, Lappin DF, Eğrilmez S, 166. Warren HS, Fitting C, Hoff E, et al. Resilience to bacterial
Buduneli N. Effect of gingival inflammation on the inflam- infection: Difference between species could be due to
matory response in patients with idiopathic uveitis. J proteins in serum. J Infect Dis 2010;201:223–232.
Clin Periodontol 2016;43:637–645. 167. Han YW, Wang X. Mobile microbiome: Oral bacteria in
152. Chu DM, Ma J, Prince AL, Antony KM, Seferovic MD, extra-oral infections and inflammation. J Dent Res 2013;
Aagaard KM. Maturation of the infant microbiome 92:485–491.
community structure and function across multiple body 168. Koren O, Spor A, Felin J, et al. Human oral, gut, and
sites and in relation to mode of delivery. Nat Med 2017; plaque microbiota in patients with atherosclerosis. Proc
23:314–326. Natl Acad Sci U S A 2011;108(suppl 1):4592–4598.
153. Ward TL, Knights D, Gale CA. Infant fungal communities: 169. Haraszthy VI, Zambon JJ, Trevisan M, Zeid M, Genco
Current knowledge and research opportunities. BMC RJ. Identification of periodontal pathogens in athero-
Med 2017;15:30. matous plaques. J Periodontol 2000;71:1554–1560.
154. Asikainen S, Chen C, Slots J. Likelihood of transmitting 170. Mahalakshmi K, Krishnan P, Krishna Baba MG, et al.
Actinobacillus actinomycetemcomitans and Porphyro­ “Association of periodontopathic anaerobic bacterial
monas gingivalis in families with periodontitis. Oral co-occurrence to atherosclerosis” — A cross-sectional
Microbiol Immunol 1996;11:387–394. study. Anaerobe 2017;44:66–72.
155. Cephas KD, Kim J, Mathai RA, et al. Comparative anal- 171. Chhibber-Goel J, Singhal V, Bhowmik D, et al. Linkages
ysis of salivary bacterial microbiome diversity in between oral commensal bacteria and atherosclerotic
edentulous infants and their mothers or primary care plaques in coronary artery disease patients. NPJ Biofilms
givers using pyrosequencing. PLoS One 2011; 6: Microbiomes 2016;2:7.
e23503. 172. Pessi T, Karhunen V, Karjalainen PP, et al. Bacterial
156. Prince AL, Ma J, Kannan PS, et al. The placental signatures in thrombus aspirates of patients with
membrane microbiome is altered among subjects with myocardial infarction. Circulation 2013; 127:
spontaneous preterm birth with and without chorioam- 1219–1228.
nionitis. Am J Obstet Gynecol 2016;214:627.e621–627. 173. Allareddy V, Prakasam S, Rampa S, et al. Impact of
e616. periapical abscess on infectious complications in
157. Prince AL, Chu DM, Seferovic MD, Antony KM, Ma J, patients undergoing extracorporeal circulation auxiliary
Aagaard KM. The perinatal microbiome and pregnancy: to open-heart surgical procedures. J Evid Based Dent
Moving beyond the vaginal microbiome. Cold Spring Pract 2017;17:13–22.
Harb Perspect Med 2015;5:a023051. 174. Wiener RC, Sambamoorthi U, Jurevic RJ. Association
158. Vanterpool SF, Been JV, Houben ML, et al. Porphyro­ of periodontitis and human papillomavirus in oral rinse
monas gingivalis within placental villous mesenchyme specimens: Results from the National Health and Nutri-
and umbilical cord stroma is associated with adverse tion Survey 2009–2012. J Am Dent Assoc 2015; 146:
pregnancy outcome. PLoS One 2016;11:e0146157. 382–389.
159. Zabor EC, Klebanoff M, Yu K, et al. Association between 175. Dayakar MM, Shipilova A, Gupta D. Periodontal pocket
periodontal disease, bacterial vaginosis, and sexual risk as a potential reservoir of high risk human papilloma
behaviours. J Clin Periodontol 2010;37:888–893. virus: A pilot study. J Indian Soc Periodontol 2016;20:
160. Pelzer E, Gomez-Arango LF, Barrett HL, Nitert MD. 136–140.
Review: Maternal health and the placental microbiome. 176. Centers for Disease Control and Prevention. Human
Placenta 2017;54:30–37. papillomavirus (HPV). http: //www.cdc.gov/hpv/.
Accessed 25 March 2018.

83
3 The Traveling Oral Microbiome

177. Vergori A, Garbuglia AR, Piselli P, et al. Oral human 191. Ahn J, Peng S, Hung CF, Roden RBS, Best SR. Prophy-
papillomavirus DNA detection in HIV-positive men: Prev- lactic immunization with human papillomavirus vaccines
alence, predictors, and co-occurrence at anal site. BMC induces oral immunity in mice. Laryngoscope 2018;128:
Infect Dis 2018;18:25. E16–E20.
178. Ylitalo N, Josefsson A, Melbye M, et al. A prospective 192. Genner J, Scheutz F, Ebbesen P, Melbye M. Antibody
study showing long-term infection with human papillo- to human papilloma virus in Danish dentists. Scand J
mavirus 16 before the development of cervical Dent Res 1988;96:118–120.
carcinoma in situ. Cancer Res 2000;60:6027–6032. 193. Vazquez-Otero C, Vamos CA, Thompson EL, et al.
179. Forman D, de Martel C, Lacey CJ, et al. Global burden Assessing dentists’ human papillomavirus-related health
of human papillomavirus and related diseases. Vaccine literacy for oropharyngeal cancer prevention. J Am Dent
2012;30(suppl 5):F12–23. Assoc 2018;149:9–17.
180. Kulkarni SS, Kulkarni SS, Vastrad PP, et al. Prevalence 194. Daley E, Dodd V, DeBate R, et al. Prevention of
and distribution of high risk human papillomavirus (HPV) HPV-related oral cancer: Assessing dentists’ readiness.
types 16 and 18 in carcinoma of cervix, saliva of patients Public Health 2014;128:231–238.
with oral squamous cell carcinoma and in the general 195. McQuillan G, Kruszon-Moran D, Flagg EW, Paulose-Ram
population in Karnataka, India. Asian Pac J Cancer Prev R. Prevalence of herpes simplex virus type 1 and type
2011;12:645–648. 2 in persons aged 14–49: United States, 2015–2016.
181. Kreimer AR, Villa A, Nyitray AG, et al. The epidemiology NCHS Data Brief 2018;(304):1–8.
of oral hpv infection among a multinational sample of 196. Cruz I, Van den Brule AJ, Steenbergen RD, et al. Prev-
healthy men. Cancer Epidemiol Biomarkers Prev 2011; alence of Epstein-Barr virus in oral squamous cell
20:172–182. carcinomas, premalignant lesions and normal muco-
182. Kreimer AR, Pierce Campbell CM, Lin HY, et al. Inci- sa—A study using the polymerase chain reaction. Oral
dence and clearance of oral human papillomavirus Oncol 1997;33:182–188.
infection in men: The HIM cohort study. Lancet 2013; 197. Marshall BJ, Warren JR. Unidentified curved bacilli in
382:877–887. the stomach of patients with gastritis and peptic ulcer-
183. Zhang C, Liu F, Pan Y, et al. Incidence and clearance of ation. Lancet 1984;1:1311–1315.
oral human papillomavirus infection: A population-based 198. Silva DG, Tinoco EM, Rocha GA, et al. Helicobacter
cohort study in rural China. Oncotarget 2017; 8: pylori transiently in the mouth may participate in the
59831–59844. transmission of infection. Mem Inst Oswaldo Cruz 2010;
184. Visalli G, Curro M, Facciola A, et al. Prevalence of human 105:657–660.
papillomavirus in saliva of women with HPV genital 199. Zou QH, Li RQ. Helicobacter pylori in the oral cavity and
lesions. Infect Agent Cancer 2016;11:48. gastric mucosa: A meta-analysis. J Oral Pathol Med
185. Wood ZC, Bain CJ, Smith DD, Whiteman DC, Antons- 2011;40:317–324.
son A. Oral human papillomavirus infection incidence 200. Hirsch C, Tegtmeyer N, Rohde M, Rowland M,
and clearance: A systematic review of the literature. J Oyarzabal O, Backert S. Live Helicobacter pylori in the
Gen Virol 2017;98:519–526. root canal of endodontic-infected deciduous teeth. J
186. Gillison ML, Broutian T, Pickard RK, et al. Prevalence of Gastroenterol 2012;47:936–940.
oral HPV infection in the United States, 2009–2010. 201. Al Sayed A, Anand PS, Kamath KP, Patil S, Preethanath
JAMA 2012;307:693–703. RS, Anil S. Oral cavity as an extragastric reservoir of
187. Sanders AE, Slade GD, Patton LL. National prevalence Helicobacter pylori. ISRN Gastroenterol 2014; 2014:
of oral HPV infection and related risk factors in the U.S. 261369.
adult population. Oral Dis 2012;18:430–441. 202. Loster BW, Czesnikiewicz-Guzik M, Bielanski W, et al.
188. McQuillan G, Kruszon-Moran D, Markowitz LE, Unger Prevalence and characterization of Helicobacter pylori
ER, Paulose-Ram R. Prevalence of HPV in adults aged (H. pylori) infection and colonization in dentists. J Physiol
18–69: United States, 2011–2014. NCHS Data Brief Pharmacol 2009;60(suppl 8):13–18.
2017;(280):1–8. 203. Matsuda R, Morizane T. Helicobacter pylori infection in
189. Meites E, Kempe A, Markowitz LE. Use of a 2-dose dental professionals: A 6-year prospective study. Heli-
schedule for human papillomavirus vaccination— cobacter 2005;10:307–311.
Updated recommendations of the Advisory Committee 204. Cooper PR, Palmer LJ, Chapple IL. Neutrophil extra-
on Immunization Practices. MMWR Morb Mortal Wkly cellular traps as a new paradigm in innate immunity:
Rep 2016;65:1405–1408. Friend or foe? Periodontol 2000 2013;63:165–197.
190. Handisurya A, Schellenbacher C, Haitel A, Senger T, 205. Ben Lagha A, Dudonné S, Desjardins Y, Grenier D. Wild
Kimbauer R. Human papillomavirus vaccination induces blueberry (Vaccinium angustifolium Ait.) polyphenols
neutralising antibodies in oral mucosal fluids. Br J target Fusobacterium nucleatum and the host inflam-
Cancer 2016;114:409–416. matory response: Potential innovative molecules for
treating periodontal diseases. J Agric Food Chem 2015;
63:6999–7008.

84
References

206. Ben Lagha A, LeBel G, Grenier D. Dual action of high- 208. Petrosino JF. The microbiome in precision medicine:
bush blueberry proanthocyanidins on Aggregatibacter The way forward. Genome Med 2018;10:12
actinomycetemcomitans and the host inflammatory 209. Giannobile WV, Kornman KS, Williams RC. Personalized
response. BMC Complement Altern Med 2018;18:10. medicine enters dentistry: What might this mean for
207. Lloyd-Price J, Mahurkar A, Rahnavard G, et al. Strains, clinical practice? J Am Dent Assoc 2013;144:874–876.
functions and dynamics in the expanded Human Micro- 210. Razzouk S, Termechi O. Host genome, epigenome, and
biome Project. Nature 2017;550:61–66. oral microbiome interactions: Toward personalized peri-
odontal therapy. J Periodontol 2013;84:1266–1271.

85
CHAPTER 4

The Role of Inflammation in


Oral-Systemic Interactions
Marcelo Freire, dds, phd, dmsc
Thomas E. Van Dyke, dds, phd

A localized inflammatory response to an injury distinct in etiology and mechanisms but have
or infection is a spatially defined and tempo- in common a programmed immune response
rally regulated condition that is self-limited. that modulates disease progression, prognosis,
If the lesion does not resolve and becomes and treatment. Periodontal tissues in particular
chronic, the acquired immune system is have an anatomical and multitissue origin, are
stimulated, including broad activation of hypervascularized, and have a unique interface
lymphocytic pathways and cell-mediated and with commensal microorganisms, maintaining
humoral immunity. The inflammatory and a controlled homeostatic response in health.
immune system can be a contributing factor Infectious and inflammatory diseases of the
in the pathology of many chronic diseases, oral cavity, such as periodontitis, result from an
including cardiovascular diseases (CVDs), imbalance of the commensal-host relationship
type 2 diabetes, rheumatoid arthritis, and that results in or may be caused by excessive
periodontal diseases. Local persistent inflam- inflammation and overgrowth of commensal
matory lesions lead to systemic dysregulation pathogens. It is becoming clear that chronic
of homeostasis, an extended response that goes oral infections and inflammation have a sys-
beyond confined localization to one tissue. The temic impact. It is relevant to the fields of
chronicity of the lesion alters molecular, cel- biology, medicine, and dentistry to understand
lular, and overall tissue responses in remote the mechanisms of the intricate molecular
regions of the body, having a transient or per- interactions that underlie the relationship
manent effect on overall health. The ability of between oral conditions that arise locally and
scientists to better understand the underlying their long-term systemic consequences.
pathology and to identify groups of patients Historically, the first descriptions of localized
susceptible to inflammatory disease through tissue response to injury and infection were
precision medicine could potentially lead to recorded by the ancient Egyptian and Greek
new, innovative treatments. cultures. Hippocrates, in the 5th century bce,
Oral conditions can significantly influence introduced terms such as edema that are still
or be influenced by local and systemic events. used to describe the local response—inflam-
Periodontal diseases, periodontic-endodontic mation.1 He referred to inflammation as an
lesions, traumatic lesions, reactionary oral early component of the healing process after
lesions (chronic irritation), premalignant tissue injury. The invention of the compound
lesions, oral cancer, and other conditions are microscope by Janssen in the 1500s and the

86
The Role of Inflammation in Oral-Systemic Interactions

subsequent improvement of its optical reso- that this would prevent or cure a plethora of
lution by Leeuwenhoek gave rise to the early local or systemic problems.3
descriptions of the tissue response to injury. The lack of scientific foundation for the
The initial concepts related to inflammation concept of focal infection eventually led to its
came from intuition (and clinical observations) rejection because there was no clear basis for
rather than careful scientific investigation. The ascribing most systemic diseases to bad teeth
controversial observations described by ancient and gums. The practice of dentistry changed,
cultures provided the framework for critical and practitioners and the community redirected
experimentation in later centuries, when it their focus to restorative procedures. However,
became clear that it was necessary to elucidate as a more scientific approach was applied to
the relationship between local responses and investigating clinical problems, it became clear
systemic health. that there are in fact situations in which oral
Still, until the middle of the 20th century, bacteria can affect distant structures, partic-
scientific and medical theory was based on ularly in the case of bacterial endocarditis in
observation and intuition rather than exper- susceptible people.
imentation and application of the scientific In the late 1980s, research evidence
method. One example is the concept that focal demonstrated a direct association between
infection or injury is the basis of systemic dis- periodontitis and systemic conditions, includ-
ease. The focal infection theory, introduced in ing coronary heart disease (CHD), stroke,
1891 by Miller, proposed a relationship between and the association of preterm birth and low–
oral and systemic infection.2 A prevailing theory birth weight babies in mothers with severe
of medicine around this time was that miasmas, periodontitis. However, the response of the
or bad smells, caused disease, in spite of Koch’s dental and medical professions this time was
discovery of the infectious nature of disease. considerably more measured than in the early
Miller emphasized that oral microorganisms part of the 20th century. There was no sugges-
or their products were the etiologic factors of a tion that elimination of oral infection would
variety of diseases in sites far removed from the cure/improve cardiovascular or other systemic
oral cavity, including arthritis, brain abscesses, inflammatory diseases. The relationship of oral
pulmonary diseases, cardiovascular and gastric disease to systemic disease was thought of in
problems, and even stupidity. terms of modifiable risk factors.
The role of oral sepsis as a cause of systemic In recent years, substantial additions to the
disease was described by William Hunter, a scientific literature recognize that inflammation
prominent British physician, who emphasized is a hallmark response to bacteria or bacte-
that dental restorations “built in, on, and remia that, as an evolutionarily programmed
around diseased teeth which form a veritable reaction to injury and infection, is a significant
mausoleum of gold over a mass of sepsis to link between oral and systemic diseases.4–6 The
which there is no parallel in the whole realm investigation of oral-systemic disease connec-
of medicine, and it is unique to oral tissues.”3 tions is a rapidly advancing area of research.
In 1919, Rosenow published a series of animal Considering the existing data, it is essential
experiments and human case reports support- to reflect on all the plausible relationships.
ing the concept of focal infection. He suggested Oral and systemic conditions may coexist, but
cooperation between dentists and physicians to they may be disorders of different origin that
ensure that the focus of oral infection would affect the outcomes of one another. Various
be eliminated completely. It became com- infectious or inflammatory conditions of the
mon practice to extract all endodontically or oral cavity, including periodontal diseases, are
periodontally involved teeth to eliminate any directly or indirectly associated with systemic
possible foci of infection, with the expectation diseases, increase the risk of development of

87
4 The Role of Inflammation in Oral-Systemic Interactions

other diseases, or vice versa. The relationship of the response. Regulation of transcription
might also be causal. This chapter examines the factors, genes, their translation, and secretion
mechanisms linking oral and systemic diseases, of proinflammatory cytokines from a variety
emphasizing unresolved inflammation as the of cells is generally dependent on nuclear fac-
key to pathogenesis. tor κB (NFκB) nuclear protein activation of
transcription. The NFκB-regulated pathways
are activated by pattern-recognition receptors
such as toll-like receptors (TLRs) that bind
Inflammation bacterial lipopolysaccharide (LPS) and other
molecules. Both inflammatory cytokines and
Innate immunity, characterized by the local chemoattractant cytokines, or chemo­­kines,
inflammatory response, is the early host are produced for autocrine, endocrine, and
response. Its purpose is to identify and elim- paracrine functions. 4,8 The cytokines are
inate infectious agents or damaged tissues. low–molecular weight proteins that modu-
As an initial and protective response to chal- late inflammation positively or negatively.
lenges presented to host tissues, inflammation Cytokines produced by resident cells, such
is characterized by vascular dilation, enhanced as epithelial cells and fibroblasts, as well as
permeability of capillaries, increased blood phagocytes in the acute and early chronic
flow, and leukocyte recruitment. Together, phases of inflammation modulate immune
these result in the five cardinal signs of inflam- cells in adaptive immunity.
mation: heat, redness, swelling, pain, and After microbial recognition, cytokines of the
loss of function. Cells control the magnitude innate response, including tumor necrosis fac-
of the inflammatory response. Polymorpho- tor α (TNF-α), interleukin (IL) 1β, and IL-6,
nuclear leukocytes (PMNs) or neutrophils are the first secreted and are collectively known
(named for their staining characteristics as the signature innate cytokines. TNF-α is a
with hematoxylin-eosin) are the first line of pleiotropic cytokine that has many func-
defense. PMNs are professional phagocytes tions, from signaling cell migration to tissue
with potent oxidative and nonoxidative kill- destruction. TNF-α upregulates the production
ing mechanisms to combat bacteria. After of IL-1β and IL-6. TNF-α is also correlated
PMN infiltration, mononuclear cells (mono- with extracellular matrix degradation and
nuclear phagocytes and macrophages) enter bone resorption through actions promoting
the site and clear cellular debris, bacteria, and the secretion of matrix metalloproteinases
apoptotic PMNs by phagocytosis without (MMPs) and receptor activator of NFκB ligand
prolonging inflammation. The degree of inflam- (RANKL) and coupled bone formation.9
mation activation is dependent on the nature Chemokines are cytokines with chemoattrac-
of the inflammatory trigger, the response, its tant functions that induce cell migration to the
localization, and its persistence.7 site of infection or injury. Once blood leuko-
The major sensors for the inflammatory cytes exit a blood vessel, they are attracted by
response are innate immune cells that migrate functional gradients of chemotactic factors.
to the injured site and resident stromal cells. Multiple cell types, including leukocytes and
Together, they trigger production of media- endothelial, epithelial, and stromal cells, pro-
tors that modulate the fate of inflammation. duce chemokines. Beyond their chemotactic
Neutrophils, macrophages, dendritic cells, role, chemokines function as messengers of
and mast cells produce low–molecular weight distinct biologic processes, including cell pro-
proteins called cytokines that control the ini- liferation, cell death, angiogenesis, and tumor
tiation of inflammation, the maintenance and metastasis. Bacterial peptides are also chemo-
regulation of its amplitude, and the duration tactic for inflammatory cells.10

88
Inflammation

The innate immune recognition of dif- leukotrienes. Excessive production of inflam-


ferent microorganisms is complex. Viral matory mediators and an exacerbated sensing
infections induce the production of inter- response to inflammatory triggers positively
ferons (IFN-α, IFN-β) and the activation of correlate with progression from acute inflamma-
cytotoxic lymphocytes (natural killer [NK] tion to chronic inflammation in many diseases.
cells); bacterial pathogens are recognized by Microorganisms that gain access to the
pattern-recognition receptors such as TLRs, blood circulation are usually eliminated by
which are expressed by tissue-resident macro- the reticuloendothelial system within min-
phages and other host cells. Binding of TLRs utes (transient bacteremia) with no clinical
induces the production of inflammatory cyto- symptoms. Local bacterial antigens that are
kines, chemokines, and proinflammatory lipid systemically dispersed trigger significant sys-
mediators such as prostaglandins. These inflam- temic inflammation. Leukocytes, endothelial
matory mediators are the major players in cells, and hepatocytes respond to bacteria and
establishing an effective inflammatory response their virulence factors with the secretion of
and clearance of bacteria. Proinflammatory proinflammatory immune mediators called
mediators, such as IL-1β, IL-6, TNF-α, and acute-phase proteins, such as C-reactive protein
prostaglandin E2 (PGE2) are produced locally (CRP), which is secreted by the liver. Persistence
in inflamed tissues. Proinflammatory cytokines of infection, with continuous exposure of the
in circulation induce systemic actions, includ- host to new antigens, results in the activation of
ing leukocytosis and acute-phase proteins in humoral immunity and antibody production.
the liver. On the other hand, parasites lead to Circulating specific antibodies form immune
the production of a different set of cytokines complexes with respective target antigens,
and cellular responses. These include IL-4, IL-5, which in turn activate the complement system
and IL-13 by mast cells and basophils. With and Fc receptors on phagocytes to amplify
continued exposure, soluble antigens react by inflammation at sites of deposition. Likewise,
circulating a specific antibody to form immune proinflammatory mediators, such as IL-1β,
complexes that further amplify inflammation IL-6, TNF-α, and PGE2, produced locally in the
at sites of deposition.5 inflamed tissues, may spill into the circulation
Locally produced lipid mediators, such as and have a systemic impact, such as induction
prostaglandins and leukotrienes, are proinflam- of endothelial dysfunction.9,11
matory signals that trigger the nervous system Although the inflammatory response is pro-
to promote fever and fatigue.10 Prostaglandins tective, failure to remove noxious materials
and leukotrienes are derived from membrane produced by neutrophils via phagocytosis,
lipids, particularly arachidonic acid. Two major failure of clearance of apoptotic inflamma-
enzyme pathways, cyclooxygenases (COXs) tory cells, or delay of apoptosis characterize
and lipoxygenases (LOs), metabolize arachi- the chronic and pathologic lesion. The incom-
donic acid. COX-1 (constitutively expressed plete elimination of leukocytes from a lesion
COX) and COX-2 (inducible COX) catalyze is observed in susceptible individuals; acute
the conversion of arachidonic acid into pros- inflammation fails to resolve, and chronic
taglandins, prostacyclins, and thromboxanes. disease and fibrosis develop. Accordingly, loss
Prostaglandins have 10 subclasses, of which of resolution and failure to return tissue to
D, E, F, G, H, and I are the most important in homeostasis results in neutrophil-mediated
inflammation. destruction and chronic inflammation with
Three LOs are important in inflammation: 5-, accumulation of proinflammatory macro-
12-, and 15-LO. LOs catalyze the formation of phages, which is directly related to human
hydroxyeicosatetraenoic acids (HETEs) from inflammatory pathologies, cancers, type 2 dia-
arachidonic acid, leading to the formation of betes, CVDs, and periodontal diseases.

89
4 The Role of Inflammation in Oral-Systemic Interactions

The natural resolution of inflammation after overgrowth of pathologic organisms, initiates


an acute inflammatory response results from chronic inflammation, which is followed by an
well-coordinated biochemical pathways that acquired immune response and tissue destruc-
are activated by a class switch of enzymes. LO tion, or periodontitis.
pathways active in the acute phase eventually Failure to remove both the trigger (patho-
result in the action of two different LOs on genic bacteria) and inflammatory cells,
the same substrate arachidonic acid molecule. mainly neutrophils, characterizes the chronic,
The double substitution leads to the forma- pathologic lesion. The rapid and complete
tion of lipoxins, a new series of eicosanoids elimination of leukocytes from an acute inflam-
that share the same backbone structure of the matory lesion is the ideal outcome following
proinflammatory leukotrienes and HETEs insult. In susceptible individuals, periodontal
but bind to distinct receptors. The active, inflammation fails to resolve, and chronic
receptor-mediated regulation halts neutrophil inflammation becomes periodontal pathology.
function, promotes neutrophil apoptosis, and Accordingly, reduced resolution and failure to
promotes the nonphlogistic (noninflammatory) return the tissues to homeostasis results ini-
accumulation of macrophages that efficiently tially in neutrophil-mediated destruction and
clear the lesion of remaining bacteria and apop- chronic inflammation followed by a complex
totic cells. immune lesion with destruction of the extra-
There are a number of resolution pathways, cellular matrix and bone, scarring, and the loss
depending on the fatty acid substrate. Omega-3 of periodontal tissue function.12,13
fatty acids in the diet are metabolized by the The presence of bacterial plaque and gin-
same enzyme pathways as arachidonic acid to givitis is prevalent in humans, affecting more
produce molecules called resolvins of the D and than 90% of the adult dentate population. The
E series (from docosahexaenoic acid and eicos- same direct correlation cannot be said for peri-
apentaenoic acid, respectively), protectins, and odontitis; despite abundant plaque deposits in
maresins. All of these factors drive resolution most people, the prevalence of moderate peri-
of inflammation through different receptors odontitis (attachment loss greater than 5 mm)
on inflammatory cells. Chronic inflammation is around 40% of the population.14 However,
results from a combination of excessive pro- this prevalence of periodontitis is not uniformly
duction of proinflammatory mediators and distributed across races, ethnicities, and socio-
a failure to produce sufficient proresolution economic groups. This being the case, despite
mediators (for review, see Serhan et al6). the universal presence of plaque, bacteria do
not appear to be the primary determinants of
the progression of gingivitis to periodontitis.
Although the popular dogma has been to accept
Periodontal Diseases that periodontitis arises from a specific subgin-
gival infection, the concept that periodontitis
The local acute periodontal inflammatory occurs when the periodontal tissues provide an
response is a reaction to the local micro- adequate ecologic environment for opportunis-
bial biofilm. The commensal flora and host tic bacteria to flourish has been presented, for
inflammatory response evolved to maintain some time, by eminent oral microbiologists as
a relationship that is symbiotic and healthy, an alternative mechanism.15–17
as observed in periodontal tissues and other The host-parasite interaction is responsi-
mucosal surfaces, including the oropharynx ble for the initiation of the gingivitis lesion,
and the gut. Disturbances of homeostasis, but what happens next is less clear. There is
characterized by increased inflammation and no definitive evidence that specific bacteria

90
Periodontal Inflammation in Oral Cancer

are responsible for the progression and mani- status in patients with oropharyngeal cancer
festation of advanced periodontitis; however, has been reported.23
specific pathogens are associated with deep The risk of cancer increases with periodon-
periodontal pockets. It can be argued that the titis, and the risk of death is higher when
specific bacteria are present as a result of the periodontitis is present. In a follow-up of sub-
disease but do not cause the disease. This is jects from NHANES I,20,24 patients diagnosed
no different than most mucosal biofilm–associ- with periodontitis had a 55% (95% confidence
ated pathologies where the complicating issue, interval [CI]: 25%–92%) increase in risk of
which still remains unresolved, is which comes death from any cancer. There was a signifi-
first—the host response or the change in the cantly increased risk for lung cancer; however,
biofilm?18 this was not evident in individuals who had
Several biologic pathways linking peri- never smoked.
odontal disease to induction of systemic The incidence of cancer consistently increased
inflammation have been identified. 17–19 In in patients with periodontitis. A larger study
health, the gingival sulcular epithelium and with a 17.7-year follow-up identified 5,720
local innate immune cells act as a natural incident cancers in 48,375 men enrolled. Men
barrier preventing bacterial penetration. In gin- who reported periodontal disease had a slightly
gival health, only a small number of bacteria, increased total cancer incidence of 14% (95%
mostly facultative anaerobes, are found in the CI: 7%–22%), which remained significant
gingival crevice and bloodstream. However, in when the analysis was limited to those who had
periodontal disease, inflamed tissue and ulcer- never been smokers.25 There was a significantly
ated subgingival pocket epithelium is ulcerated increased risk of lung, kidney, pancreatic, and
and permeable, providing bacteria a port of hematologic cancers after adjustment. The risk
entry to the circulation. for lung cancer was the only association when
the number of teeth was used as the exposure.25
Similar genetic risk factors for periodontal
Periodontal Inflammation in disease and cancers were found in a prospec-
tive study in Sweden. The study identified more
Oral Cancer than 4,000 incident cancers after a median
follow-up of 27 years.26 At baseline, the par-
A higher incidence of cancer development is ticipants were classified as having periodontal
found in individuals with chronic inflammatory disease if they reported that at least half their
conditions; this insight has driven suggestions teeth had loosened or fallen out on their own.
of a possible link with periodontal inflamma- Periodontal disease was associated with a 15%
tion.20,21 Associations between periodontal (95% CI: 1%–32%) increased risk for all can-
disease and the risk for precancerous lesions cers. Periodontal infection was also associated
relate to chronic inflammation. A possible link with increased risk of colorectal, pancreatic,
between periodontal disease and oral neoplasm and prostate cancers.
has been described. In fact, recent evidence In twin analyses, monozygotic twins with
from the National Health and Nutrition baseline periodontal disease showed a 50%
Examination Survey (NHANES) III indicated increase in total cancer risk, but in dizygotic
that periodontitis was significantly related to twins this association was markedly attenu-
tumors.22 The stratified analysis found that ated. Similar patterns emerged for digestive
this association was only present in current tract cancers, suggesting that shared genetic
smokers. Also, a stronger relationship between risk factors may partially explain associations
periodontitis and human papillomavirus (HPV) between periodontal disease and cancers.20,26

91
4 The Role of Inflammation in Oral-Systemic Interactions

Proresolution signals Proinflammatory signals

TGF-β TNF-α
IL1-Ra IL-1

Chronic inflammation
IL-4 IL-6
Health

IL-10 CCL2
GM-CSF CXCL8
Local tissue
Immunoresolvents: CRP
inflammation
Resolvins Histamine
Lipoxins Prostaglandins
Protectins
Maresins

Fig 4-1  |  Tissue-inflammatory processes link to systemic homeostasis. The nature of challenge and balance
between the response from sensors and mediators leads to distinct fates in acute inflammation. Proinflamma-
tory mediators such as IL-1β, TNF-α, and others listed are important molecules that initiate the tissue response
to injury and infection. The termination of acute processes is modulated by immunoresolvents and consequent
upregulation of anti-inflammatory cytokines. An unresolved inflammatory process is the primary step leading
to development of chronic inflammation and loss of local and systemic function. CCL2, chemokine (C-C motif)
ligand 2; CXCL8, interleukin 8; TGF-β, transforming growth factor β; IL1-Ra, interleukin 1 receptor antagonist;
GM-CSF, granulocyte-macrophage colony-stimulating factor.

Link Between Periodontal tissue destruction and osteoclast-mediated bone


loss. The etiology of periodontitis is recognized
Disease and Systemic to be primarily bacterial but multifactorial, and
Conditions some behavioral, environmental, systemic, and
genetic risk factors influence host susceptibil-
ity and disease progression. The impact of a
Uncontrolled (chronic) inflammation is a hall- dysregulated inflammatory response has the
mark of various human conditions, including potential to harm the systemic health of indi-
periodontitis, diabetes, CVDs, and premature viduals (Fig 4-1).
labor.23–25 Despite advances in knowledge of
causes and risk factors associated with chronic
periodontitis, there are no signs of decline in Periodontitis and diabetes
prevalence. According to the US Centers for Dis- Among many diseases that impact the popula-
ease Control and Prevention (CDC), 647 million tion globally, type 2 diabetes is a major public
adults worldwide had periodontal diseases as health problem. Worldwide, about 347 million
of 2012.14 Periodontal diseases, including gin- adults suffer from type 2 diabetes.28 According
givitis and periodontitis, are leukocyte-driven to the World Health Organization (WHO), the
inflammatory diseases characterized by soft prevalence may double by the year 2030, and

92
Link Between Periodontal Disease and Systemic Conditions

health care expenditures related to diabetes will disease, type 2 diabetes, and atherosclerosis.
increase significantly.29 Evidence suggests that inflammatory cells may
Inflammatory diseases such as type 2 diabetes be found within adipose tissue; perhaps the
and chronic periodontal diseases have a recipro- fat itself is actively involved in inflammatory
cal relationship. Among people diagnosed with processes, contributing to the release of proin-
type 2 diabetes, one-third present with severe flammatory mediators.
periodontal disease, and adult diabetic patients Fat tissue adipokines have many actions in
with severe periodontitis have a higher risk of sites away from their origin, including the liver,
poor glycemic control, increasing the risk of blood vessel walls, and muscles. They regulate
oral and systemic complications.28 Hyperglyce- metabolic functions, including glucose lev-
mia resulting from type 2 diabetes can impact els, insulin, and lipid metabolism. Although
PMNs and monocytes through various mecha- adipose tissue may release a host of proin-
nisms, including the accumulation of advanced flammatory cytokines, adiponectin is of interest
glycation end products (AGEs) that leads to an because it seems to be anti-inflammatory and
increase in extracellular production of super- therefore protective. For example, studies have
oxide by leukocytes and increased secretion shown that lower levels of plasma adiponectin
of proinflammatory cytokines, such as IL-1β, are found in patients with both diabetes and
insulin-like growth factor, TNF-α, and MMPs.30 CHD than in patients with diabetes without
Increased inflammation in periodontitis leads to CHD, suggesting that adiponectin may be anti-
prolonged activation of PMNs and monocytes atherogenic.18,31,33 Humans in insulin-resistant
and results in ineffective clearance of bacte- states also have lower levels of adiponectin; this
ria and prolonged inflammation, aggravating was reversed by the administration of thiazoli-
systemic diabetes regulation. Systemic inflam- dinedione, an insulin-sensitizing compound.30,31
mation in type 2 diabetes, defined by increased There also seems to be a negative relationship
circulating TNF-α, is associated with obesity between adiponectin and CRP, again suggesting
and periodontitis and has been proposed as a that adiponectin has an anti-inflammatory role.
mechanism for the connection between these Diabetic patients have a higher risk for
conditions. A case-control study demonstrated developing chronic periodontitis, and those
that periodontitis is associated with elevated with elevated hemoglobin A1c (HbA1c) levels
plasma triglycerides and total cholesterol.31 have a significantly higher prevalence of peri-
A relationship between inflammatory path- odontitis and experience more tooth loss than
ways and metabolic diseases, such as type 2 do those with better metabolic control. Acute
diabetes or insulin resistance, is one line of and chronic infections may adversely influence
active investigation.32 Attempts to understand glycemic control.28 Thus, the concept of HbA1c
the molecular pathways that regulate diabe- being adversely affected by systemic inflam-
tes and insulin resistance require the study of mation is well accepted by medical and dental
adipose tissue. For example, adipose tissue is communities.
now recognized as a biologically active endo- It is now clear that a biologically plausible
crine organ and not simply a site of inert lipid link between periodontitis and metabolic con-
storage. Adipocytes, like any stromal cells, trol has been determined. If adequate treatment
secrete cytokines, now collectively known of periodontitis can modify glycemic control,
as adipokines, into the circulation, including as suggested in some studies,28 periodontal
adiponectin, TNF-α, plasminogen activator therapy may be an essential contribution to
inhibitor type 1, and resistin.33 The realization a patient-management program that incorpo-
that adipocytes have a molecular influence on rates lifestyle changes and medications.
the entire system helps explain why obesity Periodontal therapy has shown uniform
is frequently associated with inflammatory outcomes across studies. While periodontal

93
4 The Role of Inflammation in Oral-Systemic Interactions

conditions have shown consistent results when acts as a stimulant of acute-phase reactants,
treated, the effects of the therapy in diabetic including CRP, serum amyloid A, and fibrino-
patients are not consistent. The standard treat- gen, especially in the liver.
ment outcome for diabetic patients has been Although cytokines at all steps have import-
reductions in HbA1c, because reducing HbA1c ant biologic actions, their amplification at each
has been shown to delay the development of step of the cascade makes the measurement of
diabetic complications. Although there have downstream mediators such as CRP partic-
been efforts to reduce HbA1c levels through ularly useful for clinical diagnosis. Increased
periodontal therapy, the ideal level to be high-sensitivity CRP plasma levels in patients
reached is not clear.28,33–35 It is possible that with prehypertension or established hyper-
periodontal treatment can improve HbA1c lev- tension may link these two conditions. Major
els by 0.4% to 0.5%35; this beneficial systemic depression, physical inactivity, family histories
action of periodontal therapy demonstrates the of CVD, advancing age, and male sex are other
importance of controlling local inflammatory risk factors for atherosclerotic CVD that are
conditions as part of diabetes management. commonly found in patients with periodontitis
and may serve as confounders.36
Risk factors common to CVD and peri-
Periodontitis and cardiovascular odontitis are thought to be related to increased
disease systemic inflammation.31 Diet, genetics, medi-
cations, and environmental factors influence
Periodontal disease has been linked to the course of periodontal disease, and when
CVD in cross-sectional and cohort stud- periodontal disease combines with these ele-
ies. 31,36,37 Studies report that elevated ments, they influence cardiovascular health
cell- and cytokine-mediated markers of inflam- status. In CVD studies, periodontal disease
mation, including CRP, fibrinogen, and various has been shown to be a contributing factor.37,41
cytokines, are associated with periodontal dis- Low-density lipoproteins (LDLs) are known
ease.18,30 The same proinflammatory markers to initiate and propagate an inflammatory
in periodontal disease have also been linked response.38 Once the migration of leukocytes
with atherothrombogenesis.18,19,38 When the has occurred, monocytes differentiate into mac-
progression of periodontal disease is reduced, rophages and proliferate. Fatty streaks develop;
levels of inflammatory markers common these consist of lipid-laden monocytes and
to both diseases (ie, IL-6, TNF-α, and CRP) macrophages as well as T lymphocytes, which
decrease, which might in turn decrease the risk are eventually joined by migrating smooth
of vascular disease. It is still unknown whether muscle cells from the vessel wall. If the cas-
inhibiting or reducing inflammation in general cade of events goes unchecked, the fatty streaks
or CRP in particular will lower the rate of vas- develop into advanced lesions, whereby macro-
cular effects.39 phages accumulate, a necrotic core is produced,
In several atherosclerosis studies using ani- and a fibrous cap is formed that blocks the
mal models, periodontal disease was shown lesion from the lumen.
to be a contributing factor.18,19,40 Activated The fibrous cap, which is characteristic of
immune cells in the atherogenic plaque pro- late-stage atherosclerosis, is of concern; rupture
duce proinflammatory cytokines (IFN, IL-1, of this fibrous cap is believed to underlie the
and TNF-α), which induce the production vast majority of myocardial infarctions that
of substantial amounts of IL-6. These cyto- may derive from these inflammatory processes.
kines are also produced in various tissues in Cytokines, such as IFN-γ, are involved in the
response to infection and in the adipose tis- destabilization of the plaque because they
sue of patients with metabolic syndrome. IL-6 lead to a decrease in matrix synthesis as well

94
Link Between Periodontal Disease and Systemic Conditions

as matrix degradation. In addition to these animals.31,40 These data suggest that the impact
cytokines, MMPs are involved in controlling of infection is not at the level of bacteremia
degradation; macrophages express MMPs, fur- and direct stimulation of arterial endothelium
ther contributing to degradation.31 but rather the result of the local inflammation
Periodontal inflammation causes a local inducing systemic stimulation.31
and systemic cytokine response. Several Patients with periodontitis who have two
studies have reported that elevated cell- and or more known risk factors for atherosclero-
cytokine-mediated markers of inflammation are sis should be referred by the dental team for
relevant to periodontal diseases.36,37,40 Proin- evaluation of atherosclerotic risk, which should
flammatory cytokines induce an acute-phase include physical examination and annual mea-
response in the liver characterized by elevated surement of blood pressure and blood lipid
levels of CRP and fibrinogen, which in turn profile.31 Patients with periodontitis and abnor-
promote atherogenesis. Also, gingival ulcer- mal serum lipid values, elevated levels of plasma
ation in periodontitis allows the migration of CRP (as measured by high-sensitivity CRP), or
bacteria into the blood (bacteremia), which both are recommended to follow a multifaceted
could provide a second inflammatory stimulus lifestyle-modification program to reduce the
leading to atheroma formation.42 risk of CVD. Cessation of cigarette smoking is
CVD, once characterized as a lipid disease recommended for all patients with periodonti-
because of the end-stage pathology, is now tis. Furthermore, all patients with periodontitis
known to have a significant inflammatory com- who have elevated blood pressure (> 130/80
ponent. In recent years, epidemiologic studies mmHg) should be treated according to stan-
have ascertained an association between CVD dard hypertension-management protocols and
and other inflammatory diseases, particularly should undertake lifestyle changes, including
periodontal diseases.4,36 Given the prolonged reduction of weight and dietary sodium intake,
nature of the pathogenesis of CVD, it has been as appropriate. Periodontal evaluation should
difficult to demonstrate cause and effect in be considered in patients with CVD who have
this association. To that end, the relationship signs or symptoms of a gingival disease or
between CVD and periodontitis was investi- unexplained tooth loss. Moreover, when peri-
gated longitudinally in an animal model. New odontitis is newly diagnosed in patients with
Zealand white rabbits were fed a 0.5% cho- CVD, dentists and physicians should closely
lesterol diet for 13 weeks. This protocol causes collaborate to optimize CVD risk reduction
atheromatous changes in the large arteries of and periodontal care.43
the animals.40 Periodontitis was induced with a Atherogenesis leading to atherosclerosis
ligature and Porphyromonas gingivalis in half involves highly specific cellular and molecular
the animals, while the other half maintained a responses that are described as an inflamma-
normal periodontium; all animals were fed the tory disease. Cardiovascular risk factors can
atherogenic diet. Animals that had periodon- be viewed as an injury or insult to which the
titis exhibited twice the atheroma formation body adapts, setting in motion a decades-long
of the control animals. These data provide response. Regardless of the cause, the ear-
direct longitudinal evidence that periodontal liest changes in atherosclerosis occur in the
inflammation can affect the progression of endothelium, leading to increased tissue per-
CVD. Further investigation was unable to iso- meability and cell adhesiveness, particularly of
late bacteria from the arterial lesions; however, leukocytes, as well as increased procoagulation
it was demonstrated that local inflammation properties that result in activation of vasoactive
was able to induce upregulation of the systemic molecules, cytokines, and growth factors.9
inflammation. Increased levels of circulating In the pathogenesis of atherosclerosis, mono-
CRP and IL-1 were detected in periodontitis cytes and T cells mediated by endothelial

95
4 The Role of Inflammation in Oral-Systemic Interactions

expression of adhesion molecules accumulate subjects with periodontal disease had a 1.14- to
at the injury site. Activated lymphocytes release 1.59-fold greater risk of developing CVD than
mediators such as cytokines and chemokines. did those without periodontal disease.36,37,42
Through a cascade of signals, the process lead- Although inflammatory response to injury
ing to entry of leukocytes into the endothelium and infection is essential to health, the response
involves the rolling, activation, and adherence causes problems when inflammatory processes
of leukocytes. One particular adhesion mole- are maladaptive, leading to chronic diseases
cule, vascular cell adhesion molecule 1, is of such as diabetes and CVD. New pathways by
importance in the adherence process because which local chronic inflammation can affect
it binds precisely with monocytes and T lym- large vessels need further investigation.
phocytes. Once adherent to the endothelium,
monocytes transmigrate into the intima. 9
During inflammation, selective recruitment of Periodontitis and pregnancy
leukocytes to the injured tissue regulates spe- Periodontal diseases are inflammatory responses
cific cell population migration to the tissues. to infectious pathogens that, although distant
Early proinflammatory cytokines, such as IL-1 from many organs, can affect the fetoplacental
and TNF-α, stimulate chemokine production. unit.44 Host cells activate a local inflammatory
Another cytokine, IFN-γ, is secreted by T lym- response against bacteria and their numerous
phocytes (specifically T helper cell type 1); this virulence factors (eg, LPS) to contain the lesion.
induces chemokine production directly and by However, several studies have reported that
acting with IL-1 and TNF-α. inflammatory cytokines, periodontal bacteria,
Two groups of chemokines associated and their virulence factors can enter the blood
with IFN-γ are CC and CXC. CC chemok- circulation to disseminate throughout the body.
ines induce the migration of monocytes. For Such an event triggers systemic inflammatory
example, monocyte chemoattractant protein responses and ectopic infections.36
1 is responsible for monocyte migration from Not surprisingly, local inflammation can
the bloodstream into the surrounding tissue interfere with the homeostasis of healthy
by binding to and activating CC chemokine pregnancy and labor. While a large number of
receptors, in this case CC chemokine recep- epidemiologic studies have shown a positive
tor 2. Depending on their receptor structural association between periodontal disease and
characteristics, CXC chemokines can induce pathologic outcomes of pregnancy, the results
migration of neutrophils or attract lympho- have not always been confirmed.44
cytes. Atheroma-associated cells have been In a healthy pregnancy, the mother and the
found to play a role in the movement and fetus exchange nutrients and waste through the
maintenance of activated T lymphocytes in umbilical cord that connects the placenta of the
vascular wall lesions during atherogenesis.16 mother to the fetus. In amniotic fluid, levels
The same study also found increased expres- of inflammatory mediators, including PGE2,
sion of the receptor for these chemokines, CXC TNF-α, and IL-1β, increase incrementally
chemokine receptor 3, by T lymphocytes in throughout pregnancy, reach a threshold, and
human atherosclerotic lesions. promote uterine contraction, cervical dilation,
When the progression of periodontal disease and delivery.45 It is of extreme importance that
is low, levels of inflammatory markers com- average levels of inflammatory mediators and
mon to both diseases (IL-6, TNF-α, and CRP) hormones be tightly regulated to maintain a
also decrease. This might, in turn, decrease regular course of pregnancy.44
the risk of vascular disease.36 When adjusted Evidence suggests that periodontal patho-
for risk factors, such as smoking, diabetes, gens interfere with homeostatic control of
alcohol intake, obesity, and blood pressure, pregnancy. Through DNA identification

96
Resolution of Inflammation and the Periodontal Disease–Systemic Disease Link

and immunodetection, both P gingivalis and olism, hormonal control, and inflammatory
Aggregatibacter actinomycetemcomitans mediators together influence health during
have been detected in biopsy specimens taken pregnancy and the outcome of labor.44 For
from the placentas of women diagnosed with more information on periodontal infections
preeclampsia.46 Most commonly in amniotic and pregnancy outcomes, see chapter 10.
fluids, Fusobacterium nucleatum was identified
in patients suffering from premature labor or
giving birth to babies with low birth weight.47
Although there is clear evidence that bac- Resolution of Inflammation
terial presence in diverse tissues establishes and the Periodontal Disease–
pathologic conditions during pregnancy, the
mechanisms by which the pathogens arise Systemic Disease Link
in the local tissues remain unclear.44 Recent
evidence indicates that pathogens can gain Periodontitis is an inflammatory disease that
access to the amniotic cavity by ascending is initiated by the oral microbial biofilm and
from the vagina and the cervix, by hematog- modulated by the host immune response
enous dissemination through the placenta, by and metabolism.15 This distinction implies
accidental introduction at the time of invasive that it is the host response to the biofilm
procedures (amniocentesis), and by retrograde that destroys the periodontium in the patho-
spread through the uterine tubes.44,48 Besides genesis of the disease. As the understanding
the pathogens themselves, their virulent fac- of inflammation pathways has matured, a
tors, including LPS, have been associated with better understanding of the molecular basis
triggering the same type of pathologic actions of resolution of inflammation has emerged.
to the placenta and fetus. Resolution of inflammation involves an
Localized elevated levels of maternal IL-1β, active, agonist-mediated, well-orchestrated
IL-6, PGE2, and TNF-α in the amniotic fluid return of tissue homeostasis.49 Table 4-1 details
have been associated with pregnancy alterations, the actions of immunoresolvents in acute
including premature birth. Other biomark- inflammatory disease models.
ers, such as MMPs, estriol, elastase, protease, The distinction between anti-inflammation
phospholipase, prolactin myeloperoxidase, and resolution has biologic and clinical
and tissue inhibitor of MMP 1 (TIMP-1) have relevance. Anti-inflammation involves phar-
been evaluated48 but with inconclusive results. macologic blocking of specific inflammatory
CRP, an acute-phase molecule synthesized by pathways; resolution is mediated by physi-
the liver, activates systemic inflammation and ologic pathways that restore homeostasis.6
proinflammatory cytokines; it is also associated Research suggests that chronic inflammatory
with premature birth.42 Other pregnancy com- periodontal disease consists of both failures of
plications have been associated with elevated resolution pathways to restore homeostasis and
levels of CRP, including intrauterine growth excessive response leading to the development
restriction and preeclampsia.44 of the chronic lesion.6,11 Proof-of-concept stud-
Finally, a combination of local complications ies published in the 1980s demonstrated that
places the host at risk for poor pregnancy out- pharmacologic anti-inflammatory measures
comes. For example, women with gestational reduced periodontal disease progression in ani-
diabetes mellitus have a high risk of present- mals and humans.71 However, such therapies
ing elevated serum levels of CRP, IL-6, and induce side effects related to the nonsteroi-
TNF-α. Through inflammatory pathways, dal anti-inflammatory drugs or other enzyme
these cytokines interfere with insulin-signaling inhibitors or receptor antagonists in periodon-
homeostasis. Therefore, carbohydrate metab- tal treatment.

97
4 The Role of Inflammation in Oral-Systemic Interactions

TABLE 4-1  |  Local and systemic actions of tissue inflammation


Inflammatory
disease models Immunoresolvents and their actions Reference(s)
Periodontitis/Rabbit Lipoxin A4/ATL
Prevents connective tissue and bone loss Serhan et al50
Accelerates healing of inflamed tissues Serhan et al50
Ceases infiltration of neutrophils Serhan et al50
Resolvin E1
Reduces bone loss Hasturk et al51
Regenerates lost soft tissue and bone tissue Hasturk et al52

Peritonitis/Mouse Lipoxin A4/ATL


Stops neutrophil recruitment Bannenberg et al53
Promotes lymphatic removal of phagocytes Schwab et al,54 Arita et al55
Resolvin E1
Decreases PMN infiltration Schwab et al54
Regulates chemokine and cytokine production Bannenberg et al53
Promotes lymphatic removal of phagocytes Spite et al56
Resolvin D1
Shortens resolution interval Recchiuti et al57
Regulates miRNAs Krishnamoorthy et al58
Reduces concentrations of LTB4, PGD2, PGF2, and TXA2 in exudates Norling et al59
Lowers antibiotic requirement Chiang et al60
Increases animal survival Chiang et al60
Reduces bacterial titers Chiang et al60
Protectin D1
Promotes local clearance of apoptotic cells Bannenberg et al53
Regulates lymphatic removal of phagocytes Ariel et al61,62
Modulates T-cell migration Schwab et al54
Maresin-1
Blocks infiltration of PMNs into the peritoneum Serhan et al63

Colitis/Mouse Lipoxin A4/ATL


Reduces severe colitis Aliberti et al64
Attenuates proinflammatory mediators’ gene expression Gewirtz et al65
Inhibits weight loss Gewirtz et al65
Reduces immune dysfunction Wallace and Fiorucci66
Resolvin E1
Improves animal survival rate Arita et al55
Reduces weight loss Arita et al55
Promotes LPS detoxification Campbell et al67
Stops neutrophil recruitment Ishida et al68
AT-Resolvin D1
Reduces disease’s activity index Bento et al69
Attenuates proinflammatory mediators’ gene expression Bento et al69
Attenuates neutrophil recruitment Bento et al69
Resolvin D2
Improves disease’s activity index Bento et al69
Reduces colonic PMN infiltration Bento et al69

Retinopathy/Mouse Resolvin E1/Resolvin D1/Protectin D1


Protects against neovascularization Connor et al70

ATL, aspirin-triggered 15-epi-lipoxin A4; LTB4, leukotriene B4; PGD2, prostaglandin D2; PGF2, prostaglandin F2; TXA2, thromboxane A2.
(Adapted from Freire and Van Dyke.73)

The isolation and characterization of biologic area of research into the use of endogenous lipid
agonist molecules, such as lipoxins, resolvins, mediators of resolution as potential therapeutic
protectins, and maresins, have opened a new agents for the management of inflammatory

98
References

periodontitis. Animal models of periodontitis the central role of inflammation as the driving
have revealed the potential of this therapeutic mechanism in the association between peri-
approach for prevention and treatment and odontitis and a number of systemic diseases
forced reconsideration of the understanding and conditions is emerging. A more precise
of the pathogenesis of human periodontal dis- picture is also emerging of the central role of
eases72 (see Table 4-1; for review, see Freire and inflammation in association with metabolism
Van Dyke73). and microbiome in the local and systemic
The importance of agonists of resolution in compartments. The target of periodontal ther-
eliminating inflammation has been confirmed apy, whether control of the biofilm or control
in demonstrations of control of systemic of local inflammation, is control of systemic
inflammatory disease by exogenous addition inflammation associated with chronic systemic
of excess lipoxins or resolvins.74 These findings diseases.
are observed in concert with the resolution of
periodontitis in the same animal, suggesting that
control of inflammation is central to the associa-
tion between periodontitis and systemic disease. References
The report provided support and a rationale for
the mechanism of the observed connections as 1. Granger DN, Senchenkova E. Inflammation and the
Microcirculation. San Rafael, CA: Morgan & Claypool
well as a basis for the clinical concern that peri- Life Sciences, 2010.
odontitis increases the risk for many important 2. Pallasch TJ, Wahl MJ. The focal infection theory:
systemic inflammatory diseases.51,52 Appraisal and reappraisal. J Calif Dent Assoc 2000; 28:
194–200.
3. Barnett ML. The oral-systemic disease connection. An
update for the practicing dentist. J Am Dent Assoc 2006;
137(suppl): 5S–6S.
Conclusion 4. Van Dyke TE, Kornman KS. Inflammation and factors
that may regulate inflammatory response. J Periodontol
2008; 79:1503–1507.
The link between oral and systemic inflamma- 5. Medzhitov R. Inflammation 2010: New adventures of
tory processes and its consequences are directly an old flame. Cell 2010;140:771–776.
related to the tissue response to challenge, 6. Serhan CN, Chiang N, Van Dyke TE. Resolving inflam-
mation: Dual anti-inflammatory and pro-resolution lipid
usually microbial, and inflammation. This
mediators. Nat Rev Immunol 2008;8:349–361.
biologic process is controlled by the balance 7. Majno G, Joris I. Cells, Tissues, and Disease: Principles
between mediators and sensors that amplify the of General Pathology. Oxford: Oxford University, 2004.
inflammatory process and those that regulate 8. Van Dyke TE. Cellular and molecular susceptibility deter-
minants for periodontitis. Periodontol 2000 2007; 45:
the return to homeostasis. Diseases associated
10–13.
with uncontrolled acute inflammation are char- 9. Van Dyke TE, van Winkelhoff AJ. Infection and inflam-
acterized by insufficient actions of resolution matory mechanisms. J Clin Periodontol 2013; 40(suppl
programs and inappropriate release and main- 14):S1–S7.
10. Graves DT, Oates T, Garlet GP. Review of osteoimmu-
tenance of high levels of toxic substances and nology and the host response in endodontic and
proinflammatory mediators that may result periodontal lesions. J Oral Microbiol 2011;3.
in damage to host tissues and prolong the 11. Fredman G, Oh SF, Ayilavarapu S, Hasturk H, Serhan
inflammatory response. In experimental ani- CN, Van Dyke TE. Impaired phagocytosis in localized
aggressive periodontitis: Rescue by Resolvin E1. PLoS
mal models, compelling evidence demonstrates One 2011;6:e24422.
the actions of proresolution mediators in the 12. Darveau RP. Periodontitis: A polymicrobial disruption of
regulation of local and systemic inflammatory host homeostasis. Nat Rev Microbiol 2010;8:481–490.
13. Eke PI, Thornton-Evans GO, Wei L, Borgnakke WS,
responses, elucidating the role of endogenous
Dye BA. Accuracy of NHANES periodontal examination
mediators in systemic inflammatory processes protocols. J Dent Res 2010;89:1208–1213.
and disease regulation. A clearer picture of

99
4 The Role of Inflammation in Oral-Systemic Interactions

14. Eke PI, Dye BA, Wei L, et al. Prevalence of periodontitis 31. Schenkein HA, Loos BG. Inflammatory mechanisms
in adults in the United States: 2009 and 2010. J Dent linking periodontal diseases to cardiovascular diseases.
Res 2012;91:914–920. J Clin Periodontol 2013;40(suppl 14):S51–S69.
15. Offenbacher S. Periodontal diseases: Pathogenesis. 32. Chapple IL, Genco R, Working Group 2 of the Joint
Ann Periodontol 1996;1:821–878. EFP/AAP Workshop. Diabetes and periodontal diseases:
16. Kornman KS, Van Dyke TE. Bringing light to the heat: Consensus report of the Joint EFP/AAP Workshop on
“Inflammation and periodontal diseases: A reappraisal.” Periodontitis and Systemic Diseases. J Clin Periodontol
J Periodontol 2008;79:1313. 2013;40(suppl 14):S106–S112.
17. Williams RC, Barnett AH, Claffey N, et al. The potential 33. Bharti P, Katagiri S, Nitta H, et al. Periodontal treatment
impact of periodontal disease on general health: A with topical antibiotics improves glycemic control in
consensus view. Curr Med Res Opin 2008; 24: association with elevated serum adiponectin in patients
1635–1643. with type 2 diabetes mellitus. Obes Res Clin Pract 2013;
18. Reyes L, Herrera D, Kozarov E, Roldán S, Progulske-Fox 7:e129–e138.
A. Periodontal bacterial invasion and infection: Contri- 34. Atouf F, Park CH, Pechhold K, Ta M, Choi Y, Lumelsky
bution to atherosclerotic pathology. J Clin Periodontol NL. No evidence for mouse pancreatic beta-cell
2013;40(suppl 14):S30–S50. epithelial-mesenchymal transition in vitro. Diabetes
19. Hayashi C, Papadopoulos G, Gudino CV, et al. Protec- 2007;56:699–702.
tive role for TLR4 signaling in atherosclerosis progression 35. Borgnakke WS, Ylöstalo PV, Taylor GW, Genco RJ.
as revealed by infection with a common oral pathogen. Effect of periodontal disease on diabetes: Systematic
J Immunol 2012;189:3681–3688. review of epidemiologic observational evidence. J Clin
20. Linden GJ, Herzberg MC, Working Group 4 of the Joint Periodontol 2013;40(suppl 14):S135–S152.
EFP/AAP Workshop. Periodontitis and systemic 36. Genco RJ, Van Dyke TE. Prevention: Reducing the risk
diseases: A record of discussions of Working Group 4 of CVD in patients with periodontitis. Nat Rev Cardiol
of the Joint EFP/AAP Workshop on Periodontitis and 2010;7:479–480.
Systemic Diseases. J Clin Periodontol 2013; 40(suppl 37. de Oliveira C, Watt R, Hamer M. Toothbrushing, inflam-
14):S20–S23. mation, and risk of cardiovascular disease: Results from
21. Coussens LM, Werb Z. Inflammation and cancer. Nature Scottish Health Survey. BMJ 2010;340:c2451.
2002;420:860–867. 38. Cybulsky MI, Jongstra-Bilen J. Resident intimal dendritic
22. Tezal M, Grossi SG, Genco RJ. Is periodontitis associ- cells and the initiation of atherosclerosis. Curr Opin
ated with oral neoplasms? J Periodontol 2005; 76: Lipidol 2010;21:397–403.
406–410. 39. Beck JD, Couper DJ, Falkner KL, et al. The Periodontitis
23. Tezal M, Scannapieco FA, Wactawski-Wende J, et al. and Vascular Events (PAVE) pilot study: Adverse events.
Local inflammation and human papillomavirus status of J Periodontol 2008;79:90–96.
head and neck cancers. Arch Otolaryngol Head Neck 40. Jain A, Batista EL Jr, Serhan C, Stahl CL, Van Dyke TE.
Surg 2012;138:669–675. Role for periodontitis in the progression of lipid deposi-
24. Hujoel PP, Drangsholt M, Spiekerman C, Weiss NS. An tion in an animal model. Infect Immun 2003; 71:
exploration of the periodontitis-cancer association. Ann 6012–6018.
Epidemiol 2003;13:312–316. 41. Couper DJ, Beck JD, Falkner KL, et al. The Periodontitis
25. Michaud DS, Liu Y, Meyer M, Giovannucci E, Joshipura and Vascular Events (PAVE) pilot study: Recruitment,
K. Periodontal disease, tooth loss, and cancer risk in retention, and community care controls. J Periodontol
male health professionals: A prospective cohort study. 2008;79:80–89.
Lancet Oncol 2008;9:550–558. 42. Paraskevas S, Huizinga JD, Loos BG. A systematic
26. Arora M, Weuve J, Fall K, Pedersen NL, Mucci LA. An review and meta-analyses on C-reactive protein in
exploration of shared genetic risk factors between peri- relation to periodontitis. J Clin Periodontol 2008; 35:
odontal disease and cancers: A prospective co-twin 277–290.
study. Am J Epidemiol 2010;171: 253–259. 43. Devchand PR, Arita M, Hong S, et al. Human ALX recep-
27. Miller SU. Increased human body water loss at reduced tor regulates neutrophil recruitment in transgenic mice:
ambient pressure. Aerosp Med 1962;33: 689–691. Roles in inflammation and host defense. FASEB J 2003;
28. Engebretson S, Kocher T. Evidence that periodontal 17:652–659.
treatment improves diabetes outcomes: A systematic 44. Madianos PN, Bobetsis YA, Offenbacher S. Adverse
review and meta-analysis. J Clin Periodontol 2013; pregnancy outcomes (APOs) and periodontal disease:
40(suppl 14):S153–S163. Pathogenic mechanisms. J Clin Periodontol 2013;
29. Zhang Y, Dall TM, Mann SE, et al. The economic costs 40(suppl 14):S170–S180.
of undiagnosed diabetes. Popul Health Manag 2009; 45. Haram K, Mortensen JH, Wollen AL. Preterm delivery:
12:95–101. An overview. Acta Obstet Gynecol Scand 2003; 82:
30. Taylor JJ, Preshaw PM, Lalla E. A review of the evidence 687–704.
for pathogenic mechanisms that may link periodontitis 46. Gonzales-Marin C, Spratt DA, Millar MR, Simmonds M,
and diabetes. J Clin Periodontol 2013; 40(suppl 14): Kempley ST, Allaker RP. Levels of periodontal pathogens
S113–S134. in neonatal gastric aspirates and possible maternal sites
of origin. Mol Oral Microbiol 2011;26:277–290.

100
References

47. Han YW, Redline RW, Li M, Yin L, Hill GB, McCormick 60. Chiang N, Fredman G, Bäckhed F, et al. Infection regu-
TS. Fusobacterium nucleatum induces premature and lates pro-resolving mediators that lower anti­biotic
term stillbirths in pregnant mice: Implication of oral requirements. Nature 2012;484:524–528.
bacteria in preterm birth. Infect Immun 2004; 72: 61. Ariel A, Li PL, Wang W, et al. The docosatriene protectin
2272–2279. D1 is produced by TH2 skewing and promotes human
48. Gürsoy M, Könönen E, Gürsoy UK, Tervahartiala T, T cell apoptosis via lipid raft clustering. J Biol Chem
Pajukanta R, Sorsa T. Periodontal status and neutrophilic 2005;280:43079–43086.
enzyme levels in gingival crevicular fluid during preg- 62. Ariel A, Fredman G, Sun YP, et al. Apoptotic neutrophils
nancy and postpartum. J Periodontol 2010; 81: and T cells sequester chemokines during immune
1790–1796. response resolution through modulation of CCR5
49. Van Dyke TE, Serhan CN. Resolution of inflammation: expression. Nat Immunol 2006;7:1209–1216.
A new paradigm for the pathogenesis of periodontal 63. Serhan CN, Yang R, Martinod K, et al. Maresins: Novel
diseases. J Dent Res 2003;82:82–90. macrophage mediators with potent antiinflammatory
50. Serhan CN, Jain A, Marleau S, et al. Reduced inflam- and proresolving actions. J Exp Med 2009;206:15–23.
mation and tissue damage in transgenic rabbits 64. Aliberti J, Hieny S, Reis e Sousa C, Serhan CN, Sher
overexpressing 15-lipoxygenase and endogenous A. Lipoxin-mediated inhibition of IL-12 production by
anti-inflammatory lipid mediators. J Immunol 2003; 171: DCs: A mechanism for regulation of microbial immunity.
6856–6865. Nat Immunol 2002;3:76–82.
51. Hasturk H, Kantarci A, Ohira T, et al. RvE1 protects from 65. Gewirtz AT, Collier-Hyams LS, Young AN, et al. Lipoxin
local inflammation and osteoclast-mediated bone A4 analogs attenuate induction of intestinal epithelial
destruction in periodontitis. FASEB J 2006;20:401–403. proinflammatory gene expression and reduce the sever-
52. Hasturk H, Kantarci A, Goguet-Surmenian E, et al. ity of dextran sodium sulfate-induced colitis. J Immunol
Resolvin E1 regulates inflammation at the cellular and 2002;168:5260–5267.
tissue level and restores tissue homeostasis in vivo. J 66. Wallace JL, Fiorucci S. A magic bullet for mucosal
Immunol 2007;179:7021–7029. protection...and aspirin is the trigger! Trends Pharmacol
53. Bannenberg GL, Chiang N, Ariel A, et al. Molecular Sci 2003;24:323–326.
circuits of resolution: Formation and actions of resolvins 67. Campbell EL, MacManus CF, Kominsky DJ, et al. Resol-
and protectins. J Immunol 2005;174:4345–4355. vin E1-induced intestinal alkaline phosphatase promotes
54. Schwab JM, Chiang N, Arita M, Serhan CN. Resolvin resolution of inflammation through LPS detoxification.
E1 and protectin D1 activate inflammation-resolution Proc Natl Acad Sci U S A 2010;107:14298–14303.
programmes. Nature 2007;447:869–874. 68. Ishida T, Yoshida M, Arita M, et al. Resolvin E1, an
55. Arita M, Yoshida M, Hong S, et al. Resolvin E1, an endogenous lipid mediator derived from eicosapentae-
endogenous lipid mediator derived from omega-3 eicos- noic acid, prevents dextran sulfate sodium-induced
apentaenoic acid, protects against 2,4,6-trinitrobenzene colitis. Inflamm Bowel Dis 2010;16:87–95.
sulfonic acid-induced colitis. Proc Natl Acad Sci U S A 69. Bento AF, Claudino RF, Dutra RC, Marcon R, Calixto
2005;102:7671–7676. JB. Omega-3 fatty acid-derived mediators 17(R)-
56. Spite M, Summers L, Porter TF, Srivastava S, Bhatnagar hydroxy docosahexaenoic acid, aspirin-triggered resol-
A, Serhan CN. Resolvin D1 controls inflammation initiated vin D1 and resolvin D2 prevent experimental colitis in
by glutathione-lipid conjugates formed during oxidative mice. J Immunol 2011;187:1957–1969.
stress. Br J Pharmacol 2009;158:1062–1073. 70. Connor KM, SanGiovanni JP, Lofqvist C, et al. Increased
57. Recchiuti A, Krishnamoorthy S, Fredman G, Chiang N, dietary intake of omega-3-polyunsaturated fatty acids
Serhan CN. MicroRNAs in resolution of acute inflam- reduces pathological retinal angiogenesis. Nat Med
mation: Identification of novel resolvin D1-miRNA 2007;13:868–873.
circuits. FASEB J 2011;25:544–560. 71. Howell TH. Blocking periodontal disease progression
58. Krishnamoorthy S, Recchiuti A, Chiang N, Fredman G, with anti-inflammatory agents. J Periodontol 1993;
Serhan CN. Resolvin D1 receptor stereoselectivity and 64(suppl 8S):828–833.
regulation of inflammation and proresolving microRNAs. 72. Recchiuti A, Serhan CN. Pro-resolving lipid mediators
Am J Pathol 2012;180:2018–2027. (SPMs) and their actions in regulating miRNA in novel
59. Norling LV, Dalli J, Flower RJ, Serhan CN, Perretti M. resolution circuits in inflammation. Front Immunol 2012;
Resolvin D1 limits polymorphonuclear leukocyte recruit- 3:298.
ment to inflammatory loci: Receptor-dependent actions. 73. Freire MO, Van Dyke TE. Natural resolution of inflam-
Arterioscler Thromb Vasc Biol 2012;32:1970–1978. mation. Periodontology 2000 2013;63:149–164.
74. Van Dyke TE. Inflammation and periodontal diseases:
A reappraisal. J Periodontol 2008;79:1501–1502.

101
CHAPTER 5

Obesity, Metabolic Syndrome,


and Oral Health
Ira B. Lamster, dds, mmsc
Nadia Laniado, dds, mph
Ilene Fennoy, md, mph

Obesity has been identified as a major health (skinfold thickness), magnetic resonance imag-
problem across the globe.1–3 It has been linked ing (MRI), hydrodensitometry (underwater
to the initiation and acceleration of a variety weighing), air plethysmography, bioelectric
of disorders, including cardiovascular disease impedance, and dual-energy x-ray absorpti-
(CVD) and associated hypertension, diabe- ometry (DXA).13,14 Anthropometry is difficult
tes and metabolic syndrome,4,5 obstructive to standardize in practice and therefore is not
sleep apnea,6 osteoarthritis,7 and nonalco- a useful clinical tool, though it has been used
holic fatty liver disease8 and is associated with frequently in population-based research studies
increased all-cause morbidity and mortality.9 and has been demonstrated to have validity
Globally, obesity continues to increase in pedi- against the more technologic methods men-
atric and adult age groups.10,11 The prevalence tioned above.15 Many of the other methods
of obesity is greater in adult women than in are not practical for routine clinical use due
adult men (Fig 5-1). In the United States, it to cost, radiation exposure, and time but have
has been estimated that state-specific medical been used to validate body mass index (BMI),
expenditures would be 6.9% to 10% lower in a measure calculated as weight in kilograms
the absence of obesity,12 demonstrating a signifi- divided by height in meters squared (or pounds
cant economic impact in addition to the impact divided by height in inches squared, multiplied
on health and well-being. Given the negative by 703).15,16
effects of obesity on health and an expanding Classification of weight status by BMI is
literature on the relationship between obesity established in adults as follows: BMI < 18.5
and oral disease, all oral health care profession- = underweight, ≥ 18.5 to 24.9 = normal
als must consider the impact of obesity on the weight, ≥ 25 to 29.9 = overweight, and ≥ 30
provision of oral health care services. = obese17 (Table 5-1). The obesity group has
been further classified by severity, with Class 1
obesity defined as BMI ≥ 30 and < 35, Class 2
as BMI ≥ 35 and < 40, and Class 3 as BMI ≥
Assessment of Obesity 40.17,18 In adults, these criteria are independent
of height, age, and sex. However, in children,
Obesity is defined as an increase in total body BMI requires adjustment for developmental
fat. Multiple methods exist for the measure- status and sex and is therefore plotted on sex-
ment of body fat, including anthropometry and age-specific growth charts with percentile

102
Assessment of Obesity

Fig 5-1 |  Age-standardized prevalence of obesity (BMI ≥ 30) worldwide in men (a) aged ≥ 20 years and
women (b) as of 2013. (Reprinted from Ng et al11 with permission.)

rankings.19 To further relate pediatric classifica- Distribution of fat between visceral depots
tion with that of the adult, severe obesity (Class and subcutaneous areas has been associated
2) in childhood has been identified as ≥ 120% with variation in risk for comorbidities.21,22
of the 95th percentile, and very severe obesity BMI fails to provide information on this aspect
(Class 3) as ≥ 140% of the 95th percentile.20 of obesity and as a result must be augmented

103
5 Obesity, Metabolic Syndrome, and Oral Health

TABLE 5-1  |  BMI-for-age weight status


Weight status category Percentile range BMI
Underweight < 5th < 18.5

Healthy weight 5th to < 85th 18.5–24.9

Overweight 85th to < 95th 25–29.9

Obese ≥ 95th ≥ 30

by other measures. Measurement of waist cir- 12- to 19-year-olds demonstrating the high-
cumference (WC) is used clinically to determine est prevalence (20.9%) and 2- to 5-year-olds
visceral adipose tissue23 and has been correlated demonstrating the lowest (13.9%).25 Overall,
with adverse cardiovascular outcomes. there was no significant sex-specific difference.
WC, waist-hip ratio, and waist-height ratio By ethnicity, black (22%) and Hispanic (25%)
have all been shown to correlate with cardio- children had the highest prevalence of obesity,
vascular comorbidities reflective of increased with non-Hispanic whites having a 14.1%
visceral body fat and increased cardiovascular prevalence and Asians having 11%.25 For both
risk.21 In addition, WC and waist-height ratio adults and children, there has been a slow but
have been associated with periodontal pockets, steady rise in the prevalence of obesity from a
albeit weakly.24 30.5% in adults and 13.9% in youth in 1999
to 2000 to the current measures of 39.8% in
adults and 18.5% in children.25
Worldwide, the number of men categorized
Prevalence of Obesity as overweight increased from 28.8% in 1980
to 36.9% in 2013, accompanied by an increase
Recent (2015–2016) estimates of obesity in for women from 29.8% to 38.0%.11 Both
the United States indicate a prevalence of developing and developed countries demon-
39.8% among adults, with younger adults strate this trend, with women showing more
having a lower incidence than adults aged rapidly increasing obesity than men. Children
20 to 60 years.25 Overall, there were no sig- demonstrate these same trends in overweight
nificant differences by age group between and obesity, with increases from 16.9% to 23%
men and women.25 Ethnicity, however, pro- in boys and 16.2% to 22% in girls from devel-
vided a significant difference in prevalence, oped countries between 1980 and 2013, while
with non-Hispanic Asians demonstrating an increases in developing countries rose from
incidence of 12.7%, non-Hispanic whites 8.1% to 12.9% in boys and 8.4% to 13.4%
37.9%, non-Hispanic blacks 46.8%, and His- in girls over this same time period. Prevalence
panics 47%.25 Within the ethnic groups, Asian, was higher at all ages in developed countries
Hispanic, and black women demonstrated sig- compared to developing countries. However,
nificantly more obesity than men of the same there are distinct geographic patterns for the
ethnic group, while white men and women had prevalence of overweight and obesity, with the
a similar prevalence.25 Middle East, North Africa, and Oceania and
For children in the United States, the Micronesia demonstrating some of the highest
prevalence of obesity from 2 to 19 years numbers for children and adults. Globally, 62%
was 18% for the years 2015 to 2016, with of all persons with obesity live in developing

104
Etiology of Obesity

countries, though the United States accounts content.38 Reduced-calorie diets have been
for 13% of all persons with obesity. Obesity shown to result in equal weight loss regard-
is clearly a global problem that is increasing less of the specific macronutrient emphasized.39
worldwide. Thus, low-fat and low-carbohydrate diets at the
same caloric level provide equal effectiveness
in inducing weight loss. Certain dietary com-
ponents, however, have been identified as more
Etiology of Obesity commonly associated with weight gain. These
include sugar-sweetened beverages (SSBs) and
Obesity represents a disorder with multifac- processed foods such as potato chips and pro-
torial causes. At a basic level, it represents a cessed meats.40 Individual dietary components
mismatch between energy in and energy out. have been particularly identified as problem-
However, the etiology of that imbalance has atic for key obesity-related disorders, such as
been shown to be at least 50% genetic,26,27 saturated fats for hypercholesterolemia41 and
while the remainder involves environmental, sugar for diabetes.42,43
psychologic, socioeconomic, and physiologic In general, the effect of dietary macronu-
factors.28,29 These factors can be seen to be trients on CVD represents a composite effect,
operational, beginning prenatally with ges- reflecting the relationship between nutrients
tational obesity30 and diabetes,31 associated rather than a single nutrient alone,44 as well as
with increased risk of childhood obesity, or the role of the individual’s genetic contribution.
in animal studies exposure to bisphenol A Dietary patterns are the areas of concern, not
and S,32 associated with fetal adipogenesis. In isolated macronutrients.45 Hence, saturated
cases of maternal diabetes, fetal exposure is fat alone is not the culprit but the extent to
linked to multigenerational effects,30 hypoth- which those saturated fats are replaced by
esized to be transmitted through epigenetic unsaturated fats versus simple carbohydrates.46
mechanisms33—that is, through mechanisms The increase in SSBs, therefore, is a particu-
affecting DNA expression but not the genes lar adverse dietary change associated with an
themselves. Additionally, the prenatal problem increase in cardiometabolic disease.47
of intrauterine growth restriction is associated SSB intake has been associated with
with childhood obesity and insulin resis- metabolic, cardiovascular, and oral health dis-
tance.34 Thus, the prenatal environmental orders.48,49 From 1965 to 2004, SSBs increased
experience is a contributor to both childhood dramatically as a proportion of the Ameri-
obesity and adult cardiovascular and meta- can diet.50,51 More recent data indicate some
bolic disease.35 decline, but more than 50% of the US popu-
Dietary composition represents another lation still consumes at least one SSB per day,
contributor to the obesity epidemic. High-fat with 25% consuming at least 200 kcal/day
diets have been hypothesized as responsi- from SSBs.52 Variations in consumption differ
ble for the increasing obesity of populations by age, race/ethnicity, and sex. Non-Hispanic
based on their contribution to increased energy black persons consume the greatest amount as
density.36,37 Nevertheless, epidemiologically a a percent of total calories ingested, both chil-
reduction in fat content in US diets has been dren (8.5% of kcal) and adults (8.6% of total
associated with a continued rise in the prev- kcal), while non-Hispanic white adults ingest
alence of obesity in the overall population.38 the least (5.3% of total kcal).52
This apparent paradox has been clarified by Genetic predisposition may contribute
the recognition that despite a decrease in fat to sensitivity to weight gain from SSBs, as
content, there has been an increase in total demonstrated by Qi et al.53 However, pro-
caloric intake and an increase in carbohydrate spective cohort studies and a meta-analysis of

105
5 Obesity, Metabolic Syndrome, and Oral Health

randomized controlled trials (RCTs) both docu- in sedentary behavior. The independent con-
ment increased weight gain in association with tributions of physical activity and sedentary
increased intake of SSBs,54 suggesting that this behaviors to obesity have been demonstrated
is a general phenomenon. Furthermore, efforts by Stamatakis et al66 in adults and Loprinzi
to limit SSB intake in children and adolescents et al63,64 in children. Efforts to modify lifestyle
have been associated with decreased weight behaviors for prevention or treatment of obe-
gain.55,56 sity must address both sedentary behavior and
physical activity. Emphasis on the importance
of a healthy lifestyle, which is a potential new
Physical activity practice activity for oral health care providers
Physical activity patterns are inversely cor- and is discussed later in this chapter, includes
related with rates of obesity.57 Spontaneous increased physical activity.
physical activity or nonexercise activity (ie,
activity of a fidgeting, restless nature, not
motivated by a reward-seeking goal, such
as food-seeking behavior) has been shown Pathophysiology of Obesity
to help prevent weight gain.58 Furthermore,
intervention data demonstrate that increases Obesity, inflammation, and insulin
in physical activity to at least 150 minutes
per week are effective in achieving weight loss
resistance
in those with appropriate diets compared to Adipose tissue is a vibrant, active tissue
those on the same diet without the exercise.59 involved in physiologic and immune regula-
In short, both spontaneous and exercise activ- tion.67 It is a multicellular organ composed of
ity contribute to total energy expenditure, fat cells (adipocytes and preadipocytes) and
with low energy expenditure associated with a network of connective tissue, vascular, and
increased weight gain.60 immune system cells.68,69 Adipocytes make up
The importance of physical activity is also 25% of the cell population, while the other cell
demonstrated from a financial perspective, types, referred to as the stromal-vascular com-
with 11.1% of health expenditures associated ponent, make up the remaining 75%.70 Cells
with individuals who have inadequate physi- of the adipose tissue organ can be categorized
cal activity, defined as less than 150 minutes as follows: (1) by function (white fat used
per week.61 Only 50% of adults in the United primarily as a storage organ for fuel reserves
States met these guidelines for aerobic activ- and brown fat used for heat generation), (2)
ity in 2012.62 For children aged 6 to 11 years by localization (subcutaneous versus visceral
in 2003–2006, 71% performed more than 60 fat), and (3) by cell types (adipocytes versus
minutes per day of moderate physical activity, the stromal-vascular component).70,71 Excessive
but only 9.9% of 12- to 17-year-olds engaged adipose tissue, particularly visceral adiposity, is
in at least 60 minutes per day of moderate associated with increased cardiovascular risk,
physical activity.63 Overall, youth activity has type 2 diabetes, and periodontal disease, all of
declined from 1971 to 2012 while electron- which have an inflammatory component.72–74
ics use and computer time have increased.64 Indeed, excessive adiposity or obesity is itself
Among adults in Australia, a trend toward recognized as an inflammatory condition,75
increases in physical activity has occurred along characterized by macrophage recruitment76 and
with an increase in sedentary behavior associ- increased production of tumor necrosis factor
ated with an increase in obesity.65 These results alpha (TNF-α),77 C-reactive protein (CRP),78
highlight the fact that an increase in physical interleukin 6 (IL-6),79 and a host of other cyto-
activity is not synonymous with a decrease kines (Fig 5-2).

106
Pathophysiology of Obesity

Adipose tissue and inflammation

Leptin Cytokines
Adiponectin Interleukins
IL-12 TNF-α
IFN-γ IL-6
IL-1βRa VEGF
Cathepsin S LPS
CCR2 HIF-1α
Integrin αvβ3
PAI-1
Leptin
HIF-1α
VEGF
Endothelial cell Preadipocyte

Th1 cell
Cytokines, chemokines

Adipocyte M1 macrophage Lymphocyte Th1 cell

Fig 5-2 |  Factors contributing to immunity and inflammation in obesity. Adipose tissue demonstrating both
adipocytes, preadipocytes, endothelial cells, and M1 macrophages with their secretory products, which support
inflammation. Adipocytes produce leptin, adiponectin, cytokines, and interleukins. M1 macrophages produce
interleukins (IL-12, IL-6, IL-1βRA), cytokines (interferon gamma [IFN-γ] and TNF-α), proteases such as cathepsin
S, chemokine receptor 2 (CCR2), hypoxia-inducible factor-1α (HIF-1α), and lipopolysaccharide (LPS). Endothe-
lial cells produce cell adhesion molecules like integrin αvβ3, cytokines, chemokines, HIF-1α, and plasminogen
activator inhibitor-1 (PAI-1). Preadipocytes produce growth factors such as vascular endothelial growth factor
(VEGF), leptin, HIF-1α, PAI-1, and cell adhesion molecules like integrin αvβ3.

Macrophage infiltration represents a key tion).81 Interferon gamma (IFN-γ), cytokine


element in the inflammation associated with secretion, IL-12, bacterial lipopolysaccharides
obesity.72,76,80 The stromal-vascular cells of (LPSs), and T-cell activation are all media-
visceral adipose tissue produce increased mac- tors of the classical activation pathway, while
rophage mRNA transcripts that correlate with immunosuppression of T-cell immune reactions
increased body mass in mice in both genetic through mediators such as IL-10, IL-13, and
and diet-induced obesity.76 Furthermore, both IL-4 is characteristic of the alternative path-
adipocyte size and BMI are predictors of the way.81 Obesity alters the macrophage type
numbers of macrophages in various fat depots distribution toward an increase in the proin-
of mice.76 Considering the importance of the flammatory M1 macrophages.82 These macro-
inflammatory response to the pathogenesis of phages overexpress genes for factors involved
periodontal disease, an in-depth discussion of in macrophage migration and phagocytosis,
the role of inflammation in obesity follows. such as IL-6 and CC chemokine receptor type
Macrophages are of two types: proin- 2 (CCR2).79,83 Thus, macrophages in adipose
flammatory (M1, classical activation) and tissue are a major contributor to the inflam-
anti-inflammatory (M2, alternative act­iva­ matory response seen with obesity.

107
5 Obesity, Metabolic Syndrome, and Oral Health

The adipocyte secretes adipokines, includ- correlate with weight gain or obesity.96–98
ing leptin, adiponectin, interleukins, and Adiponectin inhibits colony-forming units
cytokines,84 which have effects on the immune (CFUs) of macrophages and granulocytes and
system. Fain et al85 demonstrated that the suppresses mature macrophage functions.99
contribution by nonadipocyte cells, particu- A reciprocal relationship exists between the
larly macrophages, was greater than that of cytokines TNF-α, IL-6, and adiponectin: Pro-
the adipocyte for a variety of these cytokines, duction of TNF-α and IL-6 is inhibited by
including cathepsin S, macrophage migratory adiponectin,100,101 while TNF-α and IL-6 inhibit
inhibiting factor (MIF), and interleukin 1β adiponectin gene expression and secretion.102
receptor antagonist (IL-1βRa).85 However, The low adiponectin levels seen in obesity thus
leptin not only acts as a cytokine but has support the inflammatory state through loss of
been shown to regulate T-cell production of suppression of TNF-α and IL-6.91
additional cytokines such as stimulation of
the Th1 cell type with its proinflammatory
cytokine production over the Th2 cell type Obesity and angiogenesis
with its immunoregulatory cytokine pro- Endothelial cells contribute to the inflammatory
duction.86–88 As a cytokine, leptin activates process by coordinating the recruitment of leu-
increased numbers of and increased phagocy- kocytes into the adipose mass.103 Activation of
tosis by macrophages/monocytes, chemotaxis endothelial cells results in the production of cell
of neutrophils, and their release of oxygen adhesion molecules (CAMs), chemokines, cyto-
radicals.89,90 Adipocyte-derived leptin directly kines, and ectoenzymes. These products guide
and indirectly stimulates immune responses, leukocytes into underlying tissues through a
augmenting the effects of the non–fat cell series of steps involving (1) tethering and roll-
component of adipose tissue, resulting in an ing by adhesion molecules such as selectin, then
increased inflammatory state. (2) firm adhesion and activation by chemoat-
Adiponectin, in contrast, exhibits dual roles tractants on the endothelium and adhesion
with respect to inflammation. In autoimmune molecules such as integrins on the leukocytes
disorders, locally and systemically increased or CAMs on the endothelium, and finally (3)
adiponectin levels have been noted in the diapedesis of leukocytes through cell-to-cell
presence of inflammation.91 Serum levels have endothelial junctions involving molecules such
been described as elevated in systemic lupus as platelet/endothelial CAMs (PECAMs).103,104
erythematosus and rheumatoid arthritis.92 In Cytokines associated with inflammation such
rheumatoid arthritis, adiponectin has been as IL-6 and TNF-α are known to induce endo-
shown to bind to adiponectin receptors (Adi- thelial activation.105 As a result, the endothelial
poR) on chondrocytes in a dose-dependent cell can function as a regulator of the move-
manner and to increase mRNA expression ment of leukocytes to the underlying tissues.
of matrix metalloproteinases associated with In addition, substances secreted by micro-
increased destruction of chondrocytes.93 Both vascular endothelial cells have been shown
IL-6 and pro-matrix metalloproteinase-1, the to promote proliferation of preadipocytes,
principal mediators of inflammation in rheuma- supporting a role of the endothelium as a con-
toid arthritis, are upregulated by adiponectin.94 tributor to fat mass expansion.106,107 Integrin
In both osteoarthritis and rheumatoid arthritis, αvβ3 and plasminogen activator inhibitor 1
increased adiponectin has been found in syno- (PAI-1), produced by both endothelial cells and
vial fluid.95 preadipocytes, have been identified as key sub-
In obesity and metabolic disorders, adi- stances in the coordinated development of both
ponectin has been well documented to have endothelial cells and adipose tissue at the same
anti-inflammatory effects and to negatively site,108 while peroxisome proliferator-activated

108
Consequences of Obesity

receptor gamma (PPAR-γ), involved in adipo-


cyte differentiation, can be shown to interfere
Consequences of Obesity
with both adipose tissue development and
angiogenesis if overexpressed as a dominant
Morbidity and mortality
negative contruct.109 Furthermore, blocking Systematic review of the literature and meta-
of angiogenesis by use of vascular endothelial analysis has documented increased all-cause
growth factor receptor 2 (VEGFR2)–blocking mortality (hazard ratio = 1.18 [95% confi-
antibody resulted in a similar failure of angio- dence interval (CI): 1.12–1.25]) in association
genesis and adipose tissue development,109 as with obesity (BMI ≥ 30 kg/m2) compared to
did use of a variety of other angiogenesis inhib- those with normal weight (BMI = 18 to ≤ 25
itors.110 Adipogenesis is therefore dependent on kg/m2).9 International data spanning 32 coun-
angiogenesis. tries, evaluating individuals followed for 5
Hypoxia is considered a key initiator of years or more and excluding those who had
the endothelial activation associated with ever smoked or had preexisting chronic condi-
expanding fat tissue.111 As the fat mass tions, reaches a similar conclusion but with the
expands with increasing recruitment of leuko- rise in mortality beginning with the overweight
cytes, the demand on local oxygen supply is category and increasing with severity of obesi-
inadequate, causing relative hypoxia. Adipose ty.117 In the United States, quality-adjusted life
tissue in obese mice has been demonstrated expectancy for 18-year-olds of normal weight
to be in a hypoxic state compared to that of (BMI = 18.5 to 24.9 kg/m2) has been shown to
lean mice.112,113 Hypoxia-inducible factor-1 vary from a high of 54 years to a low of 48.2
(HIF-1), a transcription factor activated years for those with Class 2 and Class 3 obe-
under low oxygen conditions, binds to mul- sity (BMI ≥ 35 kg/m2).118 Years of life lost are
tiple genes in the cis-acting hypoxic response greatest for younger individuals, from 8.4 years
elements responsible for glucose metabolism, in men aged 20 to 39 years who are severely
angiogenesis, inflammation, cellular stress, obese compared to normal-weight men versus
extracellular matrix remodeling, and apop- 0.8 years lost in severely obese men aged 60
tosis.114 Furthermore, an increase in HIF-1α to 79 years, with similar results for women.119
transcription factor along with an increase in Not only is mortality increased with obe-
protein secretion of leptin and VEGF occurs sity, but so is disability. When disability is
when human preadipocytes are exposed to defined as limitations in activities of daily
hypoxic conditions.115 living (ADL), a gradation of increased disabil-
Macrophages in the non–fat cell compart- ity can be shown to occur as obesity severity
ment of adipose tissue also demonstrate gene increases.120 Cheng et al121 have documented
expression for HIF-1α.113,116 Under hypoxic both functional limitations (ie, limitations in
conditions, gene expression by macrophages mobility) and limitations in ADL in associa-
for TNF-α, IL-6, IL-1, MIF, and VEGF can all tion with obesity in those aged 60 years and
be demonstrated to be increased.113 With both older.121 From the National Health and Nutri-
macrophages and preadipocytes providing evi- tion Examination Survey (NHANES) data for
dence of increased cytokine production in the 1988 through 2004, the odds of impairment
hypoxic state, it is likely that hypoxia underlies increased with increasing obesity. However,
the inflammatory state seen in obesity. from 2005 to 2012 the odds stabilized for
Insulin resistance represents one of the major functional limitations and severe impairment
outcomes of this chronic inflammatory state.72 of ADL with a decline in the odds of mod-
erate impairment of ADL.122 Recent years,
therefore, have demonstrated some improve-
ment in the disability status of obese people.

109
5 Obesity, Metabolic Syndrome, and Oral Health

However, cross-sectional studies in Europe of analysis of 100,000 individuals identified


those aged between 50 and 75 years document hypertension, dyslipidemia, and osteoarthritis
a decline in the proportion of the population costs of $18 million or more each per year.138
expressing a sense of having good health from As of 2010, interventions to prevent obe-
81% for those at normal BMI to 53% for those sity through medication or physician/dietician
with Class 2 obesity, while the proportion of counseling were estimated to reduce health care
people without chronic disease declined from expenditures in the United States by as much as
62% and 65% in normal-weight men and $2 billion.139 Clearly, a major effort to reduce
women to 26% and 32%, respectively, in obesity is important to health economics.
men and women with Class 2 obesity.123 This
suggests that although the degree of disability
associated with obesity appears to be stable, Metabolic syndrome
it nevertheless impairs the sense of well-being Metabolic syndrome, a cluster of risk factors
and disease-free life expectancy. associated with increased risk of diabetes mel-
Chronic disease in association with obesity litus and CVD, is commonly associated with
involves multiple organ systems.124 Common many of the disorders described earlier as
and significant comorbidities have included consequences of obesity.140 Diagnosis of this
CVD and diabetes,125,126 nonalcoholic fatty liver syndrome is somewhat controversial.141–143 In
disease,127–129 focal glomerulosclerosis,130,131 the United States, the National Cholesterol
obstructive sleep apnea,6,132 polycystic ovarian Education Program (NCEP) Adult Treatment
syndrome,133 and gastroesophageal reflux.134 Panel III (ATPIII) criteria have been com-
Cancer has also been implicated as a disorder monly used, requiring the presence of three
influenced by the presence of obesity.135,136 or more of the following five characteristics:
abdominal obesity, hypertension, elevated
fasting blood sugar, elevated triglycerides, and
Economic impact low high-density lipoprotein (HDL) choles-
Systematic review of international data demon- terol.144 Using the NCEP/ATPIII definition, the
strates that obese individuals incur 6% to 43% prevalence of metabolic syndrome from 2003
higher health care costs than individuals of nor- to 2012 in the United States was estimated
mal weight.137 In terms of BMI levels, those at 33%, with a higher prevalence in women
with BMI 30 or above incurred 30% higher than in men.145 Both the International Diabe-
costs than those with BMI 25 and below.137 tes Federation and the National Heart, Lung,
In the United States and the United Kingdom, and Blood Institute with the American Heart
these costs have been estimated to be largely Association have considered WC a requisite
driven by a rise in prevalence of obesity with part of any definition.143 These differences can
an associated rise in the chronic comorbid con- be shown to identify different populations,
ditions of diabetes, heart disease, stroke, and particularly with respect to ethnic or racial
cancer.1 differences.143,146 Prevalence rates of macrovas-
One health system database, the Geisinger cular complications, however, increase under
system, reviewed electronic medical records all of these definitions. In addition to the major
and claims from 2004 through 2013, identi- criteria listed here, PPAR modulation and pro-
fying 21 obesity-related chronic conditions in thrombotic and proinflammatory states have
56,895 individuals.138 The incremental costs for been considered components of the syndrome,
a single condition varied from $160 per per- with the presence or absence of insulin resis-
son per year for angina to $1,663 per person tance affecting morbidity.147 Regardless of these
per year for a pulmonary embolus. Population variations in definition, metabolic syndrome

110
Obesity and the Oral Cavity

has been associated with increased left ventric- Periodontal disease and obesity
ular mass and left ventricular hypertrophy and
an increased risk of having a cardiovascular Obesity and periodontitis are conditions asso-
event and developing diabetes.146 Hence, the ciated with local and systemic inflammation.
presence of metabolic syndrome represents a Based on an understanding of the contribution
significant increased health risk. of obesity to the systemic inflammatory bur-
den and the importance of the inflammatory
response to the progression of periodontitis,
the relationship of obesity to periodontitis has
Obesity and the Oral Cavity been a focus of research.
There is evidence from animal models that
There is interest in the relationship of body obesity can affect the severity of periodontal
weight and obesity to oral diseases. This inter- destruction. In a study by Verzeletti et al,148
est derives from the recognized association of two groups of Wistar rats were established.
obesity and diabetes and the importance of the One was fed a regular diet and the other a
relationship between diabetes and oral health diet high in calories. After 90 days, silk lig-
(reviewed in chapter 6). The mechanism that is atures were placed in the crevice around the
central to the relationship between diabetes and maxillary second molars, and all animals were
periodontitis is enhanced local and systemic sacrificed 30 days later. More weight gain was
inflammation and production of inflammatory observed in the animals fed the diet high in cal-
mediators by adipocytes and macrophages in ories. Further, there was greater loss of alveolar
adipose tissue contributing to the inflammatory bone in the obese group. Similarly, in a study
burden. In addition, weight gain is often related by Cavagni et al,149 obese Wistar rats demon-
to food intake, and the intake of fermentable strated significantly greater bone loss than a
carbohydrates can have an effect on the denti- control group.
tion and other tissues in the oral cavity. Obesity
is also associated with sleep apnea, and dental
professionals are expanding their role in treat- Cross-sectional clinical studies
ing this disorder. Systematic reviews and meta-analyses have
The relationship between overweight and supported a positive association between the
obese status and different oral/dental diseases prevalence of periodontitis and overweight/
and disorders has been studied. The resulting obesity in both children and adults.74,150–152 The
research strongly suggests the need for dental majority of studies have been cross-sectional,
professionals to be aware of the association not longitudinal, in design; therefore, cause and
between oral disease and obesity, understand effect could not be determined. In these studies,
the basis of these links, and develop a pre- risk ratios and odds ratios (ORs) ranged from
ventive approach to reduce and manage the 1.35 to 1.81, with significant methodologic
general health effects associated with being heterogeneity among the studies. There were
overweight and obese. Further, oral health care inconsistencies in research design with regard
professionals need to consider a role in obesity to the indices of periodontal disease and cutoffs
management for their patients. They are well for the definitions of adiposity and periodon-
versed in delivering a nutrition message related tal disease.153 Furthermore, many studies did
to carbohydrate intake and diet activities and not consider important covariates and con-
should consider expanding their responsibility founders such as diabetes mellitus, smoking,
in regard to weight management for patients socioeconomic status (SES), hypertension, and
in their care. oral hygiene.

111
5 Obesity, Metabolic Syndrome, and Oral Health

Association between overweight/obesity and periodontitis in adults.

Study %
ID OR (95% CI) weight

Buhlin 2003 n = 96 4.54 (1.59–13.00) 9.58


Genco 2005 n = 12,367 1.45 (1.13–1.93) 21.18
Nshida 2005 n = 372 3.17 (1.79–5.61) 16.31
Saito 2005 n = 584 4.30 (2.10–8.90) 13.97
Kushiyama 2009 n = 1,070 1.09 (0.77–1.53) 29.09
Han 2009 (males) n = 96 2.50 (0.85–7.38) 9.27
Han 2009 (females) n = 102 1.47 (0.52–4.95) 9.70
Overall (I-squared = 73.0%; P < .001) 2.13 (1.40–3.26) 100.00
Note: Weights are from random effects analysis

Odds ratio 0.25


0.5 1 2 4 8 16

Fig 5-3  |  Overweight and obesity compared with normal weight (body mass index). (Adapted from Suvan et
al157 with permission.)

Regarding the biologic plausibility of the accepted clinical parameters and indices and
association between periodontitis and obesity, not on other variables such as level of oral
two explanations have been proposed. The first hygiene or number of missing teeth. A total
is an exaggerated inflammatory response. Adi- of 28 studies contributed to the calculation of
pocytes, macrophages, and other cell types in the OR of the association between periodontal
fat tissue produce a range of proinflammatory disease and obesity, which was 1.35 (95% CI:
cytokines, and the concentration of acute-phase 1.23–1.47). Stronger associations were found
proteins and the number of circulating leuko- between obesity and periodontal disease for
cytes is elevated in obesity.154 In support of this younger versus older adults, women versus
concept, elevated levels of TNF-α in gingival men, and those who did not smoke versus
crevicular fluid (GCF) have been observed smokers.
in obese patients.155 The other mechanism is A subsequent systematic review and
related to reduced sensitivity to insulin, which meta-analysis157 included 33 studies in the
is important to the linkage of obesity and the systematic review and 19 in the meta-analysis.
development of type 2 diabetes mellitus. With There was a statistically significant associa-
elevated circulating levels of proinflammatory tion between periodontitis and obesity (OR:
cytokines, blood levels of glucose would rise 1.81; CI: 1.42–2.30), overweight (OR: 1.27;
with increased production and accumulation CI: 1.06–1.151) and when overweight and
of advanced glycation end products, for a obese weight categories were combined (OR:
greater systemic inflammatory burden. This 2.13; CI: 1.40–3.26; Fig 5-3).157 Again, cause
mechanism has been associated with increased and effect could not be assessed due to the
periodontal pathology.156 cross-sectional nature of most studies.
Chaffee and Weston74 conducted a system- Examining individual studies helps to
atic review and meta-analysis of the association demonstrate the heterogeneity of the research
between chronic periodontitis and obesity. findings. Recognizing that cigarette smoking
They examined 554 citations and 70 studies is the most important environmental risk fac-
that met specific inclusion criteria. An empha- tor for periodontal disease, the relationship
sis was placed on reporting of periodontitis by between body weight (assessed by BMI) and

112
Obesity and the Oral Cavity

periodontitis in a group of nonsmoking older depths and obesity was observed; however, a
adults was examined.158 In a fully adjusted relationship was seen between obesity and
model, there was no significant association dental calculus. Unfortunately, while the larger
between BMI and periodontitis. Compared parent study was longitudinal in nature, the
to individuals with a BMI below 25, the rela- periodontal variables were only collected at
tive risk (RR) of more advanced periodontitis one time point.
for those individuals with a BMI between 25 Longitudinal data are also available from a
and 30 was 0.7 (95% CI: 0.6–0.9) and a BMI study by the Veterans Administration. An anal-
of above 30 was 1.1 (95% CI: 0.8–1.4). This ysis sought to determine whether changes in
study should be interpreted cautiously because body weight could predict the progression of
of the other risk factors that would occur in periodontitis.161 Increases in weight, WC, and
this population, including advanced age,158 as arm fat were related to changes in periodontal
well as a very broad definition of periodontitis. disease over a period of nearly 30 years. Pro-
gression of periodontal disease was defined as
an increase in probing depth greater than 3
Longitudinal studies mm or tooth loss associated with periodonti-
In a systematic review that analyzed longi- tis. Men were categorized at baseline as being
tudinal studies of obesity and periodontitis, normal weight (BMI 18.5–24.9 kg/m2), over-
overweight/obesity, weight gain, and increased weight (BMI 25–29.9 kg/m2), or obese (BMI
WC were identified as potential risk factors ≥ 30 kg/m2), and weight gain in these groups
for the development and/or worsening of was divided into tertiles.
periodontitis.159 In addition, studies with lon- In general, greater weight gain over time
ger follow-up times (> 20 years) were more was associated with greater progression of
likely to show a direct association between periodontitis. For men with normal weight
obesity and periodontitis, perhaps due to the at baseline, both the greatest weight gain and
longer exposure of the periodontium to the the greatest increase in WC were associated
bacterial biofilm with persistence of the inflam- with greater progression of periodontitis, as
matory response. Whether fat was distributed compared to those with the least weight gain
viscerally or generally affected the association; or a smaller increase in WC (Fig 5-4).161 An
individuals with visceral fat accumulation had increase in arm fat was associated with greater
a stronger association than those with high progression of periodontal disease in men in
BMI. This is consistent with the production the normal-weight group. Similar trends were
by abdominal adipose tissue of proinflamma- seen for obese men, but differences did not
tory adipocytokines such as TNF-α and IL-6.153 reach significance for this subgroup because
Similar conflicting results were reported for the number of patients was small. This is an
younger individuals. Using data from a longi- important contribution to the literature on
tudinal study,160 measures of obesity including oral changes related to excessive weight and
BMI, WC, and a history of being identified as confirms the association of weight gain and
obese or overweight in earlier examinations progression of periodontitis. The data suggest
were related to measures of periodontal dis- a concurrent increase in weight and severity of
ease, including bleeding following periodontal periodontal disease.
probing, the presence of calculus, and the num- Similar findings were reported in a study
ber of teeth with a probing depth greater than of the relationship of BMI at baseline to the
or equal to 5 mm. While there was an associa- development of periodontal disease during the
tion between gingival bleeding and obesity, this following 5 years.162 In a fully adjusted model,
relationship did not remain in a fully adjusted the hazard ratio of developing periodontitis for
model. No association between deeper probing men who were overweight (BMI 25.0–29.9)

113
5 Obesity, Metabolic Syndrome, and Oral Health

Tertile of weight change Tertile of waist circumference change Tertile of arm fat area change
18 – 18 – 18 –
16 – 16 – 16 –
Teeth with PPD event (no.)

* * **
14 – 14 – 14 –
12 – 12 – 12 –
10 – 10 – 10 –
8– 8– 8–
6– 6– 6–
≤ –0.05 kg/yr
4– > –0.05 to 0.19 kg/yr 4– ≤ 0.14 cm/yr
4– ≤ 0.25 cm2/yr
> 0.14 to 0.38 cm/yr > 0.25 to 0.5 cm2/yr
2– > 0.19 kg/yr
2– > 0.38 cm/yr 2– > 0.5 cm2/yr
0– 0– 0–















2 3 4 5 6 7 8 9









2 3 4 5 6 7 8 9 2 3 4 5 6 7 8 9
Examination no. Examination no. Examination no.

Fig 5-4 |  Mean cumulative number of teeth with probing pocket events in men who were normal weight at
baseline. (Adapted from Gorman et al161 with permission.)

was 1.30 (P < .001), and for men who were contrary to the generally accepted epidemio-
obese (BMI ≥ 30.0) the risk was 1.44 (P = logic finding that periodontal disease prevalence
.072). The risk for women was 1.70 (P < .01) is greater in men but may be partially explained
and 3.24 (P < .05), respectively. by the limitation of just using BMI as the mea-
It is becoming clear that it is important to surement of obesity, as previously discussed.
distinguish between different patterns of fat For example, males have more muscle mass
distribution. Body weight and BMI alone do and higher BMI, which may account for this
not describe body fat distribution or the RR study’s finding that there was no progression
for adverse effects of obesity. Furthermore, as in periodontitis severity for males.
mentioned by these authors, high BMI in men
is not necessarily indicative of excessive fat.163
Other measurements, such as WC and arm fat, Treatment effects
are more reliable markers for intra-abdominal The evidence of the modifying effect of obe-
visceral fat and upper body subcutaneous adi- sity on the treatment of periodontal disease is
posity. In a population-based cohort study inconclusive. In a study by Suvan et al,167 over-
of adults, investigators looked at the effects weight/obese patients with severe periodontitis
of abdominal and general adiposity on peri- were followed for 2 months after nonsurgical
odontal outcomes after 3 years and found periodontal therapy. Their combined data-
that abdominal obesity was associated with base consisted of 260 individuals ranging in
unfavorable periodontal outcomes.164 Their age from 27 to 77 years. They concluded that
findings corroborate other studies, which show obesity was an independent predictor of worse
that abdominal obesity is associated with met- periodontal treatment outcomes at 2 months,
abolic disturbances and increased risk for other measured by percentage of probing pocket
chronic diseases.165 depth greater than 4 mm (P < .012). How-
With regard to effect modification by sex, ever, they cautioned that interpretation of these
a 5-year population-based prospective study results is limited based on issues with study
found an association between obesity and design and sample size. Further, the magnitude
attachment loss only in women.166 This is of the difference was minimal.

114
Obesity and the Oral Cavity

In a study that examined the effects of scal- that both mediators were more closely associ-
ing and root planing (SRP) on the clinical ated with obesity than with periodontitis.
response and serum levels of adipocytokines A study of a panel of inflammatory mark-
in 24 obese patients with periodontitis and a ers/adipocytokines were evaluated in serum
control group of 24 nonobese patients with and GCF from individuals in four groups
periodontitis, obese patients had lower reduc- (with and without obesity, with and without
tions in probing depth after 6 months of SRP. periodontitis).171 Levels of resistin, adiponec-
In addition, the treatment did not affect the tin, leptin, TNF-α, and IL-6 were determined.
circulating levels of leptin and adiponectin in Both obesity and periodontal disease appeared
any group.168 to influence the concentration of the inflam-
In sum, a systematic review and meta-analysis matory mediators in serum and GCF. The
that included a total of 15 studies and 867 levels of adiponectin, which is considered to be
patients investigated the effect of overweight/ anti-inflammatory, were lower in the serum of
obesity on response to periodontal treatment. patients with periodontitis and were not influ-
While the evidence was determined to be of low enced by obesity. TNF-α levels were influenced
quality, the authors concluded that there was by both obesity and periodontitis. Higher levels
no difference in clinical periodontal outcomes of TNF-α were seen in obese patients versus
(clinical attachment level and probing depth) normal-weight patients when periodontitis was
but that there were significant differences in cir- not present. Periodontitis was associated with
culating levels of certain inflammatory markers higher levels of TNF-α. These data emphasize
(adiponectin and leptin) following treatment of that the effect of periodontitis and obesity on
overweight/obese patients and normal-weight local and systemic levels of inflammatory medi-
patients.169 ators is complex.171
Treatment effects can also be used to eval-
uate the influence of periodontal diseases on
Levels of inflammatory mediators local and systemic levels of inflammatory medi-
Levels of inflammatory mediators in biologic ators. The effect of conservative periodontal
fluids from patients with obesity and peri- therapy on circulating levels of inflammatory
odontitis can provide insight into the systemic mediators in obese and normal-weight indi-
and local inflammatory response. Progranulin viduals with periodontitis has been studied.172
(PGN) is a proinflammatory mediator that All individuals received a complete periodon-
has been associated with influx of polymor- tal examination, evaluation of the lipid profile,
phonuclear leukocytes and macrophages and and assessment of the levels of CRP, blood glu-
neovascularization in wound healing sites.170 cose, insulin, IL-6, TNF-α, and leptin. Insulin
CRP levels are closely linked to levels of PGN, resistance was also determined. All parameters
and elevations in BMI are associated with ele- were recorded prior to treatment and 3 months
vations in PGN. In one study, concentration of after SRP (which included extraction of hope-
PGN and CRP were evaluated in four groups less teeth) and a complete-mouth disinfection
of patients171: individuals who were not obese protocol that relied on subgingival irrigation
without periodontitis (group 1), obese but and rinsing with chlorhexidine.
without periodontitis (group 2), not obese with Both groups demonstrated a pronounced
periodontitis (group 3), and obese with peri- improvement in clinical parameters following
odontitis (group 4). The concentration of both treatment. The obese group demonstrated a
mediators was determined in serum and GCF. significant reduction in the concentration of
The concentrations of both mediators in serum circulating IL-6, TNF-α, and leptin following
and GCF were highest in group 4, followed by treatment, while the normal-weight individuals
group 2, group 3, and then group 1, indicating demonstrated a reduction in the concentration

115
5 Obesity, Metabolic Syndrome, and Oral Health

of IL-6. Further, insulin resistance decreased or periodontitis affecting the metabolic syn-
in the obese individuals. This was a small drome) could not be assessed due to a lack of
study, but it suggested that periodontal dis- longitudinal data. Further, metabolic syndrome
ease and periodontal inflammation can both includes a number of criteria that are risk fac-
adversely affect the concentration of inflam- tors for periodontitis, emphasizing the complex
matory mediators in blood as well as induce nature of these associations.
insulin resistance in patients with obesity and In a review article on metabolic syndrome
periodontitis. and periodontal disease, Lamster and Pagan175
Examining specific measures of systemic suggest that their association is the result of
inflammation has also provided evidence for systemic oxidative stress and a robust inflam-
the association of periodontitis and obesity. matory response. Furthermore, evidence
There is also preliminary evidence to sug- indicates that of the metabolic syndrome com-
gest that treatment of periodontal disease in ponents, dysglycemia and obesity are the most
patients with metabolic syndrome reduces CRP closely related to the risk of periodontitis, with
and white blood cell count and raises the level lipid levels and hypertension playing a lesser
of HDL.173 These findings suggest that patients role.
with metabolic syndrome should have a dental The relationship between metabolic syn-
evaluation, and treatment would be indicated drome and dental caries is less clear. In a review
if periodontitis is present. In addition, patients of the epidemiologic evidence for an associ-
presenting to the dental office with periodon- ation between obesity, metabolic disorders,
titis and signs and symptoms of metabolic and oral diseases, some studies have shown a
syndrome should be referred to a medical pro- positive relationship between metabolic syn-
vider for appropriate treatment. drome and caries and others have not shown
any association.153 It has been postulated that
hyperglycemia and reduced salivary flow are
Metabolic syndrome and the risk of factors associated with an increased risk of
periodontal disease dental caries in persons with these conditions.
As noted previously, metabolic syndrome is
a complex of clinical and physiologic alter-
ations that include excessive weight/obesity, Dental caries
insulin resistance, dyslipidemia, and elevated The expectation that dental caries and weight
blood pressure. Metabolic syndrome places should be correlated is based on the assump-
individuals at risk for development of diabe- tion, and in many cases the observation, that
tes mellitus and CVD. Periodontitis has been overweight individuals consume greater quan-
linked to a number of criteria that are part tities of high-caloric and cariogenic foods.
of metabolic syndrome. A systematic review Dietary patterns such as the frequency of
and meta-analysis including 20 studies exam- eating cariogenic foods have been shown to
ined the relationship of metabolic syndrome increase the risk for dental caries.176,177 For
to periodontitis.174 Most were cross-sectional young children in the primary dentition, there
in design and yielded an OR of 1.71 (95% CI: is a link between frequent consumption of
1.42–2.03). The OR increased to 2.09 (95% cariogenic foods and severe early childhood
CI: 1.28–3.44) when a more precise diag- caries (S-ECC).176 In a study that examined
nosis of metabolic syndrome was required. feeding practices and intake of sweets in Bra-
Approximately 40% of patients with metabolic zilian children aged younger than 12 months,
syndrome also presented with periodontitis. an association was found between caries inci-
However, the direction of this association dence in early childhood (38 months) and diet
(metabolic syndrome affecting periodontitis, (RR = 1.46–1.55).178 In another cross-sectional

116
Obesity and the Oral Cavity

study, it was reported that children with S-ECC cross-sectional Kuwaiti study of 8,284 chil-
(average age 3.6 years) had significantly higher dren aged 9 to 11 years reported a statistically
BMI scores than their caries-free peers.179 significant decrease in the prevalence of caries
In older children, however, studies of the in permanent teeth with increasing BMI-for-age
relationship between dental caries and BMI categories.187 A more recent cross-sectional
are not conclusive.180,181 A systematic review study of 8,000 fourth- and fifth-grade Kuwaiti
of the literature in 2012 examined 49 papers children concluded that the percentage of chil-
that were published between 2004 and dren with decayed or filled teeth was negatively
2011. The authors reported that the studies correlated with BMI.188
fell into three categories with respect to the Several cross-sectional studies have examined
association between BMI and dental caries: the association between dental caries and obe-
23 studies found no association, 17 found a sity solely in adolescent populations. A study
positive relationship, and 9 found that caries conducted in India examined 2,000 children
decreased with increasing BMI.180 Another aged 12 to 15 years who were normal weight
systematic review and meta-analysis, which (average BMI of 20.8) and overweight (average
included 14 papers, found a significant rela- BMI of 26.9) and concluded that there was
tionship between obesity and caries but not not a significant difference in the mean num-
when the permanent and primary dentitions ber of decayed, missing or filled teeth (DMFT)
were analyzed separately. Furthermore, obesity between groups (3.4 versus 3.9).189 Another
and caries were positively correlated in children study of Indian school children aged 11 to
from developed countries but not in develop- 14 years looked at the relationship between
ing countries, suggesting that socioeconomic BMI, caries, and SES. They found that in the
factors confound the association.182 high SES category, there were fewer caries in
Examining specific studies, there are reports overweight children than in normal-weight
in the literature that demonstrate a posi- children.190
tive association between obesity and dental Larger and more detailed studies in the
caries. Evaluating a cohort of 1,160 Mexi- United States using national surveys have
can children aged 4 to 5 years, a significant failed to find an association between body
association was found between caries in the weight and caries. Analyses of oral health and
primary dentition and being overweight or body measurement data from the 1999–2002
at-risk for being overweight (now referred to as NHANES indicated that for 6,000 children
obese and overweight).183 A prospective study aged between 2 and 17 years, there was no
of 403 Swedish 15-year-olds who had been statistically significant association between
followed since the age of 1 year found that caries and BMI-for-age percentile in the
snacking habits reported in early childhood primary or the permanent dentition after
were associated with increased prevalence of controlling for age, sex, race/ethnicity, and
interproximal caries.184 In a German study of poverty status. Furthermore, of all children
2,071 primary-school pupils, a significant asso- with caries, those who were overweight had
ciation between caries prevalence and BMI was fewer DMFT than their normal-weight coun-
found and remained significant after adjusting terparts.191 A later study based on the same
for age.185 NHANES database examined over 1,500 2- to
There are also several well-designed stud- 6-year-olds and found no significant associ-
ies that have shown an inverse relationship ation between overweight and caries.192 A
between obesity and caries. In a study of third study examined data from NHANES III
1,003 Iranian children, a statistically signifi- (1988–1994) for 10,180 children and from
cant positive association was found between NHANES 1999–2000 for 7,568 children. In
increased BMI and being caries free.186 A both surveys, for children aged 2 to 5 years

117
5 Obesity, Metabolic Syndrome, and Oral Health

there were no significant differences in caries two-thirds of individuals between the ages of
prevalence by weight categories. However, in 2 and 19 consume at least one SSB per day,
NHANES III, overweight children aged 6 to which is approximately 7% of the total daily
18 years were less likely to have caries than caloric intake.196 Among adults, approximately
the normal-weight group.193 50% consume at least one SSB daily, with
Clearly, there are conflicting findings in the young adults having the highest mean intake
literature with respect to caries and weight sta- and percentage of daily calories from SSBs.197
tus, suggesting that this relationship is complex Several studies have noted that there is an
and affected by a multitude of factors. Diet, association between consumption of SSBs and
eating patterns and frequency, SES, race/eth- obesity.54,198–201 Many systematic reviews, includ-
nicity, and psychosocial factors are important ing most recently a review of the association
variables that are examined in the literature. between SCBs and weight gain among children
Systematic reviews note the inconsistencies in aged younger than 12 years, have shown a sta-
the measures of weight status194 as a possible tistically significant positive association between
cause of the variation in findings. consumption and adiposity.202,203 However, in
More research is needed to address the con- a study based on data from NHANES 1999–
flicting findings in the relationship between 2004, 100% fruit juice intake was not shown to
caries and weight status. The factors that be associated with early childhood caries, and it
account for weight gain may or may not is suggested that parents should encourage other
account for caries prevalence across different healthy eating behaviors.204
populations and age groups. For example, Despite the link with obesity and the assump-
physiologic factors such as a reduced flow of tion of a relationship between SSB and caries,
whole saliva in obese children have been sug- there is a paucity of evidence linking SSB con-
gested as an explanation for increased caries in sumption and dental disease (eg, caries, tooth
these individuals.195 Identifying common risk loss, periodontitis). This may be due to the
factors associated with increased weight would multitude of modifying factors (eg, saliva flow
suggest approaches to implement health pro- and constituents, frequency of sugar intake,
motion and disease prevention for two chronic fluoride exposure, oral hygiene practices). A
conditions. Further, exploring the reasons for 2001 cross-sectional study of NHANES data
differences between groups may allow investi- that examined sugared soda consumption and
gators to develop innovative ways to decrease caries found that there were significant associ-
the negative effects of obesity and caries. ations between SSB consumption and DMFT
scores in individuals older than 25 years but
not in younger individuals.205 An in-depth sys-
Sugar-sweetened beverages tematic review in 2014, used to inform World
No discussion of the obesity epidemic, diet, Health Organization guidelines that recom-
and oral disease would be complete without mend restriction of free sugars to less than
mention of the impact of SSB consumption 10% of energy consumption, concluded that
on weight gain. SSBs are defined as soda, fruit there was moderate evidence to support a rela-
drinks, sports and energy drinks, presweetened tionship between amount of sugar consumed
coffees and teas, and other sugar-sweetened and caries, particularly in children.206
drinks. SSBs do not include diet sodas or 100% A few research reports have looked at this
fruit juice. Sugar-containing beverages (SCBs) longitudinally. In a 4-year prospective study
include both SSBs and 100% fruit juice. of the relationship between SSBs and caries
The most recent report from the US Centers in Finnish adults, it was found that despite
for Disease Control and Prevention on SSB con- individuals’ sociodemographic characteris-
sumption among US youth indicates that almost tics, adults drinking one to two and three or

118
Obesity and the Oral Cavity

more SSBs daily had 31% and 33% greater Tooth eruption and tooth
DMFT than those who did not drink SSBs,
movement
respectively.207 With regard to tooth loss
and SSB consumption, only a limited num- Evidence suggests that high body fat con-
ber of published studies are available. In a tent affects hormonal metabolism, skeletal
cross-sectional assessment of young adults growth, and onset of puberty. A longitudinal
aged 18 to 39 years, a positive association study demonstrated that higher BMI gain in
was found between SSB consumption and childhood is associated with an earlier onset
tooth loss. This relationship was observed of puberty.214,215 It appears that although early
even for those who consumed fewer than one puberty and increased height are associated
SSB per day.208 Another cross-sectional study with obesity, once puberty has been reached,
analyzed the relationship between diabetes, final adult height is similar to normal-weight
SSBs, and tooth loss. Using self-reported data children.215–217
from the Behavioral Risk Factor Surveillance Given obesity’s association with accelerated
System, the authors concluded that among skeletal growth, it is possible that hormonal
adults with diabetes mellitus, consuming two changes produced by obesity could mod-
or more SSBs per day was associated with hav- ify the timing of tooth eruption.218 Many
ing had six or more teeth extracted (OR: 1.46, cross-sectional studies have shown that over-
P < .0001).49 weight or obese children have accelerated dental
In response to the wide availability and development, which can have implications
aggressive marketing of SSBs and evidence for caries risk and orthodontic treatment.219
suggesting that these beverages contribute to For example, in a longitudinal study of 110
the obesity epidemic, many countries have Mexican schoolchildren, overweight children
implemented taxes on SSBs, including Den- had fewer carious teeth and more erupted per-
mark, France, Hungary, Ireland, Mexico, manent teeth than normal-weight children (at
Norway, and the United Kingdom. Policy-level aged 11 years, the children in the overweight
efforts have recently been initiated in several group had an average of five more permanent
US cities to tax SSBs as a primary preventive teeth than normal-weight children).220
method to control consumption.209,210 To date, In a study by Hilgers et al221 on childhood
several US cities, including Berkeley, Philadel- obesity and dental development, the dental ages
phia, San Francisco, Oakland, Boulder, and for 104 children were determined by subtract-
Seattle, have introduced taxes on SSBs.211 A ing each subject’s chronologic age from the
systematic review of the effect of taxes on calculated dental age such that a positive value
SSB consumption in middle-income countries reflected accelerated dental development and a
concluded that the taxes will reduce intake negative value reflected delayed development.
and prevent an increase in obesity prevalence, When analyzed by BMI-for-age percentile, the
but for permanent change to occur other obe- mean difference in dental age was 0.63 ± 1.31
sity interventions need to be implemented as years for normal-weight subjects, 1.51 ± 1.22
well.212 A study of the effect of SSBs on caries years for overweight subjects, and 1.53 ± 1.28
and treatment costs concluded that taxes on years for obese subjects. After adjusting for
SSBs would reduce caries and dental treatment age and sex, the authors concluded that dental
costs, especially for younger individuals and development was associated with higher BMI
those who have low income.213 percentile.221
In a cross-sectional study of 100 children
aged 8 to 12 years, Zangouei-Booshehri et
al222 studied the relationship between BMI
and dental age. Using the international BMI

119
5 Obesity, Metabolic Syndrome, and Oral Health

standards and collapsing the categories into


normal and above-normal weight groups, 30 –

Median number of permanent teeth erupted


they found that the mean eruptive age was 9.5
25 –
years for the children in the normal BMI group
and 10.0 years for the 15% of children in the 20 –
above-normal BMI group. They concluded that
15 –
increased BMI is associated with increased den-
tal eruptive age.222 10 –
In a study of 540 adolescent orthodontic
patients aged 8 to 17 years, BMI percentile, 5–
Nonobese
skeletal maturation, and dental age were Obese
0–
assessed via retrospective chart review.223 Lin-










ear and logistic regression models assessed

5–
6–
7–
8–
9–
10
11 .9
12 .9
13 2.9
–1
–1
–1
–1
5.9
6.9
7.9
8.9
9.9
the effect of weight on dental age and cervical

0
1

3.9
vertebral stage (used to assess skeletal matu- Age (y)
ration). The results demonstrated that cervical Fig 5-5 |  Median number of teeth erupted by age
vertebral stage and dental age were slightly but category and obesity status. (Reprinted from Must et
significantly greater in subjects with higher BMI al225 with permission.)
percentiles. The coefficient for the BMI percen-
tile was 0.005 year per one unit increase in
dental age (P < .001), and the OR for the effect reflect the US population, obese children aged
of the BMI percentile on the cervical vertebral 5 to 14 years had a higher average number of
method was 1.02 (P < .0001). The impact of erupted teeth during the mixed dentition period
obesity on the accuracy of determining dental than normal-weight peers. Analyses of 5,434
age has significant clinical implications for the participants found that, on average, obese chil-
timing of orthodontic treatment. dren had 1.44 more erupted permanent teeth
Looking at percent body fat as another than nonobese children after adjusting for age,
measure of obesity, a study calculated BMI sex, and race/ethnicity225 (Fig 5-5). The acceler-
percentile, used DXA to determine body ation of tooth eruption in obese children may
fat analysis, and assessed skeletal and den- also affect the sequence of tooth emergence,
tal age for 107 children. The difference with implications for orthodontic treatment.
between chronologic and skeletal-dental age The correlation between BMI-for-age percen-
was statistically significant for preobese and tile and the sequence of tooth eruption was
obese children but not for underweight and examined in a cross-sectional sample of 8,292
normal-weight children. The results demon- Kuwaiti children aged 9 to 11 years. Using
strated that, in addition to BMI percentile, the BMI percentiles, 29% of the children
there is an association between accelerated were obese and 18% were overweight. When
skeletal-dental age and percentage of body fat compared to the 53% normal/underweight
as measured by DXA.224 children, the obese and overweight children
The aforementioned studies were carried had significantly more permanent teeth, adjust-
out on convenience samples, opening the pos- ing for age and sex. The overweight and obese
sibility of biased or nongeneralizable findings. groups had an average of 1.2 and 2.7 more
However, a study based on data from three erupted teeth than the normal-weight group,
NHANES databases from 2001 to 2006 also respectively. Moreover, this study demonstrated
found that obesity was significantly associated that there was a statistically significant differ-
with tooth eruption in children. In this national ence in mean number of erupted teeth between
proportional probability sample designed to each discrete BMI category.187

120
Obesity and the Oral Cavity

Mechanisms to explain accelerated skele- disease relationship. 232,233 If masticatory


tal maturation (and early puberty) in obese function is compromised, individuals may
children appear to involve control of skeletal avoid hard-to-chew foods that contain fiber
maturation and turnover. In the literature, it and instead choose processed foods or foods
has been suggested that leptin, a hormone that are rich in carbohydrates, with only limited
produced by adipose tissue, can stimulate amounts of fiber, protein, and other important
skeletal growth and accelerate pubertal devel- nutrients. The result may be a greater caloric
opment.226,227 Alternatively, leptin might act intake than what is expended, resulting in
directly on the skeletal growth centers. Leptin weight gain.
receptors have been found in the cartilaginous Masticatory efficiency has been assessed
growth centers involved in skeletal matura- in patients with a range of dental conditions.
tion. Therefore, an obese subject may have a Participants chewed a test cylinder of silicone
mechanism of central resistance to leptin and rubber, and particle size was evaluated after 20
an increased sensitivity to leptin peripherally, chewing cycles. Swallowing trigger was also
causing increased differentiation and prolifer- determined, participants were questioned about
ation of chondrocytes, resulting in precocious their ability to chew, and an oral examination
skeletal maturation.228 Leptin was strongly was conducted to determine the number of
associated with fat mass as well as loss of eat- occluding teeth. Participants’ weights were
ing control in a study of 7- to 18-year-olds.229 assessed as BMI.
The concentration of leptin in saliva was sig- Increased BMI was associated with fewer than
nificantly higher in obese children compared 10 occluding pairs of teeth. Similarly, reduced
to nonobese children.187 masticatory function and swallowing trigger
Early tooth eruption and skeletal matura- were associated with a higher BMI. However,
tion are important considerations for pediatric eating habits and masticatory function are com-
dentists and orthodontists. When considering plex processes, and the authors emphasized
orthodontic therapy, knowledge of the stage the importance of a multidisciplinary health
of skeletal development is essential because care approach to management of overweight
treatment is often initiated during the early and obese patients. They suggest that dentists
mixed dentition to redirect growth in a more can play different roles in this effort, including
favorable manner.218 A prospective cohort counseling patients about the health hazards
study found that the number of paired teeth associated with excessive weight and the impor-
at 15 months was correlated with obesity at tance of healthy eating, as well as the need
17 years, suggesting that early primary tooth to restore masticatory function to enhance a
eruption may be an unrecognized indicator for patient’s ability to eat a healthy diet. This should
the development of obesity.230 Furthermore, be accompanied by dietary guidance.
obese orthodontic patients had a significantly Tooth loss can have several causes, including
higher rate of tooth movement at the beginning dental caries, periodontal disease, and trauma.
of orthodontic therapy than normal-weight A systematic review found a bidirectional
controls, which was associated with increased association between obesity and tooth loss.234
levels of inflammatory markers (eg, leptin and Because the studies included in the review were
resistin) in GCF.231 cross-sectional in nature, causality could not be
determined. Further longitudinal studies are
necessary to assess the direction of the associ-
Masticatory efficiency ation (ie, whether edentulism leads to obesity
The relationship between body weight or obesity leads to edentulism). In a 22-year
and masticatory efficiency may be an longitudinal study of the association of obesity
important determinant of the obesity–oral and edentulism in Swedish individuals aged 55

121
5 Obesity, Metabolic Syndrome, and Oral Health

to 84 years, the authors found a strong asso- associated with severity of OSA and may be
ciation that was more apparent in women.235 linked to sleep deprivation and activation of the
sympathetic nervous system.245 A reciprocal or
bidirectional relationship between OSA and obe-
Obesity and Obstructive sity has been described by Ong et al,244 in which
obesity may lead to OSA and OSA can have an
Sleep Apnea impact on body weight. OSA causes changes in
energy expenditure during sleep and wakeful-
Obesity, as measured by BMI, neck circumfer- ness, increases preference for energy-dense food,
ence, and waist-to-hip ratio, is the strongest alters hormonal regulation specific to appetite
risk factor for the common sleep disorder and satiety, and affects sleep duration and qual-
known as obstructive sleep apnea (OSA).236 ity, which may affect daytime physical activity
More than 70% of individuals with OSA are and increase lethargy and sleepiness.
clinically obese based on their BMI, and prev- In addition to adults, children and adoles-
alence has increased with the global rise in cents are also affected by OSA. The primary
obesity.237 Sleep apnea is characterized by com- risk factor for OSA in children is adenotonsillar
plete or partial obstruction of the upper airway hypertrophy, but underlying craniofacial anat-
with associated respiratory effort and snoring. omy and obesity are also significant risk factors.
These recurrent episodes of shallow or paused The prevalence of OSA in children has been
breathing during sleep result in reduced blood estimated at anywhere between 2% and 20%,
oxygen saturation and significant comorbidi- with obese children having a prevalence as high
ties (eg, daytime sleepiness, CVD, hypertension, as 60%.242,246 Kohler et al247 found that among
and neurocognitive deficits).238 The etiology of adolescents, there was a 3.5-fold increase in
OSA is complex, and in addition to obesity OSA risk with each standard-deviation increase
there are many established risk factors, includ- in BMI percentile. Studies have demonstrated a
ing underlying craniofacial abnormalities (eg, relationship between OSA, inflammation, and
retrognathia, micrognathia), hyoid bone posi- insulin resistance in obese and nonobese chil-
tion, nasal anatomy, and macroglossia.239 dren.248,249 Using imaging techniques, a strong
The prevalence of OSA in the US popula- relationship was found between visceral adi-
tion has increased almost 15-fold from 1993 posity and OSA, independent of BMI.250 The
to 2010.240 The Wisconsin Sleep Cohort, an authors concluded that the quantity and loca-
ongoing longitudinal study of sleep disorders, tion of adipose tissue was a factor that might
estimated the prevalence of OSA to be 9% for explain why some obese children do or do not
women and 17% for men between the ages of develop OSA.
50 to 70 years.241 Although measurement stan- Traditional nonsurgical therapies for OSA
dards and levels of apnea differ among studies, include weight loss and continuous positive
it is estimated that approximately one of every airway pressure (CPAP) in the upper airway
five adults in the United States has mild OSA during sleep. An increasingly popular and more
and one of every 15 adults has moderate OSA.242 tolerable treatment option is oral appliance
Predictors of an increase in OSA include male therapy (OAT), which involves the fabrication
sex, high BMI, increase in waist-to-hip propor- of removable mandibular repositioning appli-
tion, and increase in serum cholesterol.243 ances.251 These appliances increase the upper
Obesity appears to affect control of the upper airway volume/opening by bringing the tongue
airway by alterations in upper airway structure forward as the mandible is advanced anteriorly.
and function, reductions in resting lung volume, A recent study suggested that screening for OSA
and negative effects on respiratory drive and in a dental office was well accepted by patients
load compensation.244 Insulin resistance is also and encouraged further medical evaluation.252

122
Conclusion

A recent systematic review of the effective- Medicine of the National Academy of Sciences
ness of mandibular advancement appliances issued a report titled, “Accelerating Progress
demonstrated a reduction in the severity of in Obesity Prevention: Solving the Weight of
OSA and an increase in oxygen saturation.253 the Nation,”261 which examined strategies and
In a small crossover study, two different appli- made recommendations for the future. The
ances were shown to be effective in improving recommendations included an emphasis on
symptoms in patients affected by mild to exercise and physical activity, the importance
moderate OSA.254,255 In addition, use of a man- of availability of healthy foods and beverages,
dibular advancement appliance was associated an emphasis on obesity management as an
with improvement in measures of quality of important part of the work environment, and
life for both patients and their bed partners.256 the important role that schools should play by
A position paper from the Canadian Sleep focusing attention of younger individuals on
Society emphasized the importance of a team health and healthy living.
approach for caring for patients with OSA.257 The relationships between obesity and
Clinical care requires that a physician should specific oral diseases and disorders (peri-
make the diagnosis and that OAT provided by odontal disease, dental caries, and early tooth
dentists should be considered for appropriate eruption) are biologically plausible. The rela-
patients who request treatment for snoring tionship between obesity and periodontitis
with or without OSA. Ideally, patients who are has attracted considerable attention. When
considered candidates for OAT should have a considering causation, it was concluded that
minimum of 10 healthy, well-supported, and the cross-sectional nature of the majority of
distributed teeth and no temporomandibular studies did not allow for determination of
pain or pathology. In addition, this appliance cause and effect.74 Suvan et al157 suggested
is recommended for patients who fall in the that proinflammatory mechanisms associated
mild to moderate category of OSA only. It is with obesity (eg, increased local inflammatory
recommended that dentists who fabricate these response, reduced insulin sensitivity, elevated
appliances have advanced training in dental blood glucose levels leading to greater accu-
sleep medicine. mulation of advanced glycation end products)
Dentists can play an active role in identi- can exacerbate periodontitis. However, they
fying children and adults with possible OSA note that many confounders, including smok-
and referring them for assessment.258 Early ing and diet, can influence both obesity and
detection, referral, and coordinated care with periodontitis. A 5-year longitudinal study of
a patient’s medical provider can prevent addi- weight gain and progression of periodontitis
tional consequences of OSA and improve suggested that weight gain and periodontal dis-
quality of life.259 Long-term follow-up care ease progression occurred concurrently.161 The
and assessment of treatment outcomes, includ- relationship of obesity and periodontal disease
ing polysomnography, can be provided by an can be considered in the context of diabetes
interdisciplinary team that includes dentists mellitus and periodontitis. That relationship is
and physicians.257,260 bidirectional, but the data supporting diabetes
mellitus affecting periodontitis is stronger than
the data supporting the converse.
Analysis of the causal relationship between
Conclusion obesity and other diseases, including peri-
odontitis, has been the subject of conceptual
Obesity is now considered one of the most analysis. Two separate reports failed to find
important health problems in the United States such a causal relationship for obesity and
and across the globe. In 2012, the Institute of periodontitis.262,263 The review of the literature

123
5 Obesity, Metabolic Syndrome, and Oral Health

I. Complete medical II. Complete oral III. Oral health


health history health history examination

Follow-up care
plan

Are
expanded
Dental treatment NO services indicated from
plan the health histories and oral
examination?

YES

Provider referrals:
physicians, nurse Health promotion Health screening
practitioner, activities: tobacco activities: HbA1c,
psychologist, social cessation, dietary HIV, hepatitis C,
worker, diabetic counseling, disease- hypertension
educators, tobacco related education
quit lines

Fig 5-6  |  Integrated oral health practice patient visit. (Reprinted from Myers-Wright and Lamster265 with per-
mission.)

included here emphasized the heterogeneous patients regarding their diet and caloric intake.
nature of the studies, which were primarily It has been proposed that oral health care pro-
cross-sectional and differed in terms of mea- viders take an active role in obesity prevention
sures of periodontitis and definitions of obesity. and treatment266 (see Fig 5-6). The frequency
These reports do not diminish the importance of dental visits for a significant segment of the
of the call for greater involvement of oral population as well as oral health care provid-
health care professionals in the management ers’ understanding of an appropriate diet that
of obesity. emphasizes reduced caloric intake make this
Oral health care professionals can make a logical extension of dental practice. Treat-
important contributions in different ways264 ment of obesity requires an interprofessional
(Fig 5-6).265 Dental treatment in general and approach that includes physicians and other
treatment of periodontitis in particular may health care providers, including dieticians.265
help patients retain their dentition and thereby In the future, dentists and dental hygien-
maintain a healthy diet. Further, periodontal ists should consider providing a broader
therapy can help to reduce the systemic inflam- health message to their patients, including the
matory burden. In addition to treating oral importance of a healthy lifestyle, exercise, diet
disease and developing a preventive regimen control, smoking cessation (if necessary), and
to avoid future disease, dental providers may weight management. This new set of responsi-
fabricate appliances to treat OSA. In the larger bilities will require oral health care providers
sense, oral health care providers can advise to receive specific training and will value the

124
Clinical Considerations

importance of interprofessional practice.267 The with obesity, being overweight is not a failure
result will be closer alignment of oral health of willpower, and shaming and stigmatizing
care providers with other health care providers them does not help them lose weight. Evidence
and ultimately improved oral health and health suggests that health care providers often have
outcomes. implicit bias toward people who are obese.268,269
This chapter has reviewed the impact of obe- As oral health care providers, together with
sity/overweight on diseases of the oral cavity. our medical colleagues, we have the ability to
These associations must be included in the diagnose and treat many oral conditions that
broader discussion of obesity and health. Much are affected by obesity. After all diagnostic and
remains unknown about obesity. We know that clinical data is collected and treatment is ren-
there are overweight/obese individuals who are dered, all health care providers must deliver
healthy and underweight and normal-weight compassionate and comprehensive care to all
individuals who are not healthy. Health care patients, regardless of their weight, with respect
providers must realize that for many people and understanding.

Clinical Considerations: What You Can Take Back to Your Practice


Is there biologic plausibility for an association range of foods without the need to limit the
between oral infections and obesity?  Obesity diet to softer, carbohydrate-rich foods. In the
is an inflammatory condition. Adipose tissue larger context, oral health care providers can
contains both adipocytes and increased num- deliver a healthy lifestyle message to dental pa-
bers of macrophages, which produce inflamma- tients, including the need to eat a healthy diet,
tory mediators that contribute to the systemic exercise, and control weight.
inflammatory burden and can exacerbate the
local inflammatory response in the periodon- Is it safe to provide dental care to patients who
tal tissues. Further, obesity is an important risk are obese?  Being overweight or obese is asso-
factor for type 2 diabetes mellitus, which is a ciated with a number of important comorbid-
recognized risk factor for periodontitis. ities (eg, hypertension, cardiovascular disease).
In the absence of these comorbidities, it is safe
Are oral infections and disorders independent to provide dental services to overweight and
risk factors for the development of obesity?  obese patients.
Though not fully addressed in the literature, it
is not biologically plausible that oral infections What should a dental practitioner tell a patient
directly increase the risk of obesity. about the association between oral infections
and obesity?  The relationship between oral
Can treatment of oral infections and disorders health and obesity is complex. Obesity may ad-
reduce the risk for the development of obesity?  versely affect periodontal health, and a healthy
The provision of dental services, either preven- mouth will allow mastication of a full range of
tive or therapeutic (treatment of dental caries foods and thereby a healthy diet. An important
or periodontal disease) does not directly con- message that can be delivered by oral health
tribute to reducing obesity. However, treatment care providers is that living a healthy lifestyle,
of dental disease, elimination of pain, and es- including not smoking, exercising, controlling
tablishment of a functional dentition can im- weight, and maintaining oral health, will de-
prove mastication and allow patients to eat a crease the risk of many chronic diseases.

125
5 Obesity, Metabolic Syndrome, and Oral Health

References 15. Siervo M, Jebb SA. Body composition assessment:


Theory into practice: Introduction of multicompartment
models. IEEE Eng Med Biol Mag 2010;29:48–59.
1. Wang YC, McPherson K, Marsh T, Gortmaker SL, 16. Müller MJ, Braun W, Enderle J, Bosy-Westphal A.
Brown M. Health and economic burden of the Beyond BMI: Conceptual issues related to overweight
projected obesity trends in the USA and the UK. and obese patients. Obes Facts 2016;9:193–205.
Lancet 2011;378:815–825. 17. Obesity: Preventing and managing the global epidemic.
2. Must A, Spadano J, Coakley EH, Field AE, Colditz G, Report of a WHO consultation [editorial]. World Health
Dietz WH. The disease burden associated with over- Organ Tech Rep Ser 2000;894:i–xii,1–253.
weight and obesity. JAMA 1999; 282: 1523–1529. 18. Seidell JC, Flegal KM. Assessing obesity: Classification
3. Zhang Y, Liu J, Yao J, et al. Obesity: Pathophysiology and epidemiology. Br Med Bull 1997;53:238–252.
and intervention. Nutrients 2014;6:5153–5183. 19. Mei Z, Grummer-Strawn LM, Pietrobelli A, Goulding A,
4. Poirier P, Giles TD, Bray GA, et al. Obesity and cardio- Goran MI, Dietz WH. Validity of body mass index
vascular disease: Pathophysiology, evaluation, and compared with other body-composition screening
effect of weight loss: An update of the 1997 American indexes for the assessment of body fatness in children
Heart Association Scientific Statement on Obesity and and adolescents. Am J Clin Nutr 2002;75:978–985.
Heart Disease from the Obesity Committee of the Coun- 20. Kelly AS, Barlow SE, Rao G, et al. Severe obesity in
cil on Nutrition, Physical Activity, and Metabolism. children and adolescents: Identification, associated
Circulation 2006;113:898–918. health risks, and treatment approaches: A scientific
5. Bastien M, Poirier P, Lemieux I, Després JP. Overview statement from the American Heart Association. Circu-
of epidemiology and contribution of obesity to cardio- lation 2013;128:1689–1712.
vascular disease. Prog Cardiovasc Dis 2014; 56: 21. Barreira TV, Staiano AE, Harrington DM, et al. Anthro-
369–381. pometric correlates of total body fat, abdominal
6. Peppard PE, Hagen EW. The last 25 years of obstructive adiposity, and cardiovascular disease risk factors in a
sleep apnea epidemiology-and the next 25? Am J Respir biracial sample of men and women. Mayo Clin Proc
Crit Care Med 2018;197:310–312. 2012;87:452–460.
7. Kulkarni K, Karssiens T, Kumar V, Pandit H. Obesity and 22. Hübers M, Geisler C, Plachta-Danielzik S, Müller MJ.
osteoarthritis. Maturitas 2016;89:22–28. Association between individual fat depots and
8. VanWagner LB, Khan SS, Ning H, et al. Body mass cardio-metabolic traits in normal- and overweight chil-
index trajectories in young adulthood predict dren, adolescents and adults. Nutr Diabetes 2017;7:
non-alcoholic fatty liver disease in middle age: The e267.
CARDIA cohort study. Liver Int 2018;38:706–714. 23. Camhi SM, Bray GA, Bouchard C, et al. The relationship
9. Flegal KM, Kit BK, Orpana H, Graubard BI. Association of waist circumference and BMI to visceral, subcutane-
of all-cause mortality with overweight and obesity using ous, and total body fat: Sex and race differences.
standard body mass index categories: A systematic Obesity (Silver Spring) 2011;19:402–408.
review and meta-analysis. JAMA 2013;309:71–82. 24. Kangas S, Timonen P, Knuuttila M, Jula A, Ylöstalo P,
10. NCD Risk Factor Collaboration. Worldwide trends in Syrjälä AH. Waist circumference and waist-to-height
body-mass index, underweight, overweight, and obesity ratio are associated with periodontal pocketing—Results
from 1975 to 2016: A pooled analysis of 2416 of the Health 2000 Survey. BMC Oral Health 2017; 17:
population-based measurement studies in 128.9 million 48.
children, adolescents, and adults. Lancet 2017;390: 25. Hales CM, Carroll MD, Fryar CD, Ogden CL. Prevalence
2627–2642. of obesity among adults and youth: United States,
11. Ng M, Fleming T, Robinson M, et al. Global, regional, 2015–2016. NCHS Data Brief 2017;(288):1–8.
and national prevalence of overweight and obesity in 26. Maes HH, Neale MC, Eaves LJ. Genetic and environ-
children and adults during 1980–2013: A systematic mental factors in relative body weight and human
analysis for the Global Burden of Disease Study 2013. adiposity. Behav Genet 1997;27:325–351.
Lancet 2014;384:766–781. 27. O’Rahilly S, Farooqi IS. Genetics of obesity. Philos Trans
12. Trogdon JG, Finkelstein EA, Feagan CW, Cohen JW. R Soc Lond B Biol Sci 2006;361:1095–1105.
State- and payer-specific estimates of annual medical 28. Aronne LJ, Nelinson DS, Lillo JL. Obesity as a disease
expenditures attributable to obesity. Obesity (Silver state: A new paradigm for diagnosis and treatment. Clin
Spring) 2012;20:214–220. Cornerstone 2009;9:9–25.
13. Fosbøl MØ, Zerahn B. Contemporary methods of body 29. Popkin BM. Contemporary nutritional transition: Deter-
composition measurement. Clin Physiol Funct Imaging minants of diet and its impact on body composition.
2015;35:81–97. Proc Nutr Soc 2011;70:82–91.
14. Wang H, Chen YE, Eitzman DT. Imaging body fat: Tech- 30. Eaton SA, Aiken AJ, Young PE, Ho JWK, Cropley JE,
niques and cardiometabolic implications. Arterioscler Suter CM. Maternal obesity heritably perturbs offspring
Thromb Vasc Biol 2014;34:2217–2223. metabolism for three generations without serial program-
ming. Int J Obes (Lond) 2018;42:911–914.

126
References

31. Burlina S, Dalfrà MG, Lapolla A. Short- and long-term 47. Singh GM, Micha R, Khatibzadeh S, et al. Estimated
consequences for offspring exposed to maternal diabe- global, regional, and national disease burdens related
tes: A review. J Matern Fetal Neonatal Med 2017:1–8. to sugar-sweetened beverage consumption in 2010.
32. Pu Y, Gingrich JD, Steibel JP, Veiga-Lopez A. Circulation 2015;132:639–666.
Sex-specific modulation of fetal adipogenesis by gesta- 48. Malik VS. Sugar sweetened beverages and cardiomet-
tional Bisphenol A and Bisphenol S exposure. abolic health. Curr Opin Cardiol 2017;32:572–579.
Endocrinology 2017;158:3844–3858. 49. Wiener RC, Shen C, Findley PA, Sambamoorthi U, Tan
33. Block T, El-Osta A. Epigenetic programming, early life X. The association between diabetes mellitus,
nutrition and the risk of metabolic disease. Atheroscle- sugar-sweetened beverages, and tooth loss in adults:
rosis 2017;266:31–40. Evidence from 18 states. J Am Dent Assoc 2017;148:
34. Ong KK, Dunger DB. Perinatal growth failure: The road 500–509.e4.
to obesity, insulin resistance and cardiovascular disease 50. Enns CW, Goldman JD, Cook A. Trends in food and
in adults. Best Pract Res Clin Endocrinol Metab 2002; nutrient intakes by adults: NFCS 1977–78. CSFII 1989–
16:191–207. 91, AND CSFII 1994–95. Fam Econ Nutr Rev 1997;10:
35. Gluckman PD, Hanson MA, Pinal C. The developmental 2–15.
origins of adult disease. Matern Child Nutr 2005; 1: 51. Slining MM, Mathias KC, Popkin BM. Trends in food
130–141. and beverage sources among US children and adoles-
36. Astrup A, Astrup A, Buemann B, Flint A, Raben A. cents: 1989–2010. J Acad Nutr Diet 2013; 113:
Low-fat diets and energy balance: How does the 1683–1694.
evidence stand in 2002? Proc Nutr Soc 2002; 61: 52. Ogden CL, Kit BK, Carroll MD, Park S. Consumption
299–309. of sugar drinks in the United States, 2005–2008. NCHS
37. Ludwig DS. Lowering the bar on the low-fat diet. JAMA Data Brief 2011;(71):1–8.
2016;316:2087–2088. 53. Qi Q, Chu AY, Kang JH, et al. Sugar-sweetened bever-
38. Briefel RR, Johnson CL. Secular trends in dietary intake ages and genetic risk of obesity. N Engl J Med 2012;
in the United States. Annu Rev Nutr 2004;24:401–431. 367:1387–1396.
39. Sacks FM, Bray GA, Carey VJ, et al. Comparison of 54. Malik VS, Pan A, Willett WC, Hu FB. Sugar-sweetened
weight-loss diets with different compositions of fat, beverages and weight gain in children and adults: A
protein, and carbohydrates. N Engl J Med 2009; 360: systematic review and meta-analysis. Am J Clin Nutr
859–873. 2013;98:1084–1102.
40. Mozaffarian D, Hao T, Rimm EB, Willett WC, Hu FB. 55. de Ruyter JC, Olthof MR, Seidell JC, Katan MB. A trial
Changes in diet and lifestyle and long-term weight of sugar-free or sugar-sweetened beverages and body
gain in women and men. N Engl J Med 2011; 364: weight in children. N Engl J Med 2012; 367:
2392–2404. 1397–1406.
41. Sacks FM, Katan M. Randomized clinical trials on the 56. Ebbeling CB, Feldman HA, Chomitz VR, et al. A random-
effects of dietary fat and carbohydrate on plasma lipo- ized trial of sugar-sweetened beverages and adolescent
proteins and cardiovascular disease. Am J Med 2002; body weight. N Engl J Med 2012;367:1407–1416.
113(suppl 9B):13S–24S. 57. Bauman AE, Grunseit AC, Rangul V, Heitmann BL.
42. Imamura F, O’Connor L, Ye Z, et al. Consumption of Physical activity, obesity and mortality: Does pattern of
sugar sweetened beverages, artificially sweetened physical activity have stronger epidemiological associ-
beverages, and fruit juice and incidence of type 2 diabe- ations? BMC Public Health 2017;17:788.
tes: Systematic review, meta-analysis, and estimation 58. Kotz CM, Perez-Leighton CE, Teske JA, Billington CJ.
of population attributable fraction. BMJ 2015; 351: Spontaneous physical activity defends against obesity.
h3576. Curr Obes Rep 2017;6:362–370.
43. Siri-Tarino PW, Sun Q, Hu FB, Krauss RM. Meta-analysis 59. Jakicic JM, Otto AD, Lang W, et al. The effect of physical
of prospective cohort studies evaluating the association activity on 18-month weight change in overweight
of saturated fat with cardiovascular disease. Am J Clin adults. Obesity (Silver Spring) 2011;19:100–109.
Nutr 2010;91:535–546. 60. Piaggi P, Thearle MS, Bogardus C, Krakoff J. Lower
44. Schwingshackl L, Hoffmann G. Comparison of effects energy expenditure predicts long-term increases in
of long-term low-fat vs high-fat diets on blood lipid levels weight and fat mass. J Clin Endocrinol Metab 2013;98:
in overweight or obese patients: A systematic review E703–E707.
and meta-analysis. J Acad Nutr Diet 2013; 113: 61. Carlson SA, Fulton JE, Pratt M, Yang Z, Adams EK.
1640–1661. Inadequate physical activity and health care expendi-
45. Mozaffarian D. Dietary and policy priorities for cardio- tures in the United States. Prog Cardiovasc Dis 2015;
vascular disease, diabetes, and obesity: A compre­hensive 57:315–323.
review. Circulation 2016;133:187–225. 62. Blackwell DL, Lucas JW, Clarke TC. Summary health
46. Siri-Tarino PW, Sun Q, Hu FB, Krauss RM. Saturated statistics for U.S. adults: National health interview survey,
fat, carbohydrate, and cardiovascular disease. Am J 2012. Vital Health Stat 10 2014;(260):1–161.
Clin Nutr 2010;91:502–509.

127
5 Obesity, Metabolic Syndrome, and Oral Health

63. Loprinzi PD, Cardinal BJ, Lee H, Tudor-Locke C. Mark- 78. Weyer C, Yudkin JS, Stehouwer CD, Schalkwijk CG,
ers of adiposity among children and adolescents: Pratley RE, Tataranni PA. Humoral markers of inflam-
Implications of the isotemporal substitution paradigm mation and endothelial dysfunction in relation to
with sedentary behavior and physical activity patterns. adiposity and in vivo insulin action in Pima Indians.
J Diabetes Metab Disord 2015;14:46. Atherosclerosis 2002;161:233–242.
64. Loprinzi PD, Edwards MK. Less sitting, more physical 79. Fried SK, Bunkin DA, Greenberg AS. Omental and
activity and higher cardiorespiratory fitness: Associations subcutaneous adipose tissues of obese subjects release
with weight status among a national sample of children. interleukin-6: Depot difference and regulation by gluco-
Health Promot Perspect 2017;7:175–177. corticoid. J Clin Endocrinol Metab 1998;83:847–850.
65. Duncan MJ, Vandelanotte C, Caperchione C, Hanley 80. Guzik TJ, Skiba DS, Touyz RM, Harrison DG. The role
C, Mummery WK. Temporal trends in and relationships of infiltrating immune cells in dysfunctional adipose
between screen time, physical activity, overweight and tissue. Cardiovasc Res 2017;113:1009–1023.
obesity. BMC Public Health 2012;12:1060. 81. Goerdt S, Politz O, Schledzewski K, et al. Alternative
66. Stamatakis E, Hirani V, Rennie K. Moderate-to-vigorous versus classical activation of macrophages. Pathobiol-
physical activity and sedentary behaviours in relation to ogy 1999;67:222–226.
body mass index-defined and waist circumference- 82. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a
defined obesity. Br J Nutr 2009;101:765–773. phenotypic switch in adipose tissue macrophage polar-
67. Fantuzzi G. Adipose tissue, adipokines, and inflamma- ization. J Clin Investing 2007;117:175–184.
tion. J Allergy Clin Immunol 2005;115:911–919. 83. Lumeng CN, Deyoung SM, Bodzin JL, Saltiel AR.
68. Frühbeck G. Adipose tissue: Structure, function and Increased inflammatory properties of adipose tissue
metabolism. In: Caballero B (ed). Encyclopedia of macrophages recruited during diet-induced obesity.
Human Nutrition, ed 3. Oxford: Elsevier/Academic, Diabetes 2007;56:16–23.
2013:1–13. 84. Kershaw EE, Flier JS. Adipose tissue as an endocrine
69. Frühbeck G, Gomez-Ambrosi J, Muruzábal FJ, Burrell organ. J Clin Endocrinol Metab 2004;89:2548–2556.
MA. The adipocyte: A model for integration of endo- 85. Fain JN, Tichansky DS, Madan AK. Most of the inter-
crine and metabolic signaling in energy metabolism leukin 1 receptor antagonist, cathepsin S, macrophage
regulation. Am J Physiol Endocrinol Metab 2001;280: migration inhibitory factor, nerve growth factor, and
E827–E847. interleukin 18 release by explants of human adipose
70. Frühbeck G. Overview of adipose tissue and its role in tissue is by the non-fat cells, not by the adipocytes.
obesity and metabolic disorders. Methods Mol Biol Metabolism 2006;55:1113–1121.
2008;456:1–22. 86. Lord GM, Matarese G, Howard JK, Baker RJ, Bloom
71. Trayhurn P. Adipocyte biology. Obes Rev 2007;8(suppl SR, Lechler RI. Leptin modulates the T-cell immune
1):41–44. response and reverses starvation-induced immunosup-
72. Xu H, Barnes GT, Yang Q, et al. Chronic inflammation pression. Nature 1998;394:897–901.
in fat plays a crucial role in the development of 87. Lord GM, Matarese G, Howard JK, Bloom SR, Lechler
obesity-related insulin resistance. J Clin Investing 2003; RI. Leptin inhibits the anti-CD3-driven proliferation of
112:1821–1830. peripheral blood T cells but enhances the production
73. Liu KH, Chan YL, Chan WB, Chan JC, Chu CW. Mesen- of proinflammatory cytokines. J Leukoc Biol 2002; 72:
teric fat thickness is an independent determinant of 330–338.
metabolic syndrome and identifies subjects with 88. Rodriguez L, Graniel J, Ortiz R. Effect of leptin on acti-
increased carotid intima-media thickness. Diabetes Care vation and cytokine synthesis in peripheral blood
2006;29:379–384. lymphocytes of malnourished infected children. Clin Exp
74. Chaffee BW, Weston SJ. Association between chronic Immunol 2007;148:478–485.
periodontal disease and obesity: A systematic review 89. Loffreda S, Yang SQ, Lin HZ, et al. Leptin regulates
and meta-analysis. J Periodontol 2010;81:1708–1724. proinflammatory immune responses. FASEB J 1998;
75. Das UN. Is obesity an inflammatory condition? Nutrition 12:57–65.
2001;17:953–966. 90. Caldefie-Chezet F, Poulin A, Vasson MP. Leptin regulates
76. Weisberg SP, McCann D, Desai M, Rosenbaum M, functional capacities of polymorphonuclear neutrophils.
Leibel RL, Ferrante AW Jr. Obesity is associated with Free Radic Res 2003;37:809–814.
macrophage accumulation in adipose tissue. J Clin 91. Fantuzzi G. Adiponectin and inflammation: Consensus
Investing 2003;112:1796–1808. and controversy. J Allergy Clin Immunol 2008; 121:
77. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose 326–330.
expression of tumor necrosis factor-alpha: Direct role 92. Barbosa Vde S, Rêgo J, Antônio da Silva N. Possible
in obesity-linked insulin resistance. Science 1993;259: role of adipokines in systemic lupus erythematosus and
87–91. rheumatoid arthritis. Rev Bras Reumatol 2012; 52:
278–287.
93. Chen X, Lu J, Bao J, Guo J, Shi J, Wang Y. Adiponectin:
A biomarker for rheumatoid arthritis? Cytokine Growth
Factor Rev 2013;24:83–89.

128
References

94. Ehling A, Schäffler A, Herfarth H, et al. The potential of 110. Rupnick MA, Panigrahy D, Zhang CY, et al. Adipose
adiponectin in driving arthritis. J Immunol 2006; 176: tissue mass can be regulated through the vasculature.
4468–4478. Proc Natl Acad Sci U S A 2002;99:10730–10735.
95. Schäffler A, Ehling A, Neumann E, et al. Adipocytokines 111. Wood IS, de Heredia FP, Wang B, Trayhurn P. Cellular
in synovial fluid. JAMA 2003;290:1709–1710. hypoxia and adipose tissue dysfunction in obesity. Proc
96. Bruun JM, Lihn AS, Verdich C, et al. Regulation of Nutr Soc 2009;68:370–377.
adiponectin by adipose tissue-derived cytokines: In vivo 112. Hosogai N, Fukuhara A, Oshima K, et al. Adipose tissue
and in vitro investigations in humans. Am J Physiol hypoxia in obesity and its impact on adipocytokine
Endocrinol Metab 2003;285:E527–E533. dysregulation. Diabetes 2007;56:901–911.
97. Villarreal-Molina MT, Antuna-Puente B. Adiponectin: 113. Ye J, Gao Z, Yin J, He Q. Hypoxia is a potential risk factor
Anti-inflammatory and cardioprotective effects. for chronic inflammation and adiponectin reduction in
Biochimie 2012;94:2143–2149. adipose tissue of ob/ob and dietary obese mice. Am J
98. Arita Y, Kihara S, Ouchi N, et al. Paradoxical decrease Physiol Endocrinol Metab 2007;293:E1118–E1128.
of an adipose-specific protein, adiponectin, in obesity. 114. Wang GL, Semenza GL. General involvement of
Biochem Biophys Res Commun 1999;257:79–83. hypoxia-inducible factor 1 in transcriptional response
99. Yokota T, Oritani K, Takahashi I, et al. Adiponectin, a to hypoxia. Proc Natl Acad Sci U S A 1993; 90:
new member of the family of soluble defense collagens, 4304–4308.
negatively regulates the growth of myelomonocytic 115. Wang B, Wood IS, Trayhurn P. Hypoxia induces leptin
progenitors and the functions of macrophages. Blood gene expression and secretion in human preadipocytes:
2000;96:1723–1732. Differential effects of hypoxia on adipokine expression
100. Masaki T, Chiba S, Tatsukawa H, et al. Adiponectin by preadipocytes. J Endocrinol 2008;198:127–134.
protects LPS-induced liver injury through modulation of 116. Oda T, Hirota K, Nishi K, et al. Activation of hypoxia-
TNF-alpha in KK-Ay obese mice. Hepatology 2004;40: inducible factor 1 during macrophage differentiation.
177–184. Am J Physiol Cell Physiol 2006;291:C104–C113.
101. Ajuwon KM, Spurlock ME. Adiponectin inhibits 117. Global BMIMC, Di Angelantonio E, Bhupathiraju Sh N,
LPS-induced NF-kappaB activation and IL-6 production et al. Body-mass index and all-cause mortality:
and increases PPARgamma2 expression in adipocytes. Individual-participant-data meta-analysis of 239
Am J Physiol Regul Integr Comp Physiol 2005; 288: prospective studies in four continents. Lancet 2016;
R1220–R1225. 388:776–786.
102. Fasshauer M, Kralisch S, Klier M, et al. Adiponectin 118. Jia H, Zack MM, Thompson WW. Population-based
gene expression and secretion is inhibited by estimates of decreases in quality-adjusted life expec-
interleukin-6 in 3T3-L1 adipocytes. Biochem Biophys tancy associated with unhealthy body mass index.
Res Commun 2003;301:1045–1050. Public Health Rep 2016;131:177–184.
103. Sengenès C, Miranville A, Lolmède K, Curat CA, 119. Grover SA, Kaouache M, Rempel P, et al. Years of life
Bouloumié A. The role of endothelial cells in inflamed lost and healthy life-years lost from diabetes and cardio-
adipose tissue J Intern Med 2007;262:415–421. vascular disease in overweight and obese people: A
104. Salmi M, Jalkanen S. Cell-surface enzymes in control modelling study. Lancet Diabetes Endocrinol 2015; 3:
of leukocyte trafficking. Nat Rev Immunol 2005; 5: 114–122.
760–771. 120. Backholer K, Wong E, Freak-Poli R, Walls HL, Peeters
105. Kułdo JM, Ogawara KI, Werner N, et al. Molecular path- A. Increasing body weight and risk of limitations in activ-
ways of endothelial cell activation for (targeted) ities of daily living: A systematic review and meta-analysis.
pharmacological intervention of chronic inflammatory Obes Rev 2012;13:456–468.
diseases. Curr Vasc Pharmacol 2005;3:11–39. 121. Cheng FW, Gao X, Bao L, et al. Obesity as a risk factor
106. Aoki S, Toda S, Sakemi T, Sugihara H. Coculture of for developing functional limitation among older adults:
endothelial cells and mature adipocytes actively A conditional inference tree analysis. Obesity (Silver
promotes immature preadipocyte development in vitro. Spring) 2017;25:1263–1269.
Cell Struct Funct 2003;28:55–60. 122. Chang VW, Alley DE, Dowd JB. Trends in the relationship
107. Hutley LJ, Herington AC, Shurety W, et al. Human between obesity and disability, 1988–2012. Am J Epide-
adipose tissue endothelial cells promote preadipocyte miol 2017;186:688–695.
proliferation. Am J Physiol Endocrinol Metab 2001;281: 123. Stenholm S, Head J, Aalto V, et al. Body mass index as
E1037–E1044. a predictor of healthy and disease-free life expectancy
108. Crandall DL, Busler DE, McHendry-Rinde B, Groeling between ages 50 and 75: A multicohort study. Int J
TM, Kral JG. Autocrine regulation of human preadipo- Obes (Lond) 2017;41:769–775.
cyte migration by plasminogen activator inhibitor-1. J 124. Abdelaal M, le Roux CW, Docherty NG. Morbidity and
Clin Endocrinol Metab 2000;85:2609–2614. mortality associated with obesity. Ann Transl Med 2017;
109. Fukumura D, Ushiyama A, Duda DG, et al. Paracrine 5:161.
regulation of angiogenesis and adipocyte differentiation 125. Berenson GS. Obesity—A critical issue in preventive
during in vivo adipogenesis. Circ Res 2003; 93: cardiology: The Bogalusa Heart Study. Prev Cardiol
e88–e97. 2005;8:234–241.

129
5 Obesity, Metabolic Syndrome, and Oral Health

126. Bray GA, Clearfield MB, Fintel DJ, Nelinson DS. Over- 144. Expert Panel on Detection, Evaluation, and Treatment
weight and obesity: The pathogenesis of cardiometabolic of High Blood Cholesterol in Adults. Executive Summary
risk. Clin Cornerstone 2009;9:30–40. of the Third Report of the National Cholesterol Education
127. Attar BM, Van Thiel DH. Current concepts and manage- Program (NCEP) Expert Panel on Detection, Evaluation,
ment approaches in nonalcoholic fatty liver disease. and Treatment of High Blood Cholesterol In Adults (Adult
ScientificWorldJournal 2013;2013:481893. Treatment Panel III). JAMA 2001;285:2486–2497.
128. Cave M, Deaciuc I, Mendez C, et al. Nonalcoholic fatty 145. Aguilar M, Bhuket T, Torres S, Liu B, Wong RJ. Preva-
liver disease: Predisposing factors and the role of nutri- lence of the metabolic syndrome in the United States,
tion. J Nutr Biochem 2007;18:184–195. 2003–2012. JAMA 2015;313:1973–1974.
129. Bedogni G, Miglioli L, Masutti F, Tiribelli C, Marchesini 146. Mancia G, Bombelli M, Facchetti R, et al. Impact of
G, Bellentani S. Prevalence of and risk factors for nonal- different definitions of the metabolic syndrome on the
coholic fatty liver disease: The Dionysos nutrition and prevalence of organ damage, cardiometabolic risk and
liver study. Hepatology 2005;42:44–52. cardiovascular events. J Hypertens 2010; 28:999–1006.
130. Tsuboi N, Utsunomiya Y, Hosoya T. Obesity-related 147. Tenenbaum A, Motro M, Schwammenthal E, Fisman
glomerulopathy and the nephron complement. Nephrol EZ. Macrovascular complications of metabolic
Dial Transplant 2013;28(suppl 4):iv108–113. syndrome: An early intervention is imperative. Int J
131. Okabayashi Y, Tsuboi N, Sasaki T, et al. Glomerulopathy Cardiol 2004;97:167–172.
associated with moderate obesity. Kidney Int Rep 2016; 148. Verzeletti GN, Gaio EJ, Linhares DS, Rosing CK. Effect
1:250–255. of obesity on alveolar bone loss in experimental peri-
132. Alonso-Alvarez ML, Cordero-Guevara JA, Teran-Santos odontitis in Wistar rats. J Appl Oral Sci 2012; 20:
J, et al. Obstructive sleep apnea in obese community- 218–221.
dwelling children: The NANOS study. Sleep 2014; 37: 149. Cavagni J, Wagner TP, Gaio EJ, Rêgo RO, Torres IL,
943–949. Rösing CK. Obesity may increase the occurrence of
133. Delitala AP, Capobianco G, Delitala G, Cherchi PL, spontaneous periodontal disease in Wistar rats. Arch
Dessole S. Polycystic ovary syndrome, adipose tissue Oral Biol 2013;58:1034–1039.
and metabolic syndrome. Arch Gynecol Obstet 150. Suvan J, D’Aiuto F, Moles DR, Petrie A, Donos N. Asso-
2017;296:405–419. ciation between overweight/obesity and periodontitis in
134. Richter JE, Rubenstein JH. Presentation and epidemi- adults. A systematic review. Obes Rev 2011; 12:
o l o g y o f g a s t ro e s o p h a g e a l re fl u x d i s e a s e . e381–e404.
Gastroenterology 2018;154:267–276. 151. Moura-Grec PG, Marsicano JA, Carvalho CA,
135. Hidayat K, Yang CM, Shi BM. Body fatness at a young Sales-Peres SH. Obesity and periodontitis: Systematic
age, body fatness gain and risk of breast cancer: review and meta-analysis. Cien Saude Colet 2014;19:
Systematic review and meta-analysis of cohort studies. 1763–1772.
Obes Rev 2018;19:254–268. 152. Martens L, De Smet S, Yusof MY, Rajasekharan S.
136. Smith LA, O’Flanagan CH, Bowers LW, Allott EH, Hurst- Association between overweight/obesity and periodon-
ing SD. Translating mechanism-based strategies to tal disease in children and adolescents: A systematic
break the obesity-cancer link: A narrative review. J Acad review and meta-analysis. Eur Arch Paediatr Dent 2017;
Nutr Diet 2018;118:652–667. 18:69–82.
137. Withrow D, Alter DA. The economic burden of obesity 153. Furuta M, Yamashita Y. Epidemiological evidences for
worldwide: A systematic review of the direct costs of the association of obesity and metabolic disorders with
obesity. Obes Rev 2011;12:131–141. oral diseases. Curr Oral Health Rep 2017;4:51–58.
138. Li Q, Blume SW, Huang JC, Hammer M, Ganz ML. 154. Bistrian B. Systemic response to inflammation. Nutr Rev
Prevalence and healthcare costs of obesity-related 2007;65(12Pt2):S170–S172.
comorbidities: Evidence from an electronic medical 155. Lundin M, Yucel-Lindberg T, Dahllöf G, Marcus C,
records system in the United States. J Med Econ 2015; Modéer T. Correlation between TNFalpha in gingival
18:1020–1028. crevicular fluid and body mass index in obese subjects.
139. Cecchini M, Sassi F. Preventing obesity in the USA: Acta Odontol Scand 2004;62:273–277.
Impact on health service utilization and costs. Pharma- 156. Schmidt AM, Weidman E, Lalla E, et al. Advanced glyca-
coeconomics 2015;33:765–776. tion endproducts (AGEs) induce oxidant stress in the
140. Sherling DH, Perumareddi P, Hennekens CH. Metabolic gingiva: A potential mechanism underlying accelerated
syndrome. J Cardiovasc Pharmacol Ther 2017; 22: periodontal disease associated with diabetes. J Peri-
365–367. odontol Res 1996;31:508–515.
141. Engin A. The definition and prevalence of obesity and 157. Suvan J, D’Aiuto F, Moles DR, Petrie A, Donos N. Asso-
metabolic syndrome. Adv Exp Med Biol 2017;960:1–17. ciation between overweight/obesity and periodontitis in
142. Ko GT. Metabolic syndrome or “central obesity adults. A systematic review. Obes Rev 2011; 12:
syndrome”? Diabetes Care 2006;29:752. e381–e404.
143. Kassi E, Pervanidou P, Kaltsas G, Chrousos G. Meta-
bolic syndrome: Definitions and controversies. BMC
Med 2011;9:48.

130
References

158. Oikarinen R, Syrjälä AM, Komulainen K, et al. Body mass 173. Acharya A, Bhavsar N, Jadav B, Parikh H. Cardiopro-
index and periodontal infection in a sample of tective effect of periodontal therapy in metabolic
non-smoking older individuals. Oral Dis 2014;20: syndrome: A pilot study in Indian subjects. Metab Syndr
e25–e30. Relat Disord 2010;8:335–341.
159. Keller A, Rohde JF, Raymond K, Heitmann BL. Associ- 174. Nibali L, Tatarakis N, Needleman I, et al. Clinical review:
ation between periodontal disease and overweight and Association between metabolic syndrome and peri-
obesity: A systematic review. J Periodontol 2015;86: odontitis: A systematic review and meta-analysis. J Clin
766–776. Endocrinol Metab 2013;98:913–920.
160. de Castilhos ED, Horta BL, Gigante DP, Demarco FF, 175. Lamster IB, Pagan M. Periodontal disease and the
Peres KG, Peres MA. Association between obesity and metabolic syndrome. Int Dent J 2017;67:67–77.
periodontal disease in young adults: A population-based 176. Palmer CA, Kent R Jr, Loo CY, et al. Diet and caries-
birth cohort. J Clin Periodontol 2012;39:717–724. associated bacteria in severe early childhood caries. J
161. Gorman A, Kaye EK, Apovian C, Fung TT, Nunn M, Dent Res 2010;89:1224–1229.
Garcia RI. Overweight and obesity predict time to peri- 177. Arcella D, Ottolenghi L, Polimeni A, Leclercq C. The
odontal disease progression in men. J Clin Periodontol relationship between frequency of carbohydrates intake
2012;39:107–114. and dental caries: A cross-sectional study in Italian
162. Morita I, Okamoto Y, Yoshii S, et al. Five-year incidence teenagers. Public Health Nutr 2002;5:553–560.
of periodontal disease is related to body mass index. J 178. Chaffee BW, Feldens CA, Rodrigues PH, Vitolo MR.
Dent Res 2011;90:199–202. Feeding practices in infancy associated with caries inci-
163. Gorman A, Kaye EK, Nunn M, Garcia RI. Changes in dence in early childhood. Community Dent Oral
body weight and adiposity predict periodontitis progres- Epidemiol 2015;43:338–348.
sion in men. J Dent Res 2012;91:921–926. 179. Davidson K, Schroth RJ, Levi JA, Yaffe AB, Mittermuller
164. Nascimento GG, Peres KG, Mittinty MN, et al. Obesity BA, Sellers EA. Higher body mass index associated with
and periodontal outcomes: A population-based cohort severe early childhood caries. BMC Pediatr 2016; 16:
study in Brazil. J Periodontol 2017;88:50–58. 137.
165. Canoy D. Distribution of body fat and risk of coronary 180. Hooley M, Skouteris H, Boganin C, Satur J, Kilpatrick
heart disease in men and women. Curr Opin Cardiol N. Body mass index and dental caries in children and
2008;23:591–598. adolescents: A systematic review of literature published
166. Gaio EJ, Haas AN, Rösing CK, Oppermann RV, Albandar 2004 to 2011. Syst Rev 2012;1:57.
JM, Susin C. Effect of obesity on periodontal attachment 181. Kantovitz KR, Pascon FM, Rontani RM, Gavião MB.
loss progression: A 5-year population-based prospec- Obesity and dental caries—A systematic review. Oral
tive study. J Clin Periodontol 2016;43:557–565. Health Prev Dent 2006;4:137–144.
167. Suvan J, Petrie A, Moles DR, et al. Body mass index as 182. Hayden C, Bowler JO, Chambers S, et al. Obesity and
a predictive factor of periodontal therapy outcomes. J dental caries in children: A systematic review and
Dent Res 2014;93:49–54. meta-analysis. Community Dent Oral Epidemiol 2013;
168. Gonçalves T, Feres M, Zimmermann G, et al. Effects of 41:289–308.
scaling and root planing on clinical response and serum 183. Vázquez-Nava F, Vázquez-Rodríguez EM, Saldívar-
levels of adipocytokines in patients with obesity and González AH, Lin-Ochoa D, Martinez-Perales GM,
chronic periodontitis. J Periodontol 2015;86:53–61. Joffre-Velázquez VM. Association between obesity and
169. Papageorgiou SN, Reichert C, Jäger A, Deschner J. dental caries in a group of preschool children in Mexico.
Effect of overweight/obesity on response to periodontal Public Health Dent 2010;70:124–130.
treatment: Systematic review and a meta–analysis. J 184. Alm A, Fåhraeus C, Wendt LK, Koch G, Andersson-Gäre
Clin Periodontol 2015;42:247–261. B, Birkhed D. Body adiposity status in teenagers and
170. Pradeep AR, Priyanka N, Prasad MV, Kalra N, Kumari snacking habits in early childhood in relation to approx-
M. Association of progranulin and high sensitivity CRP imal caries at 15 years of age. Int J Paediatr Dent 2008;
concentrations in gingival crevicular fluid and serum in 18:189–196.
chronic periodontitis subjects with and without obesity. 185. Willershausen B, Moschos D, Azrak B, Blettner M.
Dis Markers 2012;33:207–213. Correlation between oral health and body mass index
171. Zimmermann GS, Bastos MF, Dias Gonçalves TE, (BMI) in 2071 primary school pupils. Eur J Med Res
Chambrone L, Duarte PM. Local and circulating levels 2007;12:295–299.
of adipocytokines in obese and normal weight individ- 186. Sadeghi M, Alizadeh F. Association between dental
uals with chronic periodontitis. J Periodontol 2013;84: caries and body mass index-for-age among
624–633. 6–11-year-old children in Isfahan in 2007. J Dent Res
172. Altay U, Gürgan CA, Ağbaht K. Changes in inflammatory Dent Clin Dent Prospects 2007;1:119–124.
and metabolic parameters after periodontal treatment 187. Tavares M, Goodson JM, Cugini M, et al. Tooth eruption
in patients with and without obesity. J Periodontol 2013; and BMI percentile in Kuwaiti fifth grade children
84:13–23. [abstract 2887]. J Dent Res 2013;92(special issue A).

131
5 Obesity, Metabolic Syndrome, and Oral Health

188. Goodson JM, Tavares M, Wang X, et al. Obesity and 203. DeBoer MD, Scharf RJ, Demmer RT. Sugar-sweetened
dental decay: Inference on the role of dietary sugar. beverages and weight gain in 2- to 5-year-old children.
PLoS One 2013;8:e74461. Pediatrics 2013;132:413–420.
189. Kottayi S, Bhat S, Hegde K, Peedikayil F, Chandru T, 204. Vargas CM, Dye BA, Kolasny CR, et al. Early childhood
Anil S. A cross-sectional study of the prevalence of caries and intake of 100 percent fruit juice: Data from
dental caries among 12- to 15-year-old overweight NHANES, 1999–2004. J Am Dent Assoc 2014; 145:
schoolchildren. J Contemp Dent Pract 2016; 17: 1254–1261.
750–754. 205. Heller K, Burt B, Eklund S. Sugared soda consumption
190. Kumar S, Kroon J, Lalloo R, Kulkarni S, Johnson NW. and dental caries in the United States. J Dent Res 2001;
Relationship between body mass index and dental 80:1949–1953.
caries in children, and the influence of socio-economic 206. Moynihan PJ, Kelly SA. Effect on caries of restricting
status. Int Dent J 2017;67:91–97. sugars intake: Systematic review to inform WHO guide-
191. Macek MD, Mitola DJ. Exploring the association lines. J Dent Res 2014;93:8–18.
between overweight and dental caries among US chil- 207. Bernabé E, Vehkalahti MM, Sheiham A, Aromaa A,
dren. Pediatr Dent 2006;28:375–380. Suominen AL. Sugar-sweetened beverages and dental
192. Hong L, Ahmed A, McCunniff M, Overman P, Mathew caries in adults: A 4-year prospective study. J Dent
M. Obesity and dental caries in children aged 2–6 years 2014;42:952–958.
in the United States: National Health and Nutrition Exam- 208. Kim S, Park S, Lin M. Permanent tooth loss and
ination Survey 1999–2002. J Public Health Dent 2008; sugar-sweetened beverage intake in U.S. young adults.
68:227–233. J Public Health Dent 2017;77:148–154.
193. Kopycka-Kedzierawski DT, Auinger P, Billings RJ, Weitz- 209. Brownell K, Farley T, Willett W, et al. The public health
man M. Caries status and overweight in 2- to 18-year-old and economic benefits of taxing sugar-sweetened
US children: Findings from national surveys. Community beverages. N Engl J Med 2009;361:1599–1605.
Dentistry and Oral Epidemiol 2008;36:157–167. 210. Chriqui JF, Chaloupka FJ, Powell LM, Eidson SS. A
194. Hayden C, Bowler JO, Chambers S, et al. Obesity and typology of beverage taxation: Multiple approaches for
dental caries in children: A systematic review and obesity prevention and obesity prevention-related reve-
meta-analysis. Community Dent Oral Epidemiol nue generation. J Public Health Policy 2013; 34:
2013;41:289–308. 403–423.
195. Modéer T, Blomberg CC, Wondimu B, Julihn A, Marcus 211. Meyer BD, Lee JY. The confluence of sugar, dental
C. Association between obesity, flow rate of whole caries, and health policy. J Dent Res 2015; 94:
saliva, and dental caries in adolescents. Obesity (Silver 1338–1340.
Spring) 2010;18:2367–2373. 212. Nakhimovsky SS, Feigl AB, Avila C, O’Sullivan G,
196. Rosinger A, Herrick K, Gahche J, Park S. Macgregor-Skinner E, Spranca M. Taxes on
Sugar-sweetened beverage consumption among US sugar-sweetened beverages to reduce overweight and
youth, 2011–2014. NCHS Data Brief 2017;(271):1–8. obesity in middle-income countries: A systematic review.
197. Rosinger A, Herrick K, Gahche J, Park S. PLoS One 2016;11:e0163358.
Sugar-sweetened beverage consumption among US 213. Schwendicke F, Thomson WM, Broadbent JM, Stolpe
adults, 2011–2014. NCHS Data Brief 2017;(270):1–8. M. Effects of taxing sugar-sweetened beverages on
198. Malik VS, Willett WC, Hu FB. Sugar-sweetened bever- caries and treatment costs. J Dent Res 2016; 95:
ages and BMI in children and adolescents: Reanalyses 1327–1332.
of a meta-analysis. Am J Clin Nutr 2009;89:438–439. 214. He Q, Karlberg J. BMI in childhood and its association
199. Ludwig DS, Peterson KE, Gortmaker SL. Relation with height gain, timing of puberty, and final height.
between consumption of sugar-sweetened drinks and Pediatr Res 2001;49:244–251.
childhood obesity: A prospective, observational analysis. 215. De Simone M, Farello G, Palumbo M, et al. Growth
Lancet 2001;357:505–508. charts, growth velocity and bone development in child-
200. Schulze MB, Manson JE, Ludwig DS, et al. hood obesity. Int J Obes Relat Metab Disord 1995;19:
Sugar-sweetened beverages, weight gain, and inci- 851–857.
dence of type 2 diabetes in young and middle-aged 216. De Leonibus C, Marcovecchio ML, Chiavaroli V, de
women. JAMA 2004;292:927–934. Giorgis T, Chiarelli F, Mohn A. Timing of puberty and
201. Te Morenga L, Mallard S, Mann J. Dietary sugars and physical growth in obese children: A longitudinal study
body weight: Systematic review and meta-analyses of in boys and girls. Pediatr Obes 2014;9:292–299.
randomised controlled trials and cohort studies. BMJ 217. Aksglaede L, Juul A, Olsen LW, Sørensen TI. Age at
2012;346:e7492. puberty and the emerging obesity epidemic. PLoS One
202. Frantsve-Hawley J, Bader JD, Welsh JA, Wright JT. A 2009;4:e8450.
systematic review of the association between consump- 218. Giuca MR, Pasini M, Tecco S, Marchetti E, Giannotti L,
tion of sugar-containing beverages and excess weight Marzo G. Skeletal maturation in obese patients. Am J
gain among children under age. J Public Health Dent Orthod Dentofacial Orthop 2012;142:774–779.
2017;77(suppl1):S43–S66.

132
References

219. Weddell LS, Hartsfield JK Jr. Dental maturity of Cauca- 236. Tishler PV, Larkin EK, Schluchter MD, Redline S. Inci-
sian children in the Indianapolis area. Pediatr Dent 2011; dence of sleep-disordered breathing in an urban adult
33:221–227. population. JAMA 2003;289:2230–2237.
220. Sánchez-Pérez L, Irigoyen ME, Zepeda M. Dental caries, 237. Malhotra A, White DP. Obstructive sleep apnoea. Lancet
tooth eruption timing and obesity: A longitudinal study 2002;360:237–245.
in a group of Mexican schoolchildren. Acta Odontol 238. Obstructive sleep apnea syndrome (780.53–0). In:
Scand 2010;68:57–64. American Academy of Sleep Medicine. The International
221. Hilgers KK, Akridge M, Scheetz JP, Kinane DE. Child- Classification of Sleep Disorders, Revised: Diagnostic
hood obesity and dental development. Pediatr Dent and Coding Manual. Westchester, IL: American Acad-
2006;28:18–22. emy of Sleep Medicine, 2001:52–128.
222. Zangouei-Booshehri M, Ezoddini-Ardakani F, Agha Aghili 239. Maciel Santos ME, Laureano Filho JR, Campos JM,
H, Sharifi A. Assessment of the relationship between Ferraz EM. Dentofacial characteristics as indicator of
body mass index (BMI) and dental age. Health 2011;3: obstructive sleep apnoea-hypopnoea syndrome in
253–257. patients with severe obesity. Obes Rev 2011; 12:
223. Mack KB, Phillips C, Jain N, Koroluk LD. Relationship 105–113.
between body mass index percentile and skeletal matu- 240. Chin K. Interactions among sleep disordered breathing,
ration and dental development in orthodontic patients. obesity, and sleep duration. Sleep Biol Rhythm 2017;
Am J Orthod Dentofacial Orthop 2013;143:228–234. 15:87–88.
224. Costacurta M, Sicuro L, Di Renzo L, Condò R, De 241. Peppard PE, Young T, Barnet JH, Palta M, Hagen EW,
Lorenzo A, Docimo R. Childhood obesity and Hla KM. Increased prevalence of sleep-disordered
skeletal-dental maturity. Eur J Paediatr Dent 2012; 13: breathing in adults. Am J Epidemiol 2013; 177:
128–132. 1006–1014.
225. Must A, Phillips SM, Tybor DJ, Lividini K, Hayes C. The 242. Young T, Peppard PE, Gottlieb DJ. Epidemiology of
association between childhood obesity and tooth erup- obstructive sleep apnea: A population health perspec-
tion. Obesity (Silver Spring) 2012;20:2070–2074. tive. Am J Respir Crit Care Med 2002; 165:
226. Halaas JL, Gajiwala KS, Maffei M, et al. Weight-reducing 1217–1239.
effects of the plasma protein encoded by the obese 243. Lee W, Nagubadi S, Kryger MH, Mokhlesi B. Epidemi-
gene. Science 1995;269:543–546. ology of obstructive sleep apnea: A population-based
227. Shalitin S, Phillip M. Role of obesity and leptin in the perspective. Expert Rev Respir Med 2008;2:349–364.
pubertal process and pubertal growth—A review. Int J 244. Ong CW, O’Driscoll DM, Truby H, Naughton MT, Hamil-
Obes Relat Metab Disord 2003;27:869–874. ton GS. The reciprocal interaction between obesity and
228. Maor G, Rochwerger M, Segev Y, Phillip M. Leptin acts obstructive sleep apnoea. Sleep Med Rev 2013; 17:
as a growth factor on the chondrocytes of skeletal growth 123–131.
centers. J Bone Miner Res 2002;17:1034–1043. 245. Caples SM, Gami AS, Somers VK. Obstructive sleep
229. Miller R, Tanofsky-Kraff M, Shomaker LB, et al. Serum apnea. Ann Intern Med 2005;142:187–197.
leptin and loss of control eating in children and adoles- 246. Narang I, Mathew JL. Childhood obesity and obstructive
cents. Int J Obes (Lond) 2014;38:397–403. sleep apnea. J Nutr Metab 2012;2012:134202.
230. Fatemifar G, Evans DM, Tobias JH. The association 247. Kohler MJ, Thormaehlen S, Kennedy JD, et al. Differ-
between primary tooth emergence and anthropometric ences in the association between obesity and obstructive
measures in young adults: Findings from a large sleep apnea among children and adolescents. J Clin
prospective cohort study. PLoS One 2014;9:e96355. Sleep Med 2009;5:506–511.
231. Saloom H, Papageorgiou S, Carpenter G, Cobourne 248. Verhulst SL, Schrauwen N, Haentjens D, et al.
M. Impact of obesity on orthodontic tooth movement Sleep-disordered breathing and the metabolic syndrome
in adolescents: A prospective clinical cohort study. J in overweight and obese children and adolescents. J
Dent Res 2017;96:547–554. Pedriatr 2007;150:608–612.
232. Katagiri S, Nitta H, Nagasawa T, et al. Reduced masti- 249. Verhulst SL, Schrauwen N, Haentjens D, et al.
catory function in non-elderly obese Japanese adults. Sleep-disordered breathing in overweight and obese
Obes Res Clin Pract 2011;5:e267–e360. children and adolescents: Prevalence, characteristics
233. Sánchez-Ayala A, Campanha NH, Garcia RC. Relation- and the role of fat distribution. Arch Dis Child 2007;92:
ship between body fat and masticatory function. J 205–208.
Prosthodont 2013;22:120–125. 250. Canapari CA, Hoppin AG, Kinane TB, Thomas BJ, Torri-
234. Nascimento GG, Leite FR, Conceição DA, Ferrúa CP, ani M, Katz ES. Relationship between sleep apnea, fat
Singh A, Demarco FF. Is there a relationship between distribution, and insulin resistance in obese children. J
obesity and tooth loss and edentulism? A systematic Clin Sleep Med 2011;7:268–273.
review and meta-analysis. Obes Rev 2016;17:587–598. 251. Mohsenin N, Mostofi MT, Mohsenin V. The role of oral
235. Österberg T, Dey DK, Sundh V, Carlsson GE, Jansson appliances in treating obstructive sleep apnea. J Am
JO, Mellström D. Edentulism associated with obesity: Dent Assoc 2003;134:442–449.
A study of four national surveys of 16,416 Swedes aged
55–84 years. Acta Odontol Scand 2010;68:360–367.

133
5 Obesity, Metabolic Syndrome, and Oral Health

252. Dillow K, Essick G, Sanders A, Sheats R, Brame J. 260. Dioguardi A, Al-Halawani M. Oral appliances in obstruc-
Patient response to sleep apnea screening in a dental tive sleep apnea. Otolaryngol Clin North Am 2016;49:
practice. J Public Health Dent 2017;77:13–20. 1343–1357.
253. Serra-Torres S, Bellot-Arcís C, Montiel-Company JM, 261. Committee on Accelerating Progress in Obesity Preven-
Marco-Algarra J, Almerich-Silla JM. Effectiveness of tion. Accelerating Progress in Obesity Prevention:
mandibular advancement appliances in treating obstruc- Solving the Weight of the Nation. Washington, DC:
tive sleep apnea syndrome: A systematic review. National Academies, 2012.
Laryngoscope 2016;126:507–514. 262. Franks PW, Atabaki-Pasdar N. Causal inference in
254. Zhou J, Liu YH. A randomised titrated crossover study obesity research. J Intern Med 2017;281:222–232.
comparing two oral appliances in the treatment for mild 263. Shungin D, Cornelis MC, Divaris K, et al. Using genetics
to moderate obstructive sleep apnoea/hypopnoea to test the causal relationship of total adiposity and
syndrome. J Oral Rehabil 2012;39:914–922. periodontitis: Mendelian randomization analyses in the
255. Borrie F, Keightley A, Blacker S, Serrant P. Mandibular Gene-Lifestyle Interactions and Dental Endpoints
advancement appliances for treating sleep apnoea/ (GLIDE) Consortium. Int J Epidemiol 2015;44:638–650.
hypopnoea syndrome. Evid Based Dent 2013; 14: 264. Tavares M, Dewundara A, Goodson JM. Obesity preven-
27–28. tion and intervention in dental practice. Dent Clin North
256. Tegelberg A, Nohlert E, Bergman LE, Andrén A. Bed Am 2012;56:831–846.
partners’ and patients’ experiences after treatment of 265. Myers-Wright N, Lamster IB. A new practice approach
obstructive sleep apnoea with an oral appliance. Swed for oral health professionals. J Evid Based Dent Pract
Dent J 2012;36:35–44. 2016;16.
257. Gauthier L, Almeida F, Arcache JP, et al. Position paper 266. Lamster IB, Wolf DL. Primary health care assessment
by Canadian dental sleep medicine professionals on the and intervention in the dental office. J Periodontol 2008;
role of different health care professionals in managing 79:1825–1832.
obstructive sleep apnea and snoring with oral appli- 267. Hein C, Williams RC. The impact of oral health on
ances. Can Respir J 2012;19:307–309. general health: Educating professionals and patients.
258. Padmanabhan V, Kavitha PR, Hegde AM. Sleep disor- Curr Oral Health Rep 2017;4:8–13.
dered breathing in children—A review and the role of a 268. Schwartz MB, Chambliss HO, Brownell KD, Blair SN,
pediatric dentist. J Clin Pediatr Dent 2010;35:15–21. Billington C. Weight bias among health professionals
259. Simmons MS, Clark GT. The potentially harmful medical specializing in obesity. Obes Res 2003;11:1033–1039.
consequences of untreated sleep-disordered breathing: 269. Phelan SM, Burgess DJ, Yeazel MW, Hellerstedt WL,
The evidence supporting brain damage. J Am Dent Griffin JM, van Ryn M. Impact of weight bias and stigma
Assoc 2009;140:536–542. on quality of care and outcomes for patients with obesity.
Obes Rev 2015;16:319–326.

134
CHAPTER 6

Associations Between
Periodontal Disease and
Hyperglycemia/Diabetes
Wenche S. Borgnakke, dds, mph, phd
Robert J. Genco, dds, phd

Burden of Periodontal of the populations studied, with greater lev-


els of periodontitis found among people with
Disease diabetes.2,3
There is recent evidence for a much greater
Periodontitis is a dental plaque (biofilm)–initi- burden of periodontitis—assessed more
ated, host-mediated disruption of the microbial accurately than in the past4—among dentate
homeostasis (relatively stable equilibrium) that adults in the United States than hitherto esti-
in susceptible individuals manifests as destruc- mated due to the first National Health and
tion of soft and hard tissues surrounding the Nutrition Examination Survey (NHANES)
teeth.1 protocol including periodontal probing at six
There is a high burden of periodontal dis- sites around all non–third molar teeth during
ease globally.2,3 While gingival bleeding was the NHANES cycles 2009 to 2014. The orig-
found to be the most prevalent sign of disease inal 2007 US Centers for Disease Control
worldwide,2 severe periodontitis was the sixth and Prevention (CDC)/American Academy
most prevalent condition in the world, not of Periodontology (AAP) case definitions for
only among the chronic conditions.3 Based on surveillance of periodontitis had only three cat-
World Health Organization (WHO) data, the egories, namely severe, moderate, and mild/no
former report estimated 10% to 15% of adults periodontitis.5 In 2012, the mild/no group was
globally to have periodontal pockets at least split in two; the definitions are displayed in
6 mm deep.2 This estimate is in accord with Table 6-1 and are referred to as the CDC/AAP
findings from a systematic review of 72 studies, case definitions for surveillance of periodonti-
which included 291,170 individuals aged 15 tis. They are increasingly considered the global
years in 37 countries, reporting the prevalence standard and were used in several studies.
of severe periodontitis to be 11.2%, which did Figure 6-1 shows the prevalence of peri-
not change during the two decades from 1990 odontitis in the 2009 to 2014 NHANES cycles,
(95% confidence interval [CI]: 10.4%–11.9%) covering all 6 years during which full-mouth
to 2010 (95% CI: 10.4%–11.9%).3 Variations periodontal probing was conducted.7 Peri-
in prevalence among countries were largely odontitis is categorized by the 2012 updated
attributed to socioenvironmental conditions CDC/AAP case definitions.6 Notably, the prev-
and behavioral risk markers as well as poor alence of mild and severe periodontitis does
oral hygiene and poor general health status not change drastically by age but remains less

135
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

TABLE 6-1  |  Case definitions proposed for population-based surveillance of


periodontitis by the CDC/AAP*6
Case (category) Definition
Severe periodontitis > 2 interproximal sites with CAL > 6 mm (not on same tooth)
AND
> 1 interproximal site with PPD > 5 mm

Moderate periodontitis > 2 interproximal sites with CAL > 4 mm (not on same tooth)
OR
> 2 interproximal sites with PPD > 5 mm (not on same tooth)

Mild periodontitis > 2 interproximal sites with CAL > 3 mm


AND
> 2 interproximal sites with PPD > 4 mm (not on same tooth)
OR
1 site with PPD > 5 mm

No periodontitis No evidence of periodontitis

*Third molars excluded. CAL, clinical attachment loss; PPD, probing pocket depth.

80
Total PD Mild PD
70 Moderate PD Severe PD
60
Prevalence (%)

50
40
30
20
10
0
30 35 40 45 50 55 60 65 70 75 80
Age (y)

Fig 6-1  |  Prevalence of peri­odon­ti­tis (PD) by age among US dentate adults aged 30
6

years and older. Total PD includes mild, moderate, and severe cases.7

than or equal to 10% for mild and less than and 10.8%, respectively.7 Almost 60% (59.9%)
or equal to 15% for severe, in contrast to the of the dentate adults older than 65 years were
moderate category, which drives the major- affected by total periodontitis, and 9.0% of
ity of the prevalence increase by age of total these cases were severe.7
periodontitis (mild, moderate, or severe). The
10,683 NHANES participants who underwent
periodontal probing represent a weighted
population of approximately 143.8 million Burden of Diabetes
noninstitutionalized US adults aged 30 years Diabetes prevalence
and older.7 The overall prevalence of total peri-
odontitis of 42.2% included 7.8% with severe Diabetes mellitus is a group of metabolic disor-
periodontitis. The corresponding figures among ders characterized by hyperglycemia that results
those with self-reported diabetes were 59.9% from defective insulin action or production

136
Burden of Diabetes

TABLE 6-2  |  Criteria for diagnosis of prediabetes and diabetes, respectively, by the American
Diabetes Association (2018)9
Prediabetes Diabetes
Fasting plasma glucose (PG) 100–125 mg/dL (IFG) 5.6–6.9 mmol/L (IFG) > 126 mg/dL > 7.0 mmol/L
or or or or
2-h PG during 75-g oral 140–199 mg/dL (IGT) 7.8–11.0 mmol/L (IGT) > 200 mg/dL > 11.1 mmol/L
glucose tolerance test or or or or
A1C: 5.7–6.4% 39–47 mmol/mol > 6.5% > 48 mmol/mol

A1C, glycated hemoglobin; fasting, no caloric intake for at least 8 hours; h, hours; IFG, impaired fasting glucose; IGT, impaired glucose tolerance.

or both. In the United States, 30.3 million or million, and 119.8 million, respectively.10
9.4% of the entire population had diabetes in Population growth and aging, as well as urban-
2015, with 30.2 million being over 18 years ization resulting in changes in diet and exercise
old, equivalent to 12.2% of US adults, includ- habits, are likely to account for the majority of
ing one out of four adults aged 65 years and this worldwide increase.
older.8 Of these adults, 23.1 million were diag- Additionally, one-third (33.9%) of US adults
nosed, whereas almost one-quarter (23.8% or (84.1 million) had prediabetes with elevated
7.2 million) were unaware of their condition.8 blood glucose levels (hyperglycemia) that pre-
Overall, type 2 diabetes accounts for 90% to dispose to diabetes, with almost half of adults
95% of the cases. Type 1 or immune-mediated aged 65 years and older being affected.8 Hyper-
diabetes, caused by autoimmune beta-cell glycemia affects about 16.2% or one in six
destruction and gestational diabetes (GDM) (21.3 million) of the 131.4 million pregnancies
diagnosed in the second or third trimester worldwide, of which 86.4% of cases are due to
of pregnancy constitute the majority of the GDM.10 A large proportion of women experi-
remaining cases.9 Other types of diabetes due encing GDM will eventually develop manifest
to other causes are rare, such as maturity-onset diabetes, as they are about 17 times more likely
diabetes of the young (MODY) and pancre- to do so 3 to 6 years postpartum compared
atic disease– and drug- or chemical-induced with women without a history of GDM.11
diabetes.9 In this chapter, we will use the term hyper-
Globally, the estimated number of adults glycemia to mean long-term elevated plasma
aged 20 to 79 years with diabetes has almost glucose levels, which represents all forms of
tripled between the years 2000 and 2017, conditions and diseases such as prediabetes,
from 151 million to 425 million. About 326.5 manifest types 1 and 2 diabetes, gestational
million of these are of working age (20 to 64 diabetes, and MODY.
years old), and up to half (212.4 million) are
unaware of their condition.10 The continued
increase in the prevalence of diabetes is esti- Diabetes case definitions
mated to reach 691 million by the year 2045.10 The American Diabetes Association current-
In 2017, the United States had the third- ly classifies prediabetes and diabetes as shown
highest number of adults aged 20 to 79 years in Table 6-2, while stating that “Prediabe-
with diabetes globally—30.2 million—com- tes should not be viewed as a clinical entity
pared with China’s 114.4 million and India’s in its own right but rather as an increased
72.9 million. By 2045, these numbers are risk for diabetes and cardiovascular disease
expected to increase to 35.6 million, 134.3 (CVD)” and “Prediabetes is associated with

137
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

obesity (especially abdominal or visceral obe- Diabetes costs


sity), dyslipidemia with high triglycerides and/
or low high-density lipoprotein (HDL) choles- In parallel with the increasing prevalence of
terol, and hypertension.”9 diabetes, the economic burden of diabetes is
predicted to escalate, particularly among those
aged 70 years and older,10 with an estimated
Diabetes complications global increase of USD 104 billion (B) between
Diabetes complications are similar for any 2017 and 2045, with these costs estimated at
type of diabetes because it is mainly hypergly- USD 727 B in 2017.10
cemia (especially of long duration) that leads In the United States, the overall cost of dia-
to the complications. Acute and chronic com- betes increased by about 25% from 2012 to
plications include dehydration, poor wound 2017.19 An estimate for the total economic cost
healing, stroke, myocardial infarction, criti- of diabetes in the United States in 2017 was
cal limb ischemia, ketoacidosis, hyperosmolar USD 327 B, of which USD 237 B was attributed
coma, kidney failure, retinopathy/blindness, to direct medical costs and the remaining USD
neuropathy, neurocognitive function/cognitive 90 B to indirect costs, distributed among absen-
decline, and serious infections possibly requir- teeism (USD 3.3 B), presenteeism (reduced
ing below-knee amputation.12 productivity while at work, USD 26.9 B),
Diabetic retinopathy is the leading cause of unemployment/long-term disability (USD
new cases of blindness among adults aged 20 37.5 B), reduced productivity when not in the
to 74 years, and diabetic kidney disease is the workforce (USD 2.3 B), and premature mortal-
leading cause of kidney failure in the United ity (USD 19.9 B).19 Diabetes greatly increases
States.12 Of all cases of blindness, the propor- annual health care costs.20 The average annual
tion that is due to diabetic retinopathy is rising, medical expenditures among people with dia-
with 1 of 39 cases globally being due to dia- betes were 2.3 times higher than what would
betic retinopathy.13 Overall, about one-third be expected in the absence of diabetes in both
of all people with diabetes have diabetic reti- 201221 and 2017,19 with the 2017 mean cost
nopathy.10 Furthermore, about one-tenth (8%) increase of between $7,510 for those younger
of individuals with newly diagnosed diabetes than 18 years to greater than $13,000 for those
have already developed diabetic neuropathy, 65 years of age and older.19 Similarly, lifetime
increasing to about 50% of those with diabe- health care expenditures were estimated to be
tes of long duration.14 Lastly, individuals with between $8,946 and $159,380 greater in people
diabetes are two to three times more likely to with diabetes, depending on demographic fac-
have CVD and up to 10 times more likely to tors and degree of obesity.17 It is estimated that
have end-stage renal disease (ESRD). the Veterans Administration could save up to $2
People with diabetes with chronically poor B a year via screening and early intervention.22
metabolic control experience morbidity, heart
disease, and stroke rates that are two to four
times higher than those in individuals without
diabetes15,16; subsequently, diabetes is found to Links Between Periodontal
shorten life expectancy by 3.3 to 18.7 years Disease and Hyperglycemia/
among US adults.17 Furthermore, pregnant
women with GDM are at high risk of trans- Diabetes
generational effects in their offspring, such as
obesity,18 diabetes, hypertension, and kidney Unfortunately, there is general lack of aware-
disease.10 ness of the links between diabetes and
periodontitis in spite of reports that a majority

138
Links Between Periodontal Disease and Hyperglycemia/Diabetes

of adults with type 2 diabetes suffer from some Mechanisms linking diabetes and
form of periodontitis.23 A considerable body of
periodontitis
evidence shows a close bidirectional associa-
tion between diabetes and periodontal disease.6 Type 2 diabetes is characterized by a systemic
Evidence for these associations is reviewed in inflammatory process that leads to insulin re-
this chapter along with its implications. While sistance and reduced pancreatic B-cell function
most of the content regards chronic periodon- and apoptosis, both of which cause increas-
titis, we use the term periodontal disease to ing hyperglycemia. Periodontitis contributes
also include gingivitis when applicable. Howev- to systemic inflammation, which adversely
er, periapical periodontitis is specified as such. affects glycemic control in patients suffering
Other periodontitis types, such as early onset/ from diabetes. The elevated systemic inflam-
aggressive periodontitis, are not addressed in mation associated with periodontitis results
this chapter due to their relatively low preva- mainly from the entry of periodontal organisms
lence and scant evidence for their association and their virulence factors into the system-
with diabetes. ic circulation. There is also strong evidence
that advanced glycation end products result-
ing from hyperglycemia lead to activation of
Shared risk markers for macrophages. Macrophages so activated then
periodontitis and diabetes produce inflammatory cytokines and reactive
oxygen species that increase the periodontal
With the current notion of inflammation being soft and hard tissue destruction found in pa-
considered the major mechanism underlying tients with diabetes24 (Fig 6-2).
the associations between periodontal disease The effects of hyperglycemia/diabetes on
and hyperglycemia, these close links become periodontitis and of periodontitis on hyper-
biologically plausible as both diseases lead to in- glycemia/diabetes are likely based on them
flammation. As well, both the modifiable23 and both contributing to a hyperinflammatory
nonmodifiable24 risk markers for the two dis- state, which may explain the bidirectional
eases are largely identical, and they are shared relationship between periodontitis and dia-
with other chronic diseases, such as athero- betes.25 Hyperlipidemia is also suggested to
sclerotic cardiovascular disease (ACVD). This play an important role in this relationship,
is why it is important to control for potential as it is associated with an increase in both
confounders that can introduce bias in analyses diseases.26 Emerging evidence for the effect
that seek to identify relationships—especially of periodontitis on diabetes27,28 suggests that
potentially causal relationships—between cer- periodontitis not only contributes to wors-
tain markers that in reality are due to other ened glycemic control but also is associated
reasons than those analyzed. with increased mortality and morbidity from
In order to avoid any finite discussion or CVD and diabetic nephropathy.29–31 However,
determination of whether there is sufficient sci- a German study found no interaction between
entific evidence for a given condition to indeed periodontitis and diabetes and therefore sug-
be considered a true risk factor, defined as a gested that these two diseases are independent
contributory cause of a condition in question, risk factors for both all-cause and CVD-related
the expression risk marker or simply marker mortality.32 There is also emerging evidence
is used throughout this chapter to express the of a role for periodontitis in the develop-
notion of an indicator or predictor or determi- ment of new cases of type 2 diabetes33,34 and
nant or driver that could be a true risk factor possibly GDM, although a 2016 systematic
involved in the causal pathogenic pathway for review of the latter35 included only three small
the disease in question. case-control36–38 and five cross-sectional studies.

139
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

Ligand RAGE
RAGE

Activation
Periodontal microbiota RAGE activation Nucleus

Exaggerated and
Impaired repair
sustained inflammation

Enhanced periodontal tissue breakdown

Fig 6-2  |  A “two-hit” model for the pathogenesis of accelerated periodontal destruction in patients with diabe-
tes mellitus. Bacterial challenge in an environment of enhanced RAGE expression, such as the periodontium of
an individual with diabetes mellitus, leads to exaggerated inflammation and impaired tissue repair, which in turn
cause accelerated and severe periodontal destruction.24 RAGE, receptor for advanced glycation end products.

Thus, prospective cohort studies are needed to Effects of hyperglycemia


ascertain any potential effect of periodontitis
on the development of GDM. Periodontitis
Poorly controlled diabetes is a risk marker for
periodontal disease. This has been known for
many years, but somehow the concept that sim-
Evidence for the ply “diabetes” without any quantifiers such as
Bidirectional Relationship “uncontrolled” or “poorly controlled” and “of
Between Periodontitis and long duration” affects periodontal tissue has
been generally accepted. In 1993, Löe summa-
Diabetes rized the scientific literature and concluded that
periodontitis should be regarded as the sixth
Diabetes and periodontal disease are complex complication of diabetes.44
chronic diseases that affect each other and the The effects of hyperglycemia on periodon-
evidence for this bidirectional relationship has tal tissues have been confirmed by several
been reviewed by several authors.24,39–43 studies of various designs, most importantly

140
Evidence for the Bidirectional Relationship Between Periodontitis and Diabetes

prospective cohort studies that allow con- periodontitis5,6 for each 1-point increase in
clusions regarding the temporal sequence mean A1c, hence regarding A1c as a linear pre-
and therefore of potential causation. That is, dictor.47 Using the A1c cut-off values of 7.0%,
hyperglycemia occurred before the periodontal 7.5%, 8.0%, 8.5%, and 9.0%, a distinctive,
disease. statistically significant dose-response pattern in
the odds for having periodontitis in the groups
Longitudinal studies.  Elevated blood glucose with A1c less than or greater than or equal to
level and an increase in inflammatory mark- the cut-off compared to the referent group
ers were associated with periodontal disease without diabetes was clearly demonstrated. For
in a cohort of individuals with diabetes fol- instance, those with diabetes with A1c greater
lowed for a period of 5 years.45 This 5-year than or equal to 7.0% had 33% greater odds
population study in Germany included 2,626 for periodontitis than the no-diabetes group;
dentate men and women aged 20 to 81 years, those with A1c greater than or equal to 9.0%
in which participants with uncontrolled type had over two-fold (2.22) greater odds of having
1 or type 2 diabetes had both greater preva- periodontitis than people without diabetes.47
lence and greater progression of both probing Using the same NHANES 2009 to 2012
pocket depth (PPD) and clinical attachment data, similar patterns were reported for total,
loss (CAL) compared to those with controlled severe, and nonsevere (moderate or mild) peri-
or no diabetes.45 odontitis,5,6 respectively, even in people with
prediabetes who are at high risk of develop-
Cross-sectional studies. Even though cross- ing diabetes.48 Compared to those without
sectional studies cannot provide information diabetes, the prevalence of total (mild, moder-
regarding the temporality of associated factors, ate, or severe) periodontitis was 43% greater
a few examples of large population studies in patients with prediabetes, 67% greater in
deserve mention. patients with controlled diabetes, and 72%
The cross-sectional Study of Health in greater in patients with uncontrolled diabetes;
Pomerania in northeastern Germany (SHIP)- the corresponding figures for severe periodon-
Trend that included 3,086 men and women titis were 66%, 112%, and 122%, respectively.
between the ages of 20 and 82 years reported Such a dose-response relationship between
that whereas prediabetes (n = 576) and increasing fasting glucose levels in prediabetes
well-controlled (glycated hemoglobin [A1c] < through diabetes and increasing prevalence of
7.0%) type 2 diabetes (n = 137) were not asso- periodontitis was also reported upon analyses
ciated with periodontitis, people with poorly of data from 9,977 participants in the 2012–
controlled (A1c > 7.0%) type 2 diabetes (n = 2013 Korea National Health and Nutrition
64) had greater mean CAL than normoglyce- Examination Survey (KNHANES).49
mic participants in the fully adjusted model.46 These and several other studies have docu-
However, this association was weaker than mented the greater incidence (development of
expected, and the association between poor new cases), prevalence, extent, and severity of
glycemic control and mean PPD was not sig- periodontal disease. The greater severity and
nificant, possibly due in part to the low number earlier onset of periodontitis in patients with
of participants having poorly controlled type type 2 diabetes were clearly demonstrated in a
2 diabetes. classic epidemiologic study of the Native Amer-
Analysis of the nationally representative icans of the Gila River Indian Community in
NHANES 2009 to 2012 data from 7,042 Arizona.50 Even though results from studies
adults aged 30 years and older suggested a among this special population group may not
dose-response relationship with a signifi- be directly generalizable to other populations,
cant increase of 18% in the odds of having they still serve to illustrate the concept.

141
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

Tooth eruption there still exists a two-fold difference in number


In a cohort of 700 children aged between 6 of missing teeth between those with and with-
and 14 years in Manhattan, half of whom had out diabetes.56 During those four decades, the
diabetes (about 98% type 1 diabetes) with number of missing teeth decreased from a mean
18% very poorly controlled (A1c > 9.5%) and of 11.2 to 6.6 teeth missing in US adults with
38% of them being “overweight and at risk diabetes versus 9.4 to 3.4 in the no-diabetes
for overweight,” children aged 10 to 14 years group.
with diabetes experienced early tooth eruption The finding of an association between tooth
compared to their peers without diabetes.51 Ex- loss and diabetes is not limited to the United
aminations of 4,876 children participating in States. A German study showed greater tooth
the NHANES 2009 to 2014 suggested that loss over 5 years in both type 1 and type 2
overweight or obese children experienced 6 diabetes when uncontrolled compared to the
to 12 months accelerated eruption of the first groups with well-controlled or no diabetes.45 In
and second molars.52 Thus, even though the the SHIP-Trend study, participants with poorly
study in Manhattan found only a weak associ- controlled type 2 diabetes had more missing
ation between obesity and early tooth eruption, teeth than normoglycemic participants (OR:
obesity status may have contributed to the ob- 2.19; 95% CI: 1.18–4.05).46 Among 24,313
served finding. middle-aged Germans, there was a clear inverse
relationship between decreasing number of
teeth present and increasing prevalence of
Tooth loss diabetes.57
The ultimate outcome of untreated periodonti- Analyses using a French database with
tis, namely loss of teeth, is greater in individuals 533,378 people—including 27,305 individuals
with hyperglycemia.53 An analysis of NHANES with type 2 diabetes—concluded that compared
2003 to 2004 data from 2,508 participants aged to people without diabetes, people with type
50 years and older showed that people with di- 2 diabetes not only had greater loss of teeth
abetes had more missing teeth (9.8 versus 6.7 at all ages, but this loss also occurred at an
teeth) and were more than twice as likely to be earlier age, peaking among 40- to 60-year-olds,
edentulous than those without diabetes upon whereas the tooth loss steadily increased with
adjustment for other factors (adjusted odds age in those without diabetes.58
ratio [aOR]: 2.25; 95% CI: 1.19–4.21) with Analyses of data from the KNHANES 2008
28% versus 14% being edentulous.53 More to 2012 study reported much greater tooth
native Hawai’ians with diabetes had lost six loss among 2,078 adults with type 2 diabetes
teeth or more compared to those without di- compared to diabetes-free participants.59 More-
abetes (OR: 1.64; 95% CI: 1.15–2.35)54; and over, those with fewer teeth present were more
among 15,945 Hispanic Americans aged 18 likely to also have diabetic retinopathy after
to 74 years, those with uncontrolled diabe- adjustment for potential confounders, includ-
tes had a greater risk for missing nine teeth ing age. For example, those with fewer than
than their normoglycemic counterparts, espe- 20 natural teeth were 8.7 times more likely to
cially among the 18- to 44-year-olds, whereas have diabetic retinopathy than those with at
no such associations were detected in those least 28 teeth.
with well-controlled diabetes or prediabetes.55 A small Japanese study reported that CVD
Even though there is a global strong trend patients with co-occurring diabetes were miss-
toward loss of fewer teeth over time, as ex- ing significantly more teeth than CVD patients
emplified in a study of NHANES data from without diabetes.60
37,609 dentate adults aged 25 years and older This knowledge may assist dental care pro-
over the 40-year period from 1971 to 2012, fessionals in suspecting possible undiagnosed

142
Evidence for the Bidirectional Relationship Between Periodontitis and Diabetes

diabetes in their patients who are not aware of clinical periodontal examinations followed the
their diabetes status. However, when assessing participants for a mean of 7.8 years.62 Eighty
studies of tooth loss, the impact of smoking (6.0%) men developed type 2 diabetes, and
must always be considered. This is unfortu- those with moderate/severe periodontitis5 at
nately not always the case. baseline had almost 70% greater risk for de-
veloping diabetes compared to men with no/
mild periodontitis at baseline (hazard ratio
Effects of periodontitis on [HR]: 1.69; 95% CI: 1.06–2.69).62 Similar-
glycemic control ly, a 2017 retrospective cohort study among
80 adults with type 2 diabetes reported that
Based on the available emerging evidence and a significant increase in A1c during the 3-year
the underlying suggested mechanisms, it is study occurred both in those with severe peri-
reasonable to suggest that periodontitis may odontitis at baseline as well as in participants
impact glycemic control and the incidence and with CAL progression during the study.63 The
complications of diabetes. As an example of greatest increase in A1c level occurred in par-
microbiologic dysbiosis, Demmer et al found by ticipants with baseline severe periodontitis and
analyzing 1,188 subgingival plaque samples for A1c less than 6.5% who actually had either pre-
11 periodontal bacteria that greater abundance diabetes or no diabetes, as per the current case
of certain periodontal bacteria was associat- definitions.9
ed with greater prevalence of prediabetes in Nonetheless, a large, well-designed, and
300 diabetes-free adults aged 20 to 55 years.61 well-executed population study concluded in
It is especially important to know whether peri- 2017 that “Contrary to the currently available
odontitis leads to elevation of blood glucose literature, no convincing evidence was found of
levels in normoglycemic individuals and people any potential association between periodontitis
with prediabetes as they are principally those and diabetes incidence or HbA1c change.”64 This
who stand to benefit most from interventions was the result of an 11-year SHIP follow-up
aimed at preventing the development of pre- study that included 2,034 German dentate
diabetes and its subsequent manifest diabetes. adults aged 20 to 81 years at baseline. A total
There is evidence that periodontitis can of 180 patients who were dentate and initially
adversely affect glycemic control in persons diabetes-free developed diabetes during the
with diabetes, further supporting the bidirec- study. However, upon controlling for potential
tional relationship of the two diseases. This confounders, importantly age and sex, neither
evidence was summarized in a 2013 systematic baseline CAL nor PPD were significantly associ-
review of the evidence in which the direction ated with diabetes incidence or with long-term
of the effect could be ascertained.27 An updated changes in A1c levels. The authors list six poten-
systematic review in 2018 reached the same tial limitations, none of which immediately
conclusions.28 No prospective cohort studies seem to indicate any strong bias that could lead
exploring the effect of periodontitis on predi- the reader to doubt these results. However, three
abetes were identified by the two systematic major circumstances need mention. First, as the
reviews,27,28 but more recent studies are reported authors point out, periodontitis cases might
and mentioned in the following sections. have been missed or misclassified by applying
the random half-mouth periodontal probing
at only four sites around each tooth. This is
Longitudinal studies because periodontitis manifests as a site-specific
A 2017 prospective cohort study among 1,331 breakdown of soft and hard periodontal tissues
initially diabetes-free men aged 58 to 72 years that does not occur in any symmetric pattern
in Northern Ireland who underwent baseline around any tooth, within an arch, or in the

143
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

entire dentition. Second, the general trend in the can improve glycemic control. Table 6-3 pro-
region over the decade during which the study vides an overview of systematic reviews with
was conducted was a decline in the number meta-analyses of such trials, mostly concluding
of new cases of diabetes along with improved that nonsurgical treatment of periodontitis in
glycemic control in people with existing diabe- patients with diabetes can result in a modest
tes, possibly due to generally improved medical reduction of HbA1c of 0.2% to 0.4% at 3
care in this part of the former East Germany. months.
Third, the prevalence of severe periodontitis5,6 For type 2 diabetes at 3 to 4 months after
also seems to have decreased during these 11 scaling and root planing (SRP), the decrease in
years: “For example, among the 40-60 years old A1c level varies between 0.18%77 and 1.03%,74
participants, 25%, 31%, and 36% were catego- while no review reports an A1c increase. Inter-
rized as severe periodontitis in SHIP-0 at HbA1c estingly, the smallest improvement was seen
levels < 5.7%, 5.7% to 6.4%, and ≥ 6.5%, with the use of adjunct antimicrobials.77 As also
respectively. The corresponding figures 10 years concluded by a 2015 Cochrane review, SRP
later (SHIP-Trend) were 19%, 22%, and 25%, without any adjunct antimicrobials seems to
respectively.”65 The latter two circumstances lead to an A1c improvement of around 0.4%.77
are described in more detail elsewhere65 and This effect is of about the magnitude that could
may be important in interpreting the findings. be expected from adding a second anti-diabetic
However, it is not possible to know whether oral medication to metformin.82
following a full-mouth, six-sites-per-tooth peri- The largest RCT conducted so far involved
odontal probing protocol and controlling for 257 subjects with type 2 diabetes and 257 sub-
both the improved diabetes prevalence and gly- jects without diabetes at five centers.83 This study
cemic control and for the decline in prevalence failed to observe any improvement in A1c level
of severe periodontitis5,6 might have produced but attracted substantial critique, such as its fail-
different results. ure to provide successful treatment that resulted
in acceptable periodontal health. At the end of
the 6-month study, a great proportion of the
Effects of nonsurgical periodontal participants had a periodontal health status suf-
treatment on glycemic control ficiently poor to be eligible for inclusion into the
study. With 72.1% of the sites showing plaque
It has long been generally accepted that acute accumulation and 41.6% showing bleeding on
and chronic infections cause elevation of the probing, and a mean of 15.7 sites with PPD
blood sugar levels as a normal part of the in- greater than or equal to 5 mm per participant,
flammatory response and thus adversely affect affecting 10.2% of the probed sites, it is rea-
glycemic control in patients with diabetes.66 sonable to expect the residual inflammation to
Glycemic control typically improves once continue to negatively impact the A1c. Hence, an
such infections are resolved. A 2015 systematic alternate conclusion would have been that due
review concluded that nonsurgical periodontal to the failure to control the participants’ peri-
treatment can reduce the level of inflammatory odontal inflammation, no conclusion could be
markers, such as C-reactive protein (CRP) and drawn regarding what the outcome would have
tumor necrosis factor alpha (TNF-α).67 There- been with successful periodontal treatment to a
fore, it is biologically reasonable to predict prespecified endpoint of periodontal health.84–87
that resolution of periodontal infections may Subsequently, several authors have reviewed
improve glycemic control in people with diabe- the systematic reviews to provide an overall
tes. Several randomized controlled trials (RCTs) assessment of the integrated evidence.88–92 Such
have addressed the question of whether, and to overviews may be termed umbrella reviews.93
what extent, nonsurgical periodontal treatment Most of these overviews of systematic reviews

144
Evidence for the Bidirectional Relationship Between Periodontitis and Diabetes

TABLE 6-3  |  Effect of nonsurgical periodontal treatment on glycemic control in type 2 diabetes:
Meta-analyses in systematic reviews of RCTs published through April 6, 2018
No. of Pooled A1c
Author (year) RCTs N change %a 95% CI P value
Teeuw et al (2010)
68
3 (+ 2 CCTs) 180 –0.40 b
–0.77; –0.04 .03
Simpson et al69 (2010) 3 244 –0.40 –0.78; –0.01 .04
Sgolastra et al (2013)
70
5 315 –0.65 –0.88; –0.43 < .05
Engebretson and Kocher71 (2013) 9 775 –0.36 −0.54; −0.19 < .0001
Liew et al72 (2013) 6 422 –0.41 –0.73; –0.09 .013
Corbella et al73 (2013)
  6-month follow-up 3 235 –0.31 –0.74; 0.11 .15
Sun et al74 (2014) 10 587 –1.21 –1.68; –0.75 .000
  3-month follow-up 6 515 –1.03 –1.7; –0.31 .003
  6-month follow-up 3 150 –1.18 –1.64; –0.72 < .001
  9-month follow-up 1 72 –1.90 –2.2; –1.6 NA
Wang TF et al (2014)
75
3 143 –0.24 –0.62; 0.14 .217
Li et al76 (2015) 9 1,082 –0.27 –0.46; –0.07 .007
Simpson et al77 (2015)
  3–4-month follow-up SRP + AM 15 1,499 –0.29 –0.48; –0.10 .003
  3–4-month follow-up: SRP only 8 547 –0.41 –0.73; –0.08 .013
  3–4-month follow-up: SRP + AM 7 952 –0.18 –0.39; 0.03 .092

  6-month follow-up: SRP + AM 5 826 0.02 –0.20; 0.16 .84


  6-month follow-up: SRP only 3 263 –0.18 –0.58; 0.22 .38
  6-month follow-up: SRP + AM 2 563 0.02 –0.18; 0.22 .83
Teshome and Yitayeh78 (2016)
  3-month follow-up: SRP + AM 7 940 –0.48 –0.78; 0.18 .002
  At end of intervention 7 940 –0.53 –0.81; –0.24 < .001
  SRP plus AM/mouthwash 3 584 –0.51 –1.0; –0.03 .04
  SRP without AM/mouthwash 4 356 –0.53 –0.87; –0.19 .002
Studies with one study including type 1 DM each: (DM 1/DM 2)
Janket et al (2005)
79
1(+4 non-RCT) 268 (7/261) –0.66c –2.2; 0.9 NS
–0.71d –2.3; 0.9 NS
Darré et al80 (2008) 9 485 (53/432) –0.46b –0.82; –0.11 .01
Corbella et al (2013)
73

  3-month follow-up 15 678 (12/666) –0.38 –0.53; –0.23 .001


Wang et al (2014)
81

  3-month follow-up 10 1,135 (39/1,096) –0.36 –0.52; –0.19 < .001


  6-month follow-up 4 754 (39/715) –0.30 –0.69; 0.09 .13
a
percentage point = actual (absolute) difference (not relative difference); b standardized mean difference; c type 2 diabetes, except seven participants
with type 1 diabetes, without adjunct antibodies; d type 2 diabetes, except seven participants with type 1 diabetes, with adjunct antibiotics.
Bolded text: statistical significance (P < .05).
AM, antimicrobials; DM1, type 1 diabetes; DM2, type 2 diabetes; N, number of study participants; NA, not available; NS, not statistically significant;
SRP, scaling and root planing (“deep cleaning”).

145
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

and meta-analyses conclude that there is evi- Screening for diabetes in the
dence to suggest that nonsurgical periodontal
dental office
treatment can lead to a small decrease in A1c
but that the quality of the original RCTs vary Diabetes mellitus is a common metabolic disor-
and their bias prevents any firm conclusion. der among patients who receive care in dental
Further studies are clearly needed in various practices. As previously discussed, periodontal
patient groups, such as those who are resis- disease adversely affects metabolic control in
tant to antidiabetic drug therapy and lifestyle individuals with diabetes. Furthermore, tooth
interventions and whose glycated hemoglobin loss from dental caries and periodontal disease
levels remain elevated. In addition, optimal can lead to poor nutrition, compromising di-
periodontal treatment aimed at reducing the etary efforts at glycemic control and weight loss
extent and severity of periodontitis is necessary in individuals with diabetes. Hence, dental care
to establish whether and which treatments help aimed at maintaining a healthy dentition free of
in affecting glycemic control. Most periodontal caries and with ideal periodontal health is nec-
intervention studies were undertaken for 3 to 6 essary for optimal care of patients with diabetes.
months, but longer studies are needed. Knowledge of the glycemic status of dental
Importantly, the group of adults aged 55 patients, especially those with periodontal dis-
years and older has been largely ignored or ease and other oral manifestations of diabetes,
sparsely represented in intervention studies.94 is important for their oral as well as overall
The original target group for a 2017 review care. Uncontrolled diabetes is associated with
was people 65 years old and older, but almost increased progression of periodontal disease.96
no evidence was available, so the change to Early diagnosis of diabetes and stabilization
a lower age limit was necessary.94 Yet it was of glycemic control can reduce the risk of
demonstrated that a combination of lifestyle complications. Furthermore, early detection
changes and dental care led to improved gly- of prediabetes and intensive lifestyle changes
cemic control and periodontal health after 6 can reduce the incidence of type 2 diabetes.
months in a small study of Thai adults aged Therefore, early diagnosis of diabetes and iden-
60 years and older with diabetes (n = 66 inter- tification of patients at high risk for diabetes (ie,
vention, n = 66 control).95 The few studies those with prediabetes) are important for the
available for review seem to suggest that older overall management of diabetes and especially
individuals with diabetes might benefit from important for patients who are also seeking
nonsurgical periodontal therapy to decrease dental care, including periodontal treatment
blood levels of A1c. and dental implants. Patterns of health care
If the potential effect on glycemic control is utilization suggest that many people obtain
reproducible and long term, nonsurgical peri- routine, preventive dental care more often than
odontal therapy could be considered a part of such medical care, so the dental office has been
the management of diabetes. This positive out- proposed as a site for screening to identify risks
come would be above and beyond the beneficial for undiagnosed systemic chronic diseases in
effects of resolving periodontal infection to pre- asymptomatic patients who are unaware of
serve the dentition, which in turn would lead to their risks and have not paid a visit to their
several desirable outcomes, among them better medical care provider within a year.97
nutrition, which would contribute indirectly In a dental clinic population, prediabetes was
to diabetes control. Studies are also needed predicted in more than 90% of patients having
to determine if the prevention or treatment four or more missing teeth, periodontal probing
of periodontal disease and possibly other oral depths greater than or equal to 5 mm at about
infections can also reduce other complications 25% of sites, and a capillary blood HbA1c level
of diabetes, including CVD and nephropathy. greater than 5.7%.98 In a dental office field trial,

146
Evidence for the Bidirectional Relationship Between Periodontitis and Diabetes

diabetes screening was carried out for 1,022 patient compliance with the referral to seek
dental patients aged 45 years and older who a medical diagnosis. Many of these barriers
had no knowledge of their diabetes status.99 can likely be overcome in the future. Capillary
More than 40% of the 1,022 dental patients blood screening for A1c is a feasible, accurate
had a capillary blood HbA1c level greater than method of screening for diabetes and predia-
5.7%, defined as hyperglycemia (see Table 6-1), betes in a dental practice. The yield could be
and were therefore referred to a physician for improved by triaging patients based on prior
evaluation. Among the patients referred and existing periodontal disease, missing teeth, and
for whom a diagnosis was returned to the den- their risk profile for diabetes.
tal office, 12% had diabetes and 23% were at Diabetes screening at clinical sites such as
high risk for diabetes (had prediabetes). Hence, dental offices and pharmacies could be of great
there seems to be the potential for detection service because about one-quarter (23.8%) of
of diabetes in one of eight—and for prediabe- individuals with manifest diabetes are unaware
tes in about one of four—dental patients aged of their condition.8 Furthermore, about 90%
45 years and older who are unaware of their of those with prediabetes are unaware of their
diabetes status.99 It should be mentioned that condition, and a large percentage of individuals
about 80% of the dental patients were seen with prediabetes are unaware of their high risk
in community health centers and about 20% for the development of diabetes. As previously
in private dental offices in Providence, Rhode mentioned, early treatment of diabetes can lead
Island, or its vicinity.99 It should be noted that to better glycemic control with fewer medica-
HbA1c is a reliable measure for preclinical tions and fewer complications. In addition,
screening for diabetes since it is also one of lifestyle modifications including loss of 7% to
the measures used in the diagnosis of diabetes 10% of weight and exercising 30 min/day for
and is not changed by eating in the previous 5 days/week in patients with prediabetes can
12 hours as are random glucose measurements. prevent the onset of diabetes at least over a
A similar study was conducted in 13 general 3-year period.101 This is one of the few chronic
dental practices in southeast Michigan among diseases for which a safe and effective regimen
1,033 dental patients aged 30 years and older has been developed to prevent its onset.
who denied having diabetes. Almost one-third Of course, patients often may not comply
(30%) of the 181 patients who were selected with lifestyle changes such as dietary changes
based on their random capillary glucose lev- or exercise regimens on a long-term basis.
els and periodontal health status actually had Dental professionals can help patients comply
hyperglycemia with 1.3% suffering from man- with these regimens because dentists often see
ifest diabetes and 28.7% with prediabetes.100 It these patients regularly; the dental team can
turned out that patients at high risk for hyper- reinforce the need for lifestyle modification to
glycemia could be identified based on only sustain diabetes prevention or management.
the following self-reported information: age, Due to the large proportion of individuals
sex, hypertension, hypercholesterolemia, and with undiagnosed diabetes and because the vast
self-reported tooth loss, weight, and height, with majority of type 2 diabetes cases—as well as
selection of those with body mass index (BMI) complications of all diabetes types—largely can
greater than or equal to 35 kg/m2. The authors be prevented or ameliorated by early diagnosis,
concluded that random capillary glucose level intervention, and management, oral health care
aided the identification but was not necessary. professionals can contribute in meaningful ways
Barriers to full implementation of HbA1c to this significant issue. Furthermore, potential
screening for prediabetes and diabetes in dental health benefits and improved quality of life, as
practices include cost, lack of insurance reim- well as actual costs associated with dental office
bursement, and, perhaps most importantly, screening for diabetes and interprofessional

147
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

management thereof, need to be rigorously calories and lower levels of carbohydrates.110


evaluated by well-designed randomized field The change in the diet likely resulted in part
clinical trials under actual practice conditions. from greater retention of teeth, and this is likely
to be associated with better overall health.
A study of largely older African American
Nutritional and dietary and American Indian adult populations showed
consequences of tooth loss that participants with fewer than 10 teeth con-
sumed less fruit, meat, beans, and oils. They
Tooth loss, especially edentulism, can result in also derived more energy from solid fat, alco-
difficulty in chewing, which leads to poorer nu- hol, and added sugar than did individuals with
trition, and in poor esthetics, a negative social 11 or more teeth. Nutritional intervention for
image, lower self-esteem, and an overall lower older individuals should certainly consider the
quality of life.102 Periodontitis and severe den- effects of tooth loss on dietary patterns, espe-
tal caries are the primary reasons for tooth loss cially in patients with diabetes.111
in adults.103 Factors such as low educational Overall, it is likely that the impact of peri-
level, low socioeconomic status, greater age, odontitis on diabetes is in part related to the
poor access to care, lack of health insurance altered diet associated with tooth loss. There-
coverage, and a history of smoking are risk fore, oral health care professionals should
markers for tooth loss. highlight to patients the importance of main-
An association between tooth loss and taining oral health with an emphasis on the
reduced consumption of important dietary con- ability to eat any and all types of foods.
stituents, including fiber, fruits, and vegetables,
has been reported,104 and tooth loss was found
to independently predict a 30% lower intake of Other oral manifestations of
both overall calories and protein, respectively, hyperglycemia
in a study of 2,749 people with chronic kid-
ney disease participating in the 1988 to 1994 Complaints of xerostomia (subjective feeling
NHANES.105 of dry mouth) may be associated with thirst,
In addition, tooth loss is associated with which is a common complaint among patients
a higher intake of refined sugars and satu- with diabetes. Xerostomia may also result
rated fatty acids106 and with consumption from sensory nerve dysfunction as a neurologic
of decreased food variety,107 and it has been complication of diabetes that can lead to hypo­
shown to severely diminish the nutritional sta- salivation (objective measure of decreased saliva
tus among elderly.108 A national study among secretion).112 Dry mouth may also be caused by
8,446 Finns followed for 13 years found the medications. If medications are determined to
missing of nine or more teeth to predict inci- be the cause of the xerostomia, the oral health
dent diabetes and even all-cause mortality.109 care professional should consider consulting
A review of the overall health and food with the patient’s physician who then could
habits among older adults in Japan also sheds change the medication or alter the dose, fre-
light on the relationship between tooth loss quency, or timing, which may provide relief for
and diet.110 A program has been in existence the patient. If this is not feasible, patients may
since 1991 to increase the number of individ- benefit from the use of sugar-free gum or candy
uals who reach the age of 80 years with 20 as well as non–alcohol-containing mouthrinses
teeth (the “80/20” program). The percentage of or saliva substitutes.
80/20 achievers increased from 10.9% in 1993 Diabetic neuropathy may moreover manifest
to 24.1% in 2005. Those who had retained in the oral cavity as a burning sensation in the
20 teeth at 80 years old consumed fewer total mouth, which in turn may impede proper oral

148
Evidence for the Bidirectional Relationship Between Periodontitis and Diabetes

Fig 6-3 |  Proportion of US adults 74–


who had a dental visit in the past year 72–
by diabetes status.114 70–

Percentage
68–
66–
64–
62– Diabetes
60– Prediabetes
58– No diabetes
56–
2004 2006 2008 2010 2012 2014

Year

hygiene. The treatment protocols for xerosto- support persons.113 Referral to a “dentist for
mia are sometimes effective in reducing this comprehensive dental and periodontal exam-
burning sensation. Diabetic neuropathy can ination” is explicitly recommended as part of
also result in taste impairment in patients the initial diabetes care management.113 As
with diabetes.112 This dysfunction can seriously well, the association recognizes that periodon-
affect the patient’s ability to maintain a proper titis is more severe in people with diabetes and
diet, which is central to glycemic control. The that periodontal infection is suggested to ad-
management of diabetic neuropathy may versely impact diabetes outcomes but that the
improve taste sensation. However, such treat- evidence for the effect of nonsurgical periodon-
ment is often ineffective. In any case, patients tal treatment is inconclusive.113
with altered taste sensation should be made Mutual referral by dental and medical profes-
aware that this is a side effect of diabetes and sionals could contribute to overall health, which
that they should make efforts to consume a diet includes both oral/dental and systemic health.
recommended to maintain healthy blood sugar People with diabetes in the United States have
levels, as well as control their weight while cop- been known for many years to visit dentists
ing with the altered taste sensation. much less frequently than their diabetes-free
counterparts. A 2018 study illustrates this sit-
uation with analyses of data collected from
Referral for dental examination by 2004 to 2014 as part of the Behavioral Risk
medical care providers Factor Surveillance System (BRFSS) conducted
via telephone surveys by the CDC in collabora-
The American Diabetes Association (ADA) tion with all states.114 The prevalence of diabetes
recognizes periodontal disease as a diabetes among the respondents increased significantly
comorbidity and recommends that patients from 6.7% in 2004 to 10.1% in 2014. The cor-
with diabetes be referred for dental exam- responding increase was from 0.8% to 1.5%
ination, preventive care, and any treatment for prediabetes. Figure 6-3 illustrates the pro-
as needed.113 Subscribing to the chronic care portion of US adults with a dental visit the year
model, it is stated in the ADA’s 2018 standards preceding the interview by their self-reported
of medical care in diabetes that dentists may diabetes status.114 Not only is the proportion
participate with physicians, nurse practitioners, of dental visits for people with known diabetes
physician assistants, nurses, dietitians, exercise much lower than among those without diabetes,
specialists, pharmacists, podiatrists, and men- but this is also the case for people with predi-
tal health professionals in the patient-centered abetes. Importantly, these data show declining
interprofessional approach with active partici- trends of having had a dental visit over the
pation also by the patient with family or other decade, namely from 71.9% to 66.5% for

149
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

diabetes-free participants versus from 66.1% Interprofessional


to 61.4% for those with diabetes. It should be
kept in mind that an estimated quarter (23.8%) Collaboration
of those with diabetes8 and almost 9 out of 10
(89%) of those with prediabetes115 are unaware Clinical pathway programs have been devel-
of their hyperglycemic state and hence are oped to share medical and dental information
unable to report belonging to those categories. to provide optimal care for patients with dia-
The association between dental visits, periodon- betes who seek periodontal care.120 By linking
titis, and diabetes may also be a reflection of an patients’ medical and dental records, it has
individual’s overall health behavior. been shown that coordinated treatment plan-
A 2016 study that used nationally repre- ning can be carried out by physicians and
sentative data from the 2008–2012 Medical dentists and that outcomes are improved when
Expenditure Panel Survey found that less than treatment is coordinated for those patients with
half of subjects had a dental visit the preceding both diabetes and dental disease.
year with great disparities by citizenship, as In another study, medical conditions asso-
only 43.6% of US-born citizens, 39.5% natu- ciated with dental conditions, including
ralized citizens, and 23.1% of noncitizens had periodontitis, were discovered using linked
at least one dental visit.116 Even though different electronic records from 2,475 patients under-
national US surveys report sizeable differences going dental treatment in a dental school
in proportion of adults with dental visits in the and medical treatment in the same university
previous year, their trends over several years hospital.121 Diabetes mellitus types 1 and 2,
tend to be comparable.117 The NHANES 2015 hypertension, hypercholesterolemia, hyper-
data indicate that 64.0% of adults aged 18 to 64 lipidemia, and conditions related to adverse
years and 62.7% of seniors aged 65 years and pregnancy outcomes were associated with
older had dental visits.118 With an even smaller periodontitis. For effective comanagement of
percentage of people with diabetes having den- patients who attend both dental school clinics
tal visits and considering the potential of good and their related medical colleges for treatment,
oral health to improve general health as well as it is important that specific patients who suffer
the diminished quality of life, including lower from these dental and medical conditions be
self-esteem, caused by poor oral health, this is a identified. This would allow treatment plans
regrettable situation on which combined efforts and management to be coordinated to promote
and initiatives by all health care professionals better overall health in patients. Another study
potentially could have a positive impact. conducted at a dental school among 2,370
A 2016 study used a sample of participants patients found such dental electronic health
of the Medicare Current Beneficiary Survey and records helpful for education, treatment plan-
found an increase in the proportion of people ning, and disease management.122
with diabetes who had preventive dental visits As previously mentioned, most risk markers
in 2011 compared to 2002, namely from 28.8% are common to periodontitis and diabetes, as
to 36.0%; this increase was comparable to that well as other chronic diseases, including CVD
among participants without diabetes, from and cancer. Therefore, modification of these
52.9% to 45.5%.119 However, the prevalence risk markers by both the dental and medi-
of preventive visits in participants with diabetes cal teams in collaboration with their mutual
was still much lower than among their counter- patients, working in a coordinated fashion,
parts without the disease, leading the authors has great potential to provide major benefits to
to the following conclusion: “Additional efforts society in reducing the morbidity and mortality
are needed to encourage people with diabetes associated with these prevalent chronic diseases
to obtain preventive dental care.”119 as well as the risk for periodontal disease. In

150
Interprofessional Collaboration

the chronic care model, collaboration by people with type 2 diabetes. Analyses of inte-
physicians, dentists, and other health care pro- grated claims data from 15,002 insureds aged
fessionals can be carried out to achieve better 18 to 64 years with newly diagnosed type 2
clinical outcomes for patients. These practice diabetes from a commercial insurance com-
models have the potential to offer great benefits. pany for dental, medical, and pharmacy care
Effective patient communication and patient showed that periodontal treatment during the
cooperation are essential for success. Lalla et al first two years after a diabetes diagnosis was
provided valuable, more detailed information linked to savings in the two following years
regarding dental and medical comanagement (third to fourth year after diagnosis) of $1,799
of patients with diabetes elsewhere.123 These for all health care, $1,577 for medical care, and
researchers collaborated with local research- $408 for diabetes care, respectively.133
ers to recalibrate their original findings made Compared to 10 other high-income coun-
in a study among a predominantly Hispanic tries, the United States spends the greatest
population living in Manhattan to a Caucasian proportion of its gross domestic product,
population in rural Wisconsin; they concluded namely almost one-fifth (17.8%) on health
that the accuracy of predicting prediabetes and care, compared to between 9.6% (Australia)
diabetes increases when incorporating dental and 12.4% (Switzerland) in the other coun-
findings with the medical information con- tries,134 and the United States has the greatest
tained in an electronic health record.124 Forty proportion of overweight or obesity (70.1%)134
statewide providers participated in a study to that is linked to type 2 diabetes. Therefore, it
further explore various collaborations and con- should be possible to institute savings on medi-
cluded that “cross-disciplinary integrated care cal care, and even small cost savings per patient
delivery (ICD) models that bridge the tradition- receiving medical care related to diabetes and
ally siloed health care domains of dentistry and its complications will add up to large amounts
medicine,” aided by mutual access to health at the population basis. Oral health care pro-
records, supported improved screening and fessionals might be able to contribute to such
referral implementation.125 Interprofessional financial savings.
patient management may be optimally carried
out in a chronic care model or in a health home
model, but with some effort it is also likely Guidelines regarding periodontal
to be successful in more traditional practice disease for medical and dental
settings.125–131
care providers and patients with or
at risk for diabetes
Cost savings Upon review of the scientific evidence, the con-
Dental care providers not only contribute to sensus of a group of dental experts in 2012
improvement of their patients’ general health was that it is appropriate to provide guide-
by screening for diabetes and other chronic lines for periodontal care in patients with
diseases and referring them for further exam- diabetes.41 These guidelines were reviewed
ination and treatment as needed. Short-term and updated in 2017 by another group of
savings in cost of health care were estimated dental experts from the European Federa-
at between $42.4 million and $102.6 million tion of Periodontology and medical experts
for medical screenings for diabetes, hyperten- from the International Diabetes Federation
sion, and hypercholesterolemia in the dental who again found scientific evidence to sup-
office.132 port recommendations for medical and dental
There is evidence that periodontal treatment professionals as well as their patients.135 These
may also lead to cost savings for health care in updated guidelines are displayed in Boxes 6-1

151
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

Box 6-1  |  Guidelines for physicians and other medical professionals for use in diabetes practice

Because of the increased risk for development of periodontitis in patients with diabetes, the following recommen-
dations are made:
• Oral health education should be provided to all patients with diabetes as part of their overall educational program.
• Patients with all forms of diabetes mellitus should be told that periodontal disease risk is increased and, if untreated,
the periodontitis has a negative impact on metabolic control and may also increase the risk of complications of
their diabetes such as cardiovascular and kidney disease.
• Patients should be advised that successful periodontal therapy may have a positive impact on their metabolic
control and diabetes complications.
• For people with diabetes, physicians should ask about a prior diagnosis of periodontal disease. If a positive diagnosis
has been made, the physician should seek to ascertain that periodontal care and maintenance are being provided.
• Investigating the presence of periodontal disease should be an integral part of a diabetes care visit. People
with diabetes should be asked about any signs and symptoms of periodontitis, including bleeding gums during
brushing or eating, loose teeth, spacing or spreading of the teeth, oral malodor and/or abscesses in the gums,
or gingival suppuration.
–– If a positive history is elicited, then a prompt periodontal evaluation should be recommended before the patient’s
scheduled annual checkup.
–– In the case of a negative history, people with diabetes should be advised to check for the above symptoms,
and if a positive sign appears, they should visit their dentist.
• For all people with newly diagnosed diabetes mellitus, referral for a periodontal examination should occur as
part of their ongoing management of diabetes. Even if no periodontitis is diagnosed initially, annual periodontal
review is recommended.
• For children and adolescents diagnosed with diabetes, annual oral screening is recommended through referral
to a dental professional.
• Patients with diabetes who have extensive tooth loss should be encouraged to pursue dental rehabilitation to
restore adequate mastication for proper nutrition.
• Patients with diabetes should be advised that other oral conditions such as dry mouth and burning mouth may
occur, and if so, they should seek advice from their dental practitioner. Also, patients with diabetes are at increased
risk of oral fungal infections and experience poorer wound healing than those who do not have diabetes.
• The physician should liaise with the dentist over diabetes management prior to the oral intervention and/or surgery
to avoid hypoglycemia and to consider its potential impact on the patient’s ability to eat.a,b

a The above guidelines also apply to people with prediabetes and metabolic syndrome.
b
These guidelines are modified from Chapple and Genco41 and also draw from http://www.aemmedi.it/files/Linee-guida_Raccomandazioni/2015/
Diabete%20e%20Parodontite_AMD_SID_SiDP.pdf
(Reproduced from Sanz et al.135)

to 6-4.135 Box 6-3 has been slightly modified the following risk factors: a first-degree relative
with further updated and expanded web ad- who has diabetes, a high-risk race/ethnicity (eg,
dresses for the patient-friendly test described African American, Hispanic, Native American,
in Box 6-3 and illustrated in Fig 6-4.135,136 The Asian American, Pacific Islander), a history of
2018 ADA professional criteria for testing CVD, hypertension (blood pressure ≥ 140/90
for diabetes and prediabetes in asymptomatic mmHg) or therapy for hypertension, hypercho-
adults9 is summarized as follows: Overweight lesterolemia (HDL cholesterol level < 35 mg/
or obese patients (BMI > 25 kg/m2 or BMI > dL [0.90 mmol/L]), hypertriglyceridemia (tri-
23 kg/m2 for Asian Americans) should be con- glyceride level > 250 mg/dL [2.82 mmol/L]),
sidered for testing if they have at least one of polycystic ovary syndrome, physical inactivity,

152
Conclusion

Box 6-2  |  Guidelines for patients with diabetes at the physician’s practice/office

Why should I have my gums checked?


If your physician has told you that you have diabetes, you should make an appointment with a dentist to have your
mouth and gums checked. This is because people with diabetes have a higher chance of getting gum disease. Gum
disease can lead to tooth loss and may make your diabetes harder to control. The earlier you seek help, the better
the outcome will be.
What should I look for that may tell me I have problems with my gums?
You may have gum disease if you have ever noticed:
• Red or swollen gums
• Bleeding from your gums or blood in the sink after you brush your teeth
• Foul taste
• Longer-looking teeth
• Loose teeth
• Increasing spaces between your teeth
• Calculus (tartar) on your teeth
If you have noticed any of these problems, it is important to see a dentist as soon as possible.
Can I have gum disease without these signs being present?
Gum disease may also be present and get worse with no apparent signs to you, especially if you smoke, so even if
you do not think you have gum disease now, you should still have annual dental checkups as part of managing your
diabetes. Your dentist will be able to pick up early signs of gum disease.
What can I do to prevent gum disease?
You need to clean your teeth and gums twice daily at home for a minimum of 2 minutes. Also, cleaning between your
teeth daily is important, and your dentist will show you how to do this. You should visit a dentist as soon as possible
for a diagnosis and advice on what you need to do. It is important to keep your mouth as healthy as possible with
regular dental care, according to the recommendations of your dentist.
What other problems with my mouth should I be looking for?
If you have diabetes, you may also suffer from dry mouth, burning mouth, or poor healing of mouth wounds.

(Reproduced from Sanz et al.135)

or other clinical conditions associated with Conclusion


insulin resistance. Patients with prediabetes
(HbA1c ≥ 5.7% [39 mmol/mol], impaired glu- Diabetes and periodontitis are common, seri-
cose tolerance, or impaired fasting glucose) ous conditions that are linked in a two-way
should be tested every year and women with mutually adverse association. The prevalence
GDM should be tested every 3 years. All other and incidence of periodontal disease are greater
patients should be tested starting at age 45. Pa- in patients with diabetes; periodontitis and its
tients with normal test results should be tested local and systemic inflammatory responses can
at least every 3 years, but more frequent testing adversely affect glycemic control in people with
should be considered based on initial results diabetes or at risk for diabetes. Their adverse
and risk status. reciprocal effects are likely based on them both
contributing to a hyperinflammatory state.

153
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

Box 6-3  |  Guidelines for oral health professionals for use in the dental practice/office for people with
diabetes mellitus

• People with diabetes should be advised that they have an increased risk for gingivitis and periodontitis. They should
also be told that if they suffer from periodontitis, their glycemic control may be more difficult to achieve, and they
are at higher risk of other complications such as eye, kidney, and cardiovascular diseases.
• Collect a careful history to highlight the type of diabetes, the duration of the disease, the presence of any
complications, and any diabetes therapy and concomitant therapies, remembering that most people with diabetes
are also being treated with anticoagulant/antiplatelet drugs, antihypertensive drugs or lipid-lowering medications.
• Ask patients how well controlled their diabetes is and when they last had their blood glucose levels checked.
Request that patients bring a copy of their last HbA1c result or that they report their latest results.
• Oral health education should be provided to all patients with diabetes. This should include individualized advice
on relevant risk factors and a tailored oral hygiene regimen, including twice-daily brushing, interdental cleaning,
and, in some cases, the use of adjunctive chemical plaque control.
• People presenting with a diagnosis of any form of diabetes mellitus should receive a thorough oral examination,
which includes a comprehensive periodontal evaluation with full-mouth pocket chart and bleeding scores.
• If no periodontitis is diagnosed initially, patients with diabetes should be placed on a preventive care regimen and
monitored regularly for periodontal changes.
• People with diabetes presenting with any acute oral/periodontal infections require prompt oral/periodontal care.
If periodontitis is diagnosed, it should be managed without delay.
• Irrespective of the level of diabetes control, nonsurgical periodontal therapy should be provided, as this may help
to improve glycemic control.
• Surgical periodontal and implant therapy is not indicated in patients who do not have acceptable diabetes control.
In well-controlled patients, the results of surgical interventions are equivalent to patients without diabetes. However,
attention should be paid to the following:
–– People with poorly controlled diabetes, who have an increased risk of postoperative infections
–– Patients managed with insulin or sulfonylureas, when the physician should be consulted about the timing of the
planned procedure and a possible change in dosage of therapy to reduce the risk of intraoperative hypoglycemia
• People with diabetes who have extensive tooth loss should be encouraged to pursue dental rehabilitation to restore
adequate mastication for proper nutrition.
• People with diabetes should also be evaluated for other potential oral complications, including dry mouth, burning
mouth, Candida infections, and dental caries.
• For children and adolescents diagnosed with diabetes, an annual oral screening for early signs of periodontal
involvement and dental caries is recommended starting as early as possible.
• Patients who present in the dental surgery/office without a diagnosis of diabetes but with risk factors for type
2 diabetes should be informed about their risk for having diabetes and referred to a physician for appropriate
diagnostic testing and follow-up care:
–– Patients’ risk may be screened for using a validated questionnaire: eg, in a Caucasian population, FindRisk
Questionnaire; https://www.idf.org/type-2-diabetes-risk-assessment/; http://www.diabetes.org/are-you-at-
risk/diabetes-risk-test/ (USA, ADA online test); http://main.diabetes.org/dorg/PDFs/risk-test-paper-version.pdf
(USA, ADA print version); https://www.diabetes.fi/files/502/eRiskitestilomake.pdf (Original FINDRISC, Finnish
Diabetes Association) (see Fig 6-4).136
–– Oral health professionals with a special interest in diabetes may wish to consider screening based on the
recommendations of the American Diabetes Association.9
–– If symptomatic (polydipsia, polyuria, polyphagia, unexplained weight loss), refer directly to a physician.

(Reproduced from Sanz et al.135)

154
Conclusion

Box 6-4  |  Guidelines for patients at the dental surgery/office who have diabetes or are found to be at risk of
diabetes

• People with diabetes have a higher chance of getting gum disease.


• You may think that you are doing well managing your gum health, but you may not be doing enough because you
have an increased risk of gum problems.
• Like diabetes, gum disease is a chronic condition and requires lifelong attention and professional care.
• You also need to clean your teeth and gums very carefully at home. Personalized advice will be provided by your
dentist. This may include the following:
–– Twice-daily brushing with either a manual or electric toothbrush
–– Cleaning between your teeth using interdental brushes where they fit; where they do not fit, flossing may be
useful
–– The use of specific dentifrices and/or mouthrinses with proven activity against dental plaque, if advised by
your dentist
• If left untreated, gum disease can lead to tooth loss and may also make your diabetes harder to control.
• Gum disease may be present and get worse with no apparent symptoms to you, so if your dentist told you that
you do not have gum disease now, you should still get regular dental checkups as part of managing your diabetes.
Your dentist will be able to pick up early signs of gum disease.
• You may have gum disease if you have ever noticed:
–– Red or swollen gums
–– Bleeding from your gums or blood in the sink after you brush your teeth
–– Foul taste
–– Longer-looking teeth
–– Loose teeth
–– Increasing spaces between your teeth
–– Calculus (tartar) on your teeth
• People with diabetes may also suffer from dry mouth, burning mouth, yeast infections of the mouth, or poor
healing of mouth wounds.
• Remember to inform your dentist about the outcome of your visits to your doctor and provide an update of the
results of your diabetes control and changes in medications.
• It is important to keep your mouth and your whole body as healthy as possible with regular dental and medical care.

(Reproduced from Sanz et al.135)

Dental management of patients with diabe- adequate before undertaking dental procedures
tes requires knowledge of the patient’s diabetic to prevent hypoglycemic episodes in the dental
status, including metabolic control and medica- office.
tions, and comorbidities. It is advised that oral Likewise, dental patients who have peri-
health care professionals coordinate the care of odontal disease and exhibit additional risk
a patient with poorly controlled diabetes with markers for diabetes, such as obesity, should be
the patient’s physician. Such patients should be made aware of the relationship between these
routinely assessed for periodontal involvement two diseases and referred to their physician for
as well as other oral complications of diabe- assessment of their diabetic status.
tes, including xerostomia, Candida infections, Early identification and early treatment of
and intraoral burning sensations. Oral health prediabetes and diabetes have many benefits
care professionals should be aware of any for the patient in terms of improved health
antidiabetic medications patients are taking and reduced risk of diabetes complications.
and ensure that their blood glucose levels are Involvement of oral health care professionals

155
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

Fig 6-4 |  FINDRISC questionnaire for assessing risk for type 2 diabetes. (Based on Lindström and
Tuomilehto.136 Design by Jaakko Tuomilehto, Department of Public Health, University of Helsinki, and
Jaana Lindström, National Public Health Institute, Finland.)

156
Conclusion

Fig 6-4  (cont) Reverse side of FINDRISC form.

157
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

Clinical Considerations: What You Can Take Back to Your Practice


What is the effect of diabetes on oral health, gical periodontal treatment can reduce the level
and what is the effect of oral health on glycemic of glycated hemoglobin, especially when signif-
control?  Hyperglycemia (abnormally high lev- icant periodontitis is present in patients with
els of blood glucose) is not merely a diagnosis poorly controlled type 2 diabetes. Moreover,
of diabetes but affects oral health by increasing extraction of terminally periodontally diseased
the risk for periodontal disease and ultimately teeth decreases the glycated hemoglobin level
tooth loss. This in turn can result in poor nutri- and thereby improves glycemic control.
tion, as well as decreased psychosocial status.
Hyperglycemia can also lead to hyposalivation Is it safe to provide dental care to patients with
and a burning sensation in the mouth. In the diabetes?  It is safe to provide dental care to
other direction, periodontitis can adversely af- patients with diabetes. It is actually very im-
fect glycemic control. portant to deliver such care because there is
evidence that poor oral health and tooth loss
Is periodontal disease an independent risk can affect diet and result in nutritional inad-
marker for the development or progression of equacies that can contribute to poor glycemic
diabetes?  Emerging evidence suggests that es- control.
pecially severe periodontitis represents a risk
marker for the development of type 2 diabe- What is the appropriate message to give to
tes and for the progression of existing type 2 the public about the association between oral
diabetes. health and diabetes?  Diabetes, especially if
poorly controlled, is an important risk mark-
What is the biologic plausibility of the associ- er for periodontal disease. Overall, the degree
ation between oral health and diabetes?  The of tooth loss in adult patients with diabetes is
bidirectional relationship between diabetes and twice that in normoglycemic individuals, and
periodontal disease is most likely related to the this loss often leads to limitation of the range
increased level of systemic inflammation that of foods that can be properly chewed. In turn,
each disease causes. These elevated systemic poor nutrition may result, which can make di-
inflammatory responses result in insulin resis- etary control of diabetes more difficult. People
tance, poor glycemic control, and aggravated with diabetes—especially if poorly controlled
destruction of periodontal soft and hard tis- and of long duration—can also suffer from xe-
sues. Importantly, the hyperglycemia is the de- rostomia or hyposalivation and a burning sen-
ciding factor affecting periodontal tissues, not sation in the mouth and are more susceptible
simply a diagnosis of diabetes. to fungal infections. In the opposite direction,
periodontal inflammation can negatively affect
Does the delivery of periodontal care contri­­- glycemic control. However, nonsurgical peri-
bute to improvement of glycemic control?  There odontal therapy can improve glycemic control.
is evidence that provision of successful nonsur-

in screening for potential hyperglycemia physicians, pharmacists, nurses, and other


could have a beneficial effect. Furthermore, health care professionals is likely to improve
the use of common electronic health records the overall health of patients with diabetes and
and interprofessional, patient-centered coor- periodontitis and thus the overall quality of life
dinated treatment planning and management of these patients.
of the common risk markers among dentists,

158
References

References 16. Singh GM, Danaei G, Farzadfar F, et al. The age-specific


quantitative effects of metabolic risk factors on cardio-
vascular diseases and diabetes: A pooled analysis.
1. Bartold PM, Van Dyke TE. Periodontitis: A host-mediated PLoS One 2013;8:e65174.
disruption of microbial homeostasis. Unlearning learned 17. Leung MY, Pollack LM, Colditz GA, Chang SH. Life years
concepts. Periodontol 2000 2013;62: 203–217. lost and lifetime health care expenditures associated
2. Petersen PE, Ogawa H. The global burden of periodontal with diabetes in the U.S., National Health Interview
disease: Towards integration with chronic disease Survey, 1997-2000. Diabetes Care 2015;38:460–468.
prevention and control. Periodontol 2000 2012; 60: 18. Zhao P, Liu E, Qiao Y, et al. Maternal gestational diabetes
15–39. and childhood obesity at age 9-11: Results of a multi-
3. Kassebaum NJ, Bernabe E, Dahiya M, et al. Global national study. Diabetologia 2016;59:2339–2348.
burden of severe periodontitis in 1990–2010: A system- 19. American Diabetes Association. Economic costs of
atic review and meta-regression. J Dent Res 2014; 93: diabetes in the U.S. in 2017. Diabetes Care 2018;
1045–1053. 41:917–928.
4. Eke PI, Thornton-Evans GO, Wei L, Borgnakke WS, 20. Hu R, Shi L, Pierre G, Zhu J, Lee DC. Diabetes and
Dye BA. Accuracy of NHANES periodontal examination medical expenditures among non-institutionalized U.S.
protocols. J Dent Res 2010;89:1208–1213. adults. Diabetes Res Clin Pract 2015;108:223–234.
5. Page RC, Eke PI. Case definitions for use in 21. American Diabetes Association. Economic costs of
population-based surveillance of periodontitis. J Peri- diabetes in the U.S. in 2012. Diabetes Care 2013; 36:
odontol 2007;78(suppl 7):1387–1399. 1033–1046.
6. Eke PI, Page RC, Wei L, Thornton-Evans G, Genco RJ. 22. Olson DE, Zhu M, Long Q, et al. Increased cardiovas-
Update of the case definitions for population-based cular disease, resource use, and costs before the clinical
surveillance of periodontitis. J Periodontol 2012; 83: diagnosis of diabetes in veterans in the southeastern
1449–1454. U.S. J Gen Intern Med 2015;30:749–757.
7. Eke PI, Thornton-Evans GO, Wei L, Borgnakke WS, 23. Weinspach K, Staufenbiel I, Memenga-Nicksch S, Ernst
Dye BA, Genco RJ. Periodontitis in US adults: National S, Geursten W, Günay H. Level of information about the
Health and Nutrition Examination Survey 2009–2014. relationship between diabetes mellitus and periodonti-
J Am Dent Assoc 2018;149:576–588. tis—Results from a nationwide diabetes information
8. Centers for Disease Control and Prevention. National program. Eur J Med Res 2013;18:6.
Diabetes Statistics Report, 2017; Estimates of Diabetes 24. Lalla E, Papapanou PN. Diabetes mellitus and periodon-
and Its Burden in the United States. https://www.cdc. titis: A tale of two common interrelated diseases. Nat
Gov/diabetes/pdfs/data/statistics/national-diabetes- Rev Endocrinol 2011;7:738–748.
statistics-report.pdf. Accessed 6 April 2018. 25. Taylor JJ, Preshaw PM, Lalla E. A review of the evidence
9. American Diabetes Association. 2. Classification and for pathogenic mechanisms that may link periodontitis
diagnosis of diabetes: Standards of medical care in and diabetes. J Periodontol 2013; 84(4 suppl):
diabetes—2018. Diabetes Care 2018; 41(suppl 1): S113–134.
S13–S27. 26. Zhou X, Zhang W, Liu X, Zhang W, Li Y. Interrelationship
10. International Diabetes Federation. IDF Diabetes Atlas, between diabetes and periodontitis: Role of hyperlipid-
ed 8. Brussels: International Diabetes Federation, 2017. emia. Arch Oral Biol 2015;60:667–674.
www.diabetesatlas.org. Accessed 6 April 2018. 27. Borgnakke WS, Ylöstalo PV, Taylor GW, Genco RJ.
11. Song C, Lyu Y, Li C, et al. Long-term risk of diabetes in Effect of periodontal disease on diabetes: Systematic
women at varying durations after gestational diabetes: review of epidemiologic observational evidence. J Clin
A systematic review and meta-analysis with more than Periodontol 2013;40(suppl 14):S135–S152.
2 million women. Obes Rev 2018;19:421–429. 28. Graziani F, Gennai S, Solini A, Petrini M. A systematic
12. American Diabetes Association. Standards of medical review and meta-analysis of epidemiologic observational
care in diabetes—2018. Diabetes Care 2018;41(suppl evidence on the effect of periodontitis on diabetes: An
1):S1–S150. update of the EFP-AAP review. J Clin Periodontol 2018;
13. Leasher JL, Bourne RR, Flaxman SR, et al. Global esti- 45:167–187.
mates on the number of people blind or visually impaired 29. Sharma P, Dietrich T, Ferro CJ, Cockwell P, Chapple IL.
by diabetic retinopathy: A meta-analysis from 1990 to Association between periodontitis and mortality in
2010. Diabetes Care 2016;39:1643–1649. stages 3-5 chronic kidney disease: NHANES III and
14. Deli G, Bosnyak E, Pusch G, Komoly S, Feher G. linked mortality study. J Clin Periodontol 2016; 43:
Diabetic neuropathies: Diagnosis and management. 104–113.
Neuroendocrinology 2013;98:267–280. 30. Shultis WA, Weil EJ, Looker HC, et al. Effect of peri-
15. Emerging Risk Factors Collaboration, Sarwar N, Gao P, odontitis on overt nephropathy and end-stage renal
et al. Diabetes mellitus, fasting blood glucose concen- disease in type 2 diabetes. Diabetes Care 2007; 30:
tration, and risk of vascular disease: A collaborative 306–311.
meta-analysis of 102 prospective studies. Lancet 2010;
375:2215–2222.

159
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

31. Chang JF, Yeh JC, Chiu YL, Liou JC, Hsuing JR, Tung 46. Kowall B, Holtfreter B, Völzke H, et al. Pre-diabetes and
TH. Periodontal pocket depth, hyperglycemia, and well-controlled diabetes are not associated with peri-
progression of chronic kidney disease: A population- odontal disease: The SHIP Trend study. J Clin
based longitudinal study. Am J Med 2017;130:61–69. Periodontol 2015;42:422–430.
e1. 47. Garcia D, Tarima S, Okunseri C. Periodontitis and glyce-
32. Kebede TG, Holtfreter B, Kocher T, et al. Association mic control in diabetes: NHANES 2009 to 2012. J
of periodontal destruction and diabetes with mortality. Periodontol 2015;86:499–506.
J Dent Res 2017;96:56–63. 48. Eke PI, Wei L, Thornton-Evans GO, et al. Risk indicators
33. Lin SY, Lin CL, Liu JH, et al. Association between peri- for periodontitis in US adults: NHANES 2009 to 2012.
odontitis needing surgical treatment and subsequent J Periodontol 2016;87:1174–1185.
diabetes risk: A population-based cohort study. J Peri- 49. Hong JW, Noh JH, Kim DJ. The prevalence and asso-
odontol 2014;85:779–786. ciated factors of periodontitis according to fasting
34. Chiu SY, Lai H, Yen AM, et al. Temporal sequence of plasma glucose in Korean adults: The 2012-2013 Korea
the bidirectional relationship between hyperglycemia National Health and Nutrition Examination Survey. Medi-
and periodontal disease: A community-based study of cine (Baltimore) 2016;95:e3226.
5,885 Taiwanese aged 35-44 years (KCIS No. 32). Acta 50. Emrich LJ, Shlossman M, Genco RJ. Periodontal
Diabetol 2015;52:123–131. disease in non-insulin-dependent diabetes mellitus. J
35. Esteves Lima RP, Cyrino RM, de Carvalho Dutra B, et Periodontol 1991;62:123–131.
al. Association between periodontitis and gestational 51. Lal S, Cheng B, Kaplan S, et al. Accelerated tooth erup-
diabetes mellitus: Systematic review and meta-analysis. tion in children with diabetes mellitus. Pediatrics 2008;
J Periodontol 2016;87:48–57. 121:e1139–e1143.
36. Chokwiriyachit A, Dasanayake AP, Suwannarong W, et 52. Pahel BT, Vann Jr WF, Divaris K, Rozier RG. A contem-
al. Periodontitis and gestational diabetes mellitus in porary examination of first and second permanent molar
non-smoking females. J Periodontol 2013;84:857–862. emergence. J Dent Res 2017;96:1115–1121.
37. Esteves Lima RP, Miranda Cota LO, Costa FO. Asso- 53. Patel MH, Kumar JV, Moss ME. Diabetes and tooth loss:
ciation between periodontitis and gestational diabetes An analysis of data from the National Health and Nutri-
mellitus: A case-control study. J Periodontol 2013;84: tion Examination Survey, 2003-2004. J Am Dent Assoc
1257–1265. 2013;144:478–485.
38. Xiong X, Elkind-Hirsch KE, Vastardis S, Delarosa RL, 54. Deguchi M, Mau MKLM, Davis J, Niederman R. Prevent-
Pridjian G, Buekens P. Periodontal disease is associated able tooth loss in Hawai’i: The role of socioeconomic
with gestational diabetes mellitus: A case-control study. status, diabetes, and dental visits. Prev Chronic Dis
J Periodontol 2009;80:1742–1749. 2017;14:e115.
39. Borgnakke WS. Hyperglycemia/diabetes mellitus and 55. Greenblatt AP, Salazar CR, Northridge ME, et al. Asso-
periodontal infection adversely affect each other. In: ciation of diabetes with tooth loss in Hispanic/Latino
Genco RJ, Williams RC (eds). Periodontal Disease and adults: Findings from the Hispanic Community Health
Overall Health: A Clinician’s Guide, ed 2. Yardley, PA: Study/Study of Latinos. BMJ Open Diabetes Res Care
Professional Audience Communications, 2014: 99–122. 2016;4:e000211.
40. Borgnakke WS, Genco RJ, Eke PI, Taylor GW. Oral 56. Luo H, Pan W, Sloan F, Feinglos M, Wu B. Forty-year
health and diabetes. In: Cowie CC, Casagrande SS, trends in tooth loss among American adults with and
Menke A, et al (eds). Diabetes in America, ed 3 [NIH without diabetes mellitus: An age-period-cohort analy-
Pub No. 17-1468]. Bethesda, MD: National Institutes sis. Prev Chronic Dis 2015;12:e211.
of Health, 2018:31.31–31.51. 57. Oluwagbemigun K, Dietrich T, Pischon N, Bergmann
41. Chapple IL, Genco R; Working group 2 of joint EFP/ M, Boeing H. Association between number of teeth and
AAP workshop. Diabetes and periodontal diseases: chronic systemic diseases: A cohort study followed for
Consensus report of the Joint EFP/AAP Workshop on 13 years. PLoS One 2015;10:e0123879.
periodontitis and systemic diseases. J Clin Periodontol 58. Mayard-Pons ML, Rilliard F, Libersa JC, Musset AM,
2013;40(suppl 14):S106–S112. Farge P. Database analysis of a French type 2 diabetic
42. Grossi SG, Genco RJ. Periodontal disease and diabetes population shows a specific age pattern of tooth
mellitus: A two-way relationship. Ann Periodontol 1998; extractions and correlates health care utilization. J
3:51–61. Diabetes Complications 2015;29:993–997.
43. Preshaw PM, Alba AL, Herrera D, et al. Periodontitis 59. Song SJ, Han K, Lee SS, Park JB. Association between
and diabetes: A two-way relationship. Diabetologia the number of natural teeth and diabetic retinopathy
2012;55:21–31. among type 2 diabetes mellitus: The Korea National
44. Löe H. Periodontal disease. The sixth complication of Health and Nutrition Examination Survey. Medicine
diabetes mellitus. Diabetes Care 1993;16: 329–334. (Baltimore) 2017;96:e8694.
45. Demmer RT, Holtfreter B, Desvarieux M, et al. The influ- 60. Aoyama N, Suzuki JI, Kobayashi N, et al. Japanese
ence of type 1 and type 2 diabetes on periodontal cardiovascular disease patients with diabetes mellitus
disease progression: Prospective results from the Study suffer increased tooth loss in comparison to those with-
of Health in Pomerania (SHIP). Diabetes Care 2012;35: out diabetes mellitus—A cross-sectional study. Intern
2036–2042. Med 2018;57:777–782.

160
References

61. Demmer RT, Jacobs Jr DR, Singh R, et al. Periodontal 76. Li Q, Hao S, Fang J, Xie J, Kong XH, Yang JX. Effect of
bacteria and prediabetes prevalence in ORIGINS: The non-surgical periodontal treatment on glycemic control
oral infections, glucose intolerance, and insulin resis- of patients with diabetes: A meta-analysis of randomized
tance study. J Dent Res 2015;94:201S–211S. controlled trials. Trials 2015;16:291.
62. Winning L, Patterson CC, Neville CE, Kee F, Linden GJ. 77. Simpson TC, Weldon JC, Worthington HV, et al. Treat-
Periodontitis and incident type 2 diabetes: A prospective ment of periodontal disease for glycaemic control in
cohort study. J Clin Periodontol 2017;44:266–274. people with diabetes mellitus. Cochrane Database Syst
63. Costa KL, Taboza ZA, Angelino GB, et al. Influence of Rev 2015;(11):CD004714.
periodontal disease on changes of glycated hemoglobin 78. Teshome A, Yitayeh A. The effect of periodontal therapy
levels in patients with type 2 diabetes mellitus: A retro- on glycemic control and fasting plasma glucose level in
spective cohort study. J Periodontol 2017;88:17–25. type 2 diabetic patients: Systematic review and
64. Kebede TG, Pink C, Rathmann W, et al. Does periodon- meta-analysis. BMC Oral Health 2016;17:31.
titis affect diabetes incidence and haemoglobin A1c 79. Janket SJ, Wightman A, Baird AE, Van Dyke TE, Jones
change? An 11-year follow-up study. Diabetes Metab JA. Does periodontal treatment improve glycemic
2018;44:243–249. control in diabetic patients? A meta-analysis of inter-
65. Kocher T, König J, Borgnakke WS, Pink C, Meisel P. vention studies. J Dent Res 2005;84:1154–1159.
Periodontal complications with hyperglycemia/diabetes 80. Darré L, Vergnes JN, Gourdy P, Sixou M. Efficacy of
mellitus: Epidemiologic complexity and clinical chal- periodontal treatment on glycaemic control in diabetic
lenge. Periodontol 2000 2018;78:59–97. patients: A meta-analysis of interventional studies.
66. Sammalkorpi K. Glucose intolerance in acute infections. Diabetes Metab 2008;34:497–506.
J Intern Med 1989;225:15–19. 81. Wang X, Han X, Guo X, Luo X, Wang D. The effect of
67. Artese HP, Foz AM, Rabelo Mde S, et al. Periodontal periodontal treatment on hemoglobin A1c levels of
therapy and systemic inflammation in type 2 diabetes diabetic patients: A systematic review and meta-analysis.
mellitus: A meta-analysis. PLoS One 2015; 10: PLoS One 2014;9:e108412.
e0128344. 82. Ibrahim M, Bahijri S, Tuomilehto J. Diabetes and Rama-
68. Teeuw WJ, Gerdes VE, Loos BG. Effect of periodontal dan. Prim Care Diabetes 2015;9:241–243.
treatment on glycemic control of diabetic patients: A 83. Engebretson SP, Hyman LG, Michalowicz BS, et al. The
systematic review and meta-analysis. Diabetes Care effect of nonsurgical periodontal therapy on hemoglobin
2010;33:421–427. A1c levels in persons with type 2 diabetes and chronic
69. Simpson TC, Needleman I, Wild SH, Moles DR, Mills periodontitis: A randomized clinical trial. JAMA 2013;
EJ. Treatment of periodontal disease for glycaemic 310:2523–2532.
control in people with diabetes. Cochrane Database 84. Merchant AT. Hemoglobin A1c levels among patients
Syst Rev 2010;(5):CD004714. with diabetes receiving nonsurgical periodontal treat-
70. Sgolastra F, Severino M, Pietropaoli D, Gatto R, Monaco ment. JAMA 2014;311:1919.
A. Effectiveness of periodontal treatment to improve 85. Chapple IL, Borgnakke WS, Genco RJ. Hemoglobin
metabolic control in patients with chronic periodontitis A1c levels among patients with diabetes receiving
and type 2 diabetes: A meta-analysis of randomized nonsurgical periodontal treatment. JAMA 2014; 311:
clinical trials. J Periodontol 2013;84:958–973. 1919–1920.
71. Engebretson S, Kocher T. Evidence that periodontal 86. Vergnes JN. Hemoglobin A1c levels among patients
treatment improves diabetes outcomes: A systematic with diabetes receiving nonsurgical periodontal treat-
review and meta-analysis. J Clin Periodontol 2013; ment. JAMA 2014;311:1920–1921.
40(suppl 14):S153–S163. 87. Engebretson SP, Hyman LG, Michalowicz BS. Hemo-
72. Liew AK, Punnanithinont N, Lee YC, Yang J. Effect of globin A1c levels among patients with diabetes receiving
non-surgical periodontal treatment on HbA1c: A nonsurgical periodontal treatment– Reply. JAMA 2014;
meta-analysis of randomized controlled trials. Aust Dent 311:1921–1922.
J 2013;58:350–357. 88. Botero JE, Rodriguez C, Agudelo-Suarez AA. Periodon-
73. Corbella S, Francetti L, Taschieri S, De Siena F, Fabbro tal treatment and glycaemic control in patients with
MD. Effect of periodontal treatment on glycemic control diabetes and periodontitis: An umbrella review. Aust
of patients with diabetes: A systematic review and Dent J 2016;61:134–148.
meta-analysis. J Diabetes Investig 2013;4:502–509. 89. D’Aiuto F, Gable D, Syed Z, et al. Evidence summary:
74. Sun QY, Feng M, Zhang MZ, et al. Effects of periodontal The relationship between oral diseases and diabetes.
treatment on glycemic control in type 2 diabetic patients: Br Dent J 2017;222:944–948.
A meta-analysis of randomized controlled trials. Chin J 90. Faggion CM Jr, Cullinan MP, Atieh M. An overview of
Physiol 2014;57:305–314. systematic reviews on the effectiveness of periodontal
75. Wang TF, Jen IA, Chou C, Lei YP. Effects of periodontal treatment to improve glycaemic control. J Periodontal
therapy on metabolic control in patients with type 2 Res 2016;51:716–725.
diabetes mellitus and periodontal disease: A
meta-analysis. Medicine (Baltimore) 2014;93:e292.

161
6 Associations Between Periodontal Disease and Hyperglycemia/Diabetes

91. Hasuike A, Iguchi S, Suzuki D, Kawano E, Sato S. 106. Elter JR, Champagne CM, Offenbacher S, Beck JD.
Systematic review and assessment of systematic Relationship of periodontal disease and tooth loss to
reviews examining the effect of periodontal treatment prevalence of coronary heart disease. J Periodontol
on glycemic control in patients with diabetes. Med Oral 2004;75:782–790.
Patol Oral Cir Bucal 2017;22:e167–e176. 107. Iwasaki M, Kimura Y, Yoshihara A, et al. Association
92. Madianos PN, Koromantzos PA. An update of the between dental status and food diversity among older
evidence on the potential impact of periodontal therapy Japanese. Community Dent Health 2015;32:104–110.
on diabetes outcomes. J Clin Periodontol 2018; 45: 108. Samnieng P, Ueno M, Shinada K, Zaitsu T, Wright FA,
188–195. Kawaguchi Y. Oral health status and chewing ability is
93. Grant MJ, Booth A. A typology of reviews: An analysis related to mini-nutritional assessment results in an older
of 14 review types and associated methodologies. adult population in Thailand. J Nutr Gerontol Geriatr
Health Info Libr J 2009;26:91–108. 2011;30:291–304.
94. Obadan-Udoh E, Jordan S, Mudah O, Borgnakke W, 109. Liljestrand JM, Havulinna AS, Paju S, Männistö S, Salo-
Tavares M. Gap analysis of older adults with type 2 maa V, Pussinen PJ. Missing teeth predict incident
diabetes receiving nonsurgical periodontal therapy. J cardiovascular events, diabetes, and death. J Dent Res
Evid Based Dent Pract 2017;17:335–349. 2015;94:1055–1062.
95. Saengtipbovorn S, Taneepanichskul S. Effectiveness of 110. Yamanaka K, Nakagaki H, Morita I, Suzaki H, Hashimoto
lifestyle change plus dental care program in improving M, Sakai T. Comparison of the health condition between
glycemic and periodontal status in aging patients with the 8020 achievers and the 8020 non-achievers. Int
diabetes: A cluster, randomized, controlled trial. J Peri- Dent J 2008;58:146–150.
odontol 2015;86:507–515. 111. Savoca MR, Arcury TA, Leng X, et al. Severe tooth loss
96. Genco RJ, Borgnakke WS. Risk factors for periodontal in older adults as a key indicator of compromised dietary
disease. Periodontol 2000 2013;62:59–94. quality. Public Health Nutr 2010;13:466–474.
97. Glick M, Greenberg BL. The potential role of dentists in 112. Borgnakke WS, Anderson PF, Shannon C, Jivanescu
identifying patients’ risk of experiencing coronary heart A. Is there a relationship between oral health and diabetic
disease events. J Am Dent Assoc 2005; 136: neuropathy? Curr Diab Rep 2015;15:93.
1541–1546. 113. American Diabetes Association. 3. Comprehensive
98. Lalla E, Kunzel C, Burkett S, Cheng B, Lamster IB. medical evaluation and assessment of comorbidities:
Identification of unrecognized diabetes and pre-diabetes Standards of medical care in diabetes—2018. Diabetes
in a dental setting. J Dent Res 2011;90:855–860. Care 2018;41(suppl 1):S28–S37.
99. Genco RJ, Schifferle RE, Dunford RG, Falkner KL, Hsu 114. Luo H, Bell RA, Wright W, Wu Q, Wu B. Trends in annual
WC, Balukjian J. Screening for diabetes mellitus in dental dental visits among US dentate adults with and without
practices: A field trial. J Am Dent Assoc 2014; 145: self-reported diabetes and prediabetes, 2004–2014. J
57–64. Am Dent Assoc 2018;149:460–469.
100. Herman WH, Taylor GW, Jacobson JJ, Burke R, Brown 115. Centers for Disease Control and Prevention. Awareness
MB. Screening for prediabetes and type 2 diabetes in of prediabetes—United States, 2005-2010. MMWR
dental offices. J Public Health Dent 2015;75:175–182. Morb Mortal Wkly Rep 2013;62:209–212.
101. Knowler WC, Barrett-Connor E, Fowler SE, et al. Reduc- 116. Wilson FA, Wang Y, Stimpson JP, McFarland KK, Singh
tion in the incidence of type 2 diabetes with lifestyle KP. Use of dental services by immigration status in the
intervention or metformin. N Engl J Med 2002; 346: United States. J Am Dent Assoc 2016; 147: 162–169.
393–403. e164.
102. Nordenram G, Davidson T, Gynther G, et al. Qualitative 117. Lin M, Li CH, Wei L, et al. Prevalence of past-year dental
studies of patients’ perceptions of loss of teeth, the visit among US adults, 1999-2010: Comparison of
edentulous state and prosthetic rehabilitation: A system- trends and estimates between the Behavioral Risk
atic review with meta-synthesis. Acta Odontol Scand Factor Surveillance System and three national surveys.
2013;71:937–951. J Public Health Dent 2017;77:105–114.
103. Thorstensson H, Johansson B. Why do some people 118. Health, United States, 2016: With Chartbook on
lose teeth across their lifespan whereas others retain a Long-Term Trends in Health. Hyattsville, MD: National
functional dentition into very old age? Gerodontology Center for Health Statistics, 2017. https://www.cdc.
2010;27:19–25. gov/nchs/data/hus/hus16.pdf. Accessed 6 September
104. Sheiham A, Steele JG, Marcenes W, et al. The relation- 2018.
ship among dental status, nutrient intake, and nutritional 119. Wiener RC, Shen C, Sambamoorthi N, Sambamoorthi
status in older people. J Dent Res 2001;80:408–413. U. Preventive dental care in older adults with diabetes.
105. Ioannidou E, Swede H, Fares G, Himmelfarb J. Tooth J Am Dent Assoc 2016;147:797–802.
loss strongly associates with malnutrition in chronic 120. Ota M, Seshima F, Okubo N, et al. A collaborative
kidney disease. J Periodontol 2014;85:899–907. approach to care for patients with periodontitis and
diabetes. Bull Tokyo Dent Coll 2013;54:51–57.

162
References

121. Boland MR, Hripcsak G, Albers DJ, et al. Discovering 129. Poudel P, Griffiths R, Wong VW, Arora A, George A.
medical conditions associated with periodontitis using Knowledge and practices of diabetes care providers in
linked electronic health records. J Clin Periodontol 2013; oral health care and their potential role in oral health
40:474–482. promotion: A scoping review. Diabetes Res Clin Pract
122. Thyvalikakath TP, Padman R, Vyawahare K, Darade P, 2017;130:266–277.
Paranjape R. Utilizing dental electronic health records 130. Myers-Wright N, Lamster IB, Jasek JP, Chamany S.
data to predict risk for periodontal disease. Stud Health Evaluation of medical and dental visits in New York City:
Technol Inform 2015;216:1081. Opportunities to identify persons with and at risk for
123. Lalla E, Hsu W, Lamster IB. Dental and medical coman- diabetes mellitus in dental settings. Community Dent
agement of patients with diabetes. In: Genco RJ, Oral Epidemiol 2018;46:102–108.
Williams RC (eds). Periodontal Disease and Overall 131. Glick M. A home away from home: The patient-centered
Health: A Clinician’s Guide, ed 2. Yardley, PA: Profes- health home. J Am Dent Assoc 2009;140:140–142.
sional Audience Communications, 2014;252–274. 132. Nasseh K, Greenberg B, Vujicic M, Glick M. The effect
124. Acharya A, Cheng B, Koralkar R, et al. Screening for of chairside chronic disease screenings by oral health
diabetes risk using integrated dental and medical elec- professionals on health care costs. Am J Public Health
tronic health record data. JDR Clin Trans Res 2018; 2014;104:744–750.
3:188–194. 133. Nasseh K, Vujicic M, Glick M. The relationship between
125. Glurich I, Nycz G, Acharya A. Status update on trans- periodontal interventions and healthcare costs and utili-
lation of integrated primary dental-medical care delivery zation. Evidence from an integrated dental, medical,
for management of diabetic patients. Clin Med Res and pharmacy commercial claims database. Health
2017;15:21–32. Econ 2017;26:519–527.
126. Bojadzievski T, Gabbay RA. Patient-centered medical 134. Papanicolas I, Woskie LR, Jha AK. Health care spending
home and diabetes. Diabetes Care 2011; 34: in the United States and other high-income countries.
1047–1053. JAMA 2018;319:1024–1039.
127. Stellefson M, Dipnarine K, Stopka C. The chronic care 135. Sanz M, Ceriello A, Buysschaert M, et al. Scientific
model and diabetes management in US primary care evidence on the links between periodontal diseases and
settings: A systematic review. Prev Chronic Dis 2013; diabetes: Consensus report and guidelines of the Joint
10:E26. Workshop on Periodontal Diseases and Diabetes by
128. Pumerantz AS, Bissett SM, Dong F, et al. Standardized the International Diabetes Federation and the European
screening for periodontitis as an integral part of multi- Federation of Periodontology. J Clin Periodontol 2018;
disciplinary management of adults with type 2 diabetes: 45:138–149.
An observational cross-sectional study of cohorts in the 136. Lindström J, Tuomilehto J. The diabetes risk score: A
USA and UK. BMJ Open Diabetes Res Care 2017; practical tool to predict type 2 diabetes risk. Diabetes
5:e000413. Care 2003;26:725–731.

163
CHAPTER 7

The Cardiovascular System


and Oral Infections
Maurizio S. Tonetti, dmd, phd, mmsc
Filippo Graziani, dds, mclindent, phd

The hypothesis that oral infections may con- of this hypothesis was confirmed by studies
tribute to atherosclerosis was formulated indicating that individuals exposed to chronic
in the mid-1980s by a group of Finnish infections have two to three times greater odds
cardiologists.1 It is relevant to attempt an of having carotid atherosclerosis.2 Chronic
understanding of the rationale for this line of obstructive pulmonary disease with infectious
research. Groundbreaking epidemiologic stud- exacerbations, chronic bronchitis, chronic
ies had identified the classic risk factors for sinusitis, and chronic or recurrent urinary tract
atherosclerosis, but cardiologists were faced infections—as well as an amorphous group
with the clinical dilemma that only one in of other infections—were all associated with
two heart attacks could be explained by the higher odds of carotid atherosclerosis. Patho-
classic set of risk factors: hypertension, dyslip- gens sustaining these chronic infections were
idemia, diabetes, and smoking. Research was considered either to have a direct effect on the
needed to identify mechanisms that explained vasculature or to act as a source of systemic
a significant portion of the burden of disease inflammation that in turn would trigger the
and, more importantly, to find ways to pre- atherosclerotic process. Indeed, epidemiologic
vent it. An interesting hypothesis that chronic studies linking systemic inflammation, athero-
infection (and later chronic inflammation) sclerosis, and cardiovascular events have shown
may contribute to the atherosclerotic process consistent associations between levels of sys-
was formulated. In this broader context, and temic inflammatory markers and increases in
given the obvious high prevalence of chronic carotid intima-media thickness, myocardial
oral infections, early associations were sought infarction, and nonhemorrhagic stroke.
between the presence of oral infections and The infection hypothesis was tested with
cardiovascular events: Subjects with acute clinical randomized controlled trials (RCTs)
cardiovascular events seemed to have poorer aimed at preventing cardiovascular events with
oral health than control subjects. The field was the use of long-acting broad-spectrum antibiot-
born. ics such as azithromycin. Early reports showed
At the turn of the 21st century, an impres- encouraging results, but later full-scale stud-
sive collection of scientific evidence stood in ies and meta-analyses showed no preventive
support of the hypothesis that chronic infec- effect from antibiotic treatment.3 The infection
tions were among the major contributors to hypothesis had failed to provide a tangible
atherosclerosis and its sequelae. The validity benefit.

164
Mechanisms

From the moment the systematic review necrosis factor alpha (TNF-α).6,7 In case-control
showed that there was no benefit in using studies, individuals with periodontitis showed
systemic antibiotics to eliminate obscure 1.65-mg/L higher concentrations of CRP than
infections to prevent cardiovascular events, unaffected subjects8–17 (Table 7-1). This is further
the hypothesis that oral infections could con- corroborated by recent evidence also showing
tribute to the overall atherosclerotic burden an increase of IL-6.18 These findings are signifi-
was seen with less favor. Supporters of the cant in several ways. First, they established that
link between oral infection and atheroscle- periodontitis is sensed by the innate acute-phase
rosis pointed out that azithromycin or other response system and leads to a chronic systemic
systemic antibiotics would fail to act specifi- inflammatory status. Second, the size of the
cally against biofilm-centered infections, such elevation in cytokines is remarkable: IL-6 and
as periodontitis.4 Indeed, the periodontal lit- CRP are important predictors of cardiovascu-
erature has shown only marginal effects from lar outcomes, and cardiovascular risk has been
the use of systemic antibiotics in the absence predicted based on their serum concentrations.
of mechanical root instrumentation (biofilm An increase of the magnitude observed for
dispersion).5 periodontitis is sufficient to shift an otherwise
This is at least part of the reason why many healthy subject to a high-risk category. These
researchers active in this field stress that peri- data support the hypothesis that periodontitis—
odontitis is a source of chronic, nonresolving as a chronic inflammatory condition driven by
inflammation and frequently play down the fact a biofilm-centered infection—contributes to the
that periodontitis is a biofilm-centered infec- systemic inflammatory load of a subject.
tion: Cardiologists these days are much less
excited about infection than about inflamma-
tion as a mechanism leading to atherosclerosis.
From an oral health standpoint, the hypoth- Mechanisms
esis of a relationship between oral health and
systemic diseases fits very well with the key Over the last two decades, a wealth of mecha-
developments of our understanding of periodon- nistic data has been used to explore potential
titis established in the late 1980s to mid-1990s: mechanisms for the association between peri-
the existence of a systemic predisposition to odontitis and atherosclerosis. Two types of
periodontitis and in general the existence of a mechanisms are primarily involved: (1) Bac-
relationship between local periodontal disease teria related to periodontal disease may enter
and systemic inflammatory and immune chal- the circulation19 and contribute directly to the
lenges. Indeed, both epidemiologic research development of atherogenesis (or thrombosis),
pointing to the existence of high-risk groups and and (2) systemic inflammation resulting from
pathophysiologic research pointing to the exis- periodontitis can contribute to atherosclerotic
tence of biochemical changes that would prime cardiovascular disease (ACVD). These data
healthy sites in diseased subjects toward sus- have been reviewed in depth by the recent
ceptibility to periodontitis seem to be consistent European Federation of Periodontology/Amer-
with the notion that the periodontal disease pro- ican Academy of Periodontology (EFP/AAP)
cess does not remain localized to the oral cavity Workshop.20,21
and that the organism as a whole senses what There is some biologic evidence of a potential
is going on in the periodontium. Accordingly, role of periodontal pathogens in the formation
a higher general inflammatory status is noted of atherosclerotic plaque. Oral and systemic
in subjects with periodontitis, as witnessed by inoculation of Porphyromonas gingivalis in
the raise of specific cytokines such as C-reactive apolipoprotein E–deficient (ie, more susceptible
protein (CRP), interleukin 6 (IL-6), and tumor to atherosclerosis) animal models has shown

165
7 The Cardiovascular System and Oral Infections

TABLE 7-1  |  Estimated difference in serum CRP concentration between individuals with
periodontitis and individuals without periodontitis from case-control studies
Periodontitis CRP
Study/subcategory n mean (SD) n Control CRP mean (SD)
Bizzarro et al /SeP8
38 3.10 (4.40) 39 1.90 (2.00)
Bizzarro et al8/MoP 53 3.10 (3.30) 39 1.90 (2.00)
Salzberg et al /GaP9
93 3.72 (2.88) 91 1.54 (2.95)
Salzberg et al9/LaP 97 2.57 (2.95) 91 1.54 (2.95)
Havemose-Poulsen et al10/GaP 27 5.00 (2.70) 25 3.00 (2.00)
Havemose-Poulsen et al10/LaP 18 5.00 (5.40) 25 3.00 (2.00)
Joshipura et al11 91 2.20 (2.25) 377 1.80 (3.00)
Buhlin et al 12
50 3.28 (4.64) 46 1.74 (1.68)
Amar et al13 17 2.30 (2.30) 21 1.00 (1.00)
Craig et al14
44 5.78 (1.07) 25 2.46 (1.44)
Noack et al15 50 4.06 (5.55) 65 1.70 (1.91)
Loos et al16 107 2.64 (3.48) 43 1.21 (1.34)
Fredriksson et al 17
17 2.62 (2.90) 17 0.87 (1.73)
Total (95% CI) 702 904
Test for heterogeneity: Chi2 = 51.41; df = 12 (P < .00001), I2 = 76.7%.
Test for overall effect: Z = 5.42 (P < .00001).
CRP, C-reactive protein; SD, standard deviation; SeP, severe periodontitis; MoP, moderate periodontitis; GaP, generalized aggressive periodontitis; LaP,
localized aggressive periodontitis; CI, confidence interval; WMD, weighted mean difference; df, degrees of freedom. (Reprinted from Paraskevas et al6
with permission.)

that together with infection of periodontal tis- specimens, the DNA of some target perio-
sues, measured as alveolar bone loss, a systemic pathogens, including Tannerella forsythia, P
response toward P gingivalis was mounted.22,23 gingivalis, and Aggregatibacter actinomyce-
Higher aortic expression of vascular cell adhe- temcomitans, was present.24 Further studies
sion molecule 1 and tissue factor was also noted confirmed these findings.25–27
in the P gingivalis–inoculated animals.22 More- • Periopathogens are able to invade endothe-
over, atherosclerotic lesions of the proximal lial cells and contribute to atherosclerosis.
aortas and aortic trees were more advanced P gingivalis has shown the capability to
and occurred earlier in P gingivalis–challenged invade several cell lines, and the majority
animals.23 Conversely, atherosclerotic lesions of P gingivalis strains may invade vascular
did not appear in the control animals despite tissues.28 Moreover, P gingivalis has been
their higher susceptibility. shown to modulate angiopoietin in human
The following findings provide further evi- aortic smooth muscle cells through gingipain
dence of the association between periodontal production.29
pathogens and atherosclerosis: • Periopathogens may trigger platelet
aggregation on their membrane. Platelet
• The DNA of periopathogens has been aggregation is characteristic of atheromas.
localized within atherosclerotic plaque. It has been shown that P gingivalis may
Assessment of 50 human endoarterectomy facilitate platelet aggregation through its
specimens showed that in at least 44% of the fimbriae and may determine a potent platelet

166
Mechanisms

WMD (random) [95% CI] Weight (%) WMD (random) 95% CI


6.40 1.20 [–0.33, 2.73]
8.11 1.20 [0.11, 2.29]
9.10 2.18 [1.34, 3.02]
9.10 1.03 [0.19, 1.87]
7.33 2.00 [0.71, 3.29]
3.53 2.00 [–0.61, 4.61]
10.16 0.40 [–0.15, 0.95]
6.98 1.54 [0.17, 2.91]
7.77 1.30 [0.13, 2.47]
9.84 3.32 [2.67, 3.97]
6.14 2.36 [0.75, 3.97]
9.38 1.43 [0.66, 2.20]
6.15 1.75 [0.14, 3.36]
100.00 1.65 [1.05, 2.24]

–4 –2 0 2 4

aggregation–inducing activity through its This triggers a systemic acute-phase inflam-


vesicles (membrane evaginations projecting matory response, characterized by increased
to the outer environment).30 levels of acute-phase proteins such as CRP
• P gingivalis accelerates the transition of and by vascular dysfunction.11,35,36
macrophages to foam cells. An important
feature in the development of early athero- The association between periodontal
sclerotic lesions is cholesterol uptake into pathogens and atherosclerosis is therefore bio-
macrophages to form foam cells. Murine logically plausible, and emphasis generally has
macrophages could be stimulated by P gingi- been given to the strength and the consistency
valis to accumulate low-density lipoproteins of the findings. A plausible biologic mecha-
to form foam cells.31 nism would suggest that bacteria or their toxic
• Periopathogens have been linked to an byproducts may easily gain access to the circu-
increased host inflammatory reaction. Sys- latory system. Indeed, episodes of bacteremia
temic inflammation is closely linked to the have been detected after normal activity such as
onset of atherosclerosis.32 Subjects affected chewing or tooth brushing. Similar to bacterial
by periodontal disease develop an intense dissemination, excessive local production of
local production of proinflammatory cyto- proinflammatory cytokines may obtain access
kines that may enter the bloodstream, as to the bloodstream and trigger a systemic
shown by the high levels of inflammatory acute-phase response. Thus, when compared
biomarkers in gingival tissues and serum.33,34 with matched periodontal healthy subjects,

167
7 The Cardiovascular System and Oral Infections

Chronic oral infection

B get to bloodstream Bacteremia Endotoxemia Proinflammatory


mediators (pocket)

Proinflammatory
Host response/ mediators (liver)
inflammation
Prothrombotic
B invade cells
mediators
B found in ath
B alive in ath Autoimmune
mediators
B induce ath

B mutants = less ath Atherothrombotic Dyslipidemia


B from ath cause disease lesion
Genetic background
Reyes et al20 Schenkein and Loos21

Fig 7-1  |  Biologically plausible mechanisms linking periodontitis and atherothrombogenesis. ath, atheroma; B,
bacteria. Dotted boxes indicate limited or no evidence. (Reprinted from Tonetti et al38 with permission.)

periodontal subjects show higher levels of components—and the effect of spillover of


CRP and IL-6, a lower number of erythrocytes, inflammatory mediators produced locally in
lower hemoglobin concentrations, higher val- the inflamed periodontal tissues on the vascu-
ues of haptoglobin, moderate leukocytosis, and lar endothelium. While it is indeed recognized
increased cholesterol, low-density lipoprotein, that the two hypotheses would overlap and
and glucose levels.37 These differences were also that bacteremia will ultimately cause inflam-
significant when the analyses accounted for a mation, a better understanding of the relative
series of possible confounding factors (age, sex, contribution of the two hypotheses may help
smoking, socioeconomic status, diabetes, body us to design better preventive or therapeutic
mass index, and alcohol use). approaches.
The two hypotheses (bacteremia and chronic A critical assessment of the mechanistic
inflammation) were reviewed at the 2012 evidence highlights that, while each piece of
Joint EFP/AAP Workshop on Periodontitis evidence taken individually is moderately inter-
and Systemic Diseases.38 Significant evidence esting, the consistency of the data across in
was identified for both hypotheses, and the vitro, ex vivo, and experimental research with a
current understanding of the mechanisms is variety of animal models provides high levels of
described in Fig 7-1. The consensus delibera- confidence regarding the existence of potential
tions on the specific position papers highlighted mechanisms. Their relevance and actual clinical
the fact that, at present, it is impossible to importance must be assessed by different types
recognize the relative contribution of a direct of studies, namely analytical epidemiology and
bacterial effect—mediated by translocation intervention trials.
and a metastatic effect of bacteria or bacterial

168
Intervention Trials

Epidemiology association was stronger in younger adults,


and there was no evidence for an association
Early studies reported associations between between periodontitis and incident coronary
various definitions of periodontitis and athero- heart disease in subjects older than 65 years; in
sclerotic vascular diseases.39 The majority of the this respect, it must be emphasized that in most
early studies were cross-sectional, thus the valid- epidemiologic studies of ACVD, single risk fac-
ity of their results was hampered by the potential tors are significant in younger subjects. Only
of reverse causality bias (cardiovascular diseases one study evaluated the association between
opening a window of susceptibility to periodon- periodontitis and a second cardiovascular
titis rather than the opposite) as well as by the event. While heterogeneity in study outcomes
use of vastly different definitions of disease, and design prevented a meta-analysis, the data
ranging from self-reporting to telephone inter- seemed remarkably consistent; indeed, partic-
view, to different protocols for partial-mouth ipants in the workshop believed that several
examinations. Furthermore, these studies did aspects of the epidemiologic evidence war-
not appropriately correct for confounding fac- ranted discussion and were sufficient to permit
tors: Periodontitis and ACVD share a variety conclusions to be made (Box 7-1).
of risk factors. Common risk factors (indica- Although it is well recognized that the epi-
tors) include cigarette smoking, obesity, male demiologic evidence of an association between
sex, stress, depression, physical inactivity, and periodontitis and ACVD has increased in
a low-quality diet. Critics have pointed out these strength and overall quality, the majority of
limitations and the need for better data analysis. the large longitudinal studies were designed for
Nonetheless, the majority of early epidemio- different purposes and the periodontal com-
logic studies have indicated an increase in risk. ponent was an additional element; hence, the
A previous meta-analysis acknowledging wide study design may be at risk of bias. Considerable
variations in study design and disease defini- debate is taking place in the scientific commu-
tion estimated that the adjusted additional risk nity about whether a better understanding of the
was about 15% higher for cerebrovascular association between periodontitis and ACVD
outcomes and about 13% higher for cardiovas- requires a specifically designed epidemiologic
cular outcomes.40 The newest evidence suggests study or if it would be a better use of the scarce
that the presence of periodontitis indicates a available resources to simply focus on the needed
34% higher risk of cardiovascular disease,41 intervention studies to establish causality. This is
and recent data supports even higher risk for a reflection of a broader unresolved debate on
incident stroke with relative risk ranging from causality, risk, and prevention between epide-
1.65 to 3.04, according to the study design.42,43 miologists and clinical investigators.
Emphasis must be placed on incident risk
estimated in longitudinal studies in which the
periodontal status had to be determined before
the observation period.44 This study design Intervention Trials
addresses the reverse causality bias. The EFP/
AAP Workshop position paper included 12 Results from definitive clinical trials are
studies (6 on coronary heart disease, 3 on cere- considered the ultimate element of proof in
brovascular disease, 2 on both coronary heart establishing the nature of the relationship
and cerebrovascular disease mortality, and 1 on between periodontitis and ACVD. The logical
peripheral arterial disease).38 All but one study framework is that if the elimination or atten-
reported positive associations between various uation of the cause is randomized through
periodontal disease measures and the incidence treatment of periodontitis (or its prevention
of ACVD, at least in specific subgroups. The at the population level), an attenuation of

169
7 The Cardiovascular System and Oral Infections

Box 7-1  |  Conclusions about epidemiologic evidence derived from the Joint EFP/AAP Workshop on
Periodontitis and Systemic Diseases38

• Risk: The outcome in all studies in the systematic review44 indicated that the occurrence of periodontitis was prior
to an incident cardiovascular event of ACVD. This higher level of evidence of association allows for statements
of risk to be made.
• Definitions of periodontitis: “Periodontitis measured using clinical attachment loss/periodontal probing depth
and/or radiographic assessment of bone loss has been associated with increased risk for various measures of
ACVD independent of established cardiovascular risk factors.” Periodontitis was not measured by self-reporting,
interview, or simplified partial-mouth recordings.
• Strength of association: “Statistically significant excess risk for ACVD in individuals with periodontitis was reported
to be independent of established cardiovascular risk factors.” The amount of excess risk adjusted for other
ACVD risk factors, however, varied by type of cardiovascular outcome and by age and sex. The risk was found
to be greater for cerebrovascular disease than for coronary heart disease and greater in males and in younger
individuals. No excess risk was reported between measures of periodontitis and incident coronary heart disease
in individuals aged older than 65 years, which supports previous findings that established individual ACVD risk
factors are weaker in older adults. This was an important point during the workshop discussions: The presence
of a degree of specificity in the association was thought to be an element that would strengthen the case for a
true relationship. In addition, the workshop consensus was that there is insufficient evidence to indicate whether
or not periodontitis is associated with the incidence of secondary cardiovascular events (a second ACVD event
after the original event). It was recognized that this finding has implications for future clinical trials and that, under
ideal circumstances, more epidemiologic evidence would be needed for the planning of such intervention trials.
Indeed, in the planning of intervention trials it is important to have a relationship between the outcome and the
exposure (primary or secondary cardiovascular event and periodontitis).
• Potential relevance: “Even low to moderate excess risk reported in studies is enough to be important from a
public health perspective because of the high prevalence of periodontitis.” Essentially, given the high burden of
periodontitis (and potentially gingivitis) in the population, even a moderate measure of risk may be relevant for a
population, because periodontitis is both preventable and treatable, and thus a large number of cases of ACVD
may be prevented by improving the oral health of the population.
• Confounding: Given the fact that chronic inflammatory diseases like cardiovascular disease and periodontitis
share several risk factors (such as smoking, diabetes, obesity, and nutrition), there is concern that the purported
association can be explained at least in part by such exposures. Indeed, the recent workshop concluded that “There
are many potentially important confounders of the association between periodontitis and ACVD risk, including
comorbidities such as diabetes and lifestyle factors such as smoking. However, established cardiovascular risk
factors do not completely explain the excess cardiovascular risk in subjects with periodontitis.” In all of the studies
reviewed, smoking status was controlled for, and in many studies excess risk was demonstrated in subjects who
had never smoked. Excess risk for periodontitis was also demonstrated in studies that controlled for diabetes.
However, excess risk could be associated with unknown confounders. The ENCODE project, a deep sequencing
project to identify all functional elements of the genome, recently found that common genetically determined
pathways underpin various complex inflammatory diseases. Therefore, such genetic determinants could be con-
founding factors. In support of this last statement, a recent report has identified a common susceptibility locus
for both cardiovascular disease and periodontitis on chromosome 9p21.3, in the long noncoding RNA ANRIL
locus.45,46 While all studies in the systematic review had to control for at least age and sex, and while smoking
was controlled for in the analyses, the potential for residual confounding in the explanation of the association
remains significant and is the result of the design of these epidemiologic studies.

170
Intervention Trials

the effect (the ACVD outcome under study) periodontal treatment regimens that included
should be observed. The design of such trials, periodontal surgery to achieve optimal control
however, is an exceedingly complex exercise. of periodontitis.49
It requires a good understanding of the most Some large studies conducted in the United
likely mechanism(s) to be targeted by the inter- States have shown limited changes in periodon-
vention, the identification of a population that tal outcomes and have been associated with
is at risk and amenable to receive treatment, no change in the systemic parameter under
and the availability of an ethically appropriate investigation. The limited results obtained
control treatment. with periodontal therapy have been ques-
tioned and have highlighted that no systemic
benefit should be expected in the absence of
Delivery of treatment an effective delivery of the intervention. The
Surprisingly little attention has been paid in current recommendation among experts seems
recent years to identifying the best possible to be to specify therapeutic targets that must be
periodontal intervention to achieve a general achieved in terms of reducing gingival inflam-
health benefit. The assumption that the best mation and probing depths rather than to
possible periodontal outcome (minimal gingi- specify a given regimen.
val inflammation and shallow pockets in the
whole of the dentition) represents the best
candidate for treatment is rational and rea- Adverse events
sonable but far from having been proven. As Shortly after the delivery of periodontal
yet, only one series of studies aimed at assess- treatment, subjects presented with a sys-
ing the optimal benefit in terms of systemic temic inflammatory response that included
inflammatory markers has been performed, by an increase in acute-phase response (CRP,
D’Aiuto et al, between 2002 and 2005.37,47,48 serum amyloid A, fibrinogen, coagulation,
These studies explored different regimens of and neutropenia) and an increase in markers
root instrumentation and the adjunctive ben- of endothelial cell damage (E-selectin, von Wil-
efit of local delivery of minocycline (with an lebrand factor, D-dimers, and flow-mediated
important anti-inflammatory effect). These dilation of the brachial artery).48,50–52 These
studies identified several important aspects in adverse events are not unexpected, because
terms of differential efficacy, adverse events, mechanical instrumentation induces significant
and timing of the effect of treatment. bacteremia and endotoxemia that are likely to
cause the observed changes. The intensity of
the observed changes, however, was surprising,
Efficacy because for several parameters they represented
Better efficacy was observed for both peri- a 10-fold increase from baseline levels. These
odontal and systemic inflammatory outcomes changes persisted for a period of up to 1 week.
after an intensive treatment regimen consisting Follow-up studies to assess the relative inten-
of complete-mouth mechanical instrumen- sity of systemic inflammatory changes after
tation, extraction of teeth with hopeless subgingival debridement or periodontal flap
prognosis, local application of minocycline surgery indicated that the greatest inflamma-
microspheres in all sites with probing depths tory response was observed immediately after
greater than 4 mm, and oral hygiene instruc- complete-mouth subgingival debridement and
tions delivered over 24 to 36 hours.37 Efficacy that, surprisingly, surgical trauma seemed to
in terms of local periodontal parameters have a smaller impact.53 A recent follow-up
paralleled changes in systemic inflammatory trial indicated that the intensity of the sys-
parameters. Some studies have employed temic inflammatory response to periodontal

171
7 The Cardiovascular System and Oral Infections

debridement is less pronounced when treatment identified systemic comorbidities (and at rel-
is delivered over multiple appointments (on a atively low risk of presenting cardiovascular
quadrant basis), and this seems an important events) and (2) individuals who have already
consideration for periodontal treatment of sub- suffered from a cardiovascular event (and are
jects at high risk for cardiovascular events.54 at a much higher risk of having new events).
Minassian et al55 explored the relevance of this In the systemically healthy population, the
systemic inflammatory response with its under- experimental rationale for treating periodon-
lying bacteremia and trauma. They looked at a titis would be to interfere with the process
large insurance claim database to examine the of atherogenesis and therefore to do a pri-
incidence rate of cardiovascular events with mary prevention trial. In such a population,
regard to their relationship to dental appoint- the adverse events of periodontal therapy
ments in which invasive dental procedures were are probably less relevant in the sense that
performed. After invasive dental treatment, a healthy individual could tolerate well the
there was a small but measurable increase in increased burden associated with periodon-
the risk of having a cardiovascular event. The tal therapy. This is the optimal population in
increased risk persisted for up to 3 months which to show prevention or delay of the ath-
after the dental appointment. erosclerotic process assessed by measuring a
variety of so-called surrogate cardiovascular
outcomes (eg, arterial elasticity, hypertension,
Timing of treatment intima-media thickness). However, these sur-
Studies that have followed the time course rogate outcomes bear relatively little weight in
of systemic responses after local periodontal the decision-making process to identify aspects
therapy show a biphasic response characterized to target with prevention; stronger evidence
by an early worsening of biochemical (CRP based on true cardiovascular events is favored
and inflammation51) and functional parame- in the cardiology community. The challenges
ters (flow-mediated dilation56). This biphasic of running a trial aimed at preventing true
response raises important issues related to the cardiovascular events in an otherwise healthy
timing of outcome assessment: For several population are formidable: The low rate of
biochemical parameters, it seems important to expected events will require either a very large
allow healing of the early acute-phase response sample size or a very long follow-up period.
before assessing the potential benefit of treat- Not unexpectedly, trials in systemically healthy
ment. Repeated episodes of treatment (either individuals with periodontitis have focused on
nonsurgical or surgical) complicate timing of surrogate markers.
sampling and outcome assessment. Running an intervention trial in a population
Recognition of the complexity of the systemic at increased risk (ie, with a previous experience
treatment effects and the fact that periodontal of a cardiovascular or cerebrovascular event)
therapy initially may bring a small added risk, poses great challenges as well. First, these
rather than a benefit, in terms of cardiovascu- individuals are normally medicated to lower
lar events raises important ethical issues in the the risk of a second event. Many of the drug
design of the proper intervention trials, specif- molecules may interfere with multiple aspects
ically in terms of the target population. of the mechanisms at the basis of a relation-
ship between periodontitis and atherosclerosis.
Second, the available periodontal intervention
Population at risk may represent a small but concrete short-term
Two types of population (and two types of risk for a secondary event.55 The advantages of
intervention trial) have been considered: (1) performing a trial in this type of population are
individuals who are free from significant associated with the higher incidence of events

172
Intervention Trials

in these populations; individuals have reached of such an approach is that it will maximize
an overall burden of atherosclerotic diseases the difference in local periodontal parame-
that put them at risk for a second event. There- ters—and, it is hoped, systemic inflammatory
fore, it is possible to decrease the sample size burden—between the test and control groups.
and/or the time of follow-up. Community care is probably one of the few
The periodontitis and vascular events (PAVE) options for studies of longer duration. Its exper-
study represents a feasibility study for such a imental appeal is based on the assumption that
secondary prevention trial.57 The trial was community periodontal therapy on one hand
remarkable because it was possible to recruit a represents the current standard of treatment
population among individuals who had recently but on the other hand falls short of the optimal
suffered from a primary cardiovascular event. level of care that can be delivered by highly
It also showed the challenge related to the specialized teams in the context of the trial. The
delivery of treatment: The experimental group experience with the PAVE study shows that,
showed moderate periodontal improvements, at least in countries like the United States, the
while the control group (left in the intention periodontal-systemic link has received such a
of the study investigators to community care, level of professional and media attention that
expected to be mainly supragingival cleaning subjects randomized to community care will
with a hygienist) received a considerable level endeavor to receive the best level of care that
of periodontal treatment.57 they can access.58 Indeed, the ethics associated
with the control group represent the biggest
challenge in running a trial in a society with
Control intervention optimal access to periodontal care.
Identification of an ethically acceptable con-
trol periodontal treatment seems to be a key
aspect in the design of a definitive trial. The Results of intervention trials
background of the ethical discussion is repre- Several RCTs have been performed to date to
sented by the current standard of periodontal assess the effect of periodontal intervention on
care available to the population. Depending systemic parameters.
on the different health care systems and the On one hand, no information is available
existence of a waiting time to receive treatment on true cardiovascular outcomes (incidence
and/or the availability of specialist care, two of cardiovascular event [ie, angina pectoris,
options have been considered. The first option myocardial infarction, stroke, death]).7 This
consists of delaying periodontal treatment until is probably due to intrinsic limitations of the
the end of a relatively short follow-up period trials’ study design. Cardiovascular events are
of the trial and delivery of a standardized pla- rare, and extremely large sample sizes would
cebo treatment (eg, supragingival cleaning and be needed to assess difference. Moreover, the
oral hygiene instructions) within the trial. The time needed to observe a clinical event would
second consists of leaving the control group to impose a long follow-up. Lastly, there are
community care. ethical concerns in designing a trial in which
Both options have limitations and advan- the control group might receive suboptimal
tages and disadvantages. Delayed treatment treatment.59
seems ethically possible only if the duration On the other hand, important effects are
of follow-up is adequately short (probably in seen in terms of cardiovascular surrogate
the range of 6 to 12 months); it also requires outcomes.7,60,61 After periodontal treatment,
adequate safety monitoring to ensure the significant reduction of inflammatory bio-
institution of standard-of-care treatment if markers is noted, reinforcing the hypothesis
periodontitis progresses. The biggest advantage that periodontitis might determine a systemic

173
7 The Cardiovascular System and Oral Infections

TABLE 7-2  |  Effect of periodontal therapy on serum CRP concentrations


Difference in experimental Difference in control group,
Study n group, mean (SD) n mean (SD)
D’Aiuto et al37 20 0.40 (1.01) 24 –0.10 (1.65)
D’Aiuto et al37
21 0.00 (2.25) 24 –0.10 (1.65)
Seinost et al67 31 0.60 (1.38) 31 0.00 (0.80)
Total (95% CI) 72 2.57 (2.95) 79
Test for heterogeneity: Chi2 = 0.57, df = 2 (P = .75), I2 = 0%.
Test for overall effect: Z = 2.32 (P = .02).
SD, standard deviation; WMD, weighted mean difference; CI, confidence interval; df, degrees of freedom. (Reprinted from Paraskevas et al6 with permission.)

inflammatory alteration. CRP, IL-6, TNF-α , inflammation, and growth and remodeling of
fibrinogen, and E-selectin are diminished after blood vessel walls. Impairment of endothe-
treatment. IL-6 diminishes by approximately lial function and integrity, called endothelial
–0.5 ng/L, TNF-α by 0.75 pg/mL, and fibrin- dysfunction, occurs in the early stages of ath-
ogen by approximately –0.5 g/L.7 CRP values erosclerosis and its progression.64,65 Endothelial
are reduced by approximately 0.4 mg/L (Table dysfunction is considered a long-term predictor
7-2), and even more in subjects with comor- of ACVD events.66 It is frequently measured as
bidities.61 Subgroup analysis also indicated flow-mediated dilation of the brachial artery
that nonsmokers and nonobese individuals using a noninvasive ultrasound technique.67
would benefit from the higher reduction of Of the RCTs aimed at assessing endothelial
systemic inflammation.7 For comparison, such dysfunction, the largest was performed by
an improvement would be sufficient to lower Tonetti et al,56 who randomized 121 healthy
the CRP-associated risk of future cardiovas- individuals suffering from severe generalized
cular events and falls within the range of CRP periodontitis to either a cycle of supragingi-
improvement that can be obtained with phar- val mechanical scaling and polishing (control)
macologic treatment with statins. or an intensive treatment regimen consisting
Traditional cardiovascular risk factors such of complete-mouth scaling and root planing,
as lipids (ie, total cholesterol and high-density extraction of hopeless teeth, and local delivery
lipoprotein [HDL]) also showed reduction after of minocycline microspheres in all pockets. At
periodontal treatment.7 The joint EFP-IDF 6 months after therapy, the intensive treatment
(International Diabetes Federation) Workshop regimen had led to an improvement in endothe-
clearly identified that, in patients with diabetes, lial function of 2.0% (95% confidence interval
periodontal treatment is capable of significant of 1.2 to 2.8; P < .001) over the control treat-
reduction of HbA1c and free blood glucose.62 ment (Fig 7-2). The observed improvement in
This is important considering that the latter endothelial function was directly associated
has been significantly associated with cardio- with the obtained improvements in periodon-
vascular events.63 tal parameters.
Furthermore, endothelial function as mea- Higashi et al68,69 extended these results of
sured through flow-mediated dilation is improved endothelial function to individuals
influenced by periodontal treatment, with a suffering from hypertension and ACVD. These
suggested improvement in dilation in a range trials are significant because they seem to sug-
of approximately 4% to 9% after treatment.60 gest that periodontal treatment may reverse the
The endothelium is a key regulator of blood early stages of atherosclerosis, even in subjects
vessel biology because it affects coagulation, with established cardiovascular disease.

174
Conclusion

WMD (random) [95% CI] Weight (%) WMD (random) 95% CI


28.84 0.50 [–0.29, 1.29]
13.38 0.10 [–1.07, 1.27]
57.78 0.60 [0.04, 1.16]
100.00 0.50 [0.08, 0.93]

–4 –2 0 2 4

IPT (n = 61)
10 CPT (n = 59)
Flow-mediated dilation of the brachial artery (%)

4
1

0
Baseline Day 1 Day 7 1 Mo 2 Mo 6 Mo

Fig 7-2  |  Effect of intensive periodontal therapy (IPT) compared with supragin-
gival debridement (community periodontal treatment [CPT]) on endothelial dys-
function. (Reprinted from Tonetti et al56 with permission.)

Conclusion and biologic mechanism, intervention trials


to date are not adequate to draw further con-
In light of the present incomplete evidence, the clusions. Nevertheless, the positive effects of
following conclusions should be drawn38: periodontal treatment on markers associated
with cardiovascular diseases are consistent.
• There is consistent and strong epidemio-
logic evidence that periodontitis imparts an The EFP/AAP Workshop consensus made
increased risk for future ACVD. the following recommendations to oral health
• The impact of periodontitis on ACVD is bio- practitioners38:
logically plausible. Translocated circulating
oral microbiota may directly or indirectly • Practitioners should be aware of the emerging
induce systemic inflammation that impacts and strengthening evidence that periodontitis
the pathogenesis of atherothrombogenesis. is a risk factor for the development of ACVD
• While in vitro, animal, and clinical studies and should advise patients of the risk.
do support the existence of the interaction

175
7 The Cardiovascular System and Oral Infections

• The rationale for prevention, diagnosis, and have to deal with the present incomplete pic-
treatment of periodontitis remains the pres- ture for years to come. At present, there seem to
ervation of the dentition and avoidance of be some firm points: The association between
the crippling effects of periodontitis-induced periodontal disease and atherosclerosis seems
alveolar bone loss and tooth loss. to be consistent. It is unclear, however, if the
• Based on the weight of the evidence, peri- link can be fully explained by modifiable risk
odontitis patients with other risk factors for factors shared between periodontitis and car-
ACVD, such as hypertension, obesity, and diovascular diseases.
smoking, who have not seen a physician Because risk factor modification is now a
within the last year should be referred for well-recognized component of periodontal
a physical. therapy, a pragmatic approach will likely see
• Modifiable lifestyle-associated risk factors periodontists and other oral health care pro-
for periodontitis (and ACVD) should be fessionals treat periodontitis and address the
addressed in the dental office and in the con- risk factors as part of the periodontal therapy.
text of comprehensive periodontal therapy This may bring general health benefits because
(eg, smoking cessation programs and advice of the control of the shared risk factors and
on lifestyle modifications such as diet and perhaps because of the control of the systemic
exercise). This goal may be better achieved in inflammatory burden contributed by peri-
collaboration with appropriate specialists and odontitis. Conversely, and this is the important
may bring health gains beyond the oral cavity. implicit point made by the recent American
• Treatment of periodontitis in patients with a Heart Association statement that critically eval-
history of cardiovascular events should fol- uated the evidence of a relationship between
low American Heart Association guidelines periodontitis and cardiovascular disease,70 it
for elective procedures. is wise for periodontists and oral health care
professionals to avoid making claims that
Besides these consensus conclusions, it seems periodontal therapy improves cardiovascular
important to recognize that the challenges of health. Such assertions are premature and may
having sufficient evidence to draw firmer con- confuse patients with cardiovascular disease,
clusions are formidable and that dental and who belong under the care of their physician
medical practitioners and their patients will or cardiologist in a medical practice.

Clinical Considerations: What You Can Take Back to Your Practice


What is the effect of oral health on cardiovascu- oral infections contribute to the development
lar disease?  Epidemiologic studies show strong or progression of cardiovascular events.
associations between poor oral health and car-
diovascular disease. This association may differ What is the biologic plausibility of the associ-
among different populations and age groups. ation between oral health and cardiovascular
disease?  Oral microbes colonize atheroscle-
Are oral infections independent risk factors for rotic lesions via bacteremia, and they may
the development of cardiovascular disease?  contribute to the eventual destabilization of
Epidemiologic studies demonstrate strong asso- the plaque. They may also lead to myocardi-
ciations between oral infections and cardiovas- al infarction or ischemic stroke. Also, oral mi-
cular disease and events. The effect is of clinical crobes contribute to the cumulative burden of
significance and is independent of other known systemic inflammatory responses that may lead
risk factors. However, we cannot yet claim that to atherosclerosis.

176
References

Does the delivery of oral health care contribute What is the appropriate message to give the
to improvement of cardiovascular disease out- public about the association between oral health
comes?  Well-designed intervention studies and cardiovascular disease?  Gum disease and
demon­ strate improvement in several estab- heart disease may go together, but scientists
lished biomarkers for cardiovascular disease. cannot yet prove that one leads to the other.
However, there are no studies showing im- The association may be due to unhealthy life-
proved clinical signs or symptoms. styles (eg, smoking, poor nutrition, sedentary
lifestyle). Adopting a healthy lifestyle in the
Is it safe to provide dental care to patients with context of oral health prevention or treatment
cardiovascular disease?
  From the perspective may bring benefits related to atherosclerosis
of disseminating oral bacteria, the transient onset and development. Microbes that usually
bacteremia caused by infrequent dental pro- live in the mouth enter the bloodstream, espe-
cedures represents much less hazard than ev- cially in the presence of gum disease. Therefore,
eryday activities such as tooth brushing and it is important for clinicians to emphasize to
flossing or mastication of hard food items. Pro- their patients that health of the mouth is part
vision of dental care to patients at risk for isch- of overall health and well-being.
emic events should be done after consultation
with the attending physician/cardiologist.

References
1. Mattila KJ, Nieminen MS, Valtonen VV, et al. Association 9. Salzberg TN, Overstreet BT, Rogers JD, Califano JV,
between dental health and acute myocardial infarction. Best AM, Schenkein HA. C-reactive protein levels in
BMJ 1989;298:779–781. patients with aggressive periodontitis. J Periodontol
2. Kiechl S, Egger G, Mayr M, et al. Chronic infections and 2006;77:933–939.
the risk of carotid atherosclerosis: Prospective results 10. Havemose-Poulsen A, Westergaard J, Stoltze K, et al.
from a large population study. Circulation 2001; 103: Periodontal and hematological characteristics associ-
1064–1070. ated with aggressive periodontitis, juvenile idiopathic
3. Andraws R, Berger JS, Brown DL. Effects of antibiotic arthritis, and rheumatoid arthritis. J Periodontol 2006;
therapy on outcomes of patients with coronary artery 77:280–288.
disease: A meta-analysis of randomized controlled trials. 11. Joshipura KJ, Wand HC, Merchant AT, Rimm EB. Peri-
JAMA 2005;293:2641–2647. odontal disease and biomarkers related to cardiovascular
4. Paju S, Pussinen PJ, Sinisalo J, et al. Clarithromycin disease. J Dent Res 2004;83:151–155.
reduces recurrent cardiovascular events in subjects 12. Buhlin K, Gustafsson A, Pockley AG, Frostegård J,
without periodontitis. Atherosclerosis 2006; 188: Klinge B. Risk factors for cardiovascular disease in
412–419. patients with periodontitis. Eur Heart J 2003; 24:
5. López NJ, Socransky SS, Da Silva I, Japlit MR, Haffajee 2099–2107.
AD. Effects of metronidazole plus amoxicillin as the only 13. Amar S, Gokce N, Morgan S, Loukideli M, Van Dyke
therapy on the microbiological and clinical parameters TE, Vita JA. Periodontal disease is associated with
of untreated chronic periodontitis. J Clin Periodontol brachial artery endothelial dysfunction and systemic
2006;33:648–660. inflammation. Arterioscler Thromb Vasc Biol 2003; 23:
6. Paraskevas S, Huizinga JD, Loos BG. A systematic 1245–1249.
review and meta-analyses on C-reactive protein in rela- 14. Craig RG, Yip JK, So MK, Boylan RJ, Socransky SS,
tion to periodontitis. J Clin Periodontol 2008; 35: Haffajee AD. Relationship of destructive periodontal
277–290. disease to the acute-phase response. J Periodontol
7. Teeuw WJ, Slot DE, Susanto H, et al. Treatment of peri- 2003;74:1007–1016.
odontitis improves the atherosclerotic profile: A systematic 15. Noack B, Genco RJ, Trevisan M, Grossi S, Zambon JJ,
review and meta-analysis. J Clin Periodontol 2014;41: De Nardin E. Periodontal infections contribute to
70–79. elevated systemic C-reactive protein level. J Periodontol
8. Bizzarro S, Van der Velden U, Ten Heggeler JM, et al. 2001;72:1221–1227.
Periodontitis is characterized by elevated PAI-1 activity.
J Clin Periodontol 2007;34:574–580.

177
7 The Cardiovascular System and Oral Infections

16. Loos BG, Craandijk J, Hoek FJ, Wertheim-van Dillen 30. Sharma A, Novak EK, Sojar HT, Swank RT, Kuramitsu
PM, Van der Velden U. Elevation of systemic markers HK, Genco RJ. Porphyromonas gingivalis platelet aggre-
related to cardiovascular diseases in the peripheral gation activity: Outer membrane vesicles are potent
blood of periodontitis patients. J Periodontol 2000;71: activators of murine platelets. Oral Microbiol Immunol
1528–1534. 2000;15:393–396.
17. Fredriksson M, Gustafsson A, Asman B, Bergström K. 31. Kuramitsu HK, Qi M, Kang IC, Chen W. Role for peri-
Hyper-reactive peripheral neutrophils in adult periodon- odontal bacteria in cardiovascular diseases. Ann
titis: Generation of chemiluminescence and intracellular Periodontol 2001;6:41–47.
hydrogen peroxide after in vitro priming and 32. Ridker PM. Intrinsic fibrinolytic capacity and systemic
FcgR-stimulation. J Clin Periodontol 1998;25:394–398. inflammation: Novel risk factors for arterial thrombotic
18. Almeida APCPSC, Fagundes NCF, Maia LC, Lima RR. disease. Haemostasis 1997;27(suppl 1):2–11.
Is there an association between periodontitis and athero- 33. Hutter JW, van der Velden U, Varoufaki A, Huffels RA,
sclerosis in adults? A systematic review. Curr Vasc Hoek FJ, Loos BG. Lower numbers of erythrocytes and
Pharmacol 2018;16:569–582. lower levels of hemoglobin in periodontitis patients
19. Tómas I, Diz P, Tobías A, Scully C, Donos N. Periodontal compared to control subjects. J Clin Periodontol 2001;
health status and bacteraemia from daily oral activities: 28:930–936.
Systematic review/meta-analysis. J Clin Periodontol 34. Offenbacher S, Farr DH, Goodson JM. Measurement
2012;39:213–228. of prostaglandin E in crevicular fluid. J Clin Periodontol
20. Reyes L, Kozarov E, Herrera D, Roldán S, Progulske-Fox 1981;8:359–367.
A. Periodontal bacterial invasion and infection: Contri- 35. Slade GD, Offenbacher S, Beck JD, Heiss G, Pankow
bution to atherosclerotic pathology. J Clin Periodontol JS. Acute-phase inflammatory response to periodontal
2013;40(suppl 14):30–50. disease in the US population. J Dent Res 2000; 79:
21. Schenkein HA, Loos BG. Inflammatory mechanisms 49–57.
linking periodontal diseases to cardiovascular diseases. 36. Wu T, Trevisan M, Genco RJ, Falkner KL, Dorn JP,
J Clin Periodontol 2013;40(suppl 14):51–69. Sempos CT. Examination of the relation between peri-
22. Lalla E, Lamster IB, Hofmann MA, et al. Oral infection odontal health status and cardiovascular risk factors:
with a periodontal pathogen accelerates early athero- Serum total and high density lipoprotein cholesterol,
sclerosis in apolipoprotein E-null mice. Arterioscler C-reactive protein, and plasma fibrinogen. Am J Epide-
Thromb Vasc Biol 2003;23:1405–1411. miol 2000;151:273–282.
23. Li L, Messas E, Batista EL Jr, Levine RA, Amar S. 37. D’Aiuto F, Parkar M, Nibali L, Suvan J, Lessem J, Tonetti
Porphyromonas gingivalis infection accelerates the MS. Periodontal infections cause changes in traditional
progression of atherosclerosis in a heterozygous apoli- and novel cardiovascular risk factors: Results from a
poprotein E-deficient murine model. Circulation 2002; randomized controlled clinical trial. Am Heart J 2006;
105:861–867. 151:977–984.
24. Haraszthy VI, Zambon JJ, Trevisan M, Zeid M, Genco 38. Tonetti MS, Van Dyke TE, Working Group 1 of the Joint
RJ. Identification of periodontal pathogens in athero- EFP/AAP Workshop. Periodontitis and atherosclerotic
matous plaques. J Periodontol 2000;71:1554–1560. cardiovascular disease: Consensus report of the Joint
25. Fiehn NE, Larsen T, Christiansen N, Holmstrup P, EFP/AAP Workshop on Periodontitis and Systemic
Schroeder TV. Identification of periodontal pathogens in Diseases. J Clin Periodontol 2013; 40(suppl 14):
atherosclerotic vessels. J Periodontol 2005;76:731–736. S24–S29.
26. Aimetti M, Romano F, Nessi F. Microbiologic analysis of 39. Beck JD, Offenbacher S. Relationships among clinical
periodontal pockets and carotid atheromatous plaques measures of periodontal disease and their associations
in advanced chronic periodontitis patients. J Periodontol with systemic markers. Ann Periodontol 2002;7:79–89.
2007;78:1718–1723. 40. Khader YS, Albashaireh ZS, Alomari MA. Periodontal
27. Pessi T, Karhunen V, Karjalainen PP, et al. Bacterial diseases and the risk of coronary heart and cerebro-
signatures in thrombus aspirates of patients with vascular diseases: A meta-analysis. J Periodontol 2004;
myocardial infarction. Circulation 2013; 127: 75:1046–1053.
1219–1228. 41. Blaizot A, Vergnes JN, Nuwwareh S, Amar J, Sixou M.
28. Dorn BR, Burks JN, Seifert KN, Progulske-Fox A. Inva- Periodontal diseases and cardiovascular events:
sion of endothelial and epithelial cells by strains of Meta-analysis of observational studies. Int Dent J 2009;
Porphyromonas gingivalis. FEMS Microbiol Lett 2000; 59:197–209.
187:139–144. 42. Lafon A, Pereira B, Dufour T, et al. Periodontal disease
29. Zhang B, Khalaf H, Sirsjö A, Bengtsson T. Gingipains and stroke: A meta-analysis of cohort studies. Eur J
from the periodontal pathogen Porphyromonas gingi- Neurol 2014;21:1155–1161,e66–67.
valis play a significant role in regulation of angiopoietin 43. Leira Y, Seoane J, Blanco M, Rodríguez-Yáñez M,
1 and angiopoietin 2 in human aortic smooth muscle Takkouche B, Blanco J, Castillo J. Association between
cells. Infect Immun 2015;83:4256–4265. periodontitis and ischemic stroke: A systematic review
and meta-analysis. Eur J Epidemiol 2017;32:43–53.

178
References

44. Dietrich T, Sharma P, Walter C, Weston P, Beck J. The 58. Couper DJ, Beck JD, Falkner KL, et al. The periodontitis
epidemiological evidence behind the association and vascular events (PAVE) pilot study: Recruitment,
between periodontitis and incident atherosclerotic retention, and community care controls. J Periodontol
cardiovascular disease. J Clin Periodontol 2013; 2008;79:80–89.
40(suppl 14):70–84. 59. Papapanou PN. Systemic effects of periodontitis:
45. Schaefer AS, Richter GM, Groessner-Schreiber B, et Lessons learned from research on atherosclerotic
al. Identification of a shared genetic susceptibility locus vascular disease and adverse pregnancy outcomes. Int
for coronary heart disease and periodontitis. PLoS Dent J 2015;65:283–291.
Genet 2009;5:e1000378. 60. Orlandi M, Suvan J, Petrie A, et al. Association between
46. Bochenek G, Häsler R, El Mokhtari NE, et al. The large periodontal disease and its treatment, flow-mediated
non-coding RNA ANRIL, which is associated with dilatation and carotid intima-media thickness: A system-
atherosclerosis, periodontitis and several forms of atic review and meta-analysis. Atherosclerosis 2014;
cancer, regulates ADIPOR1, VAMP3 and C11ORF10. 236:39–46.
Hum Mol Genet 2013;22:4516–4527. 61. Demmer RT, Trinquart L, Zuk A, et al. The influence of
47. D’Aiuto F, Nibali L, Parkar M, Suvan J, Tonetti MS. anti-infective periodontal treatment on C-reactive
Short-term effects of intensive periodontal therapy on protein: A systematic review and meta-analysis of
serum inflammatory markers and cholesterol. J Dent randomized controlled trials. PLoS One 2013;8:e77441.
Res 2005;84:269–273. 62. Sanz M, Ceriello A, Buysschaert M, et al. Scientific
48. D’Aiuto F, Parkar M, Andreou G, et al. Periodontitis and evidence on the links between periodontal diseases and
systemic inflammation: Control of the local infection is diabetes: Consensus report and guidelines of the Joint
associated with a reduction in serum inflammatory mark- Workshop on Periodontal Diseases and Diabetes by
ers. J Dent Res 2004;83:156–160. the International Diabetes Federation and the European
49. Elter JR, Hinderliter AL, Offenbacher S, et al. The effects Federation of Periodontology. J Clin Periodontol 2018;
of periodontal therapy on vascular endothelial function: 45:138–149.
A pilot trial. Am Heart J 2006;151:47. 63. Jansen H, Samani NJ, Schunkert H. Mendelian random-
50. D’Aiuto F, Nibali L, Mohamed-Ali V, Vallance P, Tonetti ization studies in coronary artery disease. Eur Heart J
MS. Periodontal therapy: A novel non-drug-induced 2014;35:1917–1924.
experimental model to study human inflammation. J 64. West S, King V, Carey TS, et al. Systems to rate the
Periodontal Res 2004;39:294–299. strength of scientific evidence. Evid Rep Technol Assess
51. D’Aiuto F, Parkar M, Tonetti MS. Acute effects of peri- (Summ) 2002;(47):1–11.
odontal therapy on bio-markers of vascular health. J 65. Ross R. The pathogenesis of atherosclerosis: A
Clin Periodontol 2007;34:124–129. perspective for the 1990s. Nature 1993;362:801–809.
52. Graziani F, Cei S, La Ferla F, Vano M, Gabriele M, Tonetti 66. Gimbrone MA Jr, Cybulsky MI, Kume N, Collins T,
M. Effects of non-surgical periodontal therapy on Resnick N. Vascular endothelium. An integrator of
glomerular filtration rate of the kidney: An exploratory pathophysiological stimuli in atherogenesis. Ann N Y
trial. J Clin Periodontol 2010;37:638–643. Acad Sci 1995;748:122–131.
53. Graziani F, Cei S, Tonetti M, et al. Systemic inflammation 67. Seinost G, Wimmer G, Skerget M, et al. Periodontal
following non-surgical and surgical periodontal therapy. treatment improves endothelial dysfunction in patients
J Clin Periodontol 2010;37:848–854. with severe periodontitis. Am Heart J 2005; 149:
54. Graziani F, Cei S, Orlandi M, et al. Acute-phase response 1050–1054.
following full-mouth versus quadrant non-surgical peri- 68. Higashi Y, Goto C, Hidaka T, et al. Oral infection-
odontal treatment: A randomized clinical trial. J Clin inflammatory pathway, periodontitis, is a risk factor for
Periodontol 2015;42:843–852. endothelial dysfunction in patients with coronary artery
55. Minassian C, D’Aiuto F, Hingorani AD, Smeeth L. Inva- disease. Atherosclerosis 2009;206:604–610.
sive dental treatment and risk for vascular events: A 69. Higashi Y, Goto C, Jitsuiki D, et al. Periodontal infection
self-controlled case series. Ann Intern Med 2010;153: is associated with endothelial dysfunction in healthy
499–506. subjects and hypertensive patients. Hypertension 2008;
56. Tonetti MS, D’Aiuto F, Nibali L, et al. Treatment of peri- 51:446–453.
odontitis and endothelial function. N Engl J Med 2007; 70. Lockhart PB, Bolger AF, Papapanou PN, et al. Peri-
356:911–920. odontal disease and atherosclerotic vascular disease:
57. Beck JD, Couper DJ, Falkner KL, et al. The periodontitis Does the evidence support an independent association?
and vascular events (PAVE) pilot study: Adverse events. A scientific statement from the American Heart Associ-
J Periodontol 2008;79:90–96. ation. Circulation 2012;125:2520–2544.

179
CHAPTER 8

The Association Between Oral


Infections and Renal Disease
Karren Komitas, dmd, phd, mdsc
Effie Ioannidou, dds, mds

The growing population and continuously of periodontitis, especially in older adults,


increasing life expectancy worldwide have are associated with various inflammatory
resulted in a dramatic growth in the propor- systemic diseases, including type 2 diabetes
tion and absolute numbers of older adults.1,2 mellitus, obesity, rheumatoid arthritis, and
In the United States, the population aged 65 others.8,9 Underlying mechanisms of these
years and older reached 45 million in 2015 associations have been extensively studied in
and is projected to reach almost 100 million various human and animal studies, which have
by 2060.3 These trends raise multiple health demonstrated that this association can be due
care concerns regarding the greater preva- to similar mechanisms of development, shared
lence of various local and systemic diseases inflammatory pathways and mediators, and
in older individuals and associated high eco- host genetic predisposition.10–12 In addition,
nomic costs.4 older adults often present with age-associated
Periodontitis is a polymicrobial multifacto- impaired adaptive immune system responses
rial inflammatory disease of tooth-supporting and persistent low-grade inflammation, thus
tissues5 and is one of the most common human making them more susceptible to microbiota of
infectious diseases. The overall prevalence of periodontal tissues.13,14 Therefore, treatment of
periodontitis in the United States is currently a systemic comorbidity can potentially alleviate
estimated at 42%,6 and its prevalence increases the severity of periodontitis, and vice versa.10
with age, reaching 62% in individuals aged 65 The term periodontal medicine has been intro-
years and older.7 It has been established that duced to define a branch of periodontology
the chronic nature of periodontitis leads to that examines possible associations between
continuous attachment loss over a long period periodontitis and systemic diseases and devel-
of time; impaired ability or inability to main- ops novel diagnostic and treatment strategies
tain proper oral hygiene, aging, smoking, poor based on the evidence of these associations.15
socioeconomic status, genetics, male sex, and The question whether periodontitis is an
decreased health literacy are among the factors independent risk factor for the development
that can contribute to the increased prevalence of a systemic disease still remains a matter of
of periodontitis in older adults.7 debate in the scientific community. The goal of
In addition, multiple lines of evidence this chapter is to assess whether current evi-
during the past two decades have demon- dence exists for an independent association
strated that the development and progression between periodontitis and renal disease, and

180
Overview of Renal Diseases

if so, to gain insight into the biologic rationale • Stage 2: Mild eGFR decrease (60–89 mL/
and mechanisms of this association. We have min/1.73 m2 )
reviewed the relevant pathophysiology and • Stage 3: Moderate eGFR decrease (30–59
predominant theories of the underlying mech- mL/min/1.73 m2 )
anisms for the development and progression • Stage 4: Severe eGFR decrease (15–29 mL/
of these diseases. We have also assessed the min/1.73 m2 )
clinical significance of these results to deter- • Stage 5: Kidney failure (eGFR < 15 mL/
mine whether improved oral hygiene and min/1.73 m2 )
periodontal treatment can reduce a risk for
renal disease, and if so, what protocols these In a recent meta-analysis of the epidemio-
strategies involve. Finally, we have aimed to logic evidence on the global prevalence of CKD
reveal gaps in our current knowledge to delin- by disease stage, stage 3 was found to have
eate the directions of future research efforts. the highest prevalence.20 CKD prevalence was
higher in developed countries with increased
numbers of elderly populations. After geo-
graphic stratification, Europe demonstrated
Overview of Renal Diseases the highest mean CKD prevalence in stages
1 to 5 of 18.38% (95% confidence interval
Chronic kidney disease (CKD) is a worldwide [CI]: 11.57–25.20). When CKD prevalence was
public health problem with global prevalence restricted to stages 3 to 5 and compared among
ranging from 8% to 16%.16 CKD has recently regions, the United States showed the highest
been ranked third in the list of diseases caus- mean CKD prevalence of 14.44% (95% CI:
ing premature mortality following AIDS and 8.52–20.36).20 This finding may result from
diabetes.17 Based on recent data, CKD is a methodologic heterogeneity in creatinine mea-
costly public health problem, with more than surements inherent in most studies. Among
4.6 million hospital outpatient visits and over the rest of the world, Iran presented with sim-
$50 billion USD in Medicare expenses (20% of ilarly high CKD prevalence due to high rates
the total Medicare spending) in 2014.18 of comorbidities such as high body mass index
CKD is defined by impaired kidney function, (BMI) and high systolic blood pressure.20
as measured by an estimated glomerular filtra- As diagnostic methods have evolved, each
tion rate (eGFR) less than 60 mL/min/1.73 m2 CKD stage has been divided into substages to
of body surface area, with or without kidney incorporate albuminuria (presence of albumin
damage, for at least 3 months.19 The eGFR in the urine), given that the levels of albumin
measures the clearance of a particular marker in the urine affect disease progression and
by the kidney, and therefore it is the standard outcomes.21 More specifically, each substage is
measure of renal excretory function and kid- defined based on specific albuminuria cutoffs:
ney health in general. Several exogenous and
endogenous clearance markers have been uti- • A1: Normal to mildly increased (< 30 mg/g
lized to assess kidney function and estimate or < 3 mg/mmol)
the eGFR, and among them serum creatinine • A2: Moderately increased (30–300 mg/g or
remains the most commonly employed marker. 2–20 mg/mmol)
There are five eGFR-based clinical stages of • A3: Severely increased (> 300 mg/g or > 30
CKD, which signify the degree of kidney dam- mg/mmol)
age and reduced kidney function:
When an eGFR is less than 15 mL/min/1.73
• Stage 1: Normal or increased eGFR (≥ 90 m2 and weekly renal urea clearance is below
mL/min/1.73 m2 ) 2.0 (a sign of severe uremia), patients are

181
8 The Association Between Oral Infections and Renal Disease

diagnosed with end-stage renal disease (ESRD) ESRD.23 This finding highlights the success of
and treated with renal replacement therapy.22 the systematic efforts for the treatment of kid-
The most recent data (2015) on the distribu- ney failure in the United States.
tion of renal replacement treatment modalities
reveal that the majority of patients are main-
tained with hemodialysis (87.3%), followed by
peritoneal dialysis (9.6%), and only 2.5% with
Epidemiology of Periodontitis
kidney transplantation.23 In the United States in Patients with CKD/ESRD
in 2015, approximately 26 million individu-
als were diagnosed with CKD, and there were Epidemiologic evidence based on large pop-
703,243 patients with ESRD on dialysis.23 ulation studies in CKD has shown that the
CKD risk factors include diabetes, hyper- prevalence of moderate and severe peri-
tension, autoimmune diseases (eg, systemic odontitis ranges from 11.5% to 14.7%.27–29
lupus erythematosus), urinary tract infections, Heterogeneity in disease cutoffs, examination
urinary stones, neoplasia, family history of methodologies (full- versus partial-mouth pro-
kidney disease, low birth weight, and demo- tocols), as well as population demographic
graphic characteristics such as race and age.21 differences do not allow the direct comparison
In addition, chronic use of nonsteroidal of periodontitis estimates in CKD with those in
anti-inflammatory medications has been a the general population total periodontitis (mild
documented risk factor for CKD.22,24 Between to severe) estimate.30 Using the National Health
25% and 40% of patients with type 1 or type 2 and Nutrition Examination Survey (NHANES)
diabetes mellitus develop diabetic nephropathy III database, our group has demonstrated a
with progressive loss of kidney function.25 As statistically significant higher prevalence of
diabetes prevalence has dramatically increased moderate periodontitis in patients with CKD
in the last few decades, CKD prevalence has also compared to non-CKD populations also
increased. The CKD progression is predicted characterized by racial disparities as shown
by several factors, including the underlying in Table 8-1.29 Overcoming the NHANES III
diagnosis, kidney function and albuminuria, limitations for periodontitis prevalence assess-
age, sex, race, hypertension, hyperlipidemia, ments,31 our group has adapted appropriate
hyperglycemia, smoking, history of cardiovas- prevalence adjustments, as described before,
cular disease (CVD), and obesity.21 Although and estimated approximately 6 million patients
CKD prevalence has increased over the last with CKD with moderate and severe periodon-
few decades, systematic efforts in glycemic titis,29 emphasizing an important public health
control and antihypertensive measures includ- problem in this special-needs population.
ing renin-angiotensin blockers have resulted Other research groups have reported the
in a stabilization of its incidence in the United prevalence of periodontitis among individ-
States.26 More specifically, in the advanced uals with ESRD ranging between 29% and
CKD stages, the adjusted prevalence of stages 64%.32–36 Despite recognition of periodontitis
3 and 4 increased during the early 2000s and as a critical public health problem,37 disease
then stabilized.26 Once the disease progressed awareness, especially in medically complex
to the ESRD stage, epidemiologic data have populations, is low.38 Poor oral health has
shown that the age-, sex-, and race-adjusted been shown to compromise chewing capac-
incidence rates for ESRD increased from 1980 ity, which affects protein intake, energy levels,
to 2001, were stable through 2006, and then and serum albumin levels in CKD/ESRD pop-
slightly declined. However, the overall ESRD ulations, thus resulting in malnutrition and
prevalence has increased by 3.4% since 2014 disease progression.39 Given the detrimental
due to longer survival among patients with effects of periodontitis on oral health, effective

182
Biologic Plausibility for the Increased Prevalence of Periodontitis in Patients with CKD/ESRD

TABLE 8-1  |  Periodontitis prevalence as stratified by race in CKD and non-CKD populations (n = 11,604)

Non-Hispanic whites Non-Hispanic blacks Mexican Americans


Non-CKD Non-CKD
CKD N= CKD N= CKD Non-CKD
N = 437 4,216 P value N = 88 3,182 P value N = 81 N = 3,600 P value
Periodontitis 12.9 (1.8) 7.5 (0.4) .001 38.9 (5.8) 14.6 (0.5) .001 37.3 (6.8) 9.0 (0.5) .001
Data represents prevalence (%) and standard deviation (SD). Notice the statistically significant difference in prevalence between CKD and non-CKD
populations in every racial group. For the entire sample, inflation-adjusted prevalence was 35.28% in CKD (data not shown). (Data from Ioannidou and
Swede.29)

treatment and control of periodontitis focused In our conceptual model, high urea levels in
on improving and maintaining oral health may the serum and gingival crevicular fluid (GCF)
need to become a priority in medically complex could alter the oral ecosystem, promoting the
renal populations. growth of periodontal pathogens, similar to
the demonstrated uremia-related changes in the
gut environment46 (Fig 8-1). More specifically,
evidence has shown that high urea levels in the
Biologic Plausibility for the gut environment resulted in a distinct microbi-
Increased Prevalence of ome in a renal failure rat model46 (see Fig 8-1).
Periodontitis in Patients with Because GCF derives primarily from serum,
increased serum urea levels are also reflected
CKD/ESRD in GCF.47,48 A variety of bacteria such as Strep-
tococcus salivarius and Actinomyces naeslundii
Periodontitis is a complex disease that involves produce urease, which catalyzes the hydrolysis
the interaction of subgingival biofilms with the of urea, resulting in ammonia, which may then
susceptible host resulting in connective tissue be protonated to produce ammonium ion and
and bone changes.14,40 In renal disease, several directly alkalize the bacterial cytoplasm49 or the
mechanisms have been proposed to contribute extracellular environment.50 Selected periodon-
to the pathogenesis of periodontitis, includ- tal pathogens such as Prevotella intermedia,
ing uremic effects on the oral environment, Fusobacterium nucleatum, Porphyromonas
uremic immunosuppression, and vitamin D gingivalis, and even Helicobacter pylori show
deficiency.34,41,42 With the progressive decline optimal growth in alkaline pH.51 Although
in glomerular filtration, uremic solutes/tox- there have been limited cross-sectional obser-
ins, typically eliminated in the urine, are vations indicating a higher frequency of red
retained in the body,43 contributing to anemia, complex periodontal bacteria in ESRD sub-
immunosuppression, inflammation, infec- jects with periodontitis than in systemically
tion, cardiovascular morbidity, and mortality. healthy controls,52,53 the role of uremia-related
The deleterious effects of uremia are most factors (urea, subgingival pH) as ecologic
pronounced in ESRD.44 As uremia becomes determinants has never been examined. In a
pronounced, it has been shown that salivary comprehensive investigation of the microbial
urea levels dramatically increase from 7.5 communities in the uremia-modified subgingi-
μmol/L to 17 to 26 μmol/L.45 val environment, Araújo et al characterized the

183
8 The Association Between Oral Infections and Renal Disease

Subgingival Host Connective Clinical


microbiome response tissue and bone presentation of
breakdown periodontitis

•  Uremic saliva •  Uremic innate •  Vitamin D


•  Urease activity and adaptive deficiency
•  Alkaline pH dysfunction •  Renal
•  Oral ecosystem •  Oxidative stress osteodystrophy
alterations •  Vitamin D •  Oxidative stress
deficiency

Fig 8-1  |  The conceptual model of modified periodontitis pathogenesis in CKD.

subgingival microbiome of patients with ESRD tuberculosis,64 reflected in a relative risk (RR)
and compared it to systemically healthy control for active tuberculosis of 7 to 2565,66 in patients
individuals using 16S rRNA gene sequencing.54 with ESRD. Thus, the uremic effects on innate
Although the study did not reveal differences and adaptive host response may also repre-
in proportions of periodontitis-associated sent a modifying factor in the pathogenesis of
taxa, there is a negative correlation between periodontitis as CKD progresses to advanced
microbiome diversity and dialysis vintage stages.
(years on dialysis). More specifically, the study As the renal function is compromised, the
showed that the years on dialysis negatively metabolic pathway of vitamin D—conversion
correlate with the diversity of the microbial to 1,25(OH)2D in the kidneys—is disturbed
communities.54 due to the inhibition of 1-α-hydroxylase activ-
Additional effects of the uremic milieu on ity (produced by the kidney).67 Vitamin D
both innate and adaptive immunity55 alter deficiency has been found to play a dual role in
host response by several mechanisms such as abnormal bone breakdown and turnover (renal
functional abnormalities of neutrophils, mono- osteodystrophy)68 and alteration of the innate
cytes/macrophages, and dendritic cells43,56–58; (monocyte activation and phagocytosis) and
abnormal neutrophil activity 59; impaired adaptive (modulation of cytokine production)
maturation of T helper cells60; and increased immunity.69
oxidative stress.61,62 As uremia increases with Figure 8-1 summarizes our conceptual
CKD progression, uremic effects become model, in which the pathogenesis of peri-
critical in ESRD.63 The innate and adaptive odontitis (shown in blue boxes) is modified
immunity effects of uremia may lead to a dis- by CKD/ESRD comorbid factors (shown in
abled immune response and, in combination white boxes). The interaction of the CKD/
with dialysis-related factors (such as membrane ESRD comorbid factors and periodontitis eti-
biocompatibility and type of vascular access), ologic events not only explains the increased
increase infection susceptibility as confirmed prevalence of periodontitis in this population
by high prevalence of chronic infection by but can also affect the response to periodontal
Chlamydia pneumoniae and Mycobacterium treatment in these populations.

184
Response of Patients with CKD to Periodontal Therapy

Periodontitis and the with a dose-response association between


CRP and periodontitis stages possibly lead-
Inflammatory Burden in ing to increased all-cause mortality rates.80 As
Patients with CKD/ESRD confirmed by analogy,81 other infections have
shown to result in increased levels of inflamma-
tory mediators in ESRD and to consequently
Within the last few decades, the high inflamma- contribute to the atherosclerotic process and
tory burden in CKD/ESRD has been attributed increased mortality.64,82,83 Therefore, the rela-
to the “uremic puzzle,” with pieces developing tionship between periodontitis and CKD/ESRD
and connecting in an intricate manner70 con- is potentially bidirectional, with CKD/ESRD
tributing to CVD mortality. We now know that not only potentiating the incidence of peri-
the complexity of the “uremic puzzle” extends odontitis but periodontitis likely contributing
past the Framingham CVD risk factors involv- to sustained inflammation and poor outcomes
ing systemic inflammation and oxidative stress in CKD/ESRD.
as variables strongly associated with poor CVD
outcomes in CKD.70 Overwhelming evidence
has indicated that the enzymatic activity of
myeloperoxidase links oxidative stress and
Response of Patients with
inflammation in uremia, thus emphasizing CKD to Periodontal Therapy
the importance of both in the atheroscle-
rotic process.71 Consequently, research has The elimination of the etiologic factors of
focused on identifying factors contributing periodontitis is the main therapeutic goal of
to systemic inflammation and oxidative stress nonsurgical periodontal therapy. Such therapy
at different levels. At an ethnic level, sev- results in biofilm disruption, reduction of clini-
eral culture-related dietary habits have been cal signs of inflammation, and improvement of
identified as anti-inflammatory, correlating with surrogate clinical periodontal parameters such
a low CVD risk.72 Dialysis-related factors such as probing pocket depth (PPD) and clinical
as membrane bioincompatibility, type of access, attachment loss (CAL).84 Importantly, periodon-
and impure dialysate have also been linked to tal status seems to have a significant impact on
systemic inflammation.73 At the patient level, oral health–related quality of life as measured
additional comorbid factors including bacterial by patient-centered outcomes.85 In terms of
infections, volume overload, failed transplant, microbiologic outcomes, the immediate effect of
and depression have been implicated in the nonsurgical periodontal therapy is the disrup-
elevated serum C-reactive protein (CRP)74 and tion and modification of the composition of the
are targeted in anti-inflammatory therapeutic bacterial biofilm, as shown by a reduction in the
efforts.63,74 total mass of subgingival plaque and decreased
In the same context, the American Heart levels and prevalence of periodontitis-associated
Association has recognized the independent species.86 Although the most drastic changes in
association between periodontitis and systemic biofilm composition occur immediately after
inflammation75 as expressed by high levels of therapy, the prevalence of red complex spe-
inflammatory biomarkers such as interleukin cies has been shown to remain low during
6 (IL-6) and CRP.76–78 Our group and others a 12-month follow-up.84 However, in the
produced evidence supporting the contribu- absence of supportive periodontal therapy, 50%
tion of periodontitis to inflammation in CKD of the originally infected sites are recolonized
as measured by CRP levels.28,42,79 Moreover, within 12 months.87 Hence, sustained improve-
the impact of periodontitis on serum CRP lev- ments after therapy are dependent on effective
els has been shown to be significant in ESRD, initial intervention and adequate maintenance.

185
8 The Association Between Oral Infections and Renal Disease

Effectiveness of Periodontal 80% reduction in periodontal lesions, which


significantly correlates with changes in inflam-
Therapy matory markers.93 Many studies94,95 have been
criticized for not achieving clinically accept-
Periodontal outcomes able endpoints prior to evaluating systemic
Nonsurgical periodontal therapy consists endpoints. When the treatment-associated oral
of scaling and root planing (SRP) under response is not pronounced, systemic effects are
local anesthesia with the primary goal to not evident.91,96
eliminate the microbial disease etiology (bac- A single 6-month randomized controlled trial
terial plaque), reduce PPD, improve CAL, (RCT) that examined the effect of periodontal
and ultimately prevent tooth loss.88 Peri- treatment on ESRD presented inconclusive
odontal treatment response is based on the results due to design limitations, including
improvement of periodontal parameters and (1) the episodic periodontal therapy approach
is maintained through maintenance visits without maintenance, as confirmed by the poor
to prevent disease relapse based on patient oral hygiene scores and declining periodontal
periodontal risk assessment.89 Studies have parameters within the observational period,
assessed the clinical effectiveness of the non- and (2) the overrepresentation of diabetic
surgical periodontal intervention in CKD and patients in the treatment arm (62%) versus
systemically healthy populations focusing on control arm (38%).91 Evidently, the episodic
periodontal outcomes. Despite the hetero- treatment approach is not effective enough
geneous study designs and methods, studies to promote optimal oral hygiene, prevent
agree that periodontal intervention is less recurrent periodontitis, and control systemic
effective in CKD/ESRD populations com- inflammation.97–99
pared to systemically healthy populations. For anti-inflammatory strategies to be
These data emphasized the need for a modi- effective, they need to consider all comorbidi-
fied periodontal therapy approach in patients ties.100,101 Oral health care needs to be a part of
with ESRD in order to achieve results similar the interprofessional collaboration taking place
to those in systemically healthy individuals.90,91 in the outpatient dialysis units, which include
Currently, only 11% of patients with CKD/ nephrologists, nurses, dietitians, social work-
ESRD have one dental visit a year,44 possibly ers, and renal access surgeons. To facilitate this,
due to low socioeconomic status and low oral general practitioners and specialists need to be
health awareness. Therefore, access to care familiar with the therapeutic needs of patients
and oral health awareness would need to be with CKD/ESRD.
addressed in this population with the develop-
ment of a multidisciplinary care model. Oral
health models especially for ESRD patients on
dialysis might need to be tailored to accommo-
Dental Management of
date patient fatigue and physical limitations. Patients with CKD/ESRD
For successful management of patients with
Systemic outcomes CKD/ESRD, a dental practitioner needs to
The systemic anti-inflammatory effect of develop a close collaboration with a nephrology
periodontal therapy is expected once a posi- team. Appropriate clinical and pharmaceuti-
tive treatment response occurs at a local/oral cal decisions and management depend on the
level with improved periodontal measures.92 severity and staging of CKD. In CKD stages 1
In systemically healthy populations, peri- to 4, the oral health practitioner has to follow
odontal intervention results in approximately kidney function as measured by an eGFR as

186
Dental Management of Patients with CKD/ESRD

well as other medical comorbidities in order to kidney transplantation. Patients on hemodial-


understand the severity of kidney disease and to ysis have vascular access to be able to connect
increase patient awareness about periodontal to the dialysis device in a frequency of three
risk and prescribe medications accordingly. For treatments per week. Patients with ESRD on
patients with CKD stage 5 or ESRD, the oral hemodialysis usually receive anticoagulation
health provider will have to know the dialysis with unfractionated heparin (UFH) depending
maintenance schedule as well as patient com- on their bleeding risk.102 In cases where the
pliance as it may affect access to dental care. patient is determined to be at a high bleeding
The majority of patients with renal diseases risk (eg, patients with clotting factor disor-
have an underlying diagnosis of either diabe- ders), heparin-free hemodialysis is performed.
tes or hypertension. Therefore, they require In the United States, UFH is administered in a
close monitoring of their vital signs (ie, blood bolus dose of 50 IU/kg at the beginning of the
pressure and pulse) as well as their glycated hemodialysis session followed by a constant
hemoglobin (HbA1c) levels. Ensuring stable fixed infusion of heparin of 800 to 1,500 IU
vital signs and controlled HbA1c levels will per hour.102 Low–molecular weight heparin
improve periodontal therapeutic outcomes. (LMWH) is used to bridge other anticoagulants
As kidney function becomes more compro- prior to an invasive procedure. UFH has a short
mised, dental management needs to be adjusted half-life of 1 hour after the last infusion, while
with more attention to drugs commonly used in LMWH’s half-life is two to four times higher.103
dentistry and metabolized and/or eliminated in The notion that dental visits for patients on
the kidney. More specifically, anti-inflammatory hemodialysis take place on the day of dialysis
drugs such as ibuprofen and aspirin should be has not been supported by evidence.104 The
avoided in patients with an eGFR less than 30 main factors to be considered when scheduling
mL/min/1.73 m2 as they may result in further dental appointments for patients on hemodi-
deterioration of kidney function, cause water alysis include the type of dental procedure,
retention, and increase the risk of gastric hem- the presence of any bleeding disorder, UFH or
orrhage. In addition, prolonged therapy with LMWH dosing, serum-activated partial throm-
anti-inflammatory medication is not recom- boplastin time (aPTT), or activated clotting
mended for patients with an eGFR less than 30 time (ACT).103 Further, uremia has been shown
mL/min/1.73 m2 . Alternatively, acetaminophen to affect platelet function, resulting in increased
could be used as an analgesic of choice in this bleeding tendency in ESRD.105 Depending on
category of patients. the type of dental procedure (surgical versus
Antibiotic therapy is another important nonsurgical), the above factors become critical.
consideration for patients with CKD/ESRD. The aPTT measures the time needed for plasma
For patients with an eGFR less than 15 mL/ to clot, assessing the intrinsic and common
min/1.73 m2 , there is a high risk for crystalluria pathways of coagulation and recommended
when penicillin is administered. When amino- for heparin therapy monitoring.102 The aPTT
glycosides, macrolides, and fluoroquinolones shows a 2.5- to 3.5-fold increase compared to
are used in patients with an eGFR less than 30 its upper limit in patients on UFH and inde-
mL/min/1.73 m2 , their dose should be reduced. pendently predicts postoperative bleeding
Antifungals is another class that should be complications following tooth extractions.106
avoided in patients with an eGFR less than 30 However, this test is performed in a hospital
mL/min/1.73 m2 .21 setting, limiting its utilization in outpatient
Once patients with CKD enter the ESRD stage units. Although there is no evidence-based
with an eGFR less than 15 mL/min/1.73 m2, recommendation on the dental management
they require renal replacement therapy, which of patients on hemodialysis, empirical evidence
includes hemodialysis, peritoneal dialysis, or has supported dental surgical visits the day of

187
8 The Association Between Oral Infections and Renal Disease

dialysis. The rationale for this practical sugges- associated with periodontitis. Although the
tion is based on the logistics of hemodialysis, exact mechanism of this association has not
its treatment duration, the anticoagulation been elucidated, there are several potential
half-life, and clearance depending on the anti- biologic hypotheses examining plausibility.
coagulant medication and dose. However, Evidence has demonstrated that periodontitis
nonsurgical dental procedures could be sched- contributes to the inflammatory burden in the
uled on the same day without any reported medically complex CKD/ESRD populations.
contraindications. In any case, the commu- Therefore, periodontitis could be considered as
nication between dental and renal providers the hidden comorbidity of CKD/ESRD.
becomes important in the decision-making Focusing on anti-inflammatory strategies
process. Because patients on peritoneal dialysis in CKD/ESRD populations, therapies aim to
receive their treatment through the abdomen control several comorbidities related to med-
via a catheter at home,102 there are no special ical and demographic characteristics as well
hemorrhagic considerations for patients on as dialysis-related factors. In this context and
peritoneal dialysis. to control local and systemic inflammation,
Currently, there is no evidence-based recom- periodontal therapeutic modalities have been
mendation to use prophylactic antibiotics in the tested with a goal to improve periodontal and
dental management of patients with ESRD. In systemic outcomes. Therefore, the prevention
addition, there is a lack of data on the dura- and treatment of periodontitis could modify
tion and effects of transient bacteremia in ESRD oral and systemic inflammatory biomarkers
populations following dental intervention. and improve the quality of life of patients with
Transient bacteremia has been shown to resolve CKD/ESRD. With this in mind, general practi-
in less than 20 minutes in systemically healthy tioners need to understand the needs of patients
individuals.107 Given the risks related to antibi- with renal diseases, emphasize the importance
otic overutilization associated with the selection of oral care, and feel confident with the dental
of resistant microorganisms and reduced antibi- management of patients with renal diseases.
otic efficacy, no prophylactic antibiotic has been Close collaboration with the renal team could
recommended for these populations. be instrumental in the selection of the appro-
priate pharmaceutical protocols according to
disease staging and residual renal function.
Additional considerations related to renal
Conclusion replacement therapy need to be discussed with
the renal team to serve a multidisciplinary care
CKD/ESRD is a global public health problem model for improved patient outcomes.
involving high health care costs and is highly

188
References

Clinical Considerations: What You Can Take Back to Your Practice


Is there a biologic plausibility for the associa- odontal therapy reduces the development of
tion between periodontal infections and renal adverse systemic outcomes in renal disease.
disease?  Although evidence is limited, there However, we know that periodontal therapy
are speculations that uremia, the main conse- could improve oral health, reduce tooth loss
quence of renal disease, could modify the oral rates, and improve oral health–related quality
environment, possibly promoting the growth of of life in patients with renal diseases.
periodontal pathogens, and also alter host in-
nate and adaptive immunity affecting infection Is it safe to provide dental care for patients with
susceptibility. In addition, renal osteodystrophy renal disease? The management of patients
manifested by abnormal bone breakdown and with renal diseases needs to follow a multidis-
turnover in patients with renal diseases may ciplinary approach, especially as these condi-
promote alveolar bone breakdown. tions become advanced. Communication with
the nephrologist and continuous updating of
Are periodontal infections independent risk fac- the medical history will be instrumental during
tors for the development of adverse systemic pharmaceutical and dental therapy.
outcomes in renal disease?  Limited evidence
has also shown that the presence of periodonti- What would a dental practitioner inform a patient
tis in patients with renal diseases may increase about the association between periodontal in-
all-cause and cardiovascular mortality rates. fections and the development of kidney disease? 
High-quality evidence has shown that peri-
Can periodontal therapy reduce the risk for the odontitis is associated with kidney disease in a
development of adverse systemic outcomes in dose-response manner. In other words, as renal
renal disease?  There is not enough high-quality disease advances, prevalence of periodontitis
evidence to support the hypothesis that peri- increases.

References
1. World Health Organization. World Report on Ageing and 7. Eke PI, Wei L, Borgnakke WS, et al. Periodontitis prev-
Health. Geneva: World Health Organization, 2015. alence in adults ≥ 65 years of age, in the USA.
2. Bloom DE. 7 billion and counting. Science 2011;333: Periodontol 2000 2016;72:76–95.
562–569. 8. Slots J. Update on general health risk of periodontal
3. Mather M, Jacobsen L, Pollard K. Aging in the United disease. Int Dent J 2003;53(suppl 3):200–207.
States. Washington, DC: Population Reference Bureau. 9. Winning L, Linden GJ. Periodontitis and systemic
2015:70. disease: Association or causality? Curr Oral Health Rep
4. Jin K, Simpkins JW, Ji X, Leis M, Stambler I. The critical 2017;4:1–7.
need to promote research of aging and aging-related 10. Borgnakke WS. Does treatment of periodontal disease
diseases to improve health and longevity of the elderly influence systemic disease? Dent Clin North Am 2015;
population. Aging Dis 2015;6: 1–5. 59:885–917.
5. Tonetti MS, Greenwell H, Kornman KS. Staging and 11. Linden GJ, Lyons A, Scannapieco FA. Periodontal
grading of periodontitis: Framework and proposal of a systemic associations: Review of the evidence. J Peri-
new classification and case definition. J Periodontol odontol 2013;84(suppl 4):S8–S19.
2018;89(suppl 1):S159–S172. 12. Kim J, Amar S. Periodontal disease and systemic condi-
6. Eke PI, Thornton-Evans GO, Wei L, Borgnakke WS, Dye tions: A bidirectional relationship. Odontology 2006; 94:
BA, Genco RJ. Periodontitis in US adults: National Health 10–21.
and Nutrition Examination Survey 2009–2014. J Am Dent
Assoc 2018;149:576–588.

189
8 The Association Between Oral Infections and Renal Disease

13. Oishi Y, Manabe I. Macrophages in age-related chronic 29. Ioannidou E, Swede H. Disparities in periodontitis prev-
inflammatory diseases. NPJ Aging Mech Dis 2016; 2: alence among chronic kidney disease patients. J Dent
16018. Res 2011;90:730–734.
14. Freire M, Van Dyke TE. The role of inflammation in 30. Eke PI, Dye BA, Wei L, Thornton-Evans GO, Genco RJ,
oral-systemic interactions. In: Glick M (ed). The CDC Periodontal Disease Surveillance workgroup. Prev-
Oral-Systemic Health Connection: A Guide to Patient alence of periodontitis in adults in the United States:
Care, ed 2. Chicago:Quintessence, 2019:102–134. 2009 and 2010. J Dent Res 2012;91:914–920.
15. Williams RC, Offenbacher S. Periodontal medicine: The 31. Eke PI, Thornton-Evans GO, Wei L, Borgnakke WS,
emergence of a new branch of periodontology. Peri- Dye BA. Accuracy of NHANES periodontal examination
odontol 2000 2000;23:9–12. protocols. J Dent Res 2010;89:1208–1213.
16. Jha V, Garcia-Garcia G, Iseki K, et al. Chronic kidney 32. Akar H, Akar GC, Carrero JJ, Stenvinkel P, Lindholm B.
disease: Global dimension and perspectives. Lancet Systemic consequences of poor oral health in chronic
2013;382:260–272. kidney disease patients. Clin J Am Soc Nephrol 2011;
17. Lozano R, Naghavi M, Foreman K, et al. Global and 6:218–226.
regional mortality from 235 causes of death for 20 age 33. Chen LP, Chiang CK, Chan CP, Hung KY, Huang CS.
groups in 1990 and 2010: A systematic analysis for the Does periodontitis reflect inflammation and malnutrition
Global Burden of Disease Study 2010. Lancet 2012; status in hemodialysis patients? Am J Kidney Dis 2006;
380:2095–2128. 47:815–822.
18. Everett B, Castel LD, McGinnis M, et al. Economic and 34. Craig RG, Kotanko P. Periodontitis and the end-stage
clinical outcomes resulting from the stage 4 chronic renal disease patient receiving hemodialysis mainte-
kidney disease case management quality improvement nance therapy. Compend Contin Educ Dent 2009; 30:
initiative. Prof Case Manag 2017;22:291–298. 544,546–552.
19. Levey AS, Eckardt KU, Tsukamoto Y, et al. Definition 35. Franek E, Blaschyk R, Kolonko A, et al. Chronic peri-
and classification of chronic kidney disease: A position odontitis in hemodialysis patients with chronic kidney
statement from Kidney Disease: Improving Global disease is associated with elevated serum C-reactive
Outcomes (KDIGO). Kidney Int 2005;67:2089–2100. protein concentration and greater intima-media thick-
20. Hill NR, Fatoba ST, Oke JL, et al. Global prevalence of ness of the carotid artery. J Nephrol 2006;19:346–351.
chronic kidney disease—A systematic review and 36. Naugle K, Darby ML, Bauman DB, Lineberger LT,
meta-analysis. PloS One 2016;11:e0158765. Powers R. The oral health status of individuals on renal
21. Inker LA, Astor BC, Fox CH, et al. KDOQI US commen- dialysis. Ann Periodontol 1998;3:197–205.
tary on the 2012 KDIGO clinical practice guideline for 37. US Department of Health and Human Services. Office
the evaluation and management of CKD. Am J Kidney of Disease Prevention and Health Promotion. Healthy
Dis 2014;63:713–735. People 2020. http://www.healthypeople.gov. Accessed
22. Levey AS, Coresh J, Balk E, et al. National Kidney Foun- 15 October 2018.
dation practice guidelines for chronic kidney disease: 38. Grubbs V, Plantinga LC, Tuot DS, Powe NR. Chronic
Evaluation, classification, and stratification. Ann Intern kidney disease and use of dental services in a United
Med 2003;139:137–147. States public healthcare system: A retrospective cohort
23. Saran R, Robinson B, Abbott KC, et al. US renal data study. BMC Nephrol 2012;13:16.
system 2017 annual data report: Epidemiology of kidney 39. Ioannidou E, Swede H, Fares G, Himmelfarb J. Tooth
disease in the United States. Am J Kidney Dis 2018;71: loss strongly associates with malnutrition in chronic
A7. kidney disease. J Periodontol 2014;85:899–907.
24. Coresh J, Selvin E, Stevens LA, et al. Prevalence of 40. Van Dyke TE, Diaz PI, Moutsopoulos N, Alekseyenko
chronic kidney disease in the United States. JAMA 2007; AV, Ioannidou E. Task force on design and analysis in
298:2038–2047. oral health research: Host-microbiome interactions in
25. Macisaac RJ, Ekinci EI, Jerums G. Markers of and risk dysbiosis. JDR Clin Trans Res 2018;3:6–9.
factors for the development and progression of diabetic 41. de Souza CM, Braosi AP, Luczyszyn SM, et al. Associ-
kidney disease. Am J Kidney Dis 2014; 63(suppl 2): ation between vitamin D receptor gene polymorphisms
S39–S62. and susceptibility to chronic kidney disease and peri-
26. Murphy D, McCulloch CE, Lin F, et al. Trends in preva- odontitis. Blood Purif 2007;25:411–419.
lence of chronic kidney disease in the United States. 42. Ioannidou E, Swede H, Dongari-Bagtzoglou A. Periodon-
Ann Intern Med 2016;165:473–481. titis predicts elevated C-reactive protein levels in chronic
27. Kshirsagar AV, Moss KL, Elter JR, Beck JD, Offenbacher kidney disease. J Dent Res 2011;90:1411–1415.
S, Falk RJ. Periodontal disease is associated with renal 43. Vanholder R, Van Laecke S, Glorieux G. The
insufficiency in the Atherosclerosis Risk In Communities middle-molecule hypothesis 30 years after: Lost and
(ARIC) study. Am J Kidney Dis 2005;45:650–657. rediscovered in the universe of uremic toxicity? J Nephrol
28. Fisher MA, Taylor GW, Shelton BJ, et al. Periodontal 2008;21:146–160.
disease and other nontraditional risk factors for CKD.
Am J Kidney Dis 2008;51:45–52.

190
References

44. Himmelfarb J, Stenvinkel P, Ikizler TA, Hakim RM. The 61. Podrez EA, Febbraio M, Sheibani N, et al. Macrophage
elephant in uremia: Oxidant stress as a unifying concept scavenger receptor CD36 is the major receptor for LDL
of cardiovascular disease in uremia. Kidney Int 2002; modified by monocyte-generated reactive nitrogen
62:1524–1538. species. J Clin Investing 2000;105:1095–1108.
45. Tomás I, Marinho JS, Limeres J, Santos MJ, Araújo L, 62. Arabi YM, Haddad S, Tamim HM, et al. Mortality reduc-
Diz P. Changes in salivary composition in patients with tion after implementing a clinical practice
renal failure. Arch Oral Biol 2008;53:528–532. guidelines-based management protocol for severe
46. Vaziri ND, Wong J, Pahl M, et al. Chronic kidney disease traumatic brain injury. J Crit Care 2010;25:190–195.
alters intestinal microbial flora. Kidney Int 2013; 83: 63. Carrero JJ, Stenvinkel P. Inflammation in end-stage renal
308–315. disease—What have we learned in 10 years? Semin
47. Ciancio SG, Golub LM, Mosovich L, Katz C, Kleinberg Dial 2010;23:498–509.
I. Urea levels in the gingival crevices of diabetic and 64. Stenvinkel P, Heimbürger O, Jogestrand T, Kärnell A,
normal adolescents. J Dent Res 1977;56:1144. Samuelsson A. Does persistent infection with Chlamydia
48. Golub LM, Borden SM, Kleinberg I. Urea content of pneumoniae increase the risk of atherosclerosis in
gingival crevicular fluid and its relation to periodontal chronic renal failure? Kidney Int 1999;55:2531–2532.
diseases in humans. J Periodontal Res 1971; 6: 65. Chia S, Karim M, Elwood RK, FitzGerald JM. Risk of
243–251. tuberculosis in dialysis patients: A population-based
49. Sachs G, Meyer-Rosberg K, Scott DR, Melchers K. Acid, study. Int J Tuberc Lung Dis 1998;2:989–991.
protons and Helicobacter pylori. Yale J Biol Med 1996; 66. Simon TA, Paul S, Wartenberg D, Tokars JI. Tuberculosis
69:301–316. in hemodialysis patients in New Jersey: A statewide
50. Scott DR, Weeks D, Hong C, Postius S, Melchers K, study. Infect Control Hosp Epidemiol 1999; 20:607–609.
Sachs G. The role of internal urease in acid resistance 67. Feinfeld DA, Sherwood LM. Parathyroid hormone and
of Helicobacter pylori. Gastroenterology 1998; 114: 1,25(OH)2D3 in chronic renal failure. Kidney Int 1988;33:
58–70. 1049–1058.
51. Marsh PD. Are dental diseases examples of ecological 68. Moe S, Drüeke T, Cunningham J, et al. Definition, eval-
catastrophes? Microbiology 2003;149:279–294. uation, and classification of renal osteodystrophy: A
52. Bastos JA, Diniz CG, Bastos MG, et al. Identification of position statement from Kidney Disease: Improving
periodontal pathogens and severity of periodontitis in Global Outcomes (KDIGO). Kidney Int 2006; 69:
patients with and without chronic kidney disease. Arch 1945–1953.
Oral Biol 2011;56:804–811. 69. Hewison M. An update on vitamin D and human immu-
53. Castillo A, Mesa F, Liébana J, et al. Periodontal and oral nity. Clin Endocrinol (Oxf) 2012;76:315–325.
microbiological status of an adult population undergoing 70. Stenvinkel P, Carrero JJ, Axelsson J, Lindholm B, Heim-
haemodialysis: A cross-sectional study. Oral Dis 2007; bürger O, Massy Z. Emerging biomarkers for evaluating
13:198–205. cardiovascular risk in the chronic kidney disease patient:
54. Araújo MV, Hong BY, Fava PL, et al. End stage renal How do new pieces fit into the uremic puzzle? Clin J
disease as a modifier of the periodontal microbiome. Am Soc Nephrol 2008;3:505–521.
BMC Nephrol 2015;16:80. 71. Himmelfarb J. Oxidative stress in hemodialysis. Contrib
55. Kato S, Chmielewski M, Honda H, et al. Aspects of Nephrol 2008;161:132–137.
immune dysfunction in end-stage renal disease. Clin J 72. Friedman AN, Moe SM, Perkins SM, Li Y, Watkins BA.
Am Soc Nephrol 2008;3:1526–1533. Fish consumption and omega-3 fatty acid status and
56. Anding K, Gross P, Rost JM, Allgaier D, Jacobs E. The determinants in long-term hemodialysis. Am J Kidney
influence of uraemia and haemodialysis on neutrophil Dis 2006;47:1064–1071.
phagocytosis and antimicrobial killing. Nephrol Dial 73. Stenvinkel P. Inflammation in end-stage renal disease:
Transplant 2003;18:2067–2073. The hidden enemy. Nephrology (Carlton) 2006;11:36–41.
57. Carracedo J, Merino A, Nogueras S, et al. On-line hemo- 74. Carrero JJ, Stenvinkel P. Persistent inflammation as a
diafiltration reduces the proinflammatory CD14+CD16+ catalyst for other risk factors in chronic kidney disease:
monocyte-derived dendritic cells: A prospective, cross- A hypothesis proposal. Clin J Am Soc Nephrol 2009;
over study. J Am Soc Nephrol 2006;17:2315–2321. 4(suppl 1):S49–S55.
58. Lim WH, Kireta S, Russ GR, Coates PT. Uremia impairs 75. Lockhart PB, Bolger AF, Papapanou PN, et al. Peri-
blood dendritic cell function in hemodialysis patients. odontal disease and atherosclerotic vascular disease:
Kidney Int 2007;71:1122–1131. Does the evidence support an independent associa-
59. Witko-Sarsat V, Gausson V, Nguyen AT, et al. AOPP– tion?: A scientific statement from the American Heart
induced activation of human neutrophil and monocyte Association. Circulation 2012;125:2520–2544.
oxidative metabolism: A potential target for 76. Ebersole JL, Cappelli D. Acute-phase reactants in infec-
N-acetylcysteine treatment in dialysis patients. Kidney tions and inflammatory diseases. Periodontology 2000
Int 2003;64:82–91. 2000;23:19–49.
60. Ando M, Shibuya A, Yasuda M, et al. Impairment of 77. Loos BG. Systemic markers of inflammation in periodon-
innate cellular response to in vitro stimuli in patients on titis. J Periodontol 2005;76(suppl 11):2106–2115.
continuous ambulatory peritoneal dialysis. Nephrol Dial
Transplant 2005;20:2497–2503.

191
8 The Association Between Oral Infections and Renal Disease

78. Noack B, Genco RJ, Trevisan M, Grossi S, Zambon JJ, 92. Armitage GC. Effect of periodontal therapy on general
De Nardin E. Periodontal infections contribute to health—Is there a missing component in the design of
elevated systemic C-reactive protein level. J Periodontol these clinical trials? J Clin Periodontol 2008; 35:
2001;72:1221–1227. 1011–1012.
79. Fisher MA, Taylor GW, Papapanou PN, Rahman M, 93. Tonetti MS, D’Aiuto F, Nibali L, et al. Treatment of peri-
Debanne SM. Clinical and serologic markers of peri- odontitis and endothelial function. N Engl J Med 2007;
odontal infection and chronic kidney disease. J 356:911–920.
Periodontol 2008;79:1670–1678. 94. Engebretson SP, Hyman LG, Michalowicz BS. Hemo-
80. Chen LP, Chiang CK, Peng YS, et al. Relationship globin A1c levels among patients with diabetes receiving
between periodontal disease and mortality in patients nonsurgical periodontal treatment—Reply. JAMA 2014;
treated with maintenance hemodialysis. Am J Kidney 311:1921–1922.
Dis 2011;57:276–282. 95. Offenbacher S, Beck JD, Moss K, et al. Results from
81. Hill AB. The Environment and disease: Association or the Periodontitis and Vascular Events (PAVE) Study: A
causation? Proc R Soc Med 1965;58:295–300. pilot multicentered, randomized, controlled trial to study
82. Stenvinkel P, Heimbürger O, Jogestrand T. Elevated effects of periodontal therapy in a secondary prevention
interleukin-6 predicts progressive carotid artery athero- model of cardiovascular disease. J Periodontol 2009;
sclerosis in dialysis patients: Association with Chlamydia 80:190–201.
pneumoniae seropositivity. Am J Kidney Dis 2002; 39: 96. Borgnakke WS, Chapple IL, Genco RJ, et al. The
274–282. multi-center randomized controlled trial (RCT) published
83. Banerjee T, Kim SJ, Astor B, Shafi T, Coresh J, Powe by the Journal of the American Medical Association
NR. Vascular access type, inflammatory markers, and (JAMA) on the effect of periodontal therapy on glycated
mortality in incident hemodialysis patients: The Choices hemoglobin (HbA1c) has fundamental problems. J Evid
for Healthy Outcomes in Caring for End-Stage Renal Based Dent Pract 2014;14:127–132.
Disease (CHOICE) study. Am J Kidney Dis 2014;64: 97. Axelsson P, Lindhe J. Effect of controlled oral hygiene
954–961. procedures on caries and periodontal disease in adults.
84. Cugini MA, Haffajee AD, Smith C, Kent RL Jr, Socransky Results after 6 years. J Clin Periodontol 1981; 8:
SS. The effect of scaling and root planing on the clinical 239–248.
and microbiological parameters of periodontal diseases: 98. Lindhe J, Westfelt E, Nyman S, Socransky SS, Haffajee
12-month results. J Clin Periodontol 2000;27:30–36. AD. Long-term effect of surgical/non-surgical treatment
85. Needleman I, McGrath C, Floyd P, Biddle A. Impact of of periodontal disease. J Clin Periodontol 1984; 11:
oral health on the life quality of periodontal patients. J 448–458.
Clin Periodontol 2004;31:454–457. 99. Wilson TG Jr, Glover ME, Malik AK, Schoen JA, Dorsett
86. Teles RP, Haffajee AD, Socransky SS. Microbiological D. Tooth loss in maintenance patients in a private peri-
goals of periodontal therapy. Periodontol 2000 2006; odontal practice.J Periodontol 1987;58:231–235.
42:180–218. 100. Seliger SL. Comorbidity and confounding in end-stage
87. Magnusson I, Lindhe J, Yoneyama T, Liljenberg B. renal disease. Kidney Int 2010;77:83–85.
Recoloni­zation of a subgingival microbiota following 101. Snaedal S, Heimbürger O, Qureshi AR, et al. Comor-
scaling in deep pockets. J Clin Periodontol 1984; 11: bidity and acute clinical events as determinants of
193–207. C-reactive protein variation in hemodialysis patients:
88. Research, Science and Therapy Committee of the Amer- Implications for patient survival. Am J Kidney Dis 2009;
ican Academy of Periodontology. Treatment of 53:1024–1033.
plaque-induced gingivitis, chronic periodontitis, and 102. Shen JI, Winkelmayer WC. Use and safety of unfrac-
other clinical conditions. J Periodontol 2001; 72: tionated heparin for anticoagulation during maintenance
1790–1800. hemodialysis. Am J Kidney Dis 2012;60:473–486.
89. Matuliene G, Studer R, Lang NP, et al. Significance of 103. Cook BW. Anticoagulation management. Semin Inter-
periodontal risk assessment in the recurrence of peri- vent Radiol 2010;27:360–367.
odontitis and tooth loss. J Clin Periodontol 2010; 37: 104. Mingarro-de-Leon A, Chaveli-Lopez B, Gavalda-Esteve
191–199. C. Dental management of patients receiving anticoag-
90. Artese HP, Sousa CO, Luiz RR, Sansone C, Torres MC. ulant and/or antiplatelet treatment. J Clin Exp Dent 2014;
Effect of non-surgical periodontal treatment on chronic 6:e155–161.
kidney disease patients. Braz Oral Res 2010; 24: 105. Molino D, De Lucia D, Gaspare De Santo N. Coagulation
449–454. disorders in uremia. Semin Nephrol 2006;26:46–51.
91. Wehmeyer MM, Kshirsagar AV, Barros SP, et al. A 106. Karsli ED, Erdogan Ö, Esen E, Acartürk E. Comparison
randomized controlled trial of intensive periodontal ther- of the effects of warfarin and heparin on bleeding caused
apy on metabolic and inflammatory markers in patients by dental extraction: A clinical study. J Oral Maxillofac
with ESRD: Results of an exploratory study. Am J Kidney Surg 2011;69:2500–2507.
Dis 2013;61:450–458. 107. Lockhart PB, Brennan MT, Sasser HC, Fox PC, Paster
BJ, Bahrani-Mougeot FK. Bacteremia associated with
toothbrushing and dental extraction. Circulation 2008;
117:3118–3125.

192
CHAPTER 9

The Association Between


Oral Infections and
Pulmonary Disease
Karren Komitas, dmd, phd, mdsc
Effie Ioannidou, dds, mds

Periodontitis is one of the most common been exposed to hazardous environmental


human inflammatory diseases, with an over- factors for longer than the general popula-
all prevalence estimated at 42% in the United tion. Therefore, a better understanding of the
States.1 Annual direct and indirect expenses mechanisms linking periodontitis and systemic
associated with periodontitis and periodontal diseases is essential for the development of
treatment worldwide are estimated at several innovative treatment approaches to minimize
hundred billion USD.2 Therefore, periodontitis the prevalence of these diseases and provide
represents a substantial health care and eco- better treatment outcomes.
nomic burden, and considerable efforts have This understanding is essential for dental
been made during the past several decades to practitioners, physicians, and other health
gain insight into the factors associated with its care personnel who depend on effective
etiology and progression. evidence-based treatment protocols. As mul-
Current scientific evidence suggests that tiple studies have explored whether or not
periodontal pathogens contained in the dental periodontitis is an independent risk factor for
plaque of a susceptible individual lead to the systemic diseases (and vice versa), the goal
development of periodontitis.3 The National of this chapter is to provide an overview of
Health and Nutrition Examination Survey pulmonary diseases (including pneumonia
(NHANES) studies have also shown that envi- and chronic obstructive pulmonary disease
ronmental factors, such as smoking, should be [COPD]) and demonstrate the current evidence
considered as risk factors for the development of the association between pulmonary disease
and progression of periodontitis.4,5 In addi- and oral infections in patients with periodonti-
tion, various contributory factors, including tis. This chapter focuses on pulmonary diseases,
but not limited to age, neglected oral hygiene, as they represent one of the most clinically
and poorly controlled systemic diseases (such significant respiratory health care entities. In
as diabetes mellitus), represent significant pre- addition, some similarities in the mechanisms
dictive covariates associated with periodontal of development and/or progression of the pul-
breakdown.3,6,7 These local, environmental, and monary diseases and periodontitis suggest that
systemic factors are especially relevant in older elimination of the etiologic factors can provide
adults, as they have increased prevalence of a substantial improvement in treatment out-
periodontitis1 and systemic diseases8 and have comes for both of these disease entities.

193
9 The Association Between Oral Infections and Pulmonary Disease

Definition, Classification, and (HAP, or nosocomial pneumonia) is health


care–associated pneumonia (HCAP). HCAP
Manifestation of Pulmonary is defined as pneumonia that occurs in any
Diseases patient who has been (1) hospitalized in an
acute care hospital for two or more days within
90 days of infection; (2) resided in a nursing
Pneumonia
home or long-term care facility; (3) received
Pneumonia is an acute infection of the lung recent intravenous antibiotic therapy, chemo-
parenchyma and is the most common lower therapy, or wound care within the past 30
respiratory tract infection.9 Clinically, it is char- days of the current infection; or (4) attended a
acterized by acute symptoms and signs of lower hospital or hemodialysis clinic.14 HAP is cate-
respiratory tract infection without other appar- gorized into ventilator-associated pneumonia
ent causes, dyspnea, cough, fever, and in some and aspiration pneumonia.
cases a headache, confusion, upper respiratory
involvement, and skin changes.10 Macro- Ventilator-associated pneumonia. Ventilator-
scopically, inflammation is characterized by associated pneumonia (VAP) is defined as
nonspecific inflammatory events, including red- pneumonia that occurs at least 48 hours after
ness, swelling, heat, pain, and loss of function. endotracheal intubation and initiation of
Microscopically, typical signs of inflammation, mechanical ventilation.15 According to a sys-
such as vasodilation, increased vascular perme­ tematic review that included nine publications,
ability, and resulting infiltration of inflammatory VAP is the most common subcategory of HAP,
cells can be observed.11,12 Chronic inflammation occurs in 10% to 20% of patients who undergo
as a result of an incomplete resolution of acute mechanical ventilation, and is associated with
inflammation is characterized predominantly by a longer hospital stay and increased financial
recruitment of macrophages and lymphocytes costs.15 The same study also showed that VAP
engaged in the elimination of necrotic debris is associated with one of the highest mortality
and apoptotic cells to prevent further spread of rates, ranging between 20% and 55%.16
persistent infection.12
Based on a patient’s age, pneumonia can be Aspiration pneumonia. Aspiration pneumonia
classified into childhood and adult categories. (AP), a commonly occurring type of HAP, is
The most recent classification of childhood an acute lung infection due to inhalation of
pneumonia from 2014 includes “pneumonia” large-volume bacteria-colonized oropharyn-
and “severe pneumonia.”13 This chapter focuses geal material that leads to the development of
on periodontitis, which in an overwhelming a radiographic infiltrate,17,18 although various
majority of cases occurs in adults, so our alternative definitions have been proposed.19
review is aimed at discussion of adult pneumo- AP constitutes approximately 5% to 15% of
nia. Although various types of classifications all pneumonia cases and affects 300,000 to
for adult pneumonia have been proposed,9 the 500,000 individuals in the United States annu-
American Thoracic Society and the Infectious ally.17,18 Up to 50% of healthy individuals can
Diseases Society of America propose the fol- aspirate small volumes of oropharyngeal secre-
lowing classification according to the site of tions during sleep, but due to the low virulence
acquisition. of these secretions and adequate cellular and
humoral host immune mechanisms, these aspi-
rations do not lead to an acute inflammatory
Hospital-acquired pneumonia response (“silent aspirations”).18,20 A study that
Health care–associated pneumonia. The first included 259 Japanese older adults receiving
category of hospital-acquired pneumonia nursing care (aged 65 to 74 years, 75 to 89

194
Definition, Classification, and Manifestation of Pulmonary Diseases

years, and 90 years and older) showed that Fusobacterium nucleatum, Prevotella mela-
the prevalence of AP is similar among younger ninogenica, Streptococcus intermedius, and
and older individuals.21 Commonly reported Clostridium species, have been identified in
risk factors for AP include male sex, intubation, bronchoalveolar lavage samples of patients
dehydration, sputum suctioning, gastroesopha- with CAP.31 In addition to bacterial pathogens,
geal reflux, and dysphagia as a consequence of viruses, including rhinovirus, influenza A or B,
various cerebrovascular and neurodegenerative and parainfluenza viruses, can also be isolated
conditions (such as Alzheimer’s disease, Par- from nasopharyngeal, oropharyngeal, or spu-
kinson’s disease, dementia, multiple sclerosis, tum samples of these patients.27,29
and stroke).21–24

Chronic obstructive pulmonary


Community-acquired pneumonia
disease
Community-acquired pneumonia (CAP) is the
second most common type of pneumonia in COPD is a chronic pulmonary disease charac-
the United States and worldwide, which can terized by persistent respiratory symptoms and
occur in any individual living in a commu- airflow limitation due to airway and/or alveolar
nity.25 Epidemiologic studies have shown that abnormalities.32 In 2010, the global prevalence
CAP remains the most common cause of death of COPD was estimated at 11.7%,33 making it
from infectious diseases among the elderly the fourth leading cause of death worldwide34;
population.26 The annual incidence of CAP is COPD is expected to become the third leading
estimated to be 25 to 44 cases per 1,000 indi- cause of death worldwide by 2030, account-
viduals,27 and it can occur at any age. A study ing for 8% of deaths.35 In the United States,
that included 46,000 older adults showed that 700,000 hospital admissions and 124,000
individuals aged 85 years and older have almost deaths occur due to COPD annually.36 Clin-
a three-fold greater incidence of CAP compared ically, COPD is manifested as dyspnea, chest
to those aged 65 to 69 years (59.3 versus 18.2 inflation, chronic cough, sputum production,
cases per 1,000 person-years, respectively).28 A wheezing, and chest tightness. Macroscopically,
recent prospective, population-based study of COPD is characterized by the destruction of
hospitalized adults with CAP showed that the lung parenchymal tissues, the breakdown of
annual incidence of CAP is 24.8 per 10,000 lung framework, increased size and numbers
adults and that adults aged 50 to 64 years, of pores of Kohn (small fenestrae in alveolar
65 to 79 years, and 80 years and older have walls), and thickening of the airway epithe-
4.0, 9.0, and 25.0 times greater incidence of lial layer due to chronic inflammation.37–39
CAP as compared to 18- to 49-year-old adults, Microscopically, it is presented as epithelial
respectively.29 In addition to age, other indepen- cell metaplasia, immune cell infiltration, and
dent CAP risk factors include male sex, COPD, peribronchial fibrosis.37,39 Smoking is consid-
bronchial asthma, history of pneumonia, con- ered the most important risk factor, whereas
gestive heart failure, and smoking.28,30 chronic exposure to environmental factors
The microbiota of older adults with CAP (such as air pollution, biomass fuel exposure,
and HCAP predominantly consists of Strep- occupational dust, and home cooking smoke)
tococcus pneumoniae (approximately 30% to and genetic factors (such as congenital abnor-
58%), Haemophilus influenzae (approximately mal lung development, accelerated aging, and
14%), Staphylococcus aureus (approximately family history of COPD) are considered con-
4% to 7%), and Pseudomonas aeruginosa tributory factors.32
(approximately 1% to 5%).27 A variety of Prior to 2017, COPD was diagnosed only
oral and periodontal pathogens, including spirometrically when a postbronchodilator

195
9 The Association Between Oral Infections and Pulmonary Disease

Tiffeneau-Pinelli index (FEV1/FVC, or a pro- Epidemiologic Association


portion of a person’s vital capacity that he or
she is able to expire in the first second of forced Between Periodontitis and
expiration to the full vital capacity) was less Pulmonary Diseases
than 0.70.32 In 2017, the Global Initiative for
Chronic Obstructive Lung Disease (GOLD) Similar to periodontitis, pulmonary diseases are
updated its diagnostic criteria and classification inflammatory diseases of polymicrobial nature,
for COPD, which also took into consideration sharing similar cellular and molecular mech-
a patient’s respiratory symptom and exacer- anisms of development and progression.43 In
bation history (which are the main targets for addition, these diseases share similar contrib-
treatment). Once the initial diagnosis of COPD utory and risk factors, including smoking and
is established spirometrically (FEV 1/FVC diseases with impaired neutrophil function,44
< 0.70), four distinct groups of COPD can thus raising a biologic plausibility for the asso-
be identified based on the severity of clinical ciation between these conditions.
symptoms using exacerbation history, mMRC During the past two decades, accumulat-
(modified British Medical Research Council ing evidence in the literature has suggested
questionnaire), and CAT (COPD Assessment increased prevalence of pulmonary diseases
Test) criteria40: (pneumonia and COPD) in patients with peri-
odontitis. Early studies have demonstrated
• Group A: Exacerbation history 0–1; mMRC that patients with AP have a higher plaque
0–1; CAT < 10 index and periodontal score.45 Further, a
• Group B: Exacerbation history 0–1; mMRC case-control study that included 22 patients
≥ 2; CAT ≥ 10 with HAP and 81 non-HAP controls (all 103
• Group C: Exacerbation history > 1; mMRC individuals had periodontitis) showed that
0–1; CAT < 10 the prevalence of periodontitis (defined as
• Group D: Exacerbation history > 1; mMRC ≤ four teeth with ≤ one site showing probing
≥ 2; CAT ≥ 10 pocket depth [PPD] ≥ 4 mm, clinical attach-
ment loss [CAL] ≥ 3 mm, and bleeding on
Based on these criteria, patients from groups probing) was significantly greater in patients
A and B do not require hospital admission; with HAP (adjusted odds ratio [OR]: 3.67;
patients from groups C and D do. Treatment of 95% confidence interval [CI]: 1.01–13.53;
COPD includes but is not limited to smoking P < .05).46 A recent 7-year prospective cohort
cessation (for current smokers), bronchodila- study that included 211 Japanese patients
tors (such as β2 agonists, antimuscarinic drugs, undergoing hemodialysis (mean patient age
and methylxanthines), and anti-inflammatory 64.4 years) demonstrated that HCAP mortal-
drugs (such as inhaled corticosteroids, gluco- ity (although the authors did not specify the
corticoids, and antibiotics).40 exact type of HCAP) is significantly associated
In addition to the GOLD classification, with the presence of periodontitis (defined as
COPD can also be classified as chronic bron- CAL ≥ 4 mm in ≥ 30% of the probed sites)
chitis, emphysema, and small-airway disease.41 (confounder-adjusted hazard ratio [HR]: 3.03;
Clinically, chronic bronchitis is characterized 95% CI: 1.14–10.64; P < .05).47 A 4-year
by cough and sputum production; emphy- study of 672 older Japanese adults (mean age
sema is characterized by enlarged distal air 80 years) has shown that the presence of 10 or
spaces distal to terminal bronchioles due to more teeth with PPD greater than 4 mm is asso-
the destruction of the airway walls, whereas ciated with significantly greater mortality from
small-airway disease is caused by obstruction AP compared to patients with AP and health-
of smaller conducting airways.41,42 ier periodontium (confounder-adjusted HR:

196
Mechanisms for Increased Association of Periodontitis and Pulmonary Diseases

3.9; 95% CI: 1.1–13.9; P < .05).24 However, Mechanisms for Increased
these conclusions should be taken with caution
considering an extremely wide CI with a low Association of Periodontitis
limit of only 1.1. Similar to studies on pneu- and Pulmonary Diseases
monia, a systematic review with meta-analysis
of 14 publications that included 3,988 patients Although the exact pathophysiologic
with COPD and 22,871 non-COPD controls mechanisms that predispose patients with peri-
showed that patients with periodontitis have odontitis to pulmonary diseases (pneumonia
significantly greater prevalence of COPD (OR: and COPD) have yet to be identified, several
2.08; 95% CI: 1.48–2.9; P < .001).48 possible mechanisms have been proposed in
Similarly, a number of studies, including the literature.55–58
large-scale studies, have assessed the prevalence
of periodontitis in patients with pulmonary dis-
eases (primarily COPD). Based on NHANES Periodontitis and pneumonia
III data that included 7,625 participants, no
Extravascular mode
significant association has been found between
COPD and periodontitis (defined at CAL ≥ 4 Direct aspiration of periodontal and/or respi-
mm), unless current smoking is included as ratory pathogens into the lower respiratory
a cofactor (OR: 3.71; 95% CI: 1.74–7.89; tract, or transmission of oral pathogens during
P < .01).49 A Korean NHANES study from mechanical ventilation and intubation. The
2010 to 2012 of 697 patients with COPD and human oral cavity is colonized with over 700
5,181 non-COPD controls demonstrated that bacterial species localized in the mucosal sur-
patients with COPD have significantly greater faces, saliva, and teeth. Saliva contains 108 to
prevalence of periodontitis (defined as PPD 109 bacteria per mL, which have a high ten-
≥ 3.5 mm) (58.1% versus 34.0%, respectively; dency to attach to the teeth and form dental
P < .001) and significantly fewer teeth (21 plaque, a highly complex and organized bio-
versus 24.5, respectively; P < .001) compared film containing over 100 different bacterial
to patients without COPD after adjust- species and therefore serving as a reservoir for
ment for other confounders.50 A nationwide opportunistic pathogens.59 The total counts of
population-based cohort study that included pathogens in the dental plaque can reach 1011
22,232 Taiwanese patients with COPD and to 1012 per gram.60
43,762 non-COPD controls reported that Because saliva is a major component of
patients with COPD have a minor but statis- AP microaspirations, salivary bacteria have
tically significant increase in the prevalence of been suggested to predispose older patients or
periodontitis (OR: 1.20; 95% CI: 1.15–1.25; those with immunocompromised conditions to
P < .001) that is not associated with sex, age, develop pneumonia. Colonization of the respi-
or comorbidities.51 Similar findings have been ratory tract with oropharyngeal bacteria is an
reported by various smaller-scale studies.52–54 undesired but often inevitable event during AP
Taken together, these studies show that or VAP61 and therefore can serve as a possible
patients with periodontitis have statistically way of dissemination of periodontal pathogens
significant greater prevalence of pulmonary dis- into the lungs. Aspirated pathogens in contact
eases, and the outcomes of pulmonary diseases with the respiratory epithelium can produce a
in patients with periodontitis are more adverse variety of inflammatory mediators, including
compared to those with healthier periodontium. reactive oxygen species and cytokines that will
In addition, patients with pulmonary diseases recruit immune cells to the site of contact and
(primarily COPD) also have statistically signif- stimulate mucus secretion by lung goblet cells,
icant greater prevalence of periodontitis. resulting in tissue inflammation.12 Early studies

197
9 The Association Between Oral Infections and Pulmonary Disease

examining sputum and transtracheal samples during pneumonia (mostly VAP and AP) can
from older adult veterans have demonstrated be triggered not only by periodontal pathogens
that in addition to respiratory pathogens (such but also by respiratory pathogens contained
as S pneumoniae and S aureus), these samples in dental plaque aspirated with saliva. Early
contain periodontitis-associated pathogens, prospective nonrandomized study of 34 older
including Prevotella intermedia and F nuclea- adults admitted to the intensive care unit (ICU)
tum.45 This suggests that periodontal pathogens demonstrated that more of these patients har-
can colonize the lungs when aspirated, thus bored S aureus and P aeruginosa in the dental
possibly resulting in inflammation and AP.17 plaque compared to a group of 25 systemi-
Using 16S and quantitative polymerase cally healthy dental patients.65 A 3-month
chain reaction (qPCR) approaches, a recent study that included 57 ICU-admitted patients
study that included 18 patients with periodon- showed that they tend to accumulate more
titis demonstrated that similar to subgingival dental plaque upon admission, and it is sig-
plaque, the saliva of these patients contained nificantly associated with the occurrence of
readily detectable counts of periodontal patho- HCAP and bacteremia.66 Using a pulsed-field
gens, including Porphyromonas gingivalis, gel electrophoresis approach, it has further
Treponema denticola, Tannerella forsythia, been demonstrated that respiratory pathogens
P intermedia, and Parvimonas micra.62 The (S aureus, P aeruginosa, and Acinetobacter
same study also reported that saliva lacks species) isolated from the supragingival
periodontal pathogens if they are not detected plaque samples are genetically indistinguish-
in the subgingival samples, suggesting that able from the respiratory pathogens isolated
pathogens contained in subgingival plaque of from lungs of ICU-admitted patients with
patients with periodontitis can shed into the VAP.67 Using a checkerboard DNA–DNA
saliva. Similarly, another study using the qPCR hybridization approach, it has been demon-
approach that included 462 patients with mod- strated that dental plaque of patients with
erate and severe periodontitis demonstrated a periodontitis contains readily detectable counts
significantly greater prevalence of salivary peri- of pneumonia-associated pathogens, such as S
odontal pathogens (P gingivalis, T forsythia, aureus.68 This may further suggest a common
and P intermedia) compared to the saliva of environmental source of infection and that the
individuals with healthy periodontium.63 dental plaque can serve as a reservoir for respi-
A recent prospective human study using the ratory pathogens.69
qPCR approach with 42 patients with pneumo- Advances in microbiologic techniques during
nia (predominantly HAP) demonstrated that the last few decades have allowed detection and
periodontal pathogens, such as P gingivalis, P quantification of microbiota with much higher
intermedia, and F nucleatum can be detected precision. Using 16S rRNA gene sequencing in
in respiratory tract specimens in 31% to 57% a group of 12 patients with VAP, it has been
of patients, suggesting a possibility that peri- shown that the 20 most abundantly detected
odontal pathogens can proliferate in the lower dental plaque pathogens are also potential
respiratory tract.64 The attempt to further respiratory pathogens, including S pneumo-
explore this possibility has been performed in a niae (the most abundant species), Enterococcus
systematic review of one case-control and four hirae, F nucleatum, S aureus, P aeruginosa, and
prospective cohort studies.57 However, the lack H influenzae.70 Similarly, a study that included
of properly designed randomized controlled 107 patients with VAP showed that S aureus
trials (RCTs) included in this systematic review and P aeruginosa have been readily detected
makes its conclusions debatable. in supragingival plaque samples.71 Interest-
Another series of studies aimed to exam- ingly, S aureus and P aeruginosa have been
ine the possibility that inflammatory changes primarily detected during hospitalization, as

198
Mechanisms for Increased Association of Periodontitis and Pulmonary Diseases

a large percentage of patients do not harbor S denture-adhered plaque, significantly lower


aureus and P aeruginosa prior to intubation, use of denture cleaners, and significantly fewer
and many of them do not harbor S aureus and dental visits compared to patients who did not
P aeruginosa after extubation.71 This suggests wear dentures during sleep (P < .05 for all
that the presence of these pathogens can be comparisons).75 These results further suggest
due to their dissemination during intubation that poor oral hygiene associated with wear-
because they are typically not associated with ing dentures while sleeping can lead to the
healthy oral microflora and represent patho- increased prevalence of AP.
gens commonly found in patients with chronic In addition to human clinical studies, several
periodontal and pulmonary diseases. A recent animal studies have shown that intratracheal
human RCT study of 16 ICU-admitted patients inoculation of periodontal pathogens (such
with VAP used DNA sequencing to evaluate as P gingivalis and T denticola) results in the
the changes in the composition of oral and development of high-mortality pneumonia,
dental plaque microbiota upon admission increased levels of proinflammatory mediators,
and demonstrated a rapid colonization of oral and activation of toll-like receptor 2 (TLR-2)
mucosa with methicillin-resistant S aureus and signaling.76–79 These results further support the
Klebsiella pneumoniae.72 biologic plausibility of AP and VAP induced by
Because a large proportion of patients with periodontal pathogens.
pneumonia are older adults, many of them Taken together, these data indicate that
wear partial dentures, thus raising concern periodontal pathogens contained in dental
about denture plaque accumulation. Using plaque and saliva can disseminate into the
a culture technique, a cross-sectional study lungs with microaspirations. In addition, older
that included 138 older Japanese adults with adult patients (especially those with dentures)
poor oral hygiene showed that dentures are tend to accumulate greater amounts of dental
populated with respiratory pathogens, predom- plaque that contains not only periodontal but
inantly Streptococcus (approximately 85% of also respiratory pathogens. Thus, these mech-
samples) and Neisseria (approximately 50%) anisms contribute to the development of AP in
species.73 A recent study using qPCR analysis susceptible individuals.
that included 130 older nonpneumonia Scot-
tish adults (mean age 70.4 years and average Damage or modification of the epithelial lin-
denture age of 4.5 years) demonstrated that ing of the lungs. It has been proposed that
in 65% of patients, partial dentures harbored periodontal pathogens could facilitate colo-
potential respiratory pathogens, including P nization of respiratory pathogens and induce
aeruginosa (most abundant), S aureus, S pneu- respiratory inflammation via several possible
moniae, H influenzae, and other species.74 mechanisms.58 For example, periodontal patho-
Interestingly, a 3-year study that included gens could modify the lining of the respiratory
524 older Japanese adults (mean age 87.8 epithelium by altering the transmembrane
years) showed that patients who wore partial receptors on the surface of target mucosal cells
dentures during night sleep have a 1.5-fold to promote adhesion of pulmonary pathogens
increase in the incidence of AP compared to promoting inflammation. Using a human-
those who did not have this habit (HR: 2.38; ized bronchial xenograft model in nude mice
95% CI: 1.25–4.56; P < .05).75 In addition, and human airway epithelial cell cultures, a
the patients who wore partial dentures during study showed that an S aureus strain deficient
night sleep had poorer oral hygiene compared of fibronectin-binding protein is decreased
to those who did not wear dentures during five-fold compared to a nonmutant S aureus
night sleep. This was evidenced by a signifi- strain.80 Because saliva contains enzymes capa-
cantly greater percentage of patients with ble of fibronectin degradation81 and fibronectin

199
9 The Association Between Oral Infections and Pulmonary Disease

is a component of the extracellular matrix,82 bacteremia may be persistent and therefore


it is suggested that during aspiration of saliva, can lead to inflammatory events in the lungs,
these enzymes are capable of damaging current evidence supporting this assumption
fibronectin and alleviating penetration of respi- is unclear.
ratory pathogens into the underlying tissues.
Current evidence for this mechanism linking Release of periodontal pathogen toxins and
periodontitis and pneumonia is uncertain. proinflammatory cytokines in the systemic
blood circulation. Although dental plaque is
considered an etiologic factor for periodonti-
Intravascular mode tis, pathogenesis and concomitant destruction
Hematogenous bacterial dissemination of of periodontal tissues are driven by the com-
periodontal pathogens during periodontal plex interaction between various types of
treatment. As discussed previously, periodontal immune cells and mediators that regulate this
pathogens can trigger respiratory inflamma- delicate process.90 Among these regulators are
tion.76,77,79 Because gingival bleeding is one of cytokines, which are secreted polypeptides
the earliest signs of gingivitis and periodonti- produced by various types of cells and exert
tis, and because bacteremia can be observed in pleiotropic effects on immunity and inflam-
patients with gingivitis and periodontitis imme- mation.90 A recent systematic review with
diately following tooth brushing,83 periodontal meta-analysis that included up to 15 studies
probing,84 ultrasonic scaling,85 and scaling and demonstrated that the levels of proinflamma-
root planing (SRP),86 the increased prevalence tory cytokines (interleukin 1 [IL-1], tumor
of pneumonia in patients with periodontitis necrosis factor alpha [TNF-α]) in gingival
can be due to hematogenous dissemination crevicular fluid are significantly increased, but
of periodontal pathogens to the lungs. This the magnitude of these increases is modest,
assumption has been supported by a system- and a high degree of bias has been observed in
atic review of nine cross-sectional studies that some publications.91 A recent cross-sectional
included 219 patients with periodontitis that study that included 239 patients with
showed that periodontal pathogens can be periodontitis demonstrated a statistically sig-
cultured from blood samples of these patients nificant association between serum levels of
following periodontal procedures.87 In addi- proinflammatory cytokines (TNF-α , interferon
tion, a study that included 25 patients with gamma [INF-γ]) and the number of teeth with
periodontitis demonstrated the presence of CAL ≥ 6 mm.92
intact P gingivalis within the blood dendritic However, the clinical significance of ele-
cells of these patients for 24 hours.88 vated serum cytokine levels in patients with
However, the clinical significance of bacter­ periodontitis and their negative impact toward
emia in the possible development of distant the development of pneumonia has yet to be
inflammation foci is unclear. Bacteremia is a understood. Similar to periodontitis, increased
transient event, even after periodontal proce- levels of proinflammatory cytokines have been
dures, and the host immune system eliminates detected in the serum of patients with CAP,
bacteria from the blood within 20 to 30 min- and they positively correlated with the sever-
utes after the procedure.86,89 A systematic ity of the respective diseases.93 However, there
review with meta-analysis of seven studies is currently no evidence in the literature that
demonstrated a lack of statistically significant demonstrates a direct causative effect of the
association between the patient’s periodon- increased serum cytokine levels in patients
tal status and bacteremia following tooth with periodontitis on the development of
brushing.83 Although it is possible that in sus- pneumonia.
ceptible older or immunocompromised patients

200
The Effects of Antimicrobial Oral Hygiene Treatment on Outcomes of Pulmonary Disease

Periodontitis and COPD A few alternative mechanisms for the asso-


ciation between periodontitis and COPD
Although the detailed underlying mechanisms have been reported. An older study suggested
of the increased association between periodon- that emphysema (one of the clinical entities
titis and COPD have not been described in the of COPD) and periodontitis are initiated by
literature, it appears that environmental factors deposition of foreign material, leading to acti-
play a crucial role in this relationship. As dis- vation of neutrophils, release of neutrophil
cussed previously, smoking is considered the proteinases, and connective tissue destruc-
most significant risk factor for both periodon- tion.100 In 2013, the Joint European Federation
titis and COPD, and it can induce inflammation of Periodontology/American Academy of Peri-
via a variety of mechanisms.94,95 Multiple studies odontology Workshop on Periodontitis and
have demonstrated that smoking is significantly Systemic Diseases proposed that hematogenous
associated with the prevalence of periodontitis95 dissemination of dental plaque organisms and
and poorer outcomes of periodontal treat- inflammatory mediators from periodontal
ment.96 In addition, a recent systematic review pockets to the lungs might occur and mod-
with meta-analysis of six studies reported ify the inflammatory status of patients with
that not only smoking but also environmental COPD.101 However, current evidence in the lit-
tobacco smoke lead to the increased prevalence erature that supports these mechanisms linking
of periodontitis (OR: 1.34; 95% CI: 0.93–1.94; periodontitis and COPD is unclear.
P > .05) and tooth loss (OR: 1.33; 95% CI: Taken together, currently available evi-
0.52–3.40; P > .05).97 Another recent system- dence in the literature suggests that the direct
atic review with meta-analysis of 42 studies aspiration of saliva containing periodontal
that included 547,391 participants showed pathogens appears to be the most plausible
that the prevalence of COPD is highest in mechanism explaining the increased prevalence
smokers and former smokers and decreased of pneumonia in patients with periodontitis.
in never-smokers.98 Interestingly, an NHANES In addition, scientific evidence suggests that
III–based study that included 7,625 participants smoking may be a key factor linking peri-
(993 patients with COPD and 6,632 non-COPD odontitis and COPD. Although an underlying
controls) demonstrated that the increased prev- biologic and physiologic rationale exists for
alence of periodontitis (defined as CAL > 4 other mechanisms reviewed earlier, the lack of
mm) in patients with COPD occurs only in cur- scientific evidence to support these assump-
rent smokers (OR: 3.71; 95% CI: 1.78–21.66; tions, especially demonstrated in the literature
P ≤ .01), especially in cases of moderate and during the past two decades, makes their role
severe COPD (OR: 6.21; 95% CI: 1.74–7.89; in linking periodontitis and pulmonary dis-
P ≤ .01), whereas no significant differences eases unlikely.
are observed for former and never-smokers.49
Nevertheless, these conclusions should be taken
with caution considering the extremely wide CI
with a low limit of only approximately 1.8. Sim- The Effects of Antimicrobial
ilarly, another systematic review that included Oral Hygiene Treatment on
five studies (1,401 patients with COPD and
1,158 non-COPD controls) demonstrated that
Outcomes of Pulmonary
the significant association between periodonti- Disease
tis (evidenced by increased plaque index) and
COPD occurs in current smokers (OR: 3.99; The oral health of older adults in nursing
95% CI: 2.58–6.16; P < .001) but not in for- homes is considered less adequate than that
mer smokers and never-smokers.99 of the general population.102 Therefore, dental

201
9 The Association Between Oral Infections and Pulmonary Disease

plaque can be an important confounding fac- patients with VAP who used 0.12% chlor-
tor for the increased prevalence of pulmonary hexidine with or without tooth brushing had
diseases in this group of patients. Considering a similar incidence of VAP (relative risk [RR]:
positive association between periodontitis and 0.77; 95% CI: 0.50–1.21; P > .05) and ICU
pulmonary diseases, various studies have aimed mortality (RR: 0.88; 95% CI: 0.70–1.10;
to examine the role of oral hygiene interven- P > .05) compared to those who followed a
tions in the prevalence of pulmonary diseases routine oral hygiene protocol.106 A study that
and associated financial cost and morbidity. included seven RCTs (a total 2,144 patients)
demonstrated that the intensified oral hygiene
protocol using antiseptics does not affect the
Pneumonia incidence of VAP-associated mortality (RR:
There is high-quality evidence in the litera- 0.96; 95% CI: 0.69–1.33; P > .05).107 A
ture that the use of chlorhexidine mouthwash study that included 5 to 18 RCTs (a total of
adjunct to mechanical oral hygiene procedures up to 2,451 patients) showed that the intensi-
exerts a substantial effect on the reduction in fied oral hygiene protocol with chlorhexidine
the amount of dental plaque in patients with mouthrinse or gel has no significant effect on
gingivitis and periodontitis.103,104 Consider- VAP-associated mortality (RR: 1.09; 95% CI:
ing these positive effects of chlorhexidine in 0.95–1.23; P > .05), duration of mechanical
patients with periodontitis, the US Centers for intervention (mean difference [MD]: –.09
Disease Control and Prevention developed an days; 95% CI: –1.73–1.55 days; P > .05), and
intensified oral hygiene protocol to minimize duration of ICU stay (MD: 0.21 days; 95%
the risk of complications of HAP in patients CI: −1.48–1.89 days; P > .05).108 Finally, a
with periodontitis.14 This protocol includes study that included five RCTs (a total of 3,490
oropharyngeal cleaning and decontamina- patients) showed that the intensified oral
tion with an antiseptic agent (most commonly hygiene protocol (performed by nursing home
chlorhexidine) as well as development and personnel or dentists/dental hygienists) in older
implementation of a comprehensive oral adults in hospitals and nursing homes leads
hygiene program for patients at high risk for to only weak reduction in the HAP-associated
developing pneumonia. mortality rate compared to routine oral care
Several systematic reviews with meta-analysis (RR: 0.80; 95% CI: 0.49–1.31; P > .05).109
have examined the effect of the intensified oral Interestingly, this study also showed that the
hygiene protocol on the incidence of pneumo- outcome of the intensified oral hygiene proto-
nia and pneumonia-associated morbidity and col on incidence of HAP is almost three-fold
mortality and reported contradictory results. as efficient when performed by dentists/den-
The first set of studies showed that intensified tal hygienists (RR: 0.43; 95% CI: 0.25–0.75;
oral hygiene did not have significant clinical P < .05) compared to nursing home person-
effects on patients with pneumonia compared nel (RR: 1.20; 95% CI: 0.97–1.48; P > .05).
to control subjects who followed the routine Although these studies failed to demonstrate
oral hygiene protocol. A study that included positive effects of the intensified oral hygiene
four RCTs (a total of 1,202 patients) reported protocol, almost all outcomes of these studies
that the use of 0.12% to 0.2% chlorhexi- did not reach statistical significance.
dine mouthwash or gel does not significantly On the other hand, another set of systematic
impact the incidence of HAP (OR: 0.42; 95% reviews with meta-analysis showed the positive
CI: 0.16–1.06; P > .05) or HAP-associated outcomes of the intensified oral hygiene pro-
mortality (OR: 0.77; 95% CI: 0.28–2.11; tocol in patients with pneumonia compared
P > .05).105 Another study that included four to control subjects who followed the routine
RCTs (a total of 828 patients) showed that oral hygiene protocol. A study that included

202
Conclusion

five RCTs demonstrated that the intensified oral Taken together, despite the discrepancy in
hygiene protocol significantly decreased the the scientific evidence, a majority of systematic
incidence of HAP (OR: 3.0; 95% CI: 2.1–4.4; reviews show that the intensified oral hygiene
P < .05).110 Another study that included seven protocol using chlorhexidine (but not povi-
RCTs (a total of 2,144 patients) showed that the done iodine) results in a statistically significant
intensified oral hygiene protocol using chlor- reduction in the incidence of pneumonia (HAP
hexidine resulted in significant reduction in the and VAP). However, the effectiveness of this
incidence of VAP (RR: 0.56; 95% CI: 0.39– protocol in reducing HAP- or VAP-associated
0.81; P < .05)107; however, this study failed mortality is still unclear.
to demonstrate any benefit of intensified oral
hygiene with antiseptics on VAP-associated mor-
tality. A study that included 18 RCTs (a total of COPD
2,451 patients) showed that the intensified oral A series of studies have also examined the
hygiene protocol with chlorhexidine mouthrinse effect of improved oral hygiene on clinical
or gel significantly reduced the incidence of VAP manifestations of COPD. A 2-year RCT study
(RR 0.75; 95% CI 0.62–0.91; P < .01),108 but that included 59 Chinese patients with COPD
this study also failed to demonstrate any effect and periodontitis reported that both supragin-
of this protocol on VAP-associated mortality. gival scaling and SRP significantly decreased
Another study that included 12 RCTs (a total the incidence of frequent COPD exacerbations
of 2,341 patients) also showed that the inten- compared to control patients who received no
sified oral hygiene protocol with chlorhexidine periodontal treatment (15% versus 3.0% versus
significantly reduced the incidence of VAP (RR: 66.7%, respectively; P < .05).114 However, the
0.72; 95% CI: 0.55–0.94; P < .05), but the baseline mean PPD of the patients in each group
use of povidone iodine antiseptic mouthwash was approximately 3.0 mm, thus questioning
had no effect (RR: 0.39; 95% CI: 0.11–1.36; the poor periodontal status of these patients.
P > .05).111 A study that included 13 RCTs (a Another study that included 40 patients with
total of 1,640 patients) showed that the inten- COPD and moderate and severe periodonti-
sified oral hygiene protocol with chlorhexidine tis demonstrated that SRP does not result in
significantly reduced the incidence of VAP only substantial improvement in periodontal health
at 2% concentration (RR: 0.53; 95% CI: 0.31– (thus questioning the clinical effectiveness of
0.91; P < .05), but not at 0.12% (RR: 1.0; 95% this treatment) but significantly reduced exac-
CI: 0.51–1.99; P > .05) or 0.20% (RR: 0.63; erbation frequency rate compared to untreated
95% CI: 0.32–1.22; P > .05) concentration.112 controls.115 Taken together, these results sug-
A study that included four RCTs (a total of gest that improved periodontal health leads to
1,009 patients) on patients with AP reported improved clinical outcomes of COPD.
that the intensified oral hygiene protocol pro-
vided by nursing home personnel and dentists/
dental hygienists is associated with a reduced
risk for the development of HAP (RR: 0.61; Conclusion
95% CI: 0.40–0.91; P < .05) and decreased
mortality (RR: 0.41; 95% CI: 0.23–0.71; P In summary, accumulating evidence in the liter-
< .01) compared to the control group that ature suggests an existing association between
included patients performing oral hygiene care periodontitis and pulmonary diseases (pneumo-
themselves.113 This set of studies showed pos- nia and COPD). Among the different categories
itive outcomes of the intensified oral hygiene of pneumonia, AP appears to have the great-
protocol, and a majority of the outcomes of est evidence for this association. However, as
these studies reached statistical significance. critical readers, we need to interpret these data

203
9 The Association Between Oral Infections and Pulmonary Disease

with caution, as this association may or may to reduce financial costs and inpatient hospital
not apply to a wide population of patients. It days and improve the quality of life of these
appears that in a majority of cases this associ- patients. However, the intensified oral hygiene
ation exists in older or immunocompromised protocol may not be effective in reducing the
adults (eg, hospitalized patients, smokers) and mortality rates in patients with HAP and VAP,
that systemically healthy patients with periodon- suggesting that when patients have advanced
titis are not more prone to pulmonary diseases cases of pneumonia with severe inflammation,
than systemically healthy individuals with no the contribution of periodontal pathogens to
periodontitis. Thus, periodontitis can predispose the total inflammatory burden of these patients
a susceptible individual to the development of is not clinically relevant.
pulmonary diseases or their more severe clinical The periodontal health of patients with pul-
manifestation, but no current data that support monary diseases (pneumonia and COPD) can
a causative association between these two con- be improved by cooperation between patients
ditions exist. Most likely, this causality will and dental professionals. The improved oral
never be established, as in this case periodon- health of these patients leads to a decreased
titis should always precede the occurrence of incidence of pneumonia (specifically, differ-
pulmonary diseases, which is not supported by ent categories of HAP) and COPD, especially
any current epidemiologic study. Nevertheless, a when performed professionally by dentists
temporal relationship between periodontitis and and dental hygienists compared to hospital/
pulmonary diseases (pneumonia and COPD) nursing home personnel. Oral self-care and
can be established, as many examples provided periodontal procedures appear to be safe for
in this chapter (eg, use of ventilators, stay in patients with pulmonary diseases unless these
ICU) are not chronic in nature. patients have medical conditions that would
We have reviewed several mechanisms pre- contraindicate this treatment. Therefore, based
viously proposed in the literature that could on the current evidence in the literature, med-
explain the increased association between peri- ical and dental personnel should discuss the
odontitis and pulmonary diseases. Based on importance of oral hygiene maintenance for
the current scientific evidence, we have con- improved outcomes of pulmonary diseases with
cluded that a direct aspiration of saliva and patients and rigorously implement it as a part
oropharyngeal secretions containing periodon- of their routine treatment protocol. In addition,
tal pathogens or mechanical dissemination of patients should not wear removable dentures
these pathogens during intubation provide the during sleep because this is when plaque
most plausible explanation for the increased accumulates, resulting in greater numbers of
association of periodontitis with AP and VAP, periodontal pathogens and increased incidence
respectively. The increased association between of AP. Equally important, medical and dental
periodontitis and COPD appears to be due to personnel should be educated about the bene-
detrimental effects of smoking, which is the fits of improved oral hygiene for better clinical
main risk factor for both diseases. outcomes and quality of life for these patients.
We have also shown that periodontitis is However, we should remember that despite
associated with greater complications/mortal- the observed clinical benefits, no current evi-
ity among patients with pulmonary diseases. A dence that periodontal treatment can prevent
majority of systematic reviews also suggest that the development of pulmonary diseases exists.
the intensified oral hygiene protocol in the risk This review also highlights some gaps in
populations provides a statistically significant our current understanding of the association
reduction in the incidence of pulmonary dis- between periodontal and pulmonary diseases.
eases. This signifies a high importance of oral A majority of studies that examined the asso-
hygiene for patients with pulmonary diseases ciation between periodontitis and pneumonia

204
Clinical Considerations

included patients with HAP (AP and VAP) examining associations between periodontitis
but not CAP. Although several systematic and pulmonary diseases used various defini-
reviews with meta-analysis were included in tions of periodontitis, making interpretation
this chapter, some of these studies reported of the results more challenging. Therefore,
and acknowledged the poor quality and biased an accurate and consistent definition of
data representation of the original publications. periodontitis, properly designed controlled
Some studies included a limited number of interventional studies, and a new series of sys-
patients, and therefore their outcomes should tematic reviews are needed to make current
be interpreted with caution. In addition, studies conclusions more scientifically solid.

Clinical Considerations: What You Can Take Back to Your Practice


Is there a biologic plausibility for an association Several studies have demonstrated that adjunc-
between oral infections and disorders and pul- tive periodontal treatment significantly im-
monary diseases?  Among the proposed mech- proves outcomes of pulmonary disease treat-
anisms for the association between periodon- ment and decreases the incidence of treatment
titis and pulmonary diseases, aspiration of complications. However, there is insufficient ev-
periodontal and/or respiratory pathogens of idence to conclude that periodontal treatment
oropharyngeal secretions and their mechan- leads to the reduction in pneumonia-associated
ical dissemination during intubation are con- mortality. In addition, although few studies
sidered the most plausible mechanisms for the have reported positive outcomes of periodontal
increased prevalence of aspiration pneumo- treatment on the incidence of chronic obstruc-
nia and ventilator-associated pneumonia, re- tive pulmonary disease, higher-quality studies
spectively. Among the proposed mechanisms of are needed to validate this conclusion.
the increased association between periodonti-
tis and chronic obstructive pulmonary disease, Is it safe to provide dental care for patients with
smoking and smoking-associated detrimental pulmonary diseases?  Evidence in the literature
effects are considered the most plausible mech- reports improved outcomes of the intensified
anism linking these diseases. oral hygiene protocol in patients with pulmo-
nary diseases. Therefore, intensified oral self-
Are oral infections and disorders independent care and professional periodontal care appear
risk factors for the development of adverse sys- to be safe and highly recommended for pa-
temic outcomes of pulmonary diseases?  Peri- tients with pulmonary diseases, unless other lo-
odontitis is associated with the increased prev- cal and/or systemic conditions contraindicate
alence of pulmonary diseases, even after adjust- these procedures (this should be established in
ment for other potential confounding factors. consultation with a physician).
In addition, adjunctive periodontal treatment
significantly improves the outcomes of pulmo- What should a dental practitioner inform a pa-
nary disease treatment. Although a temporal re- tient about the association between oral infec-
lationship between periodontitis and pulmo- tions and disorders and the development of sys-
nary diseases can be established, there is no evi- temic outcomes of pulmonary diseases?  Cur-
dence in the literature that supports a causative rent evidence in the literature has demonstrated
association between these two conditions. Thus, that patients with pulmonary diseases have bet-
periodontitis cannot currently be considered an ter outcomes of the pulmonary disease treat-
independent risk factor for pulmonary diseases. ment if they have no concurrent periodontitis.
Therefore, these patients should be informed
Can treatment of oral infections and disorders that the maintenance of adequate oral hygiene
reduce the risk for the development of adverse might improve the outcomes of the pulmonary
systemic outcomes of pulmonary diseases?  disease treatment.

205
9 The Association Between Oral Infections and Pulmonary Disease

References 17. DiBardino DM, Wunderink RG. Aspiration pneumonia:


A review of modern trends. J Crit Care 2015;30:40–48.
18. Marik PE. Aspiration pneumonitis and aspiration pneu-
1. Eke PI, Thornton-Evans G, Wei L, Borgnakke WS, Dye monia. N Engl J Med 2001;344:665–671.
B, Genco RJ. Periodontitis in US adults: National Health 19. Komiya K, Ishii H, Kadota J. Healthcare-associated
and Nutrition Examination Survey 2009-2014. J Am pneumonia and aspiration pneumonia. Aging Dis 2015;
Dent Assoc 2018;149:576–588. 6:27–37.
2. Tonetti MS, Jepsen S, Jin L, Otomo-Corgel J. Impact 20. Ramsey D, Smithard D, Kalra L. Silent aspiration: What
of the global burden of periodontal diseases on health, do we know? Dysphagia 2005;20:218–225.
nutrition and wellbeing of mankind: A call for global 21. Manabe T, Teramoto S, Tamiya N, Okochi J, Hizawa N.
action. J Clin Periodontol 2017;44:456–462. Risk factors for aspiration pneumonia in older adults.
3. Ramseier CA, Anerud A, Dulac M, et al. Natural history PLoS One 2015;10:e0140060.
of periodontitis: Disease progression and tooth loss over 22. DeLegge MH. Aspiration pneumonia: Incidence, mortal-
40 years. J Clin Periodontol 2017;44:1182–1191. ity, and at-risk populations. JPEN J Parenter Enteral
4. Eke PI, Wei L, Thornton-Evans GO, et al. Risk indicators Nutr 2002;26(suppl 6):S19–S24.
for periodontitis in US adults: NHANES 2009 to 2012. 23. Marik PE. Pulmonary aspiration syndromes. Curr Opin
J Periodontol 2016;87:1174–1185. Pulm Med 2011;17:148–154.
5. Tomar SL, Asma S. Smoking-attributable periodontitis 24. Awano S, Ansai T, Takata Y, et al. Oral health and mortal-
in the United States: Findings from NHANES III. National ity risk from pneumonia in the elderly. J Dent Res 2008;
Health and Nutrition Examination Survey. J Periodontol 87:334–339.
2000;71:743–751. 25. Wunderink RG, Waterer GW. Community-acquired
6. Zenthöfer A, Baumgart D, Cabrera T, et al. Poor dental pneumonia. N Engl J Med 2014;370:1863.
hygiene and periodontal health in nursing home resi- 26. Hoyert DL, Kung HC, Smith BL. Deaths: Preliminary
dents with dementia: An observational study. Odontology data for 2003. Natl Vital Stat Rep 2005;53:1–48.
2017;105:208–213. 27. Janssens JP, Krause KH. Pneumonia in the very old.
7. Ziukaite L, Slot DE, Van der Weijden FA. Prevalence of Lancet Infect Dis 2004;4:112–124.
diabetes mellitus in people clinically diagnosed with 28. Jackson ML, Neuzil KM, Thompson WW, et al. The
periodontitis: A systematic review and meta-analysis of burden of community-acquired pneumonia in seniors:
epidemiologic studies. J Clin Periodontol 2018;45: Results of a population-based study. Clin Infect Dis
650–662. 2004;39:1642–1650.
8. Freid VM, Bernstein AB, Bush MA. Multiple chronic 29. Jain S, Self WH, Wunderink RG, et al. Community-
conditions among adults aged 45 and over: Trends over acquired pneumonia requiring hospitalization among
the past 10 years. NCHS Data Brief 2012;(100):1–8. U.S. adults. N Engl J Med 2015;373:415–427.
9. Mackenzie G. The definition and classification of pneu- 30. Kaplan V, Angus DC, Griffin MF, Clermont G, Scott
monia. Pneumonia (Nathan) 2016;8:14. Watson R, Linde-Zwirble WT. Hospitalized community-
10. Prina E, Ranzani OT, Torres A. Community-acquired acquired pneumonia in the elderly: Age- and sex-related
pneumonia. Lancet 2015; 386: 1097–1108. patterns of care and outcome in the United States. Am
11. Bordon J, Aliberti S, Fernandez-Botran R, et al. Under- J Respir Crit Care Med 2002;165:766–772.
standing the roles of cytokines and neutrophil activity 31. Yamasaki K, Kawanami T, Yatera K, et al. Significance
and neutrophil apoptosis in the protective versus dele- of anaerobes and oral bacteria in community-acquired
terious inflammatory response in pneumonia. Int J Infect pneumonia. PLoS One 2013;8:e63103.
Dis 2013;17: e76–83. 32. Macisaac RJ, Ekinci EI, Jerums G. Markers of and risk
12. Moldoveanu B, Otmishi P, Jani P, et al. Inflammatory factors for the development and progression of diabetic
mechanisms in the lung. J Inflamm Res 2009;2:1–11. kidney disease. Am J Kidney Dis 2014; 63(suppl 2):
13. World Health Organization. Revised WHO Classification S39–S62.
and Treatment of Childhood Pneumonia at Health Facil- 33. Adeloye D, Chua S, Lee C, et al. Global and regional
ities. Geneva: World Health Organization, 2014. estimates of COPD prevalence: Systematic review and
14. Healthcare Infection Control Practices Advisory Commit- meta-analysis. J Glob Health 2015;5:020415.
tee, CDC. Guidelines for preventing healthcare- 34. Lozano R, Naghavi M, Foreman K, et al. Global and
associated pneumonia, 2003 recommendations of the regional mortality from 235 causes of death for 20 age
CDC and the Healthcare Infection Control Practices groups in 1990 and 2010: A systematic analysis for the
Advisory Committee. Respir Care 2004;49:926–939. Global Burden of Disease Study 2010. Lancet 2012;
15. Mehta A, Bhagat R. Preventing ventilator-associated 380:2095–2128.
infections. Clin Chest Med 2016;37:683–692. 35. Mathers CD, Loncar D. Projections of global mortality
16. Safdar N, Dezfulian C, Collard HR, Saint S. Clinical and and burden of disease from 2002 to 2030. PLoS Med
economic consequences of ventilator-associated pneu- 2006;3:e442.
monia: A systematic review. Crit Care Med 2005;33: 36. Corbridge S, Wilken L, Kapella MC, Gronkiewicz C. An
2184–2193. evidence-based approach to COPD: Part 1. Am J Nurs
2012;112:46–57.

206
References

37. Yoshikawa A, Sato S, Tanaka T, et al. Breakdown of 52. Si Y, Fan H, Song Y, Zhou X, Zhang J, Wang Z. Asso-
lung framework and an increase in pores of Kohn as ciation between periodontitis and chronic obstructive
initial events of emphysema and a cause of reduction pulmonary disease in a Chinese population. J Periodon-
in diffusing capacity. Int J Chron Obstruct Pulmon Dis tol 2012;83:1288–1296.
2016;11:2287–2294. 53. Brook I, Frazier EH. Immune response to Fusobacterium
38. Kobayashi Y, Uehara T, Kawasaki K, et al. Three- nucleatum and Prevotella intermedia in the sputum of
dimensional analysis of alveolar wall destruction in the patients with acute exacerbation of chronic bronchitis.
early stage of pulmonary emphysema. Clin Anat 2015; Chest 2003;124:832–833.
28:227–234. 54. Terashima T, Chubachi S, Matsuzaki T, et al. The asso-
39. Kim V, Rogers TJ, Criner GJ. New concepts in the patho- ciation between dental health and nutritional status in
biology of chronic obstructive pulmonary disease. Proc chronic obstructive pulmonary disease. Chron Respir
Am Thorac Soc 2008;5:478–485. Dis 2017;14:334–341.
40. Vogelmeier CF, Criner GJ, Martinez FJ, et al. Global 55. Gomes-Filho IS, Passos JS, Seixas da Cruz S. Respi-
strategy for the diagnosis, management, and prevention ratory disease and the role of oral bacteria. J Oral
of chronic obstructive lung disease 2017 report. GOLD Microbiol 2010;2.
executive summary. Am J Respir Crit Care Med 2017; 56. Peter KP, Mute BR, Doiphode SS, Bardapurkar SJ,
195:557–582. Borkar MS, Raje DV. Association between periodontal
41. MacNee W. Pathogenesis of chronic obstructive pulmo- disease and chronic obstructive pulmonary disease: A
nary disease. Proc Am Thorac Soc 2005;2:258–266. reality or just a dogma? J Periodontol 2013; 84:
42. Tuder RM, Petrache I. Pathogenesis of chronic obstruc- 1717–1723.
tive pulmonary disease. J Clin Investing 2012; 122: 57. Azarpazhooh A, Leake JL. Systematic review of the
2749–2755. association between respiratory diseases and oral
43. Yu H, Lin M, Wang X, Wang S, Wang Z. Toll-like receptor health. J Periodontol 2006;77:1465–1482.
4 polymorphism is associated with increased suscep- 58. Scannapieco FA. Role of oral bacteria in respiratory
tibility to chronic obstructive pulmonary disease in Han infection. J Periodontol 1999;70:793–802.
Chinese patients with chronic periodontitis. J Oral Sci 59. Larsen T, Fiehn NE. Dental biofilm infections- An update.
2016;58:555–560. APMIS 2017;125:376–384.
44. Hobbins S, Chapple IL, Sapey E, Stockley RA. Is peri- 60. Hirschfeld J, Kawai T. Oral inflammation and bacteremia:
odontitis a comorbidity of COPD or can associations Implications for chronic and acute systemic diseases
be explained by shared risk factors/behaviors? Int J involving major organs. Cardiovasc Hematol Disord Drug
Chron Obstruct Pulmon Dis 2017;12:1339–1349. Targets 2015;15:70–84.
45. Loesche WJ, Lopatin DE. Interactions between peri- 61. Park DR. The microbiology of ventilator-associated
odontal disease, medical diseases and immunity in the pneumonia. Respir Care 2005;50:742–763.
older individual. Periodontol 2000 1998;16:80–105. 62. Belstrøm D, Sembler-Møller ML, Grande MA, et al.
46. Gomes-Filho IS, Santos CM, Cruz SS, et al. Periodontitis Microbial profile comparisons of saliva, pooled and
and nosocomial lower respiratory tract infection: Prelim- site-specific subgingival samples in periodontitis
inary findings. J Clin Periodontol 2009;36:380–387. patients. PLoS One 2017;12:e0182992.
47. Iwasaki M, Taylor GW, Awano S, et al. Periodontal 63. Salminen A, Kopra KA, Hyvärinen K, et al. Quantitative
disease and pneumonia mortality in haemodialysis PCR analysis of salivary pathogen burden in periodon-
patients: A 7-year cohort study. J Clin Periodontol 2018; titis. Front Cell Infect Microbiol 2015;5:69.
45:38–45. 64. Nagaoka K, Yanagihara K, Harada Y, et al. Quantitative
48. Zeng XT, Tu ML, Liu DY, Zheng D, Zhang J, Leng W. detection of periodontopathic bacteria in lower respira-
Periodontal disease and risk of chronic obstructive tory tract specimens by real-time PCR. J Infect
pulmonary disease: A meta-analysis of observational Chemother 2017;23:69–73.
studies. PLoS One 2012;7:e46508. 65. Scannapieco FA, Stewart EM, Mylotte JM. Colonization
49. Hyman JJ, Reid BC. Cigarette smoking, periodontal of dental plaque by respiratory pathogens in medical
disease, and chronic obstructive pulmonary disease. J intensive care patients. Crit Care Med 1992; 20:
Periodontol 2004;75:9–15. 740–745.
50. Chung JH, Hwang HJ, Kim SH, Kim TH. Associations 66. Fourrier F, Duvivier B, Boutigny H, Roussel-Delvallez M,
between periodontitis and chronic obstructive pulmo- Chopin C. Colonization of dental plaque: A source of
nary disease: The 2010 to 2012 Korean National Health nosocomial infections in intensive care unit patients. Crit
and Nutrition Examination Survey. J Periodontol 2016; Care Med 1998;26:301–308.
87:864–871. 67. Heo SM, Haase EM, Lesse AJ, Gill SR, Scannapieco
51. Shen TC, Chang PY, Lin CL, et al. Risk of periodontal FA. Genetic relationships between respiratory pathogens
diseases in patients with chronic obstructive pulmonary isolated from dental plaque and bronchoalveolar lavage
disease: A nationwide population-based cohort study. fluid from patients in the intensive care unit undergoing
Medicine (Baltimore) 2015;94:e2047. mechanical ventilation. Clin Infect Dis 2008; 47:
1562–1570.

207
9 The Association Between Oral Infections and Pulmonary Disease

68. Fritschi BZ, Albert-Kiszely A, Persson GR. Staphylococ- 83. Tomás I, Diz P, Tobías A, Scully C, Donos N. Periodontal
cus aureus and other bacteria in untreated periodontitis. health status and bacteraemia from daily oral activities:
J Dent Res 2008;87:589–593. Systematic review/meta-analysis. J Clin Periodontol
69. El-Solh AA, Pietrantoni C, Bhat A, et al. Colonization of 2012;39:213–228.
dental plaques: A reservoir of respiratory pathogens for 84. Daly CG, Mitchell DH, Highfield JE, Grossberg DE,
hospital-acquired pneumonia in institutionalized elders. Stewart D. Bacteremia due to periodontal probing: A
Chest 2004;126:1575–1582. clinical and microbiological investigation. J Periodontol
70. Marino PJ, Wise MP, Smith A, et al. Community analysis 2001;72:210–214.
of dental plaque and endotracheal tube biofilms from 85. Kinane DF, Riggio MP, Walker KF, MacKenzie D, Shearer
mechanically ventilated patients. J Crit Care 2017; 39: B. Bacteraemia following periodontal procedures. J Clin
149–155. Periodontol 2005;32:708–713.
71. Sands KM, Wilson MJ, Lewis MAO, et al. Respiratory 86. Forner L, Larsen T, Kilian M, Holmstrup P. Incidence of
pathogen colonization of dental plaque, the lower bacteremia after chewing, tooth brushing and scaling
airways, and endotracheal tube biofilms during mechan- in individuals with periodontal inflammation. J Clin Peri-
ical ventilation. J Crit Care 2017;37:30–37. odontol 2006;33:401–407.
72. Tuon FF, Gavrilko O, Almeida S, et al. Prospective, 87. Horliana AC, Chambrone L, Foz AM, et al. Dissemination
randomised, controlled study evaluating early modifica- of periodontal pathogens in the bloodstream after peri-
tion of oral microbiota following admission to the odontal procedures: A systematic review. PLoS One
intensive care unit and oral hygiene with chlorhexidine. 2014;9:e98271.
J Glob Antimicrob Resist 2017;8:159–163. 88. Carrion J, Scisci E, Miles B, et al. Microbial carriage
73. Sumi Y, Miura H, Michiwaki Y, Nagaosa S, Nagaya M. state of peripheral blood dendritic cells (DCs) in chronic
Colonization of dental plaque by respiratory pathogens periodontitis influences DC differentiation, atherogenic
in dependent elderly. Arch Gerontol Geriatr 2007; 44: potential. J Immunol 2012;189:3178–3187.
119–124. 89. Maharaj B, Coovadia Y, Vayej AC. An investigation of
74. O’Donnell LE, Smith K, Williams C, et al. Dentures are the frequency of bacteraemia following dental extraction,
a reservoir for respiratory pathogens. J Prosthodont tooth brushing and chewing. Cardiovasc J Afr 2012; 23:
2016;25:99–104. 340–344.
75. Iinuma T, Arai Y, Abe Y, et al. Denture wearing during 90. Freire M, Van Dyke TE. The role of inflammation in
sleep doubles the risk of pneumonia in the very elderly. oral-systemic interactions. In: Glick M (ed). The
J Dent Res 2015;94(suppl 3):28S–36S. Oral-Systemic Health Connection: A Guide to Patient
76. Benedyk M, Mydel PM, Delaleu N, et al. Gingipains: Care, ed 2. Chicago:Quintessence, 2018:86–101.
Critical factors in the development of aspiration pneu- 91. Stadler AF, Angst PD, Arce RM, Gomes SC, Oppermann
monia caused by Porphyromonas gingivalis. J Innate RV, Susin C. Gingival crevicular fluid levels of cytokines/
Immun 2016;8:185–198. chemokines in chronic periodontitis: A meta-analysis.
77. Kimizuka R, Kato T, Ishihara K, Okuda K. Mixed infec- J Clin Periodontol 2016;43:727–745.
tions with Porphyromonas gingivalis and Treponema 92. Kampits C, Montenegro MM, Ribeiro IW, et al. Periodon-
denticola cause excessive inflammatory responses in a tal disease and inflammatory blood cytokines in patients
mouse pneumonia model compared with monoinfec- with stable coronary artery disease. J Appl Oral Sci
tions. Microbes Infect 2003;5:1357–1362. 2016;24:352–358.
78. Hajishengallis G, Wang M, Bagby GJ, Nelson S. Impor- 93. Bacci MR, Leme RC, Zing NP, et al. IL-6 and TNF-alpha
tance of TLR2 in early innate immune response to acute serum levels are associated with early death in
pulmonary infection with Porphyromonas gingivalis in community-acquired pneumonia patients. Braz J Med
mice. J Immunol 2008;181:4141–4149. Biol Res 2015;48:427–432.
79. Okuda K, Kimizuka R, Abe S, Kato T, Ishihara K. Involve- 94. Palmer RM, Wilson RF, Hasan AS, Scott DA. Mecha-
ment of periodontopathic anaerobes in aspiration nisms of action of environmental factors—Tobacco
pneumonia. J Periodontol 2005; 76(suppl 11): smoking. J Clin Periodontol 2005;32(suppl 6):180–195.
2154–2160. 95. Nikota JK, Stämpfli MR. Cigarette smoke-induced
80. Mongodin E, Bajolet O, Cutrona J, et al. Fibronectin- inflammation and respiratory host defense: Insights from
binding proteins of Staphylococcus aureus are involved animal models. Pulm Pharmacol Ther 2012; 25:
in adherence to human airway epithelium. Infect Immun 257–262.
2002;70:620–630. 96. Heasman L, Stacey F, Preshaw PM, McCracken GI,
81. Gibbons RJ, Etherden I. Fibronectin-degrading enzymes Hepburn S, Heasman PA. The effect of smoking on
in saliva and their relation to oral cleanliness. J Peri- periodontal treatment response: A review of clinical
odontol Res 1986;21:386–395. evidence. J Clin Periodontol 2006;33:241–253.
82. Bonnans C, Chou J, Werb Z. Remodelling the extracel- 97. Akinkugbe AA, Slade GD, Divaris K, Poole C. Systematic
lular matrix in development and disease. Nat Rev Mol review and meta-analysis of the association between
Cell Biol 2014;15:786–801. exposure to environmental tobacco smoke and peri-
odontitis endpoints among nonsmokers. Nicotine Tob
Res 2016;18:2047–2056.

208
References

98. Kamal R, Srivastava AK, Kesavachandran CN. 107. Chan EY, Ruest A, Meade MO, Cook DJ. Oral decon-
Meta-analysis approach to study the prevalence of tamination for prevention of pneumonia in mechanically
chronic obstructive pulmonary disease among current, ventilated adults: Systematic review and meta-analysis.
former and non-smokers. Toxicol Rep 2015; 2: BMJ 2007;334:889.
1064–1074. 108. Hua F, Xie H, Worthington HV, Furness S, Zhang Q, Li
99. Tan L, Wang H, Pan C, Zhao J. Periodontal health and C. Oral hygiene care for critically ill patients to prevent
chronic obstructive pulmonary disease stratified by ventilator-associated pneumonia. Cochrane Database
smoking: A meta-analysis. Int J Clin Exp Med 2016;9: Syst Rev 2016;10:CD008367.
23190–23197. 109. Sjögren P, Wårdh I, Zimmerman M, Almståhl A, Wikström
100. Travis J, Pike R, Imamura T, Potempa J. The role of M. Oral care and mortality in older adults with pneumonia
proteolytic enzymes in the development of pulmonary in hospitals or nursing homes: Systematic review and
emphysema and periodontal disease. Am J Respir Crit meta-analysis. J Am Geriatr Soc 2016;64:2109–2115.
Care Med 1994;150(6pt2):S143–146. 110. Scannapieco FA, Bush RB, Paju S. Associations
101. Linden GJ, Herzberg MC. Periodontitis and systemic between periodontal disease and risk for nosocomial
diseases: A record of discussions of working group 4 bacterial pneumonia and chronic obstructive pulmonary
of the Joint EFP/AAP Workshop on Periodontitis and disease. A systematic review. Ann Periodontol 2003;8:
Systemic Diseases. J Clin Periodontol 2013; 40(suppl 54–69.
14):S20–S23. 111. Labeau SO, Van de Vyver K, Brusselaers N, Vogelaers
102. Weening-Verbree L, Huisman-de Waal G, van Dussel- D, Blot SI. Prevention of ventilator-associated pneumo-
dorp L, van Achterberg T, Schoonhoven L. Oral health nia with oral antiseptics: A systematic review and
care in older people in long term care facilities: A system- meta-analysis. Lancet Infect Dis 2011;11:845–854.
atic review of implementation strategies. Int J Nurs Stud 112. Villar CC, Pannuti CM, Nery DM, Morillo CM, Carmona
2013;50:569–582. MJ, Romito GA. Effectiveness of intraoral chlorhexidine
103. James P, Worthington HV, Parnell C, et al. Chlorhexidine protocols in the prevention of ventilator-associated
mouthrinse as an adjunctive treatment for gingival pneumonia: Meta-analysis and systematic review.
health. Cochrane Database Syst Rev 2017; 3: Respir Care 2016;61:1245–1259.
CD008676. 113. Kaneoka A, Pisegna JM, Miloro KV, et al. Prevention of
104. Richards D. Chlorhexidine mouthwash plaque levels healthcare-associated pneumonia with oral care in indi-
and gingival health. Evid Based Dent 2017;18:37–38. viduals without mechanical ventilation: A systematic
105. Pineda LA, Saliba RG, El Solh AA. Effect of oral decon- review and meta-analysis of randomized controlled trials.
tamination with chlorhexidine on the incidence of Infet Control Hosp Epidemiol 2015;36:899–906.
nosocomial pneumonia: A meta-analysis. Crit Care 114. Zhou X, Han J, Liu Z, Song Y, Wang Z, Sun Z. Effects
2006;10:R35. of periodontal treatment on lung function and exacer-
106. Gu WJ, Gong YZ, Pan L, Ni YX, Liu JC. Impact of oral bation frequency in patients with chronic obstructive
care with versus without toothbrushing on the prevention pulmonary disease and chronic periodontitis: A 2-year
of ventilator-associated pneumonia: A systematic review pilot randomized controlled trial. J Clin Periodontol 2014;
and meta-analysis of randomized controlled trials. Crit 41:564–572.
Care 2012;16:R190. 115. Kucukcoskun M, Baser U, Oztekin G, Kiyan E, Yalcin F.
Initial periodontal treatment for prevention of chronic
obstructive pulmonary disease exacerbations. J Peri-
odontol 2013;84:863–870.

209
CHAPTER 10

Periodontal Infections and


Adverse Pregnancy Outcomes
Yiorgos A. Bobetsis, dds, phd
Wenche S. Borgnakke, dds, mph, phd
Panos N. Papapanou, dds, phd

For the past two decades, the association Burden of Disease Study identified neonatal
between periodontal diseases and adverse disorders and specifically preterm birth (PTB),
pregnancy outcomes has been the focus of defined as the live birth of an infant before
investigation in a variety of studies, ranging completion of the 37th gestational week, as
from experimental animal models to epidemio- significant contributors to global mortality.1
logic association studies and intervention trials Every year, more than 15 million babies (1 in
in humans. Yet several uncertainties remain, every 10 live births) are born too soon around
and oral health care professionals and the the world,2 and about 1 million children die
public alike are exposed to frequently con- each year as a result of complications of PTB.1,3
tradictory information that must be clarified. Surviving PTB infants often face multiple life-
This chapter addresses the following specific long challenges, including respiratory distress,
questions: impaired motor skills, cognitive and intellec-
tual impairment, psychiatric diseases, and
• What is the effect of pregnancy on the peri- social, visual, and learning difficulties; some
odontal tissues? of these complications even prevent living
• What is the biologic plausibility of the asso- independently.4,5
ciation between periodontal diseases and PTB largely occurs for three reasons: (1)
adverse pregnancy outcomes? medically indicated PTB, (2) spontaneous PTB,
• Are periodontal diseases independent risk and (3) preterm premature rupture of mem-
factors for the development of adverse preg- branes. The rate of PTB varies greatly, from
nancy outcomes? 5% to 18% among babies born across 184
• Does the delivery of periodontal therapy in countries.6 Among 39 countries with a Very
pregnant women contribute to an improve- High Human Development Index ranking, the
ment in pregnancy outcomes? PTB rate ranged from a low of 5.3% (Lat-
• Is periodontal therapy during pregnancy safe via) to a high of 14.7% (Cyprus).5 The United
for mother and child? States ranked 37th on this list (Fig 10-1) but
• What is the appropriate message to give the 54th highest among 184 countries.7 Based on
public about this association? the most recent global data, the United States
had the 6th highest number of PTBs among the
The impact of adverse pregnancy outcomes world’s 184 countries, surpassed only by India,
on society cannot be overestimated. The Global China, Nigeria, Pakistan, and Indonesia.7

210
Periodontal Infections and Adverse Pregnancy Outcomes

14.7
14.0
15
12.0
11.5
10.9
10.5
9.2
9.2
10

8.6
8.0
8.0
8.0
7.9

7.8
7.8

7.7
7.6
7.6
7.6
7.4
7.5
7.4
7.3
7.1

6.7
6.7
6.7

6.6
6.5
6.4
6.3
6.0
5.9
5.9
5.7
5.7
5.5
5.5
5.3
5

0
Cyprus
Bahrain
United States
Singapore
Austria
Qatar
Germany
South Korea
Hungary
Israel
Netherlands
Argentina
Belgium
United Kingdom
Canada
Portugal
Australia
United Arab Emirates
New Zealand
Slovenia
Spain
Switzerland
Czech Republic
Chile
Poland
France
Denmark
Greece
Italy
Ireland
Slovakia
Norway
Sweden
Japan
Estonia
Lithuania
Finland
Croatia
Latvia
Preterm birth rate (per 100 live births)

Fig 10-1  |  Preterm birth rates in 2010 for 39 countries with a Very High Human Development Index ranking.
(Reprinted from Chang et al5 with permission.)

Grade Range
A 8.1 or less
B 8.2 – 9.2
C 9.3 – 10.3
D 10.4 – 11.4
F 11.5 or greater

Fig 10-2  |  US preterm birth rates and grades by state.2

The prevalence of PTB in the United States countries in Northern Europe (around 5%).
has declined since its peak of 12.9% in 2006 The US PTB rate of live births is highest for
to a rate of 9.85% in 2016,8 earning the United non-Hispanic black infants (13.4%), followed
States a grade of C on a scale from A to F from by Hispanic (9.1%) and non-Hispanic white
the March of Dimes.2 Infants in the United (8.9%) infants.8 Large geographic disparities
States are born preterm at almost double the in these rates exist within the United States, as
rate of infants in the western industrialized illustrated in Fig 10-2.

211
10 Periodontal Infections and Adverse Pregnancy Outcomes

Box 10-1  |  Risk factors/markers of preterm birth and low birth weight9

The diverse nature of the exposures and phenotypic characteristics associated with PTB have led to the proposal
to classify PTB as a syndrome, the development of which depends on the interaction of environmental exposures
and genotypic characteristics:9,10

• Young maternal age • Depression


• African American or Afro-Caribbean maternal • Obesity-related preeclampsia
genotype • Diabetes
• Intrauterine infections • Hypertension
• Interpregnancy interval of < 6 months • Low prepregnancy body mass index
• Drugs and heavy alcohol use • Low socioeconomic status
• Tobacco use • Low educational status
• Multiple gestation • Fetal genotype
• In vitro fertilization • History of previous preterm birth
• Psychologic and social stress

PTB babies are further subdivided into the Effects of Pregnancy on the
categories extremely preterm (born before 28
weeks), severely premature (between 28 and 31 Periodontal Tissues
weeks), moderately premature (between 32 and
33 weeks), and near term (between 34 and 36 While dental plaque is the primary etiology
weeks). Additional neonatal disorders or preg- for the development of periodontitis, the
nancy complications include low birth weight, presence of bacterial biofilms adjacent to the
defined as having a weight at birth of less than gingival margin is necessary but not always
2,500 g (5 lb 8 oz), approximately one-third sufficient for the initiation and progression of
less than the average weight of a term infant the disease. A number of systemic factors may
(3.5 kg/7 lb); intrauterine growth restriction increase the susceptibility of the host to the
or small for gestational age, defined as weight microbial challenge, contribute to enhanced
of less than the 10th percentile at any given gingival inflammation and bacterial dysbio-
gestational age; preeclampsia (ie, high maternal sis, and hence affect the extent and severity of
blood pressure [> 140/90 mmHg] and signifi- periodontitis. Several hormones, including the
cant proteinuria [300 mg/24 h]) after the 20th sex steroids, have an influence on the cellular
gestational week; eclampsia (ie, occurrence of components of the periodontal tissues and may
life-threatening maternal seizures); and gesta- interfere with the mechanisms involved in the
tional diabetes (ie, glucose intolerance with pathobiology of periodontitis. Fluctuations in
onset or first recognition during pregnancy, the levels of these hormones in physiologic or
most commonly during the second trimester). nonphysiologic conditions may result in signifi-
Several risk factors have been associated cant alterations in the periodontium, especially
with adverse pregnancy outcomes, some of in the presence of preexisting plaque-induced
which involve infectious or inflammatory path- gingival inflammation.
ways (Box 10-1).

212
Effects of Pregnancy on the Periodontal Tissues

Natural sex steroids, also known as gonadal • Estrogen-mediated suppression of leuko-


steroids, are steroid hormones produced by the cyte production in the bone marrow and
gonads (ovaries or testes), by adrenal glands, or inhibition of polymorphonuclear leukocyte
by conversion from other sex steroids in other chemotaxis and phagocytosis.16
tissues, such as the liver or adipose depots. The • Progesterone-induced reduction of the
main classes of sex steroids are androgens, anti-inflammatory effects of glucocorti-
estrogens, and progestogens, of which the most coids, either directly via receptor binding
important human derivatives include testoster- on osteoblasts or indirectly by antagonizing
one, estradiol, and progesterone, respectively. glucocorticoid receptors.17
In general, androgens are considered male sex
hormones because they have masculinizing Therefore, periods of hormonal flux during
effects, while estrogens and progestogens are puberty, menstruation, pregnancy, menopause,
considered female sex hormones; however, all hormone replacement therapy, or use of contra-
three types are present in both sexes at differ- ceptives have been associated with periodontal
ent levels. manifestations.
Because periodontal tissues possess receptors There is a significant rise in the amount of
for sex steroids, researchers have extensively sex steroids produced during the course of
studied the effects of these hormones on the pregnancy; secreted near-term levels of estra-
periodontium, which include the following: diol, estriol, and progesterone reach 20.0, 80.0,
and 300.0 mg/d, respectively, compared with
• An impact on the degree of keratinization normal secretion levels of 0.6, 3.0, and 19.0
of the gingival epithelium, and thus its bar- mg/d, respectively, in nonpregnant women.18
rier function as a first line of defense against This temporary hormonal elevation correlates
bacterial pathogens.11 with an increase in the prevalence, extent, and
• Involvement in vascular functions such as severity of gingival inflammation, commonly
angiogenesis and vascular permeability. referred to as pregnancy gingivitis, which occurs
Specifically, higher estrogen levels stimu- in approximately 50% of pregnant women19,20
late angiogenesis, while elevated circulating and usually affects interdental papillae of ante-
progesterone enhances capillary permeability rior areas. The severity of gingival inflammation
and dilation, resulting in increased gingival is accentuated from the second gestational
exudate that may facilitate the recruitment month and as pregnancy progresses under
of inflammatory cells in the gingival area consistent plaque colonization.21 After the 8th
and the crevicular fluid. The enhanced vas- gestational month, and especially after delivery,
cular permeability may also be partly due to gingival inflammation subsides and reverts to
stimulating effects of progesterone on pros- the levels of the first trimester. This spontaneous
taglandin synthesis.12 improvement in the absence of periodontal
• Involvement in periodontal connective tissue therapy correlates closely with the concomi-
turnover,13 because estrogen has been shown tant dramatic reduction in the secretion of sex
to enhance the proliferation of fibroblasts steroids.22 However, despite this exacerbated
and the production and maturation of col- inflammatory response and accompanying
lagen, while progesterone has the opposite increases in sulcular depth, gingival crevicular
effects.14 fluid (GCF) flow, and bleeding on probing,13
• An effect of estrogen on salivary peroxi- loss of clinical attachment is infrequent.21
dases, which are active against a variety of In a small percentage of pregnant women (at
microorganisms. Estrogen changes the redox frequencies reported in the literature ranging
potential of the salivary peroxidases.15 between 0.5% and 10.0%), the combined stim-
ulatory effects of sex steroids on angiogenesis

213
10 Periodontal Infections and Adverse Pregnancy Outcomes

bacteria during the second trimester.24,25 In


comparison with nonpregnant patients, preg-
nant patients harbor higher levels of Prevotella
intermedia, Prevotella nigrescens, and Por-
phyromonas gingivalis,26 and colonization by
the aforementioned species was shown to be
enhanced throughout gestation.27–29 Although
the exact mechanisms underlying these shifts
are unclear, hormone-enhanced inflammation
and gingival bleeding appear to increase the
bioavailability of nutrients, augment bacte-
Fig 10-3  |  Clinical presentation of pregnancy gran-
uloma or epulis.
rial growth, and facilitate the proliferation of
hemolytic bacteria, such as P gingivalis.30
Local immunologic modifications may also
underlie the occurrence of pregnancy gingivi-
and extracellular matrix can lead to an exag- tis. Sex steroids were shown to decrease the
gerated inflammatory response to dental production of interleukin 6 (IL-6) by fibro-
plaque and the formation of a localized mass of blasts and to reduce the levels of plasminogen
highly vascularized granulation tissue, referred activator inhibitor 2 in the GCF.12,31,32 High
to as pregnancy granuloma or epulis (Fig 10-3). levels of estrogen and progesterone inhibit
This lesion commonly arises from the proximal polymorphonuclear leukocyte chemotaxis and
gingival tissues of the maxillary anterior, can phagocytosis and suppress antibody and T-cell
reach dimensions that usually do not exceed responses because of the reduction of B lym-
2 cm in diameter, has a pedunculated base, phocytes and CD3 and CD4 cells,33 suggesting
and clinically and histologically resembles a that pregnant women may experience a state of
pyogenic granuloma, or rather a telangiectatic compromised immunity that may also facilitate
granuloma or a capillary hemangioma, because bacterial growth.
it is usually not purulent. It is characterized by
rapid growth and a bright red, hyperemic, and
edematous appearance because of increased
vascularization and may show ulceration of Biologic Plausibility
its thin epithelial lining. The lesion is typically
not painful but may bleed spontaneously or The hypothesis that a number of local or
when mechanically stimulated. systemic pathologic conditions, such as tonsil-
Occasionally, surgical removal of the gran- litis, pneumonia, endocarditis, and septicemia,
uloma may be necessary, especially when it originated from oral foci of infection was first
interferes with mastication or normal speech, formulated by Miller in 1891,34 but lack of sci-
but full excision should be accompanied by entific evidence condemned the theory of focal
thorough debridement and meticulous oral infection to dormancy. Almost 100 years later,
hygiene to minimize the risk of recurrence. the landmark experimental studies in the preg-
However, in most cases, its removal is best nant hamster by Collins et al35 paved the way
deferred until after parturition, when there is for an intense research effort focusing on the
often considerable regression in its size.23 role of periodontal infection and inflammation
In parallel to the clinical manifestations, in adverse pregnancy outcomes.
pregnancy results in altered periodontal micro- To better appreciate the pathogenic mecha-
bial colonization profiles, including an increase nisms that may underlie the association between
in the proportion of anaerobic over aerobic periodontal infection and inflammation and

214
Biologic Plausibility

adverse pregnancy outcomes, some basic fetal inflammatory responses, which are both
knowledge of the physiology of normal and linked to adverse pregnancy outcomes.43 Several
complicated pregnancies is essential. After con- non–genital tract infections, such as pyelo­­
ception, the fetus is nourished by the mother nephritis, asymptomatic bacteriuria, pneumonia,
through the vessel-rich placenta and the umbili- and appendicitis, have also been associated with,
cal cord. Provided with the necessary nutrients, and probably predispose to, PTB.44
the fetus grows in the amniotic fluid contained Periodontal diseases are also infectious, and
by the amniotic sac. The walls of this cavity although they occur at a distance from the
consist of the amnion and the chorion, both of fetoplacental unit, inflammatory mediators
which are attached to the uterus through the produced at the gingiva, periodontal patho-
decidua (the uterine lining or endometrium) gens, and bacterial byproducts may enter the
and the myometrium (the middle layer of the blood circulation and disseminate throughout
uterine wall). As the fetus grows, satisfaction the body. This low-grade bacteremia may trig-
of its increasing needs for nutrients and coping ger the induction of systemic inflammatory
successfully with the decreasing space are crit- responses and/or the establishment of ectopic
ical for the survival of both mother and fetus. infections.45,46
As pregnancy progresses, amniotic fluid A mechanistic model potentially explaining
levels of prostaglandin E2 (PGE2) and inflam- the biologic association between periodontal
matory cytokines such as tumor necrosis factor infection and inflammation and adverse preg-
alpha (TNF-α) and interleukin 1β (IL-1β) nancy outcomes46 is illustrated in Fig 10-4. The
rise steadily until a critical threshold level is model consists of two alternative pathways, one
reached, at which point they induce rupture direct and one indirect. In a direct pathway,
of the amniotic sac membranes, uterine con- periodontal bacteria and/or their byproducts
traction, cervical dilation, and delivery.36 Thus, disseminate to the fetoplacental unit, where
normal parturition is controlled by inflam- they establish an ectopic infection and/or trig-
matory signaling. This process represents a ger a local inflammatory response that results
triggering mechanism that can be modified by in the elevation of inflammatory cytokines
external stimuli, including infection and inflam- and mediators that contribute to pregnancy
matory stressors. complications. In an indirect pathway, inflam-
In the medical literature, high levels of inflam- matory cytokines and mediators produced in
matory mediators (such as IL-1β, IL-6, TNF-α , the periodontal tissues in response to peri-
and PGE2)37–40 in the amniotic fluid and the odontal pathogens enter the blood circulation
serum have been associated with various preg- and reach (1) the fetoplacental unit, where
nancy complications. C-reactive protein (CRP), they contribute to an enhanced concentration
an acute-phase reactant synthesized by the liver of these mediators in this compartment, and
in response to proinflammatory cytokines and (2) the liver, where they stimulate a systemic
hence a marker of systemic inflammation, has inflammatory response through the production
also been associated with an increased risk for of acute-phase reactants. In turn, these media-
PTB, low birth weight, and preeclampsia.41,42 tors enter the blood circulation and reach the
In addition, genitourinary tract infections fetoplacental unit, where they exacerbate intra-
have been implicated in intrauterine infections. uterine inflammation.
Microorganisms gain access to various sites Current evidence indicates that the majority
of the fetoplacental compartment primarily of intrauterine infections originate in the lower
by ascending from the vagina and cervix or genital tract, with the infectious agents ascend-
by hematogenous dissemination through the ing into the otherwise sterile womb.47 Hence,
placenta. This microbial invasion is frequently it is possible that periodontal pathogens may
associated with intra-amniotic inflammation and reach the amniotic space as a result of ascending

215
10 Periodontal Infections and Adverse Pregnancy Outcomes

Fig 10-4 |  Biologic pathways associating periodontal infection and inflammation with ad-
verse pregnancy outcomes. TLR-4, toll-like receptor 4; LBW, low birth weight. (Reprinted
from Madianos et al46 with permission.)

infection following oral-genital transfer. How- the chamber resembles a monoinfection rather
ever, this third pathway is probably the least than a mixed, biofilm-mediated infection such
likely, because from an ecologic point of view it as periodontitis. The discussion that follows
is difficult for oral bacteria to establish colonies summarizes key research findings that support
in the vagina as a result of oral-genital contact the biologic plausibility of each of the two
due to colonization resistance. pathways.
The direct and indirect pathways have been
extensively studied in human and animal mod-
els. The two more common animal models Direct pathway
used include the bacteremia infection model, Probably the strongest evidence in favor of
in which periodontal pathogens are injected a hematogenous dissemination of periodon-
in the circulation to mimic bacteremia from tal pathogens to the amniotic cavity derives
periodontal pathogens in humans, and the from two case reports of women with adverse
chamber model, in which periodontal patho- pregnancy outcomes. In the first case, the same
gens are injected in a subcutaneously implanted clonal type of uncultivable oral Bergeyella was
chamber to mimic a distant site of infection identified in the subgingival plaque as well as
and inflammation such as periodontal infec- the amniotic fluid of a woman with PTB, while
tion. Although these experimental models are no Bergeyella was detected in the mother’s vag-
meant to simulate a periodontal infection in inal tract.48 In the second case, an oral strain
a simplified and reproducible manner, they of Fusobacterium nucleatum was identified as
have several key limitations. The amount of the cause of a term stillbirth and was isolated
bacteria injected in the circulation may not from the lung and stomach of the infant. The
correspond to a transient bacteremia induced same clonal type of F nucleatum was present
by periodontal infection, and the injection of in the subgingival plaque of the mother but not
a single bacterial species in the circulation or in her vaginal or rectal microflora.49

216
Biologic Plausibility

Immunologic studies in humans also support women who suffered a fetal loss, while higher
the direct pathway. In these studies, serum from IgG levels against P gingivalis were associ-
the umbilical cord was collected, and levels of ated with more low–birth weight infants.53
immunoglobulin (Ig) M and IgG were evaluated These seemingly contradictory findings from
against specific periodontal pathogens. The idea seroepidemiologic studies may indicate that
behind these experiments is that fetal exposure low levels of maternal serum IgG antibodies
to periodontal pathogens and/or their byprod- to periodontal bacteria signify inadequate
ucts would result in specific antibody responses protection against the disseminating oral
against these bacteria. Presence of IgM antibod- pathogens, allowing them to translocate to the
ies in the umbilical cord blood would suggest fetoplacental unit and contribute to pregnancy
direct exposure of the fetus in utero, because the complications. Alternatively, elevated mater-
fetus is not immunocompetent and the size of nal serum IgG could indicate either an increase
the IgM molecule prevents its passage through in systemic exposure by oral pathogens or a
the placental barrier, precluding the possibility hyperinflammatory phenotype, which may
that the antibodies are of maternal origin. In predispose these women to an increased fetal
contrast, IgG antibodies against the periodontal inflammatory response and injury.
pathogens would be of maternal origin, indicat- Additional evidence indicating that peri-
ing a chronic exposure of the mother to these odontal pathogens may translocate to the
bacteria or their byproducts. fetoplacental unit derives from studies that
Studies evaluating IgM antibodies against detected bacterial DNA in this compartment.
specific periodontal bacteria have revealed a Indeed, DNA from several periodontal patho-
higher prevalence of umbilical cord IgM sero- gens, such as P gingivalis, Aggregatibacter
positivity for one or more organisms of the red actinomycetemcomitans, F nucleatum, and
complex (P gingivalis, Tannerella forsythia, and Bergeyella species has been identified in amni-
Treponema denticola) or the orange complex otic fluid, placenta, and even neonatal gastric
(Campylobacter rectus, F nucleatum, Pepto- aspirates obtained from complicated pregnan-
streptococcus micros, P intermedia, and P cies.48,54 A study that investigated placental
nigrescens) in women who experienced preterm microbiome profiles using a comparative 16S
or vaginal bleeding.50,51 The concomitant pres- ribosomal RNA–based and whole-genome shot-
ence of these bacteria in the dental plaque of gun metagenomic approach showed that they
pregnant women with pregnancy complica- were most similar to the human oral microbiome
tions implies a hematogenous dissemination rather than to the microbiome of the vagina, the
of these pathogens to the amniotic cavity and gut, or the skin.55 Corroborating findings have
fetal exposure. been obtained from experiments with animal
However, findings from maternal serum IgG models. Specifically, using the chamber model
antibody levels are not always consistent. For in mice, researchers found DNA from P gin-
example, data from a large obstetrics and peri- givalis and C rectus in the maternal liver and
odontal therapy trial indicated that mothers uterus of infected pregnant animals and in the
who delivered preterm had significantly lower placenta of fetuses showing intrauterine growth
serum IgG levels against P gingivalis or other restriction.56,57 However, intravenous infection
red complex bacteria than did mothers who of mice with F nucleatum was restricted inside
delivered at term.52 In another study, the high- the uterus without spreading systemically.58
est rate of PTB occurred in mothers with low Nevertheless, to fully appreciate the signifi-
or no IgG responses to red complex species cance of these findings, it must be remembered
and high IgG responses to orange complex that periodontal pathogens have been detected
species.50 Other studies have found elevated in human amniotic fluid and fetoplacental tis-
serum IgG antibodies against F nucleatum in sues in normal pregnancies as well.59 Hence, it

217
10 Periodontal Infections and Adverse Pregnancy Outcomes

is still unclear which factors determine whether in the inflammatory infiltrate, predominantly by
fetal exposure to periodontal bacteria will con- neutrophils, and in decidual necrosis.63
tribute to pregnancy complications. It is likely C rectus infection in mice induces major
that varying levels of host susceptibility to peri- alterations in the structure of the placenta,
odontal pathogens may render some pregnant as indicated by the decrease in the size of the
women more vulnerable to adverse pregnancy labyrinth.63 This may be partly due to the
outcomes than others. attenuation in the expression of genes related
Another possibility is that variation in vir- to placental and fetal growth, such as placental
ulence properties within a single species may growth factor (PGF) and insulin-like growth
enable some, but not all, strains to colonize the factor 2 (IGF2).64 Because the labyrinth is the
amniotic cavity and induce pregnancy compli- area of the placenta where the exchange of
cations. This notion is further supported by nutrients and waste between the mother and
mechanistic studies. Indeed, in mice, F nuclea- the fetus takes place, its diminished volume
tum was shown to reach the placental blood may result in insufficient nutrition for the fetus,
vessels after hematogenous dissemination resulting in restricted fetal growth and low
and to invade the endothelial cells lining the birth weight. Furthermore, structural damage
vessels, cross the endothelium, proliferate in in the placenta may disrupt the normal blood
the surrounding tissues, and finally spread flow between the fetus and the mother, affect-
to the amniotic fluid.58 Intravenous injection ing maternal blood pressure, and may thus lead
of P gingivalis into pregnant rats resulted in to preeclampsia.
strain-dependent colonization in the placenta.60 Finally, C rectus infection in mice results in
In vitro studies showed that specific C rectus an increased rate of neonatal mortality. In the
strains were able to invade human trophoblast surviving pups, C rectus can be detected in the
cells.61 Thus, it appears that the ability of bac- brain and may induce a local inflammatory
teria to invade the fetoplacental unit may be an response, which is accompanied by increased
important property in the context of induction apoptosis and defects in nerve myelination.63
of adverse pregnancy outcomes. Comparable effects have also been reported
Infection of the placenta with periodontal in humans; neonates exposed to both C rectus
pathogens may result in the induction of local and P gingivalis infection are twice as likely
inflammatory responses, including elevation of to be admitted to the neonatal intensive care
IL-2 and interferon γ and reduction in IL-10.56 unit.65 It has also been established that preterm
These responses are probably mediated by the infants demonstrate an increased risk for
release of major components of the bacterial developing neurodevelopmental, behavioral,
cell wall, such as lipopolysaccharide or outer and learning problems. Thus, fetal exposure
membrane vesicles, and are dependent on the to periodontal pathogens may induce tissue
potency of these virulence factors and the damage to fetal organs, and depending on the
inflammatory profile of the host. In pregnancy, extent of this damage, the fetus may die or the
the innate proinflammatory immune response neonate may demonstrate an increased risk of
is strictly regulated within the uterus to pre- perinatal mortality or morbidity. Detrimental
vent immunologic rejection of the fetal allograft. exposures during fetal development may man-
However, the local increase in proinflammatory ifest themselves later in life.
mediators may disrupt this delicate balance and
elicit an inflammatory burden that may contrib-
ute to preterm rupture of the membranes and Indirect pathway
uterine contraction, which in turn may lead to Patients with periodontal infection and/or
miscarriage or PTB.62 Histologically, this inflam- inflammation show an increased production
matory response is accompanied by an increase of proinflammatory cytokines and mediators

218
Epidemiologic Studies of Maternal Periodontitis and Adverse Pregnancy Outcomes

from periodontal tissues. Once released, these that showed that women giving birth to preterm,
cytokines may diffuse in the GCF or enter low–birth weight babies had significantly greater
the blood circulation and reach the fetopla- mean levels of clinical attachment loss than did
cental unit. Inflammatory cytokines (such mothers with full-term, normal–birth weight
as IL-1β, IL-6, and TNF-α) could then stim- babies,68 a large number of publications have
ulate the production of prostaglandins in the reported on the association between maternal
chorion and exacerbate cervical ripening and periodontal status and pregnancy outcomes. As
uterine contraction, leading to an increased risk expected, a number of studies corroborated and
for PTB. However, limited evidence is available extended these early observations, while others
to date to suggest an association between these failed to detect an association. Several systematic
cytokines in the GCF, serum, or amniotic fluid reviews have been carried out so far on this topic,
and pregnancy complications in women with including one by Ide and Papapanou,69 which
periodontitis.66 was recently updated by Petrini et al70 to include
Another possibility is that proinflammatory additional publications appearing between June
cytokines released in the maternal circulation, 2012 and April 2017. Predefined criteria were
along with disseminated bacteria or bacterial used to evaluate the existing evidence associat-
byproducts from the periodontal tissues, may ing maternal periodontitis, defined broadly and
induce a low-grade systemic inflammation by encompassing a variety of conditions ranging
stimulating hepatic production of acute-phase from gingivitis to aggressive periodontitis, and
reactants, such as CRP and fibrinogen. Elevated three primary pregnancy complications: PTB,
plasma CRP could then augment the inflam- low birth weight, and preeclampsia. The review69
matory responses at the fetoplacental interface discussed in detail a number of features related
through complement activation, tissue damage, to the methodology used and the overall quality
and induction of proinflammatory cytokines. of the studies, all of which could conceivably
Although elevated levels of CRP have been have an impact on the strength of the association
associated with PTB, intrauterine growth between the putative exposure under investiga-
restriction, preeclampsia, and gestational dia- tion (ie, poor maternal periodontal status) and
betes mellitus (GDM), there are only a few the outcome (ie, adverse pregnancy outcomes).
studies evaluating the association between These study features included the type of peri-
pregnancy complications and CRP levels in odontal examination performed (whether it was
pregnant women with periodontitis, and the based on a complete-mouth or partial-mouth
findings are inconsistent.67 Thus, significantly examination); the consistency in the timing of
less evidence is available to support the indirect the examination with respect to gestational
than the direct pathway. age (antepartum or postpartum); whether the
study examiners were blinded with respect to the
occurrence of the particular adverse pregnancy
outcome, excluding the possibility of examina-
Epidemiologic Studies tion bias; and whether additional exposures
of Maternal Periodontitis with known or suspected roles in pregnancy
and Adverse Pregnancy outcome were also considered in the data anal-
yses along with maternal periodontitis. In this
Outcomes context, accounting for the role of confounding
factors (ie, additional exposures associated with
Observational studies both periodontitis and adverse pregnancy out-
comes) was deemed to be critically important.
Since the first case-control study of pregnant or Another issue highlighted in the review
postpartum women published in the mid-1990s was the use of continuous versus categorical

219
10 Periodontal Infections and Adverse Pregnancy Outcomes

definitions of periodontitis: While some stud- that studies that used categorical definitions
ies defined maternal periodontal status using of periodontitis were more likely to detect
continuous variables (eg, mean probing depth statistically significant associations between
and clinical attachment loss, percentage of sites maternal periodontitis and adverse pregnancy
with bleeding on probing), other studies used outcomes than studies that used continuous
categorical (dichotomous) definitions to clas- measures of periodontitis. Likewise, studies
sify the women as having periodontitis or being using a case-control design were more likely
periodontally healthy. Given the absence of to detect associations than the more robust
universally accepted definitions of periodontitis prospective cohort studies. Nevertheless, and
in general and in women of childbearing age despite the aforementioned shortcomings in
in particular, the threshold values used across the existing evidence, the systematic review
studies to define a case of periodontitis varied concluded that there is a positive association
substantially. between poor maternal periodontal status and
Finally, because of the generally low prev- all three adverse pregnancy outcomes exam-
alence of adverse pregnancy outcomes in the ined (PTB, low birth weight, and preeclampsia).
population, the majority of the studies used a With respect to the magnitude of the adverse
case-control design rather than the preferred effect, the review identified it as modest but
longitudinal prospective cohort design. The as independent of other exposures. In other
latter is much better suited for the assessment words, the identified association between peri-
of interactions among different exposures but odontitis and adverse pregnancy outcomes
requires recruitment of considerably larger cannot solely be ascribed to common risk fac-
numbers of participants and is therefore more tors that are also more prevalent in women
demanding logistically. with periodontitis, such as poor socioeconomic
The review reiterated that there is a high status, young maternal age, certain race or eth-
degree of variability among the study popula- nicity characteristics, and tobacco smoking.
tions involved in the available studies as well as Thus, the epidemiologic evidence from the
in the methodologies used for recruitment and association studies available so far is largely
periodontitis assessment. More than 50 contin- consistent with the biologically plausible role
uous parameters and 14 different definitions of of periodontitis as a systemic stressor that was
cases (not specific to pregnancy) were applied described in the first part of this chapter. This
in studies exploring relationships between peri- conclusion is also corroborated by a recently
odontal disease and preterm delivery and/or published overview of systematic reviews72 that
low–birth weight infants.71 Therefore, it is dif- reported positive associations between peri-
ficult to compare the findings of these reports odontal disease and PTB (relative risk [RR]:
with regard to the prevalence of periodontal 1.6; 95% confidence interval [CI]: 1.3–2.0; 17
disease among pregnant women. To illustrate studies, 6,741 participants), low birth weight
the importance of choice of case definitions, (RR: 1.7; 95% CI: 1.3–2.1; 10 studies, 5,693
Manau et al71 calculated the prevalence of peri- participants) and preeclampsia (odds ratio
odontal disease to be between 2.2% and 70.8% [OR]: 2.2; 95% CI: 1.4–3.4; 15 studies, 5,111
when 14 different case definitions were applied participants). Based on the above figures, the
to the same data from 1,296 pregnant women. estimated population-attributable fractions for
Ide and Papapanou69 found that poten- periodontal disease were between 5% and 38%
tially detrimental exposures that may be for PTB, 6% and 41% for low birth weight,
shared between periodontitis and adverse and 10% and 55% for preeclampsia.
pregnancy outcomes did not appear to have A limited number of studies73–76 suggest a
been adequately accounted for in all stud- potential association between maternal peri-
ies. Another observation that emerged was odontitis and GDM, a condition characterized

220
Epidemiologic Studies of Maternal Periodontitis and Adverse Pregnancy Outcomes

by first-onset glucose intolerance and incident similar periodontal treatment after delivery.
hyperglycemia during pregnancy.77 GDM may Table 10-1 summarizes the key features and
reverse postpartum or develop to prevalent type study outcomes of RCTs with a sample size of
2 diabetes mellitus and is associated with sig- at least 100 women.
nificant maternal and fetal morbidity.78 Given The data from these studies have been
that infectious and/or inflammatory stimuli included in meta-analyses in several systematic
have been associated with the pathogenesis reviews94–96 that concluded that nonsurgical
of GDM,78,79 a link between maternal peri- treatment of periodontitis during the second
odontitis and GDM appears to be biologically trimester does not result in a decreased rate
plausible. Two recent systematic reviews80,81 of preterm delivery or have a positive impact
reported statistically significant positive asso- on birth weight. Although individual trials
ciations between the two conditions based on that reported improved gestational outcomes
a limited number of studies but cautioned that in pregnant women allocated to the active
the degree of heterogeneity with respect to clin- intervention arm do exist, the findings of the
ical definitions and analytical methodology in higher-quality studies and the outcomes of the
the underlying studies was high and therefore aggregate analyses are unequivocal and do not
does not allow firm conclusions. support this notion. A thoughtful discussion of
the potential reasons underlying these diverse
findings was published by Michalowicz et al.93
Intervention studies Given that the larger, high-quality RCTs were
Given the positive association between peri- consistent in documenting that nonsurgical
odontitis and adverse pregnancy outcomes, periodontal therapy provided during the second
an obvious question arises that is of major trimester has no positive effect on gestational
interest for both patients and clinicians: Can outcomes, these authors questioned whether
treatment of maternal periodontitis result in there is a need for additional studies of similar
improved gestational outcomes? The public design. Nevertheless, a recent systematic review
health implications of such a possibility are of the effectiveness of periodontal therapy to
obviously profound, because approximately improve adverse pregnancy outcomes97 pointed
half the variance in the prevalence of PTB and out that although periodontal treatment was
low birth weight is unexplained by the cur- found to have no significant effects on PTB or
rently established risk factors. Therefore, even low birth weight overall, data analyses from
if a relatively small proportion of the cases studies in populations with high occurrence
could be ascribed to maternal periodontitis, (≥ 20%) of PTB and low birth weight indi-
and this risk turned out to be modifiable by cated that periodontal treatment seemed to
means of periodontal therapy, the benefits reduce the risk of PTB (OR: 0.42; 95% CI:
would be substantial. 0.24–0.73) and low birth weight (OR: 0.32;
This question is arguably one of the most 95% CI: 0.15–0.67) in these cohorts, although
thoroughly investigated in the field of dental trial sequential analyses showed that firm evi-
medicine: At least 15 randomized controlled dence was not reached.
trials (RCTs) reported thus far have collectively A frequent criticism of the available RCTs is
enrolled in excess of 7,000 pregnant women that the periodontal treatment rendered failed
with periodontitis or gingivitis. Approximately to result in adequate improvement of mater-
half these women were randomized to receive nal periodontal status to the extent needed
scaling and root planing (SRP), occasionally to impact pregnancy outcomes. Although
accompanied by adjunctive antimicrobial this is indeed a possibility, given that peri-
pharmacotherapies, prior to completion of the odontal inflammation was not eliminated in
second trimester, while the other half received most studies, the subgroups of individuals in

221
10 Periodontal Infections and Adverse Pregnancy Outcomes

TABLE 10-1  |  RCTs with a total sample size of > 100 that examined the effect of nonsurgical
periodontal treatment during gestation on preterm birth
Statistically
improved
Randomized Study pregnancy
Study; country N Intervention quality* outcome
López et al82 (2002); Chile 400 SRP + 0.12% chlorhexidine mouthrinse† Low Yes

Jeffcoat et al83 (2003); USA 366 SRP and adjunctive systemic metronidazole High No
in approximately half of the women in the
treatment arm

López et al84 (2005); Chile 870‡ SRP + 0.12% chlorhexidine mouthrinse Low Yes

Michalowicz et al85 (2006); USA 823 SRP High No

Offenbacher et al86 (2006); USA 109 SRP Low Yes

Tarannum and Faizuddin87 (2007); India 200 SRP + 0.2% chlorhexidine mouthrinse Low Yes

Newnham et al88 (2009); Australia 1,082 SRP + 0.12% chlorhexidine mouthrinse High No

Offenbacher et al89 (2009); USA 1,806 SRP High No

Macones et al90 (2010); USA 759 SRP High No

Oliveira et al91 (2011); Brazil 246 SRP Low No

Weidlich et al92 (2013); Brazil 303 SRP High No

*As assessed by Michalowicz et al.93



18% of the women in the test group also received adjunctive systemic antibiotics (amoxicillin and metronidazole).

Includes exclusively women with gingivitis.
SRP, scaling and root planing.

whom a pronounced resolution of periodontal periodontal therapy indicated that interven-


inflammation was achieved did not appear to tions that are not likely to induce bacteremia
experience the best pregnancy outcomes. On the and a rise in systemic inflammation may
other hand, the induction of bacteremia98 and improve pregnancy outcomes.
the increase in systemic inflammation99 shown
to occur in conjunction with instrumentation
of the periodontal tissues may have posed a sig-
nificant challenge to the fetoplacental unit and
Dental and Periodontal
negated the positive effects of the subsequent Therapy During Pregnancy
improvement in gingival inflammation.
It has therefore been argued that the ideal Dentists have a general reluctance to treat
timing of delivery of the periodontal inter- pregnant women, for reasons ranging from the
vention is prior to conception. Obviously, the relative paucity of objective data to support
logistics involved in the conduct of such a trial safety claims regarding specific dental proce-
are complicated, and the concept cannot be dures or use of adjunctive therapeutic agents
tested easily. However, it is noteworthy that to a genuine concern that they may cause harm
a nonrandomized trial100 that tested the effect to the pregnant woman or the fetus. In some
of an antiseptic oral mouthrinse as the sole countries, these attitudes are likely further

222
Dental and Periodontal Therapy During Pregnancy

promoted by a fear of litigation. Conversely, it treatment, tooth extraction, and tooth resto-
is known through surveys in a variety of pop- ration, with or without exposure to diagnostic
ulations that a relative minority of pregnant radiation) during pregnancy in a prospective
women (between 25% and 50%) receive any observational study. The authors followed 210
dental care, including prophylaxis, even when pregnant women exposed to dental local anes-
insured. All types of dental services decrease thetics (approximately half of whom were in
during pregnancy and increase after pregnancy, the first trimester) and compared them with
compared with prepregnancy usage.101 Utiliza- 794 pregnant women who were not exposed
tion of dental services is still lower for pregnant to any teratogens. The rate of major anom-
women of low socioeconomic status102 or alies was not significantly different between
women of certain cultural backgrounds in the groups (4.8% versus 3.3%, P = .3), and
whom the fear of harm to the fetus as a result no differences in the rate of miscarriage, ges-
of dental care is deeply rooted.103 tational age at delivery, or birth weight were
The first study that addressed issues of safety observed.
of dental treatment in pregnant women as part Based on a thorough review of the existing
of an RCT was the Obstetrics and Periodontal scientific evidence, a national consensus state-
Therapy study,104 which randomly assigned 823 ment developed by the national Oral Health
women with periodontitis to receive SRP, either Care During Pregnancy Expert Workgroup106
at 13 to 21 weeks’ gestation or up to 3 months concluded that “Oral health care, including
after delivery. All women were evaluated for use of radiographs, pain medication, and local
essential dental treatment needs, defined as anesthesia, is safe throughout pregnancy.” The
the presence of moderate to severe caries and statement calls for interprofessional collabora-
fractured or abscessed teeth, and 351 women tion between health care providers throughout
received essential dental care prior to the the pregnancy and on delivery to ensure the
completion of the second trimester. The study best possible health of the mother and child.
demonstrated that the rates of serious adverse All health care providers, including physicians
events, defined as spontaneous abortions or and their assistants, nurses, midwives, nurse
stillbirths, fetal or congenital anomalies, and practitioners, dentists, and dental hygienists,
PTBs, were not statistically different between are encouraged to educate themselves regard-
groups of women that received essential dental ing oral health and dental care in pregnancy,
care alone, essential dental care combined with to provide pertinent oral health education, and
nonsurgical periodontal therapy, or no dental to collaborate to ensure that pregnant women
treatment during pregnancy. Thus, this study and new mothers attain and maintain the best
provided high-quality evidence demonstrating possible oral health.
that dental and periodontal treatment that also The national consensus statement also con-
involved use of local anesthetics is safe during tains step-by-step guidance targeting prenatal
the second trimester. These findings have been health care professionals, oral health care pro-
further corroborated by the additional large fessionals, pregnant women, and new mothers.
RCTs mentioned earlier (see Table 10-1), Moreover, it provides a user-friendly table-form
none of which reported a statistically signif- overview of pharmaceutical agents with their
icant higher incidence of adverse pregnancy indications, contraindications, and special con-
outcomes among women who received peri- siderations. Last, the statement provides the
odontal therapy during gestation than among address for the National Maternal and Child
those who received periodontal treatment Oral Health Resource Center’s website with
after delivery. A more recent study105 evalu- links to informative brochures in English and
ated the safety of exposure to local anesthetics in nine additional languages with tips for good
as part of dental care (including endodontic oral health during pregnancy.

223
10 Periodontal Infections and Adverse Pregnancy Outcomes

The statement also introduces the dental periodontal infection and inflammation may
home concept, developed by the American contribute to an increased incidence of PTB,
Association of Public Health Dentistry107 and low birth weight, preeclampsia, and possibly
supported by the American Association of Pedi- GDM. Meta-analyses of the available epide-
atric Dentists (AAPD). The dental home model miologic association studies also support the
delineates an ongoing relationship between the notion that maternal periodontal infections
dentist and the patient, inclusive of all aspects have a negative impact on pregnancy outcomes,
of oral health care, delivered in a comprehen- independent of known confounders. However,
sive, continuously accessible, coordinated, and the strength of this association is rather modest
family-centered way. Pregnant women should and may vary in different populations.
be encouraged to obtain periodontal and The effect of maternal periodontal treatment
restorative treatment to attain and maintain during pregnancy on the incidence of adverse
good oral health throughout their pregnancy pregnancy outcomes has been investigated in
and beyond.108 several high-quality RCTs, all of which have
Because teenage mothers are among those at administered nonsurgical periodontal therapy
the highest risk for preterm delivery, the AAPD prior to the completion of the second trimes-
has developed guidelines on oral health care for ter. The preponderance of evidence from these
pregnant adolescents.109 The advice is aimed at trials indicates that periodontal therapy does
not only the future mother but also the den- not improve pregnancy outcomes. Thus, at first
tal professionals who must be familiar with glance, the findings from the mechanistic and
state statutes that govern consent for care for epidemiologic association studies on the one
a pregnant woman who is not categorized as hand and those from the intervention studies
an adult of legal age. If a pregnant adolescent’s on the other appear to be conflicting. However,
parents are unaware of the pregnancy and a strict and appropriate interpretation of the
state laws require parental consent for dental RCTs is that the particular intervention tested
treatment, the practitioner should encourage (ie, nonsurgical periodontal therapy adminis-
the adolescent to inform her parents so that tered after the first trimester and prior to the
the appropriate informed consent for dental completion of the second trimester) does not
treatment can occur. The AAPD recommends result in improved pregnancy outcomes.
incorporating positive youth development that This conclusion is clearly not synonymous
goes beyond traditional care and suggests that a with the notion that maternal periodontal
strong interpersonal relationship be cultivated infections are unrelated to adverse pregnancy
between the adolescent and her dental care pro- outcomes. What the RCTs suggest is that any
viders. Through positive youth development, increased risk for adverse pregnancy outcomes
the dentist can promote healthy lifestyles, teach documented in the mechanistic and epidemio-
positive patterns of social interaction, and pro- logic association studies cannot be reversed by
vide a safety net in times of need. the particular interventions performed.
The possible effects of different interventions
with respect to timing or intensity have not been
examined. Therefore, it is unknown whether
Conclusion alternative therapeutic approaches may yield
improved outcomes. However, the primary
The association between poor maternal reason RCTs are conducted is to identify the
periodontal status and adverse pregnancy best therapies, to update standards of care, and
outcomes is biologically plausible, and mul- to inform stakeholders responsible for public
tiple lines of evidence support a number of health policy. RCTs are not designed to accept
mechanistic links through which maternal or refute a hypothesized causal relationship

224
Conclusion

between an exposure and an outcome. Instead, the most compliant women or in those whose
they adopt a strategy that minimizes the effects periodontal inflammation was most effectively
of confounders and focus on whether any risk suppressed, recommendations regarding prena-
conferred by the exposure can be modified by tal care policy cannot be based on subgroup
means of the tested intervention. In the case of findings but must adhere to the intent-to-treat
maternal periodontal infections and adverse principle that is the cornerstone of RCTs.
pregnancy outcomes, it may be argued that The possibility that the optimal time point
the central question from a public health to provide periodontal interventions in women
point of view—that is, whether treatment of with periodontitis is prior to conception is cer-
pregnant women with periodontitis will result tainly compatible with current understanding
in improved pregnancy outcomes—has been of the effects of maternal periodontal infection
addressed by several high-quality trials, and and inflammation on the fetoplacental unit.
the answer is no. Indeed, it is known that periodontal therapy
It is probably not meaningful to focus on the results in a substantial, short-term increase in
fact that periodontal treatment in the existing systemic inflammation.110 Furthermore, find-
RCTs improved but clearly did not eliminate ings from an RCT of the effects of periodontal
periodontal inflammation in the treated women treatment on vascular endothelial function
and thus to advocate that more intensive or demonstrated that periodontal therapy results
effective interventions would have resulted in immediate, significant impairment of endo-
in improved outcomes. Indeed, the pragmatic thelial function that is restored to pretreatment
therapeutic approach adopted by the available levels within approximately 2 months after
RCTs (ie, full-mouth SRP followed by regu- intervention, while the beneficial effects of
lar periodontal maintenance until delivery) therapy manifest at 6 months after interven-
is probably what the majority of pregnant tion.111 This timeline may be too extended to
women can tolerate and accommodate within translate into any tangible improvements in the
the busy prenatal period. The improvement in context of pregnancy outcomes. Therefore, res-
periodontal status that is typically achieved by toration of maternal periodontal health prior
this type of intervention in pregnant women to conception makes biologic sense. However,
falls short of eliminating (or even substantially a formal assessment of the potential effects of
suppressing) gingival inflammation. While one periodontal treatment administered prior to
can speculate about what other types of inter- conception in an RCT is logistically difficult,
ventions would have achieved or focus on and such a study may not be available in the
pregnancy outcomes occurring in the subset of near future.
women who were the most compliant with the In the meantime, while it is not justified to
research protocol or those whose periodontal recommend that pregnant women be treated
health improved the most, it is the response periodontally for the sole purpose of improv-
that occurs in the majority of women that is ing pregnancy outcomes, maternal periodontal
interesting from a public health point of view, therapy is safe for the mother and the unborn
not what occurs in small subgroups that may child. It improves oral health and is an indis-
be atypical and may not represent the source pensable part of a holistic approach geared
population. Although from a biologic point of toward the advancement of general health,
view, valuable conclusions can still be drawn risk factor control, and enhancement of
based on the pregnancy outcomes achieved in health-promoting behaviors.

225
10 Periodontal Infections and Adverse Pregnancy Outcomes

Clinical Considerations: What You Can Take Back to Your Practice


What is the effect of pregnancy on periodontal What is the biologic plausibility of the associa-
health?  The significant increase in sex ste- tion between oral health and adverse pregnancy
roids during pregnancy affects the periodon- outcomes?  Oral bacteria and bacterial by-
tal tissues via multiple mechanisms, and their products can gain access to the fetoplacental
net effect is manifested as an increase in the unit via hematogenous spread, where they may
prevalence, extent, and severity of gingival elicit inflammatory responses that contribute to
inflammation (pregnancy gingivitis). These adverse pregnancy outcomes.
changes are infrequently associated with loss of
connective tissue attachment and usually revert Does the delivery of oral health care contribute
after parturition. to an improvement of pregnancy outcomes? 
Well-conducted studies providing periodontal
Are oral infections independent risk factors therapy during the second trimester of preg-
for the development of adverse pregnancy nancy have shown no accompanying improve-
outcomes?  Epidemiologic studies demonstrate ments in adverse pregnancy outcomes.
an association between poor maternal peri-
odontal health and adverse pregnancy out- Is dental care during pregnancy safe for both
comes (preterm birth, low birth weight, and mother and child?  Dental care during pregnan-
preeclampsia). The magnitude of this effect is cy has been shown to be safe for both moth-
moderate but independent of other confound- er and child.
ers. Emerging evidence suggests that maternal
periodontitis may be associated with increased What is the appropriate message to give the
risk for gestational diabetes, but the findings public about the association between oral health
need to be substantiated in larger studies in di- and pregnancy?  Oral health is an integral part
verse populations. of overall health and should be maintained at
all times, pregnancy included.

References 5. Chang HH, Larson J, Blencowe H, et al. Preventing


preterm births: Analysis of trends and potential reduc-
tions with interventions in 39 countries with very high
1. Global Burden of Disease (GBD) 2015 Mortality and human development index. Lancet 2013;381:223–234.
Causes of Death Collaborators. Global, regional, and 6. World Health Organization. Preterm Birth Fact Sheet.
national life expectancy, all-cause mortality, and http://www.who.int/ news-room/ fact-sheets/ detail/
cause-specific mortality for 249 causes of death, 1980– preterm-birth. Updated 19 February 2018. Accessed
2015: A systematic analysis for the Global Burden of 30 August 2018.
Disease Study 2015. Lancet 2016; 388:1459–1544. 7. Blencowe H, Cousens S, Oestergaard MZ, et al.
2. March of Dimes. 2017 Premature Birth Report Card. National, regional, and worldwide estimates of preterm
https://www.marchofdimes.org/materials/Premature birth rates in the year 2010 with time trends since 1990
BirthReportCard-United-States-2017.pdf. Accessed 29 for selected countries: A systematic analysis and impli-
August 2018. cations. Lancet 2012;379:2162–2172.
3. Liu L, Oza S, Hogan D, et al. Global, regional, and 8. Martin JA, Hamilton BE, Osterman MJK. Births in the
national causes of under-5 mortality in 2000–15: An United States, 2016. NCHS Data Brief 2017;(287):1–8.
updated systematic analysis with implications for the 9. Goldenberg RL, Culhane JF, Iams JD, Romero R. Epide-
Sustainable Development Goals. Lancet 2016;388: miology and causes of preterm birth. Lancet 2008; 371:
3027–3035. 75–84.
4. Saigal S, Doyle LW. An overview of mortality and 10. Villar J, Papageorghiou AT, Knight HE, et al. The preterm
sequelae of preterm birth from infancy to adulthood. birth syndrome: A prototype phenotypic classification.
Lancet 2008;371:261–269. Am J Obstet Gynecol 2012;206:119–123.

226
References

11. Croley TE, Miers C. Epithelial changes in the oral mucosa 28. Gürsoy M, Haraldsson G, Hyvönen M, Sorsa T, Paju-
resulting from a variation in hormone stimulus. J Oral kanta R, Könönen E. Does the frequency of Prevotella
Med 1978;33:86–89. intermedia increase during pregnancy? Oral Microbiol
12. Miyagi M, Aoyama H, Morishita M, Iwamoto Y. Effects Immunol 2009;24:299–303.
of sex hormones on chemotaxis of human peripheral 29. Carrillo-de-Albornoz A, Figuero E, Herrera D,
polymorphonuclear leukocytes and monocytes. J Peri- Bascones-Martínez A. Gingival changes during preg-
odontol 1992;63:28–32. nancy: II. Influence of hormonal variations on the
13. Soory M. Targets for steroid hormone mediated actions subgingival biofilm. J Clin Periodontol 2010;37:230–240.
of periodontal pathogens, cytokines and therapeutic 30. Fujiwara N, Tsuruda K, Iwamoto Y, et al. Significant
agents: Some implications on tissue turnover in the increase of oral bacteria in the early pregnancy period
periodontium. Curr Drug Targets 2000;1:309–325. in Japanese women. J Investig Clin Dent 2017;8.
14. Mealey BL, Moritz AJ. Hormonal influences: Effects of 31. Ferris GM. Alteration in female sex hormones: Their
diabetes mellitus and endogenous female sex steroid effect on oral tissues and dental treatment. Compen-
hormones on the periodontium. Periodontol 2000 2003; dium 1993;14:1558–1564,1566.
32:59–81. 32. Kinnby B, Matsson L, Astedt B. Aggravation of gingival
15. Kimura S, Elce JS, Jellinck PH. Immunological relation- inflammatory symptoms during pregnancy associated
ship between peroxidases in eosinophils, uterus and with the concentration of plasminogen activator inhibitor
other tissues of the rat. Biochem J 1983;213:165–169. type 2 (PAI-2) in gingival fluid. J Periodontal Res 1996;
16. Ito I, Hayashi T, Yamada K, Kuzuya M, Naito M, Iguchi 31:271–277.
A. Physiological concentration of estradiol inhibits poly- 33. Raber-Durlacher JE, van Steenbergen TJ, Van der
morphonuclear leukocyte chemotaxis via a receptor Velden U, de Graaff J, Abraham-Inpijn L. Experimental
mediated system. Life Sci 1995;56:2247–2253. gingivitis during pregnancy and post-partum: Clinical,
17. Gallagher JC, Kable WT, Goldgar D. Effect of progestin endocrinological, and microbiological aspects. J Clin
therapy on cortical and trabecular bone: Comparison Periodontol 1994;21:549–558.
with estrogen. Am J Med 1991;90:171–178. 34. Miller WD. The human mouth as a focus of infection.
18. Soory M. Hormonal factors in periodontal disease. Dent Dental Cosmos 1891;33:689–709.
Update 2000;27:380–383. 35. Collins JG, Windley HW 3rd, Arnold RR, Offenbacher
19. Machuca G, Khoshfeiz O, Lacalle JR, Machuca C, S. Effects of a Porphyromonas gingivalis infection on
Bullón P. The influence of general health and inflammatory mediator response and pregnancy
socio-cultural variables on the periodontal condition of outcome in hamsters. Infect Immun 1994; 62:
pregnant women. J Periodontol 1999; 70:779–785. 4356–4361.
20. Gürsoy M, Gürsoy UK, Sorsa T, Pajukanta R, Könönen 36. Haram K, Mortensen JH, Wollen AL. Preterm delivery:
E. High salivary estrogen and risk of developing preg- An overview. Acta Obstet Gynecol Scand 2003; 82:
nancy gingivitis. J Periodontol 2013;84:1281–1289. 687–704.
21. Tilakaratne A, Soory M, Ranasinghe AW, Corea SM, 37. Inglis SR. Biochemical markers predictive of preterm
Ekanayake SL, de Silva M. Periodontal disease status delivery. Infect Dis Obstet Gynecol 1997;5:158–164.
during pregnancy and 3 months post-partum, in a rural 38. Gücer F, Balkanli-Kaplan P, Yüksel M, Yüce MA, Türe
population of Sri-Lankan women. J Clin Periodontol M, Yardim T. Maternal serum tumor necrosis factor-alpha
2000;27:787–792. in patients with preterm labor. J Reprod Med 2001;46:
22. Bieri RA, Adriaens L, Spörri S, Lang NP, Persson GR. 232–236.
Gingival fluid cytokine expression and subgingival bacte- 39. Greig PC, Murtha AP, Jimmerson CJ, Herbert WN,
rial counts during pregnancy and postpartum: A case Roitman-Johnson B, Allen J. Maternal serum
series. Clin Oral Investig 2013;17:19–28. interleukin-6 during pregnancy and during term and
23. Wang PH, Chao HT, Lee WL, Yuan CC, Ng HT. Severe preterm labor. Obstet Gynecol 1997;90:465–469.
bleeding from a pregnancy tumor. A case report. J 40. von Minckwitz G, Grischke EM, Schwab S, et al. Predic-
Reprod Med 1997;42:359–362. tive value of serum interleukin-6 and -8 levels in preterm
24. Kumar PS. Sex and the subgingival microbiome: Do labor or rupture of the membranes. Acta Obstet Gynecol
female sex steroids affect periodontal bacteria? Peri- Scand 2000;79:667–672.
odontol 2000 2013;61:103–124. 41. Pitiphat W, Gillman MW, Joshipura KJ, Williams PL,
25. Paropkari AD, Leblebicioglu B, Christian LM, Kumar PS. Douglass CW, Rich-Edwards JW. Plasma C-reactive
Smoking, pregnancy and the subgingival microbiome. protein in early pregnancy and preterm delivery. Am J
Sci Rep 2016;6:30388. Epidemiol 2005;162:1108–1113.
26. Carrillo-de-Albornoz A, Figuero E, Herrera D, Cuesta P, 42. Tjoa ML, van Vugt JM, Go AT, Blankenstein MA, Oude-
Bascones-Martínez A. Gingival changes during preg- jans CB, van Wijk IJ. Elevated C-reactive protein levels
nancy: III. Impact of clinical, microbiological, during first trimester of pregnancy are indicative of
immunological and socio-demographic factors on gingi- preeclampsia and intrauterine growth restriction. J
val inflammation. J Clin Periodontol 2012;39:272–283. Reprod Immunol 2003;59:29–37.
27. Jensen J, Liljemark W, Bloomquist C. The effect of 43. Romero R, Mazor M. Infection and preterm labor. Clin
female sex hormones on subgingival plaque. J Peri- Obstet Gynecol 1988;31:553–584.
odontol 1981;52:599–602.

227
10 Periodontal Infections and Adverse Pregnancy Outcomes

44. Goldenberg RL, Culhane JF, Johnson DC. Maternal 60. Bélanger M, Reyes L, von Deneen K, Reinhard MK,
infection and adverse fetal and neonatal outcomes. Clin Progulske-Fox A, Brown MB. Colonization of maternal
Perinatol 2005;32:523–559. and fetal tissues by Porphyromonas gingivalis is
45. Han YW, Wang X. Mobile microbiome: Oral bacteria in strain-dependent in a rodent animal model. Am J Obstet
extra-oral infections and inflammation. J Dent Res 2013; Gynecol 2008;199:86.e81–87.
92:485–491. 61. Arce RM, Diaz PI, Barros SP, et al. Characterization of
46. Madianos PN, Bobetsis YA, Offenbacher S. Adverse the invasive and inflammatory traits of oral Campylo-
pregnancy outcomes (APOs) and periodontal disease: bacter rectus in a murine model of fetoplacental growth
Pathogenic mechanisms. J Clin Periodontol 2013; restriction and in trophoblast cultures. J Reprod Immunol
40(suppl 14):S170–S180. 2010;84:145–153.
47. Han YW. Oral health and adverse pregnancy outcomes— 62. Africa CW. Oral colonization of Gram-negative anaer-
What’s next? J Dent Res 2011;90:289–293. obes as a risk factor for preterm delivery. Virulence 2011;
48. Han YW, Ikegami A, Bissada NF, Herbst M, Redline RW, 2:498–508.
Ashmead GG. Transmission of an uncultivated Bergeye- 63. Offenbacher S, Riche EL, Barros SP, Bobetsis YA, Lin
lla strain from the oral cavity to amniotic fluid in a case D, Beck JD. Effects of maternal Campylobacter rectus
of preterm birth. J Clin Microbiol 2006;44:1475–1483. infection on murine placenta, fetal and neonatal survival,
49. Han YW, Fardini Y, Chen C, et al. Term stillbirth caused and brain development. J Periodontol 2005; 76:
by oral Fusobacterium nucleatum. Obstet Gynecol 2133–2143.
2010;115:442–445. 64. Bobetsis YA, Barros SP, Lin DM, Arce RM, Offenbacher
50. Madianos PN, Lieff S, Murtha AP, et al. Maternal peri- S. Altered gene expression in murine placentas in an
odontitis and prematurity. Part II: Maternal infection and infection-induced intrauterine growth restriction model:
fetal exposure. Ann Periodontol 2001;6:175–182. A microarray analysis. J Reprod Immunol 2010; 85:
51. Boggess KA, Moss K, Murtha A, Offenbacher S, Beck 140–148.
JD. Antepartum vaginal bleeding, fetal exposure to oral 65. Jared H, Boggess KA, Moss K, et al. Fetal exposure to
pathogens, and risk for preterm birth at <35 weeks of oral pathogens and subsequent risk for neonatal inten-
gestation. Am J Obstet Gynecol 2006;194:954–960. sive care admission. J Periodontol 2009;80:878–883.
52. Ebersole JL, Novak MJ, Michalowicz BS, et al. Systemic 66. Stadelmann P, Alessandri R, Eick S, Salvi GE, Surbek
immune responses in pregnancy and periodontitis: D, Sculean A. The potential association between gingival
Relationship to pregnancy outcomes in the Obstetrics crevicular fluid inflammatory mediators and adverse
and Periodontal Therapy (OPT) study. J Periodontol pregnancy outcomes: A systematic review. Clin Oral
2009;80:953–960. Investig 2013;17:1453–1463.
53. Dasanayake AP, Boyd D, Madianos PN, Offenbacher 67. Pitiphat W, Joshipura KJ, Rich-Edwards JW, Williams
S, Hills E. The association between Porphyromonas PL, Douglass CW, Gillman MW. Periodontitis and plasma
gingivalis-specific maternal serum IgG and low birth C-reactive protein during pregnancy. J Periodontol
weight. J Periodontol 2001;72:1491–1497. 2006;77:821–825.
54. Ercan E, Eratalay K, Deren O, et al. Evaluation of peri- 68. Offenbacher S, Katz V, Fertik G, et al. Periodontal infec-
odontal pathogens in amniotic fluid and the role of tion as a possible risk factor for preterm low birth weight.
periodontal disease in pre-term birth and low birth J Periodontol 1996;67:1103–1113.
weight. Acta Odontol Scand 2013;71:553–559. 69. Ide M, Papapanou PN. Epidemiology of association
55. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, between maternal periodontal disease and adverse
Versalovic J. The placenta harbors a unique microbiome. pregnancy outcomes—Systematic review. J Clin Peri-
Sci Transl Med 2014;6:237ra65. odontol 2013;40(suppl 14):S181–S194.
56. Lin D, Smith MA, Champagne C, Elter J, Beck J, Offen- 70. Petrini M, Gürsoy M, Gennai S, Graziani F. Biological
bacher S. Porphyromonas gingivalis infection during mechanisms between periodontal diseases and preg-
pregnancy increases maternal tumor necrosis factor nancy complications. A systematic review and meta-
alpha, suppresses maternal interleukin-10, and analysis of epidemiological association between adverse
enhances fetal growth restriction and resorption in mice. pregnancy outcomes and periodontitis: An update of
Infect Immun 2003;71:5156–5162. the review by Ide & Papapanou (2013). Oral Health &
57. Yeo A, Smith MA, Lin D, et al. Campylobacter rectus Pregnancy 2013:1–41. www.efp.org/publications/proj-
mediates growth restriction in pregnant mice. J Peri- ects/ oralhealthandpregnancy/ reports/ review-
odontol 2005;76:551–557. biological-mechanisms-8-6.pdf. Accessed 17 January
58. Han YW, Redline RW, Li M, Yin L, Hill GB, McCormick 2018.
TS. Fusobacterium nucleatum induces premature and 71. Manau C, Echeverria A, Agueda A, Guerrero A, Eche-
term stillbirths in pregnant mice: Implication of oral verria JJ. Periodontal disease definition may determine
bacteria in preterm birth. Infect Immun 2004; 72: the association between periodontitis and pregnancy
2272–2279. outcomes. J Clin Periodontol 2008;35:385–397.
59. Katz J, Chegini N, Shiverick KT, Lamont RJ. Localization 72. Daalderop LA, Wieland BV, Tomsin K, et al. Periodontal
of P. gingivalis in preterm delivery placenta. J Dent Res disease and pregnancy outcomes: Overview of system-
2009;88:575–578. atic reviews. JDR Clin Trans Res 2018;3:10–27.

228
References

73. Novak KF, Taylor GW, Dawson DR, Ferguson JE 2nd, 90. Macones GA, Parry S, Nelson DB, et al. Treatment of
Novak MJ. Periodontitis and gestational diabetes melli- localized periodontal disease in pregnancy does not
tus: Exploring the link in NHANES III. J Public Health reduce the occurrence of preterm birth: Results from
Dent 2006;66:163–168. the Periodontal Infections and Prematurity Study (PIPS).
74. Xiong X, Elkind-Hirsch KE, Vastardis S, Delarosa RL, Am J Obstet Gynecol 2010;202:147.e141–148.
Pridjian G, Buekens P. Periodontal disease is associated 91. Oliveira AM, de Oliveira PA, Cota LO, et al. Periodontal
with gestational diabetes mellitus: A case-control study. therapy and risk for adverse pregnancy outcomes. Clin
J Periodontol 2009;80:1742–1749. Oral Investig 2011;15:609–615.
75. Esteves Lima RP, Miranda Cota LO, Costa FO. Asso- 92. Weidlich P, Moreira CH, Fiorini T, et al. Effect of nonsur-
ciation between periodontitis and gestational diabetes gical periodontal therapy and strict plaque control on
mellitus: A case-control study. J Periodontol 2013;84: preterm/low birth weight: A randomized controlled clin-
1257–1265. ical trial. Clin Oral Investig 2013;17:37–44.
76. Chokwiriyachit A, Dasanayake AP, Suwannarong W, et 93. Michalowicz BS, Gustafsson A, Thumbigere-Math V,
al. Periodontitis and gestational diabetes mellitus in Buhlin K. The effects of periodontal treatment on preg-
non-smoking females. J Periodontol 2013;84:857–862. nancy outcomes. J Clin Periodontol 2013;40(suppl 14):
77. Buchanan TA, Xiang AH. Gestational diabetes mellitus. S195–S208.
J Clin Investig 2005;115:485–491. 94. Polyzos NP, Polyzos IP, Mauri D, et al. Effect of peri-
78. Buchanan TA, Xiang AH, Page KA. Gestational diabetes odontal disease treatment during pregnancy on preterm
mellitus: Risks and management during and after preg- birth incidence: A meta-analysis of randomized trials.
nancy. Nat Rev Endocrinol 2012;8:639–649. Am J Obstet Gynecol 2009;200:225–232.
79. Wolf M, Sauk J, Shah A, et al. Inflammation and glucose 95. Boutin A, Demers S, Roberge S, Roy-Morency A, Chan-
intolerance: A prospective study of gestational diabetes dad F, Bujold E. Treatment of periodontal disease and
mellitus. Diabetes Care 2004;27:21–27. prevention of preterm birth: Systematic review and
80. Esteves Lima RP, Cyrino RM, de Carvalho Dutra B, et meta-analysis. Am J Perinatol 2013;30:537–544.
al. Association between periodontitis and gestational 96. Iheozor-Ejiofor Z, Middleton P, Esposito M, Glenny AM.
diabetes mellitus: Systematic review and meta-analysis. Treating periodontal disease for preventing adverse birth
J Periodontol 2016;87:48–57. outcomes in pregnant women. Cochrane Database Syst
81. Abariga SA, Whitcomb BW. Periodontitis and gestational Rev 2017;6:CD005297.
diabetes mellitus: A systematic review and meta-analysis 97. Schwendicke F, Karimbux N, Allareddy V, Gluud C.
of observational studies. BMC Pregnancy Childbirth Periodontal treatment for preventing adverse pregnancy
2016;16:344. outcomes: A meta- and trial sequential analysis. PLoS
82. López NJ, Smith PC, Gutierrez J. Periodontal therapy One 2015;10:e0129060.
may reduce the risk of preterm low birth weight in 98. Zhang W, Daly CG, Mitchell D, Curtis B. Incidence and
women with periodontal disease: A randomized magnitude of bacteraemia caused by flossing and by
controlled trial. J Periodontol 2002;73:911–924. scaling and root planing. J Clin Periodontol 2013; 40:
83. Jeffcoat MK, Hauth JC, Geurs NC, et al. Periodontal 41–52.
disease and preterm birth: Results of a pilot intervention 99. D’Aiuto F, Parkar M, Tonetti MS. Acute effects of peri-
study. J Periodontol 2003;74:1214–1218. odontal therapy on bio-markers of vascular health. J
84. López NJ, Da Silva I, Ipinza J, Gutiérrez J. Periodontal Clin Periodontol 2007;34:124–129.
therapy reduces the rate of preterm low birth weight in 100. Jeffcoat M, Parry S, Gerlach RW, Doyle MJ. Use of
women with pregnancy-associated gingivitis. J Peri- alcohol-free antimicrobial mouth rinse is associated
odontol 2005;76:2144–2153. with decreased incidence of preterm birth in a high-
85. Michalowicz BS, Hodges JS, DiAngelis AJ, et al. Treat- risk population. Am J Obstet Gynecol 2011; 205:
ment of periodontal disease and the risk of preterm 382.e381–386.
birth. N Engl J Med 2006;355:1885–1894. 101. Jiang P, Bargman EP, Garrett NA, Devries A, Springman
86. Offenbacher S, Lin D, Strauss R, et al. Effects of peri- S, Riggs S. A comparison of dental service use among
odontal therapy during pregnancy on periodontal status, commercially insured women in Minnesota before,
biologic parameters, and pregnancy outcomes: A pilot during and after pregnancy. J Am Dent Assoc 2008;
study. J Periodontol 2006;77:2011–2024. 139:1173–1180.
87. Tarannum F, Faizuddin M. Effect of periodontal therapy 102. Milgrom P, Lee RS, Huebner CE, Conrad DA. Medicaid
on pregnancy outcome in women affected by periodon- reforms in Oregon and suboptimal utilization of dental
titis. J Periodontol 2007;78:2095–2103. care by women of childbearing age. J Am Dent Assoc
88. Newnham JP, Newnham IA, Ball CM, et al. Treatment of 2010;141:688–695.
periodontal disease during pregnancy: A randomized 103. Ressler-Maerlender J, Krishna R, Robison V. Oral health
controlled trial. Obstet Gynecol 2009;114:1239–1248. during pregnancy: Current research. J Womens Health
89. Offenbacher S, Beck JD, Jared HL, et al. Effects of (Larchmt) 2005;14:880–882.
periodontal therapy on rate of preterm delivery: A 104. Michalowicz BS, DiAngelis AJ, Novak MJ, et al. Exam-
randomized controlled trial. Obstet Gynecol 2009;114: ining the safety of dental treatment in pregnant women.
551–559. J Am Dent Assoc 2008;139:685–695.

229
10 Periodontal Infections and Adverse Pregnancy Outcomes

105. Hagai A, Diav-Citrin O, Shechtman S, Ornoy A. Preg- 108. American Academy of Pediatric Dentistry. Council on
nancy outcome after in utero exposure to local Clinical Affairs. Guideline on perinatal oral health care.
anesthetics as part of dental treatment: A prospective Reference Manual 37; 6: 140–145. http: / / www.aapd.
comparative cohort study. J Am Dent Assoc 2015;146: org/media/Policies_Guidelines/G_PerinatalOralHealth-
572–580. Care.pdf. Accessed 30 August 2018.
106. Oral Health Care During Pregnancy Expert Workgroup. 109. American Academy of Pediatric Dentistry. Council on
Oral Health Care During Pregnancy: A National Consen- Clinical Affairs, Committee on the Adolescent. Guideline
sus Statement. http: / / www.mchoralhealth.org/PDFs/ on oral health care for the pregnant adolescent. Refer-
OralHealthPregnancyConsensus.pdf. Accessed 31 ence Manual 37; 6: 159–165. http: / / www.aapd.org/
August 2018. media/Policies_Guidelines/G_Pregnancy.pdf. Accessed
107. Oral Health Policy and Advocacy Committee, Health 30 August 2018.
Home Subcommittee. Toward a Comprehensive Health 110. Graziani F, Cei S, Orlandi M, et al. Acute-phase response
Home: Integrating the Mouth to the Body. http://www. following full-mouth versus quadrant non-surgical peri-
aaphd.org/ assets/ resolution-statements/ aaphd%20 odontal treatment: A randomized clinical trial. J Clin
final%20health%20home%20resolution%20-%20 Periodontol 2015;42:843–852.
last%20revision%20oct%202011.pdf. Accessed 30 111. Tonetti MS, D’Aiuto F, Nibali L, et al. Treatment of peri-
August 2018. odontitis and endothelial function. N Engl J Med 2007;
356:911–920.

230
CHAPTER 11

Oral Infections and Cancer


Dominique S. Michaud, scd

Over the past 50 years, the number of heart periodontal disease (and oral microbiota) and
disease deaths in the United States has rapidly cancer risk is vital to adequately address bias.
declined while the number of cancer deaths has
gradually increased.1 In 2016, 595,930 people
died of cancer and 633,842 died of heart dis-
ease, and in coming years the number of cancer Shared Risk Factors for
deaths is predicted to surpass those from heart
disease.1 Cancers of the lung, breast, prostate,
Periodontal Disease and
and colon (including rectum) are the most Cancer
common cancers and combined account for
45% of all cancer deaths in the United States.2
Implications for causality
Although the total number of cancer deaths Establishing causality between any risk fac-
(absolute) is increasing, cancer death rates have tor and cancer risk is extremely challenging
been slowly decreasing since the 1990s,1 with because of the long latency period often
the most notable decreases occurring for lung, observed between exposure and cancer man-
colorectal, and prostate cancers.2 No changes ifestation. For most known carcinogens, the
in cancer death rates, even increases, have been evidence needed to confirm causality took
observed for other cancers.2 decades to accumulate and required hundreds
There are large racial and ethnic dispari- of studies to address bias (including confound-
ties in cancer incidence and death rates in the ing), temporality, and biologic mechanisms.
United States.2 Disproportionate poverty can Even for cigarette smoking, one of the stron-
explain a portion of these disparities,2 but dif- gest known cancer risk factors, three decades
ferences in behaviors (and risk factor profiles) elapsed between the first reports of an associa-
also account for some of these differences. tion3 and acceptance of causality.4 The biggest
Understanding which risk factors are caus- challenge in evaluating potential causality
ally related to cancer risk is critical to address between periodontal disease and cancer risk
cancer prevention. Established risk factors for is being able to rule out bias that arises from
the most common cancers are discussed in unbalanced prevalence of risk factors in indi-
the following sections. Considering these fac- viduals with periodontal disease and those
tors when studying the relationship between without periodontal disease. Consequently,

231
11 Oral Infections and Cancer

it is important to understand that numerous and periodontal disease development and pro-
behavioral factors and health conditions are gression is strong (for more detail, see chapter
associated with both periodontal disease and 5). Given that obesity appears to be a risk fac-
cancer. tor for periodontal disease as well as for many
cancer types, observational studies examining
associations between periodontal disease and
Smoking, obesity, and type 2 cancer need to account for differences in sub-
diabetes ject adiposity, in addition to smoking history,
to provide unbiased results.
Cigarette smoking is estimated to account Type 2 diabetes has been associated with
for 30% of all cancer deaths and more than several cancers, including pancreatic and co­
80% of lung cancer cases.5 Cigarette smok- lorectal cancers, as well as with periodontal
ing has been causally associated with 13 types disease (see chapter 6). Type 2 diabetes is a
of cancer.6 The strongest associations are complex disease that shares many risk factors
observed for lung cancer, with relative risks with cancer, including obesity, smoking, phys-
(RRs) ranging between 15 and 30, depend- ical inactivity, and diet.10 Although causality
ing on smoking intensity.7 Similarly, smoking has not been established for type 2 diabetes
has been estimated to account for a substan- and cancer,10 it should be considered as a po-
tial proportion of periodontal disease cases tential confounder in analyses on periodontal
(approximately 40%), and RRs for smoking disease and cancer, as it is unclear whether lack
and periodontal disease range between 3 and of adjustment in observational studies will re-
7.8 Former smokers experience higher risks sult in bias.
than never-smokers for both lung cancer and
periodontal disease.7,8 Tobacco smoke is known
to contain numerous carcinogens, but smoking Genetic susceptibility
also influences other important biologic mech- There is some evidence that dental conditions,
anisms, including the immune response, which including periodontal diseases, have a genetic
impacts both the development of cancer and component. The best studies to address this
periodontal disease.8 Consequently, to avoid directly are those that compare periodon-
bias, all observational studies evaluating the tal disease prevalence in monozygotic (MZ)
association between periodontal disease and and dizygotic (DZ) twins. Three large twin
cancer must comprehensively account for cur- studies have reported higher concordance for
rent smoking status, as well as for smoking adult periodontitis in MZ compared to DZ
duration and intensity; unfortunately, not all twins11–13; after adjusting for environmental
published studies on this topic have done so, factors, the heritability component has been
limiting the interpretation of results. estimated to be between 30% and 50%.12,13
Although associations for obesity and cancer In addition, aggressive periodontitis has been
are not as strong in magnitude as those for ciga- reported to aggregate in families and is also
rette smoking, with most RRs ranging between likely to have a strong genetic component.14
1.5 and 3, obesity is estimated to account for A number of genome-wide association stud-
approximately 8% of incident cancer cases and ies (GWASs) have been conducted to identify
has been causally related to cancer at 13 ana- genetic variants associated with periodontal
tomical sites by the International Agency for disease.15–17 However, results from these studies
Research on Cancer (IARC).9 The association have not been reproducible (each identifying
between obesity and periodontal disease is not unique genetic loci). As periodontal disease
causally established, but the evidence for a pos- definitions are complex (and not consistent
itive association between obesity, weight gain, across studies), it is possible that the lack of

232
Biologic Plausibility for an Association Between Periodontal Disease and Cancer

consistency across findings underlie the com- and clearly demonstrated how DNA mutations
plexity of the disease rather than evidence arise from the integration of viruses into human
for a lack of genetic contribution. In a recent DNA. In 1994, the IARC reported that there
GWAS, unique genetic variants were identified was sufficient evidence on Helicobacter pylori
for six periodontal complex traits character- and stomach cancer to classify this bacterium
ized by dysbiotic microbial communities and as a Class I carcinogen. The extensive research
inflammatory markers15; this suggests that dif- that has been conducted on H pylori, partly to
ferent periodontal disease traits have different establish causal associations with cancer, has
underlying molecular mechanisms, thus argu- revealed how bacteria can cause inflammation,
ably making it more difficult to identify genetic induce oxidative stress and DNA damage,
variants when using one clinical definition. and increase cell proliferation.19 Over the past
The possibility that associations reported decade, the number of studies on bacteria and
between periodontal diseases and cancer risk cancer has exploded, with some of the earliest
are explained by shared genetic factors has research arising from interest in microbiota and
been examined in one twin study.18 The authors oral cancer.20,21 Biologic plausibility, impor­
reported an attenuated association between tant in establishing causality, has solidified
periodontal disease and cancer among MZ with mechanistic studies demonstrating how
twins, suggesting genetic factors may partially bacteria can impact the immune response and
explain the observed association for cancer risk interact with human cells to activate signaling
in DZ twins. However, it should be noted that pathways that can lead to carcinogenesis.22
the definition of periodontal disease in the twin Fusobacterium nucleatum and Porphyromonas
study was based on tooth mobility and not gingivalis are two oral bacterial species that
clinical measurements.18 Given that a number have been recently associated with cancer risk.
of genetic variants linked to periodontal dis- The plausible mechanisms for these two species
eases have been identified in genes associated are discussed in more detail in the following
with the immune response,15 it is important sections.
to acknowledge the possibility that shared
genetic susceptibility may account for some of
the associations observed between periodontal Fusobacterium species and cancer
disease and cancer risk in other studies; unfor- Over the past 5 years, there has been tremen-
tunately, adjusting for genetic susceptibility in dous growth in the interest in Fusobacterium
statistical analyses is not easily achieved, as the species and how they may play a role in can-
genetic factors remain mostly unknown. cer, especially colorectal cancer. Fusobacterium,
particularly F nucleatum, has been detected
in a high proportion of colon cancer tissue
(higher than in normal colon tissue).23–25 Mice
Biologic Plausibility for infected with F nucleatum have an increase in
an Association Between tumor growth and tumor burden compared to
Periodontal Disease and those mice not infected.23,24,26,27 Infection with
F nucleatum using mouse models has also been
Cancer shown to promote tumor progression in the
oral cavity.28 In a recent study, antimicrobial
Up through the 1980s, known associations treatment in mice with xenografts of human
between infection and carcinogenesis were primary colorectal adenocarcinoma positive
largely limited to viruses and parasites; decades for Fusobacterium reduced Fusobacterium load
of research on viruses and cancer development and led to a reduction in cancer cell prolifera-
have established numerous causal associations tion and overall tumor growth.25

233
11 Oral Infections and Cancer

In experimental studies, immune response Observational Studies Linking


and signaling pathways have been shown to be
affected by proteins produced by F nucleatum. Periodontal Disease to Cancer
For example, F nucleatum has been shown
to inhibit antitumor immunity by natural All cancers
killer cells through the production of protein
Fap2.29 Bacteria with a mutated Fap2 gene, Despite the heterogeneity in measures used
and thereby inability to produce this protein, to ascertain periodontal disease status, asso-
can no longer inhibit natural killer cells from ciations reported in observational studies
recognizing and removing tumor cells.29 F evaluating risk for all cancers combined have
nucleatum has also been shown to promote been surprisingly consistent. To date, findings
cell proliferation by activating the β -catenin from eight unique cohort studies have been
signaling pathway,30,31 an important pathway published with RRs ranging between 1.14
in colorectal cancer. These data provide sup- and 1.5518,40–46 (only one study was not statis-
port for a causal link between F nucleatum tically significant, and the number of cancers
and cancer, although these results need to be was small in that cohort41). Unfortunately,
evaluated in conjunction with human studies three of these studies did not adjust for smok-
to determine causality. ing status42–44; after removing these studies, the
highest RR is 1.24.46
Associations for periodontal disease and can-
Porphyromonas gingivalis and cer risk among never-smokers have not been as
pancreatic cancer strong; either weak45,47 or null41 associations
were reported in three of the four cohort stud-
One of the keystone bacteria for periodontal ies with separate results on never-smokers.41,47,48
disease, P gingivalis,32 has been implicated in It is unclear if these findings indicate that the
pancreatic carcinogenesis (discussed later) and overall associations are confounded by smok-
has been extensively studied due to its unique ing status or if there is an interaction between
ability to evade the immune response.33 P gin- smoking and periodontal disease status such
givalis has been shown to evade the immune that only smokers are at higher risk of can-
response by modification of its lipid A, the cer with periodontal disease. The differences
biologic core of bacterial lipopolysaccha- observed between smokers and never-smokers
ride, universally recognized by the toll-like (for periodontal disease) may also be due to
receptor 4–myeloid differentiation factor 2 heterogeneity of types of cancers represented
(TLR-4–MD-2) complex.34–36 In addition, P in the smoking groups, given that associations
gingivalis has been shown to alter dendritic between periodontal disease and cancer vary
cell maturation37,38 and promote expansion by cancer type.
of myeloid-derived suppressor cells (MDSCs),39
providing further support for its active role in
immune evasion and suppression. Head and neck cancers
Research on how bacteria may impact can- It is not surprising that interest in the role of
cer is in its infancy, and much more work is oral health (including periodontal disease)
needed to elucidate their role in carcinogenesis. and cancer risk was first raised with oral can-
Examining how the microbiome impacts the cers. A large number of studies have reported
innate immune response and causes inflamma- positive associations between tooth loss,
tion is becoming an important field in cancer oral health, and oral cancers, most of which
research. are case-control studies.49 The quality of these
studies, however, has varied, and it is difficult to

234
Observational Studies Linking Periodontal Disease to Cancer

identify causal associations with retrospective 33% increased risk of lung cancer for indi-
study designs. Case-control studies have also viduals with a history of periodontal disease
addressed the potential link between periodon- compared to those with no periodontal disease
tal disease and head and neck cancers, including (95% confidence interval [CI]: 1.19–1.49).49
oral cancers. Periodontal disease has been mea- In a cohort study using dental examinations
sured with different levels of accuracy; however, for the classification of periodontal disease sta-
more recent studies have included measures of tus that was published after the meta-analysis
bone loss due to periodontitis50,51 and peri- was conducted, a two-fold increase in risk of
odontal pocket depth.52 Overall, findings are lung cancer was observed in participants with
inconsistent, with earlier studies showing baseline severe periodontitis compared to those
weak inverse associations53 and more recent with no or mild periodontitis.47 In that study,
studies reporting strong associations, with a higher risk was also noted among those par-
four- to five-fold higher risks for severe peri- ticipants who were edentulous (hazard ratio
odontitis.50,51 Associations from cohort studies [HR]: 2.60; 95% CI: 1.65–4.08) compared to
have been weaker, with RRs ranging between those with no or mild periodontitis. While the
1.10 and 1.20; however, numbers of cases were association persisted among never-smokers, the
small overall, and thus analyses could not be number of cases was small, and no associations
done on oral cancers separately.44,45,48 were observed among black participants.47
Given that head and neck cancers have Figure 11-1 provides the meta-analysis for all
different causes, with human papillomavirus cohort studies (including the newest study).
(HPV)–positive cancers forming a distinct Results for associations between periodon-
group, there is growing interest in the rela- tal disease and lung cancer have varied among
tionship between periodontal disease and never-smokers (with null results in two cohort
persistence of HPV infections in the oral cav- studies48,57), but particularly interesting is the
ity. HPV has been identified in 26% of gingival possibility that smokers may be at greater
biopsies of subjects with periodontal pockets, risk of developing lung cancer if they have
suggesting that periodontal disease may play periodontal disease. An interaction between
a role in persistence of HPV infection.54 One smoking and periodontal disease was observed
study reported higher HPV-positive oral tumors for women with higher smoking intensity and
among individuals with periodontal disease.55 periodontal disease compared to those with
Moreover, a recent study reported that His- higher smoking dose and no periodontitis in
panic individuals (with no history of cancer) the Women’s Health Initiative (WHI) Obser-
with severe periodontal disease had a close to vational Study cohort (HR: 1.29; 95% CI:
three-fold higher risk of having HPV-positive 1.95–3.87).57 The number of missing teeth has
oral saliva than those without periodontal also been associated with a higher risk of lung
disease.56 While the direction of association cancer in two out of four cohort studies.40,43,58,59
cannot be determined for HPV and periodon-
tal disease, these findings highlight the need to
stratify results for periodontal disease and head Colorectal cancer
and neck cancer by cancer site. Studies reporting associations between peri-
odontal disease and colorectal cancer risk
have been less consistent and weaker than
Lung cancer those for lung cancer but deserve attention
Some of the strongest associations between because of the known link between inflam-
periodontal disease and cancer have been mation and colorectal cancer, as well as the
observed for lung cancer. A meta-analysis con- findings on F nucleatum (discussed previously).
ducted on smoking-adjusted RRs estimated a Although F nucleatum is not considered a

235
11 Oral Infections and Cancer

Study Participants Adjusted RR (95% CI)


Hujoel, 2003 Both sexes 1.73 (1.01, 2.97)
Michaud, 2008 Male 1.36 (1.15, 1.60)
Arora, 2010 Both sexes 1.41 (0.81, 2.46)
Mai, 2014 Female 1.25 (1.06, 1.48)
Mai, 2016 Female 4.61 (0.55, 38.70)
Michaud, 2018 Both sexes 1.79 (1.21, 2.65)
Overall (I-squared = 0.0%; P = .426) 1.36 (1.22, 1.51)

0.3 0.5 1 2 5 10 20 40

Fig 11-1  |  Meta-analysis of cohort studies on periodontal disease and lung cancer.

keystone pathogen for periodontal disease, it Pancreatic cancer


is commonly found in the oral cavity and may
disseminate more easily to the gut in individu- To date, positive associations between peri-
als with periodontal disease. odontal disease and pancreatic cancer risk or
In a recent meta-analysis, having a history of mortality have been reported in six prospec-
periodontal disease was estimated to increase tive cohort studies18,42,43,47,62,63 (although one
the risk of colorectal cancer by 47% compared study defined periodontal disease using a tooth
to those with no periodontal disease (although mobility measure18). In the first exploratory
this association was not statistically significant; study to examine periodontitis and pancreatic
95% CI: 0.95–2.29).49 Results from the ARIC cancer, individuals with periodontitis at base-
cohort (published after the meta-analysis) line had a higher risk of fatal pancreatic cancer
were of similar magnitude for colorectal can- compared to those with healthy periodontium
cer (HR: 1.51; 95% CI: 0.95–2.42 for severe (RR: 1.77; 95% CI: 0.85–1.85); unfortunately,
periodontitis compared to no or mild periodon- results were not presented with adjustment
titis).47 Moreover, results were stronger among for cigarette smoking.43 A statistically sig-
never-smokers (HR: 2.12; 95% CI: 1.00–4.47 nificant 64% increase in risk of pancreatic
for severe periodontitis versus no or mild peri- cancer was observed in a second study on
odontitis), and edentulous individuals had a periodontal disease, after adjusting for age,
three-fold higher risk of colorectal cancer in the smoking, diabetes, and body mass index.47 In
same cohort.47 Three additional studies have a follow-up analysis of this cohort (with 10
been published since the meta-analysis was additional years of follow-up), never-smokers
conducted; a positive association was reported with periodontal disease at baseline had a 57%
in a retrospective cohort study conducted in higher risk of pancreatic cancer compared to
Taiwan using insurance records of periodontal those with no periodontal disease.48 Similar
treatment (RR: 1.64; 95% CI: 1.50–1.80),60 positive associations were reported in four
but no association was observed in the WHI additional cohort studies with data on peri-
cohort study (RR: 0.98; 95% CI: 0.82–1.17),45 odontitis,18,42,62,63 although smoking was not
and only a modest association for moderate/ adjusted in two of these studies.42,62 Although
severe periodontal disease was noted in the data were not available on periodontal disease
Nurses’ Health Study cohort (RR: 1.22; 95% status, a study conducted in Sweden linking
CI: 0.91–1.63).61 oral health examinations to cancer registries

236
Future Directions and Potential Opportunities for Cancer Prevention or Early Detection

with an average of 28.7 years of follow-up measuring bacterial DNA in saliva in cancer
observed a two-fold increase in pancreatic can- patients,67–69 the cohort analysis was unique in
cer risk among individuals who were reported that saliva samples had been obtained up to 10
as having “unacceptable dental plaque” on the years prior to cancer diagnosis.66
baseline dental examination.64 Only one cohort
study to date has reported no association
between periodontal disease (self-reported)
and pancreatic cancer.45 In contrast to reports Future Directions and
on the assaociation of periodontal disease and Potential Opportunities for
pancreatic cancer, findings on tooth loss (a pos-
sible marker of past severe periodontitis) and
Cancer Prevention or Early
pancreatic cancer have been inconsistent but Detection
largely null.49
Several studies have examined the rela- New studies conducting research on oral health
tionship between oral bacteria previously and cancer have increased rapidly in the past
associated with periodontal disease and risk of decade and have made substantial contribu-
pancreatic cancer to explore the potential role tions to this field. However, many questions
of bacteria. The association between antibodies remain, including whether periodontal disease
to periodontal pathogens and risk of pancreatic prevention and/or treatment can reduce cancer
cancer was first examined in a large prospec- incidence and mortality, whether periodontal
tive cohort study using blood samples stored disease associations with cancer vary by race/
on over 385,000 men and women at baseline ethnicity, and whether therapeutic opportu-
(ie, prior to cancer diagnosis).65 A greater than nities for cancer treatment may arise from
two-fold increase in risk of pancreatic cancer microbial research. No intervention study has
was reported among those subjects with high been conducted to examine if treatment of peri-
levels of antibodies to a pathogenic strain of odontal disease reduces the risk of cancer; such
P gingivalis (OR: 2.38; 95% CI: 1.16–4.90; a study would require an extremely large sam-
comparing greater than 200 ng/mL versus less ple size and a very long follow-up period (if the
than 200 ng/mL) after adjusting for known risk study were even ethically feasible). An observa-
factors, including smoking.65 In contrast, no tional study using the Taiwan National Health
associations were detected for other oral bac- Insurance system database compared incidence
teria tested in that study. In a separate cohort cancer rates among subjects who received peri-
study, individuals with antibodies to P gingiva- odontal disease treatment to those who did
lis (greater than 69.1 EU compared to less than not receive treatment in the population.70 The
69.1) were associated with a three-fold higher main assumption in this analysis was that the
risk of orodigestive cancer mortality (RR: 3.03; comparison group, randomly selected from the
95% CI: 0.99–9.31); unfortunately, small case Taiwanese population (age- and sex-matched),
numbers did not allow for a separate analysis had a high prevalence of untreated periodontal
on pancreatic cancer.63 In a separate cohort disease; however, the prevalence of periodontal
analysis (combining data from two large pro- disease in Taiwan is estimated to be approx-
spective cohort studies), a 1.6-fold increased imately 20%.71 Consequently, the inverse
risk was reported for the presence of P gin- associations reported in that study, including
givalis taxa measured directly in saliva using for gastrointestinal and lung cancers, should
16S RNA genes, and a two-fold increased risk be interpreted with caution because they do
was observed for higher mean of Aggregati- not directly demonstrate that treatment of peri-
bacter actinomycetemcomitans taxa, another odontal disease reduces cancer risk and also did
periodontal pathogen.66 Unlike other studies not adjust for smoking. Observational studies

237
11 Oral Infections and Cancer

with strong study designs are urgently needed treatment of periodontitis would translate to
to address the critical question of whether pre- a reduction in cancer burden. In animal mod-
vention or treatment for periodontal disease els, oral bacterial infections promote tumor
will impact cancer development, recurrence, growth,23–25 suggesting that these exposures
or mortality. may have a late effect in carcinogenesis, but
Although a number of small intervention it is less clear in humans when these bacteria
studies on periodontal disease suggested that disseminate to the different body sites.
nonsurgical treatment could improve glyce- If the associations with oral infections are
mic control in diabetic patients, findings from indeed causal, prevention of periodontal dis-
a large randomized controlled trial did not ease will likely have a more pronounced impact
confirm the earlier findings.72 The controversy on cancer than treatment, given the long
over the effectiveness of periodontal therapy latency periods seen with cancer. With RRs
on glycemic control in diabetic patients, an between 1.2 and 1.5 for periodontal disease
example where the data were originally very and cancer, the number of preventable can-
promising, should serve as a cautionary note cers from removing periodontal disease as an
when evaluating the potential impact of peri- “exposure” (through prevention of periodontal
odontal therapy on cancer (given the current disease) could be high given that periodontal
paucity of data in this field). Cancer develop- disease is a highly prevalent condition. Improv-
ment, from tumor initiation to diagnosis, takes ing our understanding of the relationship
place over several decades, and exposures that between periodontal disease and other risk
are known to cause cancer often occur decades factors as they relate to cancer risk, as well as
prior to diagnosis. Because at this time we do the identification of potential bacteria that may
not know if oral infections and associated be involved in carcinogenesis, may also provide
immune responses impact cancer initiation or new opportunities for early cancer detection
progression, it is difficult to predict whether (through biomarker discovery).

Clinical Considerations: What You Can Take Back to Your Practice


Is there a biologic plausibility for an associa- Can treatment of oral infections and disorders
tion between oral infections and cancer?  Ex- reduce the risk for the development of cancer? 
perimental studies have provided important Causality has not been clearly established for
clues on how oral bacteria cause inflammation, periodontal disease and cancer; therefore, we
modulate the immune response, and impact cannot be certain at this time that treatment
key cancer pathways. Whether associations be- will reduce the risk of cancer. More research is
tween oral infections and cancer are causal re- needed to provide an answer to this question.
mains to be determined, but biologic plausibil-
ity is strong. What should a dental practitioner inform a pa-
tient about the association between oral infec-
Are oral infections and disorders independent risk tions and cancer?  Dental practitioners should
factors for the development of cancer?  Yes, oral be able to inform patients that there is a pos-
infections and periodontal disease appear to be sibility that periodontal disease increases the
independent risk factors for the development of risk of cancer, particularly oral, lung, colorec-
cancer as associations have remained strong in tal, and pancreatic cancers, but it is too early to
studies that have adjusted for known cancer risk know if periodontal treatment can reduce the
factors. In addition, associations for certain can- risk of these cancers.
cers remain positive among never-smokers, ar-
guing against confounding by smoking.

238
References

References 18. Arora M, Weuve J, Fall K, Pedersen NL, Mucci LA. An


exploration of shared genetic risk factors between peri-
odontal disease and cancers: A prospective co-twin
1. Weir HK, Anderson RN, Coleman King SM, et al. Heart study. Am J Epidemiol 2010;171:253–259.
disease and cancer deaths—Trends and projections in 19. Salama NR, Hartung ML, Müller A. Life in the human
the United States, 1969–2020. Prev Chronic Dis 2016; stomach: Persistence strategies of the bacterial patho-
13: E157. gen Helicobacter pylori. Nature Rev Microbiol 2013;11:
2. Cancer Facts & Figures 2018. Atlanta: American Cancer 385–399.
Society, 2018. 20. Hooper SJ, Crean SJ, Fardy MJ, et al. A molecular
3. Hoffman FL. Cancer and smoking habits. Ann Surg analysis of the bacteria present within oral squamous
1931; 93:50–67. cell carcinoma. J Med Microbiol 2007;56:1651–1659.
4. Parascandola M. Cigarettes and the US Public Health 21. Hooper SJ, Wilson MJ, Crean SJ. Exploring the link
Service in the 1950s. Am J Public Health 2001; 91: between microorganisms and oral cancer: A systematic
196–205. review of the literature. Head Neck 2009; 31:
5. Islami F, Goding Sauer A, Miller KD, et al. Proportion 1228–1239.
and number of cancer cases and deaths attributable to 22. Gagnaire A, Nadel B, Raoult D, Neefjes J, Gorvel JP.
potentially modifiable risk factors in the United States. Collateral damage: Insights into bacterial mechanisms
CA Cancer J Clin 2018;68:31–54. that predispose host cells to cancer. Nat Rev Microbiol
6. Warren GW, Alberg AJ, Kraft AS, Cummings KM. The 2017;15:109–128.
2014 Surgeon General’s report: “The health conse- 23. Kostic AD, Gevers D, Pedamallu CS, et al. Genomic
quences of smoking—50 years of progress”: A paradigm analysis identifies association of Fusobacterium with
shift in cancer care. Cancer 2014;120:1914–1916. colorectal carcinoma. Genome Res 2012;22:292–298.
7. IARC Working Group on the Evaluation of Carcinogenic 24. Castellarin M, Warren RL, Freeman JD, et al. Fusobac-
Risks to Humans. Tobacco smoke and involuntary terium nucleatum infection is prevalent in human
smoking. IARC Monogr Eval Carcinog Risks Hum 2004; colorectal carcinoma. Genome Res 2012;22:299–306.
83:1–1438. 25. Bullman S, Pedamallu CS, Sicinska E, et al. Analysis of
8. Johnson GK, Slach NA. Impact of tobacco use on peri- Fusobacterium persistence and antibiotic response in
odontal status. J Dent Educ 2001;65:313–321. colorectal cancer. Science 2017;358:1443–1448.
9. Colditz GA, Peterson LL. Obesity and cancer: Evidence, 26. Kostic AD, Chun E, Robertson L, et al. Fusobacterium
impact, and future directions. Clin Chem 2018; 64: nucleatum potentiates intestinal tumorigenesis and
154–162. modulates the tumor-immune microenvironment. Cell
10. Giovannucci E, Harlan DM, Archer MC, et al. Diabetes Host Microbe 2013;14:207–215.
and cancer: A consensus report. Diabetes Care 2010; 27. Ahn J, Sinha R, Pei Z, et al. Human gut microbiome and
33:1674–1685. risk for colorectal cancer. J Natl Cancer Inst 2013;105:
11. Corey LA, Nance WE, Hofstede P, Schenkein HA. 1907–1911.
Self-reported periodontal disease in a Virginia twin popu- 28. Binder Gallimidi A, Fischman S, Revach B, et al. Peri-
lation. J Periodontol 1993;64:1205–1208. odontal pathogens Porphyromonas gingivalis and
12. Mucci LA, Björkman L, Douglass CW, Pedersen NL. Fusobacterium nucleatum promote tumor progression
Environmental and heritable factors in the etiology of in an oral-specific chemical carcinogenesis model.
oral diseases—A population-based study of Swedish Oncotarget 2015;6:22613–22623.
twins. J Dent Res 2005;84:800–805. 29. Gur C, Ibrahim Y, Isaacson B, et al. Binding of the Fap2
13. Michalowicz BS, Diehl SR, Gunsolley JC, et al. Evidence protein of Fusobacterium nucleatum to human inhibitory
of a substantial genetic basis for risk of adult periodon- receptor TIGIT protects tumors from immune cell attack.
titis. J Periodontol 2000;71:1699–1707. Immunity 2015;42:344–355.
14. Meng H, Ren X, Tian Y, et al. Genetic study of families 30. Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW.
affected with aggressive periodontitis. Periodontol 2000 Fusobacterium nucleatum promotes colorectal carcino-
2011;56:87–101. genesis by modulating E-cadherin/β-catenin signaling
15. Offenbacher S, Divaris K, Barros SP, et al. Genome-wide via its FadA adhesin. Cell Host Microbe 2013; 14:
association study of biologically informed periodontal 195–206.
complex traits offers novel insights into the genetic basis 31. Chen Y, Peng Y, Yu J, et al. Invasive Fusobacterium
of periodontal disease. Hum Mol Genet 2016; 25: nucleatum activates beta-catenin signaling in colorectal
2113–2129. cancer via a TLR4/P-PAK1 cascade. Oncotarget 2017;
16. Divaris K, Monda KL, North KE, et al. Exploring the genetic 8:31802–31814.
basis of chronic periodontitis: A genome-wide association 32. Hajishengallis G. Periodontitis: From microbial immune
study. Hum Mol Genet 2013;22:2312–2324. subversion to systemic inflammation. Nat Rev Immunol
17. Munz M, Willenborg C, Richter GM, et al. A genome-wide 2015;15:30–44.
association study identifies nucleotide variants at 33. Tribble GD, Kerr JE, Wang BY. Genetic diversity in the
SIGLEC5 and DEFA1A3 as risk loci for periodontitis. oral pathogen Porphyromonas gingivalis: Molecular
Hum Mol Genet 2017;26:2577–2588. mechanisms and biological consequences. Future
Microbiol 2013;8:607–620.

239
11 Oral Infections and Cancer

34. Akira S, Takeda K, Kaisho T. Toll-like receptors: Critical 49. Michaud DS, Fu Z, Shi J, Chung M. Periodontal disease,
proteins linking innate and acquired immunity. Nat Immu- tooth loss, and cancer risk. Epidemiol Rev 2017; 39:
nol 2001;2:675–680. 49–58.
35. Miller SI, Ernst RK, Bader MW. LPS, TLR4 and infectious 50. Tezal M, Sullivan MA, Hyland A, et al. Chronic periodon-
disease diversity. Nat Rev Microbiol 2005;3:36–46. titis and the incidence of head and neck squamous cell
36. Reife RA, Coats SR, Al-Qutub M, et al. Porphyromonas carcinoma. Cancer Epidemiol Biomarkers Prev 2009;
gingivalis lipopolysaccharide lipid A heterogeneity: Differ- 18:2406–2412.
ential activities of tetra- and penta-acylated lipid A 51. Moergel M, Kämmerer P, Kasaj A, et al. Chronic peri-
structures on E-selectin expression and TLR4 recogni- odontitis and its possible association with oral squamous
tion. Cell Microbiol 2006;8:857–868. cell carcinoma—A retrospective case control study.
37. Miles B, Scisci E, Carrion J, Sabino GJ, Genco CA, Head Face Med 2013;9:39.
Cutler CW. Noncanonical dendritic cell differentiation 52. Moraes RC, Dias FL, Figueredo CM, Fischer RG. Asso-
and survival driven by a bacteremic pathogen. J Leukoc ciation between chronic periodontitis and oral/
Biol 2013;94:281–289. oropharyngeal cancer. Braz Dent J 2016;27:261–266.
38. Miles B, Zakhary I, El-Awady A, et al. Secondary 53. Winn DM, Blot WJ, McLaughlin JK, et al. Mouthwash
lymphoid organ homing phenotype of human myeloid use and oral conditions in the risk of oral and pharyngeal
dendritic cells disrupted by an intracellular oral patho- cancer. Cancer Res 1991;51:3044–3047.
gen. Infect Immun 2014;82:101–111. 54. Hormia M, Willberg J, Ruokonen H, Syrjänen S. Marginal
39. Su L, Xu Q, Zhang P, Michalek SM, Katz J. Phenotype periodontium as a potential reservoir of human papillo-
and function of myeloid-derived suppressor cells mavirus in oral mucosa. J Periodontol 2005;76:358–363.
induced by Porphyromonas gingivalis infection. Infect 55. Tezal M, Sullivan Nasca M, Stoler DL, et al. Chronic
Immun 2017;85:e00213-17. periodontitis-human papillomavirus synergy in base of
40. Michaud DS, Liu Y, Meyer M, Giovannucci E, Joshipura tongue cancers. Arch Otolaryngol Head Neck Surg
K. Periodontal disease, tooth loss, and cancer risk in 2009;135:391–396.
male health professionals: A prospective cohort study. 56. Ortiz AP, González D, Vivaldi-Oliver J, et al. Periodontitis
Lancet Oncol 2008;9:550–558. and oral human papillomavirus infection among Hispanic
41. Mai X, LaMonte MJ, Hovey KM, et al. Periodontal adults. Papillomavirus Res 2018;5:128–133.
disease severity and cancer risk in postmenopausal 57. Mai X, LaMonte MJ, Hovey KM, et al. History of peri-
women: The Buffalo OsteoPerio Study. Cancer Causes odontal disease diagnosis and lung cancer incidence
Control 2016;27:217–228. in the Women’s Health Initiative Observational Study.
42. Heikkilä P, But A, Sorsa T, Haukka J. Periodontitis and Cancer Causes Control 2014;25:1045–1053.
cancer mortality: Register-based cohort study of 68,273 58. Ansai T, Takata Y, Yoshida A, et al. Association between
adults in 10-year follow-up. Int J Cancer 2018; 142: tooth loss and orodigestive cancer mortality in an
2244–2253. 80-year-old community-dwelling Japanese population:
43. Hujoel PP, Drangsholt M, Spiekerman C, Weiss NS. An A 12-year prospective study. BMC Public Health 2013;
exploration of the periodontitis-cancer association. Ann 13:814.
Epidemiol 2003;13:312–316. 59. Tu YK, Galobardes B, Smith GD, McCarron P, Jeffreys
44. Chung SD, Tsai MC, Huang CC, Kao LT, Chen CH. A M, Gilthorpe MS. Associations between tooth loss and
population-based study on the associations between mortality patterns in the Glasgow Alumni Cohort. Heart
chronic periodontitis and the risk of cancer. Int J Clin 2007;93:1098–1103.
Oncol 2016;21:219–223. 60. Hu JM, Shen CJ, Chou YC, et al. Risk of colorectal
45. Nwizu NN, Marshall JR, Moysich K, et al. Periodontal cancer in patients with periodontal disease severity: A
disease and incident cancer risk among postmeno- nationwide, population-based cohort study. Int J
pausal women: Results from the Women’s Health Colorectal Dis 2018;33:349–352.
Initiative Observational Cohort. Cancer Epidemiol 61. Momen-Heravi F, Babic A, Tworoger SS, et al. Periodon-
Biomarkers Prev 2017;26:1255–1265. tal disease, tooth loss and colorectal cancer risk: Results
46. Michaud DS, Lu J, Peacock-Villada AY, et al. Periodontal from the Nurses’ Health Study. Int J Cancer 2017;140:
disease assessed using clinical dental measurements 646–652.
and cancer risk in the ARIC study. J Natl Cancer Inst 62. Chang JS, Tsai CR, Chen LT, Shan YS. Investigating the
2018;110:843–854. association between periodontal disease and risk of
47. Michaud DS, Joshipura K, Giovannucci E, Fuchs CS. pancreatic cancer. Pancreas 2016;45:134–141.
A prospective study of periodontal disease and pancre- 63. Ahn J, Segers S, Hayes RB. Periodontal disease,
atic cancer in US male health professionals. J Natl Porphyromonas gingivalis serum antibody levels and
Cancer Inst 2007;99:171–175. orodigestive cancer mortality. Carcinogenesis 2012;33:
48. Michaud DS, Kelsey KT, Papathanasiou E, Genco CA, 1055–1058.
Giovannucci E. Periodontal disease and risk of all 64. Huang J, Roosaar A, Axéll T, Ye W. A prospective cohort
cancers among male never smokers: An updated anal- study on poor oral hygiene and pancreatic cancer risk.
ysis of the Health Professionals Follow-up Study. Ann Int J Cancer 2016;138:340–347.
Oncol 2016;27:941–947.

240
References

65. Michaud DS, Izard J, Wilhelm-Benartzi CS, et al. Plasma 69. Torres PJ, Fletcher EM, Gibbons SM, Bouvet M, Doran
antibodies to oral bacteria and risk of pancreatic cancer KS, Kelley ST. Characterization of the salivary microbi-
in a large European prospective cohort study. Gut 2013; ome in patients with pancreatic cancer. PeerJ 2015;3:
62:1764–1770. e1373.
66. Fan X, Alekseyenko AV, Wu J, et al. Human oral micro- 70. Hwang IM, Sun LM, Lin CL, Lee CF, Kao CH. Periodontal
biome and prospective risk for pancreatic cancer: A disease with treatment reduces subsequent cancer
population-based nested case-control study. Gut 2018; risks. QJM 2014;107:805–812.
67:120–127. 71. Yu HC, Su NY, Huang JY, Lee SS, Chang YC. Trends
67. Olson SH, Satagopan J, Xu Y, et al. The oral microbiota in the prevalence of periodontitis in Taiwan from 1997
in patients with pancreatic cancer, patients with IPMNs, to 2013: A nationwide population-based retrospective
and controls: A pilot study. Cancer Causes Control study. Medicine (Baltimore) 2017;96:e8585.
2017;28:959–969. 72. Engebretson S. Periodontal disease and glycemic
68. Farrell JJ, Zhang L, Zhou H, et al. Variations of oral control in diabetics. Evid Based Dent 2014;15:93–94.
microbiota are associated with pancreatic diseases
including pancreatic cancer. Gut 2012;61:582–588.

241
CHAPTER 12

Oral Manifestations of
Systemic Diseases
Alessandro Villa, dds, phd, mph
Sook-Bin Woo, dmd, mmsc

The general dentist in daily practice encounters is in a unique position to facilitate an early
a variety of oral mucosal disorders, most of diagnosis. Early diagnosis and management
which are straightforward and easy to diag- can often diminish the morbidity associated
nose. However, even a banal-appearing lesion with a systemic disease, improve well-being,
may be a manifestation of systemic disease or and reduce health care costs.
side effect of treatment for systemic disease. This chapter presents eight common oral
Moreover, the oral signs may be the first pre- conditions (Box 12-1) that are encountered
sentation of a systemic disorder, and the dentist in routine general dental practice. For each

Box 12-1  |  Common oral conditions that are encountered in routine general dental practice

Oral ulcers Gingivitis –– Heavy metal pigmentation


• Traumatic ulcer • Conventional gingivitis –– Others
• Recurrent aphthous ulcer/ • Desquamative gingivitis and • Intrinsic pigmentation
stomatitis hypersensitivity reactions –– Melanin
• Aphthous-like ulcers from • HIV/AIDS-associated linear –– Blood pigment
systemic disease gingivitis and necrotizing Poorly or nonhealing extraction
• Drug-induced oral ulcers ulcerative gingivitis sockets and osteonecrosis
• Viral infections Diffuse gingival hyperplasia • Dry socket
• Erythema multiforme • Drug-induced gingival hyperplasia • Infection
• Deep fungal infections • Acute leukemia • Osteoradionecrosis
• Vesiculobullous disease • Gingival fibromatosis • Drug-induced osteonecrosis
White/red lesions • Destructive membranous • Metastatic tumor to the jaw
• Frictional/factitial keratosis periodontal disease Xerostomia and hyposalivation
• Leukoplakia Pigmented lesions • Hyposalivation associated with
• Oral lichen planus/lichenoid • Extrinsic pigmentation normal glands
lesions –– Medication-induced • Hyposalivation associated with
• Candidiasis pigmentation damaged glands
• Coated/hairy tongue –– Foods and dentifrices Burning mouth syndrome (oral
• Migratory glossitis –– Amalgam tattoo dysesthesia syndrome)

242
Oral Ulcers

condition, the discussion first presents one or


two “most likely” diagnoses based on the fre-
quency of occurrence and then offers a detailed
description of other conditions that may look
similar but represent the oral presentation of
a systemic condition or drug effect. The differ-
ences in history, clinical findings, and diagnostic
tests that may help distinguish between each of
them are also presented.

Oral Ulcers
Fig 12-1 |  Aphthous ulcer with grayish-white fibrin
pseudomembrane and erythematous halo.
An ulcer is a loss of epithelium over an area of
the mucosa covered by a yellow or whitish-gray
fibrin pseudomembrane. Traumatic ulcers are
the most common type, followed by aphthous
ulcers. However, oral ulcers associated with reactions, and immunologic factors; many cases
systemic diseases, use of some medications, are idiopathic.2 However, many systemic con-
infections in immunocompromised patients, ditions may present with aphthous-like ulcers,
vesiculobullous diseases, and immune-mediated and patients must be carefully questioned and
diseases such as erythema multiforme may evaluated for such systemic conditions.
resemble idiopathic aphthous ulcers. RAS generally presents as shallow oval or
round ulcers covered by a grayish-white fibrin
pseudomembrane with an erythematous halo
Traumatic ulcer (Fig 12-1). They occur on the nonkeratinized
Traumatic ulcerations are caused by sharp or mucosa and resolve completely between epi-
broken teeth, rough fillings, acute biting of the sodes. Except for major RAS, they rarely occur
mucosa, and friction from ill-fitting dentures. on the hard palatal mucosa, dorsum of the
As such, the side of the tongue, buccal mucosa, tongue, or attached gingiva. The frequency of
and lower lip are the most frequently affected episodes varies from monthly to only a few
sites, and removal of traumatic factors leads times a year. A minority of women develop
to resolution. minor RAS just before their menses.
There are four main forms: minor, major,
herpetiform, and severe (Table 12-1). Biopsy,
Recurrent aphthous ulcers/ usually performed to rule out other conditions
stomatitis (such as bullosing disorders), shows an ulcer
with nonspecific acute and chronic inflamma-
Idiopathic recurrent aphthous stomatitis (RAS), tion. RAS is not caused by a virus or other
popularly known as canker sores, affects 10% infectious agent.
to 20% of the population and usually first man- Treatment, whether idiopathic or related to
ifests in childhood or adolescence; the frequency systemic disease, depends on the severity and
of episodes usually diminishes with age.1 The frequency of the lesions and centers around
etiology is multifactorial, and some factors pain control, definitive treatment of the ulcers,
include trauma, smoking cessation, hormonal and reducing the frequency of outbreaks.
changes, stress, genetics, some hypersensitivity Patients who experience one or two episodes

243
12 Oral Manifestations of Systemic Diseases

TABLE 12-1  |  Presentation of recurrent aphthous stomatitis

Minor* Major Herpetiform

Size and number of ulcers < 1 cm; usually 1 to 5 ulcers ≥ 1 cm; usually 1 ulcer < 0.5 cm; usually 10 ulcers or more

Duration 7 to 10 d Months 7 to 10 d

Scarring No Yes/No No

*There is a severe variant of minor RAS in which an affected individual is almost never without ulcers.

TABLE 12-2  |  Topical immunosuppressive agents and pain medications

Immunosuppressive agents Instructions

Triamcinolone, 0.1% in methylcellulose paste Apply to affected site three to four times a day; methylcellulose is an effective
(for solitary lesions only) covering agent, although triamcinolone is only of moderate potency.

Clobetasol, 0.05% gel or cream Dry area and apply to affected site three to four times a day (on gauze if
Bethametasone, 0.05% gel or cream appropriate). Do not consume food or drink for 20 min after application.
Fluocinonide, 0.05% gel or cream OR
Fluocinolone, 0.05% gel or cream Place gel in stent and wear for 15 to 30 min two to three times a day (for
Tacrolimus, 0.1% ointment extensive gingival lesions).

Dexamethasone elixir, 0.5 mg/5 mL Dispense 300 mL; swish 5 mL for 3 to 5 min (timed) and spit out three to four
Compounded clobetasol, 0.05% solution times a day. Do not consume food or drink for 20 min after use.
Tacrolimus solution, 5 mg/mL

Intralesional steroid injection with Administer 10 to 15 mg triamcinolone per cm2 of ulceration.


triamcinolone

Pain medications

Benzydamine hydrochloride, 0.15% Swish and spit out 5 to 15 mL three to four times a day when in pain.
Dyclonine hydrochloride, 1% Viscous lidocaine may be mixed in equal volume with diphenhydramine,
Viscous lidocaine, 2% aluminum/magnesium, and bismuth subsalicylate.

Benzocaine, 10% to 20% Apply to the affected area three to four times a day when in pain.

a year probably just require reassurance and incorporated in a mucoadhesive, is sometimes


the use of over-the-counter local anesthetics effective. More frequent or severe outbreaks
(such as 10% benzocaine) or mucoadhesive may be treated with topical corticosteroids and
agents such as methylcellulose paste (Orabase other immunosuppressive agents, and larger,
Paste, Colgate-Palmolive) and polyvinylpyrro- recalcitrant ulcers or those of major RAS may
lidone sodium hyaluronate (Gelclair, Helsinn require intralesional therapy such as triamcin-
Healthcare). Amlexanox (OraDisc A, ULURU), olone injections at a dosage of 10 to 15 mg/
a prescription anti-inflammatory agent cm2 of ulcer size (Table 12-2). The same topical

244
Oral Ulcers

management strategies are used for ulcers asso-


ciated with other disorders. Systemic therapy
for severe outbreaks is with prednisone (usually
at 0.75 to 1 mg/kg) and maintenance therapy
with pentoxifylline, colchicine, azathioprine,
mycophenolate, or thalidomide.

Aphthous-like ulcers from


systemic disease
Several systemic conditions have been associ-
ated with aphthous-like ulcers, and these ulcers
resolve when systemic disease is controlled, Fig 12-2  |  Swelling of the lips in a patient with oro-
facial granulomatosis.
unlike idiopathic RAS for which there is no
specific etiologic agent. Aphthous-like lesions
may occur in patients deficient in folic acid,
iron, or vitamins B1, B2, B6, or B12; these are
readily diagnosed with a blood test, and ulcers
typically resolve or improve with repletion,
although not always. Management of intestinal disease involves
Inflammatory bowel diseases such as Crohn the use of systemic medications (eg, corticoste-
disease and ulcerative colitis often lead to oral roids, salicylates, immunosuppressive agents,
ulcers. Patients experience abdominal pain, and biologic agents such tumor necrosis factor
diarrhea, fever, fatigue, weight loss, and anemia, alpha [TNF-α] inhibitors) or surgical resection.
and these can be elicited by a careful history, Gluten-sensitive enteropathy (also known
although oral findings may precede systemic as celiac disease or celiac sprue) is an autoim-
symptoms. Crohn disease is a granulomatous mune disease of the small intestines caused by
inflammatory disorder of unknown etiology, a hypersensitivity reaction to gliadin, a gluten
although genetic, immunologic, environmental, protein. Almost all patients have the HLA-DQ2
microbial, dietary, and vascular factors have allele and some the HLA-DQ8 allele. A spe-
been implicated. It affects the ileum and large cific association was found with chromosome
bowel in more than 90% of patients, although 15q26, chromosome 5q, and possibly 11q.
any part of the gastrointestinal tract may be Aphthous-like ulcers are seen in 3% to 61%
affected.3 of patients, and dental enamel defects have
Crohn disease is strongly associated with been reported.4 If this condition is suspected,
aphthous ulcers that generally have a linear blood tests for antigliadin, antiendomysial,
appearance if located in the oral vestibule. and anti–tissue transglutaminase antibodies
Other findings include angular cheilitis; swell- should be performed. Signs and symptoms are
ing of the lips (Fig 12-2), face, and gingiva; and relieved on a gluten-free diet. Because of mal-
edema of the oral mucosa resulting in fissuring, absorption after iliectomy, patients may develop
or a “cobblestone” appearance. In addition to aphthous-like ulcers from vitamin B12 deficiency.
aphtheiform ulcers, patients with ulcerative Hypersensitivities to other foods such as eggs
colitis may present with pustular and ulcerative or chocolate may cause aphthous ulcers and a
eruptions in the oral cavity referred to as pyosto- periodic fever syndrome (periodic fever, aph-
matitis vegetans. The pustules often coalesce and thae, pharyngitis, adenopathy [PFAPA]), which
break down, leaving superficial erosions. is usually seen in the first and second decade.5

245
12 Oral Manifestations of Systemic Diseases

Behçet disease is a systemic vasculitis associ- Drug-induced oral ulcers


ated with the HLA-B51 gene. It is characterized
by aphthous-like ulcers, genital ulcers, ocular Drug-induced oral ulcers must be consid-
lesions (especially anterior uveitis), and skin ered in older adults with a sudden onset
lesions (such as erythema nodosum). Cardio- of aphthous-like ulcers because idiopathic
vascular, musculoskeletal, gastrointestinal, and RAS usually has an onset in the first and
neurologic symptoms are variably present. second decade of life. Several medications,
Onset is usually during the third and fourth especially those used in oncology settings,
decade of life, and it is more prevalent in indi- have been associated with ulcers in the oral
viduals from Turkey, Japan, the Middle East, cavity. Targeted therapies and biologic agents
and other Asian countries.6,7 MAGIC (mouth are used increasingly in the treatment of can-
and genital ulcers with inflamed cartilage) cers, autoimmune diseases, and even chronic
syndrome combines features of both relaps- mucosal disease. Dentists should be familiar
ing polychondritis and Behçet disease with with the names of these medications as they
major aphthous ulcers and, rarely, multifocal may encounter patients taking these medica-
neurologic abnormalities. Oral ulcers in these tions. These ulcers do not represent aphthous
patients are treated using the same approach ulcers but are rather considered aphthous-like
used for RAS. Treatment for the disease ulcers because they resolve when the drug is
includes the use of anti–TNF-α biologic agents, withdrawn and recur only when the patient
such as etanercept and infliximab. is rechallenged with the drug. Therefore, a
Cyclic neutropenia is a rare inherited condi- detailed drug history is extremely important
tion due to mutations in the gene for neutrophil when evaluating abrupt-onset oral ulcers.
elastase. These patients usually have a circu- Oral mucositis, ulcerative or otherwise,
lating neutrophil count that fluctuates and still represents a major acute complication
severe neutropenia occurring every 3 weeks of cancer chemotherapy. The most stomato-
and lasting for 3 to 5 days. During these toxic agents include antimetabolites, such as
periods, patients are susceptible to bacterial capecitabine, fluorouracil, and methotrexate;
infections and may report a history of recur- alkylating agents, such as cyclophosphamide
rent fever, lymph node enlargement, malaise, and melphalan; antimitotics, such as paclitaxel,
aphthous-like ulcers, and pharyngitis. Other vinblastine, and vincristine; and anthracycline
oral findings include gingivitis, gingival ulcers, antibiotics, such as daunorubicin, doxorubicin,
and periodontitis. Blood tests during these epi- carboplatin, cisplatin, and hydroxyurea. Muco-
sodes reveal low neutrophil counts. Treatment sal soreness and sensitivity generally begin about
is mostly with topical steroids (see Table 12-1) 3 to 5 days after the first infusion, oral erythema
because of its relapsing-remitting nature. or ulcers develop a few days later, and resolu-
The prevalence of oral complications in tion occurs in approximately 15 days.8 Lesions
patients with HIV/AIDS has decreased since involve mostly nonkeratinized sites and tend to
the advent of antiretroviral therapy. Patients be more diffuse than discrete (Fig 12-3). Ulcers
affected by HIV/AIDS often present with major that affect the keratinized mucosa of the hard
aphthous-like oral ulcers that may affect both palate, gingiva, or dorsal tongue during chemo-
nonkeratinized and keratinized epithelium. All therapy are likely caused by herpes infections
such ulcers in patients with HIV/AIDS must be (discussed later). Topical anesthetics (such as
biopsied to rule out infectious etiologies before viscous lidocaine or morphine rinses for severe
a diagnosis of aphthous-like ulcer can be con- cases) or systemic or narcotic pain medications
firmed. Topical and systemic therapy is often help relieve symptoms until the ulcers heal.
required for resolution. These ulcers resolve if Methotrexate is an immune-modulating
the patient is adequately treated for HIV/AIDS. and antimetabolite drug. Methotrexate at low

246
Oral Ulcers

Fig 12-3  |  Oral mucositis after radiation therapy in- Fig 12-4 |  mTOR inhibitor oral ulcer of the left
volving large ulcers of the tongue. tongue. (Courtesy of Dr Ryan Trowbridge, Division of
Dermatology, Brigham and Women’s Hospital, Bos-
ton, Massachusetts.)

doses is used to control chronic inflammatory often experience a burning sensation while
disorders such as psoriasis and rheumatoid brushing their teeth with a toothpaste contain-
arthritis. At higher doses, methotrexate acts ing SLS. The denaturing effect of SLS on the
as a chemotherapeutic agent in the treatment mucin layer overlying the epithelium may result
of non-Hodgkin lymphoma, leukemia, and in inflammation of the oral mucosa, sloughing,
some solid tumors. Oral lesions associated and ulcers. SLS has also been shown to be a
with methotrexate toxicity are typically char- predisposing factor for RAS, although this is
acterized by dose-dependent erythema and equivocal since this may represent a contact
ulceration (broadly termed mucositis). Folate irritation rather than true RAS.9
supplementation is an effective way to reduce Finally, nonsteroidal anti-inflammatory
such mucosal damage. Treatment of these drugs (NSAIDs) such as piroxicam, indometh-
lesions is with topical corticosteroids and, if acin, and ibuprofen may cause oral ulcers. For
necessary, dose reduction. SLS- or medication-induced lesions, discontin-
Mammalian target of rapamycin (mTOR) uation leads to resolution.
inhibitors are a group of agents prescribed
for the management of soft tissue sarcomas
(temsirolimus and ridaforolimus), advanced Viral infections
metastatic renal cell carcinoma (everolimus), Herpes simplex virus 1 (HSV-1), also known
and immunosuppression following organ as human herpesvirus 1 (HHV-1), is one of
transplantation (sirolimus and everolimus). the most common viral infections that pro-
The side effect of mTOR inhibitors of most duces oral ulcers as part of primary herpetic
significance to dentists is that of discrete gingivostomatitis, recrudescent oral disease,
aphthous-like oral ulcers (Fig 12-4), clini- mucocutaneous orofacial disease, and ocular
cally distinct from the more diffuse ulcers and disease. Occasionally oral lesions are caused
erythema of chemotherapy-induced muco- by HSV-2, a virus historically considered spe-
sitis. This condition has been termed mTOR cific for the anogenital area but no longer so
inhibitor-associated stomatitis and occurs because of changing sexual practices. Primary
within 10 days of start of the drug. Management herpetic gingivostomatitis typically occurs
is with topical or intralesional corticosteroids. in children and young adults, although most
Sodium lauryl sulfate (SLS) is the most com- primary HSV infections are asymptomatic or
monly used detergent in dentifrices. Patients exhibit only very mild symptoms.

247
12 Oral Manifestations of Systemic Diseases

TABLE 12-3  |  Herpes virus infections

Type Clinical presentation Location of ulcers in the oral cavity

HHV-1 (HSV-1) Primary herpetic Painful clusters of 1- to 5-mm oral Keratinized and nonkeratinized mucosa
gingivostomatitis ulcers, flulike symptoms

Recrudescent herpetic Lip prodrome of tingling, followed by Junction of lip vermilion and skin (most
infection in healthy a papule, blister, clustered vesicles, common), intraoral keratinized mucosa
individuals and ulcer, scabbing, and resolution (less common)
on lip vermilion; oral ulcers

Recrudescent herpetic infec- Clustered vesicles, coalescent Keratinized and nonkeratinized mucosa
tion in immunocompromised ulcers, or single aphthous-like oral
patients ulcers

HHV-2 (HSV-2) Same as HSV-1 Usually found on skin and mucosa below
the waist; may also be found in the mouth

HHV-3 (VZV) Primary varicella zoster virus Flulike symptoms and pruritic Skin, intraoral (uncommon)
(chicken pox) papular skin rash followed by vesicle
formation; oral form (uncommon):
0.1- to 0.5-cm painful oral ulcers

Shingles (herpes zoster Skin shingles; oral lesions resemble Facial or oral shingles along distribution
infection) recrudescent HSV; usually unilateral of trigeminal nerve

HHV-4 (EBV) Primary EBV infection Infectious mononucleosis

Recrudescent EBV infection Asymptomatic white papules and Lateral or dorsal tongue (most common
plaques with vertical lines (hairy sites)
leukoplakia) often secondarily
infected with Candida

HHV-5 (CMV) Primary CMV infection Asymptomatic or infectious


mononucleosis–like symptoms or
mild flulike symptoms

Recrudescent CMV infection Mostly in immunocompromised Oral mucosa


patients; large (usually > 1 cm),
painful, penetrating ulcers

HSV, herpes simplex virus; VZV, varicella zoster virus; EBV, Epstein-Barr virus; CMV, cytomegalovirus.

Seroepidemiologic evidence reveals that 50% illness, trauma, emotional stress, or menses
to 60% of adults in the United States have may reactivate the latent virus in the ganglia,
circulating antibodies to the virus, although resulting in viral replication and transport via
few would remember experiencing a primary nerves to the skin or mucosal surfaces.
infection.10 Following an incubation period HSV reactivation results in either asympto­
of 2 to 20 days, some patients will develop matic shedding in bodily fluids or secretions or
flulike symptoms (such as fever, lethargy, and the development of recurrent clinical lesions
headache) 2 or 3 days before the onset of a (recrudescent HSV). Asymptomatic shedding
sore throat and painful oral ulcers. These remains one of the most important causes of
resolve within 1 to 2 weeks. The virus then transmission. Differences in presentation are
establishes latency within the sensory ganglion. presented in Table 12-3, and a typical ulcer
Subsequently, trigger factors such as sunlight, is shown in Fig 12-5. Hydration, nutritional

248
Oral Ulcers

a b

c d

Fig 12-5 |  Recurrent HSV infection in a cancer patient. (a) Large ulcer of the tongue dorsum and coated
tongue; (b) primary HSV infection of the palatal gingiva presenting as clustered ulcers; (c) herpes labialis; and (d)
recrudescent HSV infection presenting as clustered ulcers of the hard palate.

support, topical and systemic pain control, and between the two. Treatment of primary VZV
use of antipyretics are important management is reported in Table 12-4.
strategies. Antiviral therapy reduces conta- Infection with cytomegalovirus (CMV), or
giousness, and this is presented in Table 12-4.11 human herpesvirus 5 (HHV-5), remains one
Primary varicella zoster virus (VZV) infec- of the major complications after solid organ
tion usually occurs in childhood and begins transplantation and may result in severe pul-
with flulike symptoms and a pruritic papu- monary, gastrointestinal, or neurologic disease.
lar rash followed by vesicle formation. These Seroepidemiologic studies show that 60% to
lesions eventually become crusted and scabs 65% of the adult population in the United
fall off in 1 to 3 weeks. In the oral cavity, pri- States has antibodies to CMV.12 Primary CMV
mary VZV infection presents as small (0.1- to infection may be asymptomatic or present with
0.5-cm), painful ulcers that heal in approxi- infectious mononucleosis–like or mild flulike
mately 2 weeks. Reactivation of VZV infection symptoms. Recrudescent CMV in the oral
leads to oral shingles. These may resemble cavity occurs mostly in immunocompromised
recrudescent HSV except they are almost patients as large (usually larger than 1-cm),
always unilateral. A culture differentiates painful ulcers on the oral mucosa.

249
12 Oral Manifestations of Systemic Diseases

TABLE 12-4  |  Antiviral agents for the treatment of oral viral infections

Topical agents Dosage and instructions

Acyclovir, 5% ointment or cream Apply to lesions every 2 h for 7 d


Penciclovir, 1% cream
Docosanol, 10% cream

Systemic agents

Acyclovir, 400- or 800-mg tablet Primary HSV infection:


400 mg three times a day for 7 to 10 d
800 mg two to five times a day for 7 to 10 d
Recurrent HSV infection:
400 mg three times a day for 5 d
800 mg two times a day for 5 d

Valacyclovir, 500- or 1,000-mg Primary HSV infection:


caplet 1 g two times a day for 7 to 10 d
Recurrent HSV infection:
500 mg two times a day for 3 d
Labial HSV infection:
2 g at first sign of recurrence, then 2 g 12 hours later
Prophylaxis (when precipitating event is known):
2 g two times on the day of the dental procedure or event, then 1 g two times the next day
(up to 1 to 3 d after the event or procedure)
Immunosuppressed individuals:
1 g daily
VZV infection:
1 g three times a day for 7 to 14 d

Famciclovir, 125- or 250-mg tablet Primary HSV infection:


250 mg three times a day for 7 to 10 d
Recurrent HSV infection:
125 mg two times a day for 5 d
VZV infection:
500 mg three times a day for 7 to 14 d

VZV, varicella zoster virus.

Because CMV, unlike HSV, infects cells deep and definitive therapy is with ganciclovir, val-
in the connective tissue, a swab culture may not ganciclovir, or cidofovir.
be diagnostic. A biopsy is the most useful tool Epstein-Barr virus (EBV)–associated muco-
for the diagnosis of CMV, and the presence of cutaneous ulcers are large ulcers of the oral
virus can be detected by immuohistochemistry mucosa, gastrointestinal tract, and/or skin
and in situ hybridization techniques. CMV can that occur either in immunocompromised or
also be detected in blood by testing for CMV immunosenescent individuals.13 They resolve
antigen on white blood cells or polymerase on reducing immunosuppression or sponta-
chain reaction studies to detect CMV DNA. neously over time.
Management is directed toward pain control,

250
Oral Ulcers

Coxsackievirus (a group of enteroviruses) or bullseye appearance and typically affect the


infection is associated with two diseases most extremities (especially the palms). The ocular
commonly seen in children: hand, foot, and and genital areas can also be affected.
mouth disease and herpangina. The incuba- EM is self-limiting, lasting from 2 to 4 weeks,
tion period is 3 to 7 days, followed by flulike and lesions heal without scarring. The history
symptoms in some cases. Herpangina is char- and clinical presentation are usually sufficient
acterized by multiple small, coalescent ulcers for diagnosis, and a biopsy is confirmatory. Cul-
on the tonsillar pillars and the soft palate. tures for HSV and a biopsy should be obtained
Hand, foot, and mouth disease has a similar when the clinical presentation is atypical. If M
oral manifestation with cutaneous vesicles of pneumoniae infection is a suspected trigger, the
the hands and feet. Both diseases occur as sea- evaluation includes a chest radiograph, poly-
sonal outbreaks among young schoolchildren. merase chain reaction testing of throat swabs,
The infection is typically self-limiting, resolv- and serologic tests. Therapy includes topical
ing within 7 to 10 days. Coxsackievirus is also and systemic analgesics, skin wound care, and
implicated in lymphonodular pharyngitis that hospital-based supportive care.16
causes a sore throat. Treatment includes pain Stevens-Johnson syndrome is a separate
management, hydration, and soft diet until entity from EM and considered an epidermal
signs and symptoms resolve. necrolytic disease. The more severe form is
toxic epidermal necrolysis, usually associated
with the use of medications such as allopurinol,
Erythema multiforme imidazole antifungals, anticonvulsants, sulfon-
Erythema multiforme (EM) is a self-limiting amides, or NSAIDs. EM major, Stevens-Johnson
mucocutaneous hypersensitivity reaction, syndrome, and toxic epidermal necrolysis man-
usually to an infectious agent, with an acute ifest with similar extensive mucosal ulcers,
onset; however, some cases may be recurrent. erosions, and hemorrhagic crusts on the lips.
From a dermatologic point of view, EM can Skin blistering is always present. Treatment
be divided into minor and major forms. EM includes thalidomide, intravenous immuno-
minor is mainly cutaneous (less than 10% globulin, and supportive care. The mortality
skin involvement), with minimal or no muco- rate is 6% to 20%.17
sal involvement. EM major involves either the
skin with at least one mucosal site or a sin-
gle mucosal site with extensive involvement. Deep fungal infections
However, some cases may involve only the oral Deep fungal infections are invasive and often
cavity. Up to 70% of the cases of EM repre- lead to necrotic ulcers of the oral cavity;
sent a hypersensitivity reaction to a recent HSV systemic dissemination is common.18 These
infection, to asymptomatic HSV reactivation, include aspergillosis and mucormycosis in
to Mycoplasma pneumoniae infection, and less patients with cancer and diabetes mellitus; coc-
commonly, to the use of certain medications.14,15 cidioidomycosis and histoplasmosis, which are
The most common disease-precipitating med- endemic in the southwestern United States and
ications include NSAIDs, sulfonamides, and the Ohio-Mississippi valleys, respectively; and
anticonvulsants. blastomycosis, which is endemic in many areas
Patients with EM may report fever, head- of Central and South America.
ache, malaise, and sore throat a few days or 1 Mucormycosis appears as black, necrotic,
to 2 weeks prior to the onset of lesions. Oral and/or fungating lesions that resem-
lesions present as painful ulcers with crusting ble malignancy; patients with diabetes
of the lips. Skin lesions are symmetric, erythem- mellitus (especially if ketoacidotic), neutrope-
atous, and edematous papules with a target nia, or malignancy and individuals who have

251
12 Oral Manifestations of Systemic Diseases

Diagnosis is made after biopsy and histo-


pathologic examination, as well as culture
and serologic and polymerase chain reaction
studies. Imaging studies are recommended
to assess the extent of systemic involvement.
Treatment requires surgical debridement and
aggressive systemic antifungal therapy for all
such infections.18

Vesiculobullous disease
Fig 12-6 |  Mucous membrane pemphigoid: des-
Similar to the vesicles of HSV, the blisters
quamative gingivitis with erythematous attached gin-
giva. of autoimmune vesiculobullous disease rup-
ture, creating shallow erosions that over time
become covered by fibrin, forming an ulcer.
undergone organ transplantation may develop Necrotic, whitish, ragged epithelial tags from
rhinocerebral mucormycosis, and the mortality the ruptured blister roof may be seen piled up
rate is 60% to 90%.19,20 at the edges of such erosions and ulcers.
Aspergillosis (caused by Aspergillus fumiga- Mucous membrane pemphigoid (MMP)
tum or Aspergillus flavus) is a fungal infection is an autoimmune vesiculobullous disease
characterized by noninvasive and invasive associated with increased frequency of the
forms. Noninvasive aspergillosis affects an HLA-DQB1*0301 allele. It is characterized
immunocompetent host and may present as a by autoreactive antibodies, usually immu-
“fungus ball” (mycetoma) within the maxillary noglobulin G (IgG) and sometimes IgA, that
sinus or a hypersensitivity to the fungus, leading target antigens in the epithelium-attachment
to allergic fungal sinusitis. Invasive infections complex, resulting in subepithelial blistering
tend to affect immunocompromised patients and mucosal fragility.21 MMP may involve the
such as those with leukemia, and the clinical larynx, nose, eyes, and genital mucosa. One
presentation is similar to that of mucormycosis. of the major complications is conjunctival
Vascular involvement leads to necrosis. scarring that can lead to blindness (hence the
The primary infection of histoplasmosis is older term, cicatricial pemphigoid). Scarring
usually mild and presents as a self-limiting may also involve the larynx and esophagus but
pulmonary disease. Immunosuppressed indi- fortunately is uncommon in the oral mucosa.
viduals may develop a disseminated form with The oral mucosal lesions almost always
multiorgan involvement (lung, liver, spleen, affect the gingiva, leading to bright red or des-
meninges, and adrenal glands). Oral lesions are quamative gingivitis (Fig 12-6). This develops
usually present in patients with disseminated when epithelium peels off the gingiva, leaving
histoplasmosis and begin with an erythematous only exposed connective tissue, which is the
area that eventually forms a papule and then basis for a positive Nikolsky sign (slight rub-
an ulcer. bing leads to dislodgement of the epithelium).
Blastomycosis is characterized by a pul- Ulcers may develop on the gingiva and else-
monary infection with mild symptoms. If where in the mouth, such as the palatal, buccal,
untreated, the symptoms worsen, and the and tongue mucosa. These ulcers differ from
microorganism spreads to the mucosa, skin, RAS because they are more diffuse and tend
and bone, forming indurated ulcers. Oral not to heal completely. Biopsy of a perilesional
lesions include verrucous ulcers and radiolu- area for histopathologic examination and direct
cent bone lesions. immunofluorescence studies are essential for a

252
White/Red Lesions

Fig 12-7  |  Custom tray to hold topical immunosup- Fig 12-8 |  Pemphigus vulgaris: ulcers of the left
pressive agent covering the involved gingiva in a pa- buccal mucosa.
tient with MMP presenting as desquamative gingivitis.

definitive diagnosis; direct immunofluorescence also be present (Fig 12-8). The most commonly
shows deposition of IgG, C3, and sometimes affected oral sites are the hard and soft palatal
IgA at the basement membrane zone. mucosa (likely because of physiologic trauma
Management of desquamative gingivitis from swallowing), gingiva (also presenting
includes topical treatment with corticosteroids as desquamative gingivitis), buccal mucosa,
or tacrolimus delivered in a custom tray that and tongue. Diagnosis is established through
covers the involved gingiva (Fig 12-7). This perilesional tissue biopsy for histopathologic
technique may also be used for management evaluation and direct immunofluorescence,
of other gingival inflammatory conditions. which shows intercellular deposition of IgG.
Systemic agents such as prednisone, dap- Oral PV should be treated with a combina-
sone, azathioprine, tetracycline, nicotinamide, tion of prednisone, mycophenolate, and topical
mycophenolate, and cyclophosphamide may steroid therapy. For severe cases and if the skin
be required for management of refractory oral is involved, specialist referral is indicated; in
MMP. When the skin is involved, the diagnosis such cases, treatment generally involves pred-
is usually bullous pemphigoid. nisone, rituximab (a monoclonal antibody),
Pemphigus vulgaris (PV) is an uncommon intravenous immunoglobulin, or extracorpo-
autoimmune vesiculobullous disorder that real photopheresis.
affects mainly older adults and was fatal in the
presteroid era. PV has a predilection for peo-
ple of Ashkenazi Jewish descent, who exhibit
a high frequency of the HLA-DRB*0402 White/Red Lesions
allele whereas non-Jewish persons exhibit
HLA-DQB*0503 more frequently. Autoanti- White papules (less than 5 mm) and plaques
bodies, mostly IgG, are directed toward the (greater than 5 mm) are extremely common
interepithelial cementing substance, leading to in the mouth, and often they have a red or
intraepithelial blistering.22 Oral ulcers, pres- erythematous component. The most common
ent in 95% of patients, are often the first sign is frictional keratosis from chronic chewing or
of PV, with skin lesions developing later. Oral nibbling, which occurs on the buccal mucosa,
lesions are characterized by painful, irregular, lateral tongue, and lower lip mucosa, often
erythematous depressed erosions with whitish bilaterally on the retromolar pad and on eden-
tissue tags at the edges; yellow ulcerations may tulous mucosa.

253
12 Oral Manifestations of Systemic Diseases

concomitant OLP, while only 10% to 15% of


patients with OLP have concomitant cutaneous
lesions.26
OLP may be idiopathic or associated with
a variety of local and systemic conditions.
Amalgam and composite restorations may
cause localized contact lichenoid hyper-
sensitivity reactions, possibly to mercury.
Some cases are caused by a hypersensitivity
to medications such as thiazide diuretics,
angiotensin-converting enzyme inhibitors,
Fig 12-9  |  Leukoplakia of the left buccal mucosa in
beta blockers, gold salts, sulfasalazine, sul-
a patient with active oral chronic GVHD.
fonylureas, and penicillamine. New biologic
agents, such as TNF-α inhibitors, may also
Leukoplakia cause lichen planus–like eruptions.
Patients with hypothyroidism, including
Leukoplakia (and its variants verrucous, Hashimoto thyroiditis, also develop OLP, but it
nodular, and erythroleukoplakia) is the most is unclear whether it is thyroid disease that pre-
common premalignant lesion and is defined disposes to OLP or the medications used to treat
by the World Health Organization as “a white it, such as levothyroxine. Hepatitis C virus infec-
plaque of questionable risk having excluded tion is associated with the development of OLP
(other) known diseases or disorders that carry in individuals living in areas of southern Europe,
no increased risk for cancer.”23 While this is particularly Italy, Portugal, and Spain, and there
generally associated with cigarette smoking, may be a genetic or ethnic predilection.25
it is seen with some frequency in patients who OLP most commonly affects the buccal
have a history of another cancer, are immu- mucosa and tongue bilaterally and can present
nocompromised from systemic disease or use with three distinct forms: reticular/keratotic
of immunosuppressive medications, or who (classic), erosive/erythematous, and ulcerative.
have chronic graft-versus-host disease (GVHD) The reticular/keratotic form, characterized
after treatment with allogeneic hematopoietic by Wickham striae (Fig 12-10), is the most
stem cell transplantation for leukemia or other common form and is often asymptomatic.
bone marrow failure syndromes24 (Fig 12-9). Occasionally patients report discomfort and
Patients with dyskeratosis congenita are also describe the buccal mucosa as “rough,” “thick,”
prone to develop leukoplakia and squamous or “tight.” The erosive/erythematous form
cell carcinoma. typically presents as desquamative gingivitis.
Patients may complain of sensitivity and dis-
comfort while eating acidic, spicy, or crunchy
Oral lichen planus/lichenoid foods. The ulcerative form is the most severe
lesions and presents as shallow ulcerations that have a
yellow fibrin membrane on the surface. Often,
Oral lichen planus (OLP) is a fairly common, patients exhibit a combination of the three
chronic, immune-mediated inflammatory forms at different sites or at different times.
mucocutaneous condition that occurs in 1% to Most patients exhibit characteristic bilateral
2% of adults.25 Middle-aged women are twice and usually symmetric distribution of lesions,
as likely to be affected as men. OLP can affect typically involving the buccal mucosa, dorsum
any mucosal surface. Approximately 70% of and ventral surfaces of the tongue, and/or gin-
individuals with cutaneous disease develop giva. A biopsy specimen should be obtained

254
White/Red Lesions

for histopathologic examination when the


presentation is not typical (such as unilateral
presentation or lack of reticulations).
Treatment is aimed at relieving pain and
resolving or reducing erythema and ulcer-
ations. The primary therapies include topical
corticosteroids and topical tacrolimus (see
Table 12-2). Occasionally systemic cortico-
steroids and immunomodulating agents (such
as the antimalarial drug hydroxychloroquine)
may be necessary. Malignant transformation to
Fig 12-10 |  Oral lichen planus: Typical white lacey
squamous cell carcinoma may occur in approx- reticulations with an ulcer surrounded by an erythem-
imately 0.1% to 0.2% of cases of OLP, and atous halo.
patients should be followed on a regular basis.27
Two other conditions may present with oral
lesions similar to OLP: lupus erythematosus in up to 20% of patients and are similar to
and chronic GVHD. Lupus erythematosus is those seen in SLE.29 A biopsy obtained from
an autoimmune connective tissue disorder with lesional tissue exhibits a positive lupus band
many subtypes characterized by distinct clini- test in 90% of active lesions. Because there is
cal features, serologic findings, and patterns of no systemic involvement, serologic studies are
humoral and cellular autoimmunity. Women not helpful.
are nine times more commonly affected than For both SLE and DLE, topical and systemic
men, and the onset is in young adulthood. Two therapy for oral involvement depends on the
common presentations often associated with severity of the lesions (see Table 12-2). Sys-
oral findings are systemic lupus erythematosus temic treatment includes immunosuppressive
(SLE) and discoid lupus erythematosus (DLE). or immunomodulatory therapy.
Common signs and symptoms of SLE include Chronic GVHD remains a significant com-
weight loss, arthritis, fever, fatigue, and gen- plication of allogeneic hematopoietic cell (bone
eral malaise. Clinically, 30% to 60% of these marrow) transplantation and continues to be
patients may present with an erythematous the leading cause of nonrelapse mortality. The
“butterfly” rash over the malar area. The kid- oral cavity is affected in around 80% of cases,
neys, heart, skin, nervous system, and joints are and mucosal lesions resemble those of OLP and
commonly involved. Often, patients affected are characterized by erythema, reticulation,
by SLE develop secondary Sjögren syndrome, and ulceration.30 Topical therapy is similar to
resulting in dry mouth. Oral lesions develop in that for OLP (see Table 12-2).
up to 44% of patients, and these may be red
with white reticulations (lichenoid) or ulcer-
ative. A biopsy obtained from normal-appearing Candidiasis
mucosa for direct immunofluorescence shows a Oral candidiasis is the most common fungal
positive result for a lupus band (linear deposi- infection encountered in dental patients. Can-
tion of IgG at the basement membrane zone). dida albicans is a commensal organism of the
Serology is positive for antinuclear antibodies oral cavity in approximately 20% to 30% of
(ANA, positive in more than 95% of patients), individuals.31 It is an opportunistic organism,
anti–double-strand DNA antibodies, anti-Smith and candidiasis develops when the normal flora
antibodies, and antiribonucleoprotein.28 is altered, in hyposalivation, or when there is an
DLE is characterized by scaly, atrophic, and immune dysfunction; it also commonly overlies
red plaques on the skin. Oral lesions develop dysplastic lesions.32 Local factors include use

255
12 Oral Manifestations of Systemic Diseases

Fig 12-11  |  Pseudomembranous candidiasis: white, Fig 12-12 |  Coated hairy tongue: hyperplastic fili-
curdy papules and plaques with mild erythema. form papillae forming a matted area.

of topical immunosuppressive agents, hypos- fluconazole, 100 to 200 mg daily for 7 to 10


alivation, wearing of dentures, and smoking. days, is extremely effective because patients
Contributing systemic factors include diabetes usually exhibit good compliance. Dentures
mellitus, anemia, immunosuppression (eg, HIV should be soaked overnight in an antibacterial
or AIDS), antibiotic use, infancy, advanced age, solution such as 0.12% to 2% chlorhexidine
and endocrine dysfunction. digluconate or 3% sodium hypochlorite diluted
Candidal infections present in three main in water (1:10 solution), and they should not
forms: pseudomembranous, erythematous, be worn overnight.
and hyperplastic. Pseudomembranous candi- Finally, candidal infections also cause angu-
diasis (thrush) is characterized by thick, white, lar cheilitis, which manifests as bilateral, bright
cheesy papules and plaques that can be rubbed red erythematous fissures at the corners of the
off, leaving, in some cases, a bleeding area mouth. Angular cheilitis is managed with top-
(Fig 12-11). The erythematous form of can- ical nystatin/triamcinolone or clotrimazole/
didiasis is mainly encountered on the palatal betamethasone cream or ointment. Aggravat-
mucosa under dentures, presenting as diffuse ing factors include poor support of the perioral
erythema demarcated by the shape of the den- musculature from reduced vertical dimension
ture base. Tongue lesions show atrophy of the and hematinic deficiencies.
filiform papillae with erythema and a smooth
tongue surface. Hyperplastic candidiasis is an
uncommon chronic variant that presents as Coated/hairy tongue
white plaques that do not rub off, suggestive Coated/hairy tongue is a benign, painless con-
of leukoplakia. dition characterized by hyperplastic filiform
Oral candidiasis is treated with topical and papillae and the accumulation of keratin on the
systemic antifungal agents. The most com- dorsum (Fig 12-12). It is often mistaken for can-
monly used topical agents include clotrimazole didiasis, but treatment with antifungal agents
troches (10 mg), which are dissolved in the is invariably unsuccessful. A positive culture
mouth, four times a day for 7 to 10 days; and for Candida (normally present in 20% to 30%
nystatin suspension (100,000 U/mL), which of the population as a commensal)31 is indica-
is swished around the mouth in a dose of 5 tive of carriage rather than candidal infection.
mL four times a day. Systemic therapy using The two common etiologic mechanisms are

256
Gingivitis

increased retention and reduced exfoliation of


keratin of the filiform papillae, which become
elongated and “hairy.” Increased retention is
related to dehydration and hypo­salivation so
that there is less watery saliva and proportion-
ally more mucinous, sticky saliva. Precipitating
factors include a recent history of illness and
antibiotic therapy, radiation therapy, smoking,
use of alcoholic mouthrinses, and polyphar-
macy (especially diruretics, antihistamines, and
anticholinergic medications such as for bladder
Fig 12-13  |  Migratory glossitis: white, arcuate lines
control). Poor diets in which meals are com- enclosing erythematous mucosa.
posed primarily of soft rather than coarse foods
(such as fresh fruits and vegetables) also lead
to reduced keratin exfoliation, and therefore
coated tongue is frequently seen in patients
convalescing from illness.
The tongue has a white, coated, matted, or mouthrinse preparation. Topical corticoste-
hairy appearance and may become pigmented roid treatment is usually necessary because of
from food metabolic products of resident bac- the chronic, relapsing-resolving nature of the
teria. Patients may complain of a pasty, stale disorder.
taste in the mouth and even gagging because
the coating tends to localize to the posterior
tongue near the circumvallate papillae. Man-
agement is directed toward hydration, stopping
Gingivitis
use of alcoholic or dehydrating mouthrinses, Conventional gingivitis
smoking cessation, improved diet, and/or gen-
tle brushing or scraping of the tongue two or The most common gingival pathology
three times a day. in dentistry is plaque-induced gingivitis.
Plaque-induced gingivitis is a common inflam-
matory disease of the gingiva resulting from
Migratory glossitis production of inflammatory cytokines in
Migratory glossitis (also known as geographic response to plaque bacterial lipopolysaccha-
tongue, migratory stomatitis, and erythema rides located at the gingival margin. Gingivitis
areata migrans) is a chronic resolving-relapsing is common in adolescents and pregnant women
condition that occurs in 1% to 2% of the because of hormonal changes. Progesterone
population,33 mainly in adults. It presents associated with pregnancy seems to increase the
as well-demarcated, patchy erythematous permeability of gingival blood vessels and ren-
depapillation of the dorsum of the tongue with der the area more sensitive to physical, bacterial,
a raised, linear or ringlike, white peripheral rim and chemical irritants. Conventional gingivitis
(Fig 12-13). Migratory glossitis is associated is characterized by erythema, edema, bleeding
with fissured tongue in 25% of cases as well upon provocation, tenderness, and sensitivity of
as with atopy (history of asthma, eczema, hay the gingiva. Patients who are mouthbreathers
fever, or food intolerance) and psoriasis.34 have a unique pattern of gingivitis, with a red
Some cases are associated with the HLA-Cw6 and swollen anterior facial gingiva.
allele. Management includes pain control with Good oral hygiene and mechanical plaque
viscous lidocaine and diphenhydramine as a removal reduce or eliminate the inflammation,

257
12 Oral Manifestations of Systemic Diseases

thereby allowing gingival tissue to heal.35 Some Plasma cell gingivitis is reversible and resolves
patients may require antibiotic therapy with completely with avoidance of the allergen.
low-dose doxycycline or metronidazole, espe- However, treatment with topical immunosup-
cially if there is underlying periodontitis. pressive agents speeds the healing process. A
patch test may help to identify the causative
agent. However, some hypersensitivity reactions
Desquamative gingivitis and may be of a nonspecific nature, and a biopsy
hypersensitivity reactions may not exhibit sheets of plasma cells.

Desquamative gingivitis is a common chronic


condition of adults with a female predilection, HIV/AIDS-associated linear
most often representing OLP, MMP (discussed gingivitis and necrotizing
earlier), plasma cell gingivitis, and other hyper-
ulcerative gingivitis
sensitivity reactions.36 Other immunobullous
disorders such as PV (discussed earlier), linear HIV/AIDS-associated linear gingivitis is charac-
IgA disease, and epidermolysis bullosa aquisita terized by a 2- to 3-mm linear band of marked
are less common conditions that may also pres- erythema along the gingival margin that does
ent in this fashion, although the last two do not not respond to conventional oral hygiene pro-
present in the oral cavity without concomitant cedures and represents a form of erythematous
skin findings. candidiasis. Treatment is with antifungal medi-
Desquamative gingivitis is characterized by cations together with mechanical debridement.
fiery red, denuded attached gingiva, sometimes Immunocompromised individuals are also at
ulcerated (with a yellow fibrin pseudomem- higher risk of developing necrotizing ulcerative
brane), primarily on the buccal or facial aspect gingivitis.37 This condition presents as ulcer-
(see Fig 12-6). The gingiva is painful, and oral ated and necrotic, edematous and hemorrhagic
hygiene procedures may be challenging. Plaque interdental papillae and marginal gingiva and
accumulation secondary to poor oral hygiene is a result of a polymicrobial infection by spi-
further aggravates the pain, erythema, and pro- rochetes, Fusobacterium species, Treponema
pensity for gingival bleeding. The patient’s diet species, and Prevotella intermedia. These
may also be limited because of pain. Patients patients may also report pain and halitosis.
with desquamative gingivitis respond well to Treatment includes use of antibiotics, daily
topical corticosteroid treatment applied within rinses with chlorhexidine gluconate, and
custom trays. debridement under local anesthesia.
Plasma cell gingivitis is a contact hypersensi-
tivity reaction to contactants such as flavoring
agents (eg, cinnamic aldehyde) in toothpaste,
chewing gum, or candies and presents as des- Diffuse Gingival Hyperplasia
quamative gingivitis. Plasma cell gingivitis may
be accompanied by glossitis and cheilitis; some- Diffuse gingival hyperplasia has been asso-
times the term plasma cell orificial mucositis ciated with multiple factors, including poor
is used because lesions may involve the upper oral hygiene, as a long-term sequela to con-
airway. Examination of a biopsy specimen ventional gingivitis, hormonal changes (such as
reveals sheets of plasma cells that raise the sus- pregnancy and puberty), adverse drug effects,
picion for a plasma cell malignancy, but in situ and hereditary conditions. Gingival hyperpla-
hybridization studies for light chain restriction sia is caused by a proliferation of fibroblasts
show the cells to be polyclonal, typical for an and an accumulation of connective tissue fibers
inflammatory process. and extracellular matrix. Bacterial plaque and

258
Diffuse Gingival Hyperplasia

Fig 12-14 |  Drug-induced gingival hyperplasia: dif- Fig 12-15 |  Mandibular and maxillary gingival en-
fuse dense, fibrotic gingival hyperplasia secondary to largement in a patient with acute myeloid leukemia.
nifedipine and cyclosporine use. (Courtesy of Dr Hani
Mawardi, Harvard School of Dental Medicine, Boston,
Massachusetts.)

hormones, such as progesterone, may exac- sulphamethoxazole and erythromycin. This is


erbate this process. Clinically the gingiva is mediated by decreased cationic influx of folate
overgrown either diffusely or multifocally resulting in a reduction in metalloproteinases
throughout the mouth. When the cause is and collagenases (leading to accumulation of
hereditary or syndromic, the tissue tends to matrix) and/or an increase in fibroblast activity.
be densely fibrotic, while hyperplastic gingiva Not all patients taking these medications
caused by other conditions tends to be edem- develop gingival hyperplasia, and it is more
atous and tender and bleeds easily. Good oral common in patients with poor oral hygiene.39
hygiene practices may resolve this condition if There may be a genetic predisposition, perhaps
it is mild, but once there is significant fibrosis, related to drug metabolism.
surgery is usually required to remove the excess Unlike plaque-induced gingival hyperplasia,
tissue. the drug-induced form tends to be less erythem-
atous, more fibrotic, and less inflamed (Fig
12-14), although inflammation often results
Drug-induced gingival hyperplasia if pseudopockets form. The gingival enlarge-
Gingival hyperplasia is commonly induced ment may resolve on discontinuation of the
by three main classes of drugs: anticonvul- medication if the condition is mild. For more
sants (phenytoin and less commonly sodium severe cases, excision of the hyperplastic tissue
valproate, primidone, vigabatrin, and phe- is required together with maintenance of good
nobarbital), calcium channel blockers (such oral hygiene and daily use of chlorhexidine
as amlodipine, bepridil, diltiazem, felodip- rinses. A low dose of doxycycline may reduce
ine, nicardipine, nifedipine, nimodipine, the risk of recurrence.
nitrendipine, and verapamil), and immuno-
suppressants (such as cyclosporine and rarely
tacrolimus).38 Drug-induced gingival enlarge- Acute leukemia
ment associated with long-term phenytoin use Acute myeloid leukemia is the most serious
occurs in approximately 15% to 50% of cases. systemic condition associated with diffuse gin-
For calcium channel blockers the prevalence gival enlargement. The leukemic cells infiltrate
ranges from 10% to 20%, and for cyclosporine the tissues, resulting in gingival enlargement
the prevalence is approximately 27%. There (Fig 12-15). Bone marrow involvement by leu-
are rare cases associated with trimethoprim kemia reduces the body’s ability to form the

259
12 Oral Manifestations of Systemic Diseases

normal components of blood, such as platelets, The growth is multifocal, diffuse, and nodular
resulting in petechiae and ecchymosis of the or smooth surfaced. The enlargement is slow
mucosa and gingiva and spontaneous bleeding. growing and becomes more prominent during
Lack of red blood cells leads to anemia, fatigue, the eruption of both primary and permanent
and pallor of the mucosa, and poorly func- teeth. The gingival enlargement often leads to
tioning cancerous white cells lead to recurrent impaction and displacement of teeth, diaste-
infections in the mouth that may exacerbate mas, and malocclusion. Patients may also have
periodontal disease. Teeth may become mobile, difficulty speaking and painful mastication.
and suppuration may be noted around the gin- Treatment for both hereditary and syndromic
giva. In addition, systemic symptoms such as gingival hyperplasia includes surgical removal
night sweats, recurrent infections, or lethargy of excess gingival tissue with recontouring.
may be present.
Dentists play an important role in the diag-
nosis of leukemia by taking a careful history, Destructive membranous
performing a biopsy of the gingiva or refer- periodontal disease (ligneous
ring the patient for a biopsy, and obtaining a
gingivitis and periodontitis)
complete blood count to establish a prelimi-
nary diagnosis. Gingival hyperplasia typically Ligneous gingivitis, or destructive membra-
resolves with effective chemotherapy. nous periodontal disease, is a rare condition
characterized by generalized nodular gingival
enlargement with or without ulceration. It is
Hereditary and syndromic gingival associated with plasminogen deficiency that
hyperplasia leads to the formation of fibrin deposits that
accumulate in the connective tissue. Ligne-
Gingival fibromatosis is a diffuse gingival ous gingivitis is seen in approximately one of
fibrous overgrowth that occurs either in com- three patients with severe homozygous plas-
bination with rare syndromes or as an isolated minogen deficiency and is often accompanied
disease from spontaneous mutation. Some by ligneous conjunctivitis. Additional clinical
associated syndromes are Cowden syndrome manifestations include involvement of the
(associated with trichilemmomas, oral muco- mucosa of the female genital tract, nasophar-
sal papillomatosis, acral and palmoplantar ynx, and tracheobronchial tree. Some patients
keratoses, and increased risk for breast and may benefit from topical and systemic cortico-
thyroid carcinoma), Zimmerman-Laband steroids or systemic warfarin.
syndrome (ear, nose, bone, and nail
defects), Murray-Puretic-Drescher syndrome
(juvenile hyaline fibromatosis with nodular/
papular skin lesions), Rutherford syndrome Pigmented Lesions
(corneal dystrophy), and Cross syndrome
(microphthalmia, hypopigmentation, mental Pigmentations of the teeth and oral mucosa
retardation, and athetosis). occur from pigments of either extrinsic or
Hereditary gingival fibromatosis is a rare intrinsic origin.
gingival hereditary condition that usually
develops during childhood and is transmit-
ted through both autosomal-dominant and Extrinsic pigmentation
autosomal-recessive modes. This disease is char- Extrinsic pigmentation is almost always caused
acterized by firm, pink gingival enlargement by local factors such as tobacco products, tea,
that is nonhemorrhagic and asymptomatic. coffee, and chlorhexidine mouthrinse, and

260
Pigmented Lesions

traumatically implanted amalgam or graphite the oral cavity, darkening of the mucosa may
particles cause readily recognizable tattoos. occur because the grayish-black color of the
underlying bone shows through the mucosa.
However, breakdown products of minocycline
Medication-induced pigmentation may be noted within the connective tissue.
Several medications, including antibiotics of the Antimalarials, such as chloroquine, hydroxy-
tetracycline family, antimalarials, oral contra- chloroquine, quinacrine, and amodiaquine,
ceptive agents, antipsychotics (phenothiazines have anti-inflammatory and immunosuppres-
such as chlorpromazine), and chemothera- sive functions. Chloroquine is also widely used
peutic agents may cause oral pigmentation.40 as an adjunct in the treatment of autoimmune
While many chemotherapeutic agents have diseases. Long-term use may cause diffuse
been reported to cause pigmentation, some of grayish-black pigmentation of the palatal
these may represent postinflammatory hyper- mucosa, as does imatinib, a tyrosine kinase
melanosis at sites where mucositis and ulcers inhibitor that is used as a first-line treatment
had developed (such as the buccal mucosa, for chronic myelogenous leukemia. Skin and
tongue, and lips) and should not be consid- nails may also be affected.
ered true medication-induced pigmentation. Estrogens, either alone or in combination
Three mechanisms have been proposed to with progesterone, have also been reported to
explain drug-induced pigmentation. First, drug cause oral pigmentation caused by an increase
metabolites that are pigmented are deposited in the activity of melanogenic enzymes. Oral
in the connective tissue causing pigmenta- involvement most often involves the palatal
tion. Second, the pigmentation is caused by mucosa and facial gingiva. Upon cessation of
breakdown products of the drug that chelate the medication, the oral and cutaneous lesions
with melanin or iron. Third, the drug (or drug (melasma or chlorasma) slowly resolve.
metabolite) stimulates an increase in melanin
production. Diagnosis of drug-induced pig-
mentation is typically made from a history Heavy metal pigmentation
of exposure to medications, as noted above. Poisoning with heavy metals such as lead,
Oral lesions present as diffuse, painless, sym- mercury, silver, platinum, arsenic, or bismuth
metric, bluish-gray macular pigmentations of presents as a dark-colored or black band along
the hard palatal mucosa that may occur con- the gingival margin, likely caused by deposition
comitantly with skin and nail pigmentation. of sulfides of heavy metals within the gingiva.
Tetracycline is a broad-spectrum antibiotic These are mostly of historical interest, and it
used to treat bacterial infections. It chelates is extremely rare to see oral manifestations of
to bone and teeth and results in brown or such heavy metal toxicity now.
gray discoloration, with fluorescence under
ultraviolet or other light sources. Minocycline
is a long-acting compound with powerful Intrinsic pigmentation
anti-inflammatory properties that is widely The two most common pigmented lesions
used to treat dermatologic conditions such as in the oral cavity caused by intrinsic agents
acne, as well as systemic infections and peri- arise from melanin deposition and blood or its
odontal disease. Tetracycline and minocycline breakdown products.
pigmentation typically occurs after long-term
use and involves the skin, thyroid, nails, con-
junctiva, sclera, and oral mucosa; bone and Melanin
teeth developing during the time of adminis- The most common melanotic lesion is the
tration are discolored in a similar fashion. In solitary melanotic macule, which presents

261
12 Oral Manifestations of Systemic Diseases

as a brown to black macule on the lip, pal- “peppered” on the skin around periorificial
atal mucosa, or gingiva. Multiple melanotic sites such as the mouth, nose, and eyes; on the
macules may occur in an idiopathic fash- extremities; and in the oral cavity. Peutz-Jeghers
ion or be related to syndromes such as syndrome is also associated with short bowel
Addison disease and lentigines syndromes syndrome, small intestine intussusception, and
including neurofibromatosis, Peutz-Jeghers anemia. Management consists of surveillance
syndrome, McCune-Albright syndrome, and of intestinal polyps.
Carney complex. McCune-Albright syndrome is characterized
Melanotic macules are seen in Addison dis- by fibrous dysplasia of bone, irregularly shaped
ease (primary adrenocortical insufficiency), a café-au-lait skin macules (resembling the coast
condition where the adrenal cortex does not of Maine) and precocious puberty. Oral mela-
produce enough glucocorticoids and sometimes notic macules may occur in some individuals
mineralocorticoids as well, leading to compen- although these are rare.
satory overproduction of adrenocorticotropic Carney complex is an autosomal-dominant
hormone (ACTH). Most cases are autoimmune, family of disorders characterized by multi-
but it can also be caused by infection or other ple lentigines of the skin and oral mucosa,
etiologies. It is typically seen in middle-aged cardiac and skin myxomas, and endocrine
or young adults, and patients report fatigue, hyperactivity.
dizziness, hypotension, and abdominal pain. Laugier-Hunziker syndrome is a rare
β -melanocyte-stimulating hormone is integral acquired disorder characterized by multiple
to the ACTH molecule, so whenever levels of melanotic macules of the oral mucosa and
ACTH are high, regardless of the etiology, mel- occasionally of the genital mucosa, conjunc-
anosis results. Patients present with generalized tiva, esophagus, and acral surfaces as well as
bronze coloration of the genitalia and skin brown melanotic bands on the nails (melanon-
(especially flexural areas) and sites of trauma. ychia). Lesions may be treated with a laser as
The oral mucosa may exhibit a sudden onset necessary.
of melanotic macules. Diagnosis is made by
observation of clinical signs and symptoms,
confirmation of elevated levels of ACTH, and Blood pigment
the inability to produce cortisol on stimulation Pigmentations may occur from vascular lesions
using synthetic ACTH. or from extravasated blood and their break-
Neurofibromatosis is a group of genetic dis- down products.
orders with autosomal-dominant inheritance;
the most common form is neurofibromatosis Petechiae and ecchymoses. The most common
type 1 (NF-1, von Recklinghausen disease of oral lesions related to blood pigment are those
the skin). NF-1 is characterized by pigmented from trauma, namely petechiae (less than 5
lesions that are usually present at birth. These mm, clustered and pinpoint), ecchymoses, and
lesions appear as freckles and tan to brown hematomas, which tend to be deeper. The onset
café-au-lait macules with smooth edges (resem- of frequent and extensive petechiae and ecchy-
bling the coast of California). Other clinical moses in the mucosa and of the skin raises the
manifestations include oral and skin neurofi- specter of a blood dyscrasia such as leukemia
bromas and Lisch nodules in the eyes. where the platelets are reduced in number and/
Peutz-Jeghers syndrome is a rare autosomal- or function or thrombocytopenia where plate-
dominant disorder characterized by intesti- lets are reduced in number such as from aplastic
nal polyposis and multiple mucocutaneous anemia or adverse effects of medications (such
melanocytic macules. Brown macules are gen- as antiplatelet aggregation medications). A sim-
erally several millimeters wide and typically ple complete blood count usually establishes

262
Poorly Healing or Nonhealing Extraction Sockets and Osteonecrosis

the diagnosis of low platelet count. Coagulop- insipidus, and cardiomyopathy. Brown to gray
athies caused by dysfunction of the intrinsic or diffuse macules of the palatal mucosa and gin-
extrinsic pathway generally lead to deep tissue giva occur in approximately 15% to 20% of
rather than mucosal bleeding. patients.41 Phlebotomy and chelation therapy
are the preferred treatment for these patients.
Vascular lesions. Vascular lesions may lead
to pigmented lesions that appear as small
red, blue, or purple blebs or nodules on the
mucosa that often blanch on pressure. Pregnant
women may develop these lesions (granuloma Poorly Healing or Nonhealing
gravidarum), often on the gingiva. If small, they Extraction Sockets and
usually regress postpartum, but they may also
fibrose and require excision. Vascular lesions Osteonecrosis
may bleed excessively when traumatized and
are readily excised or ablated with a laser. After dental extractions, a blood clot is formed
within 24 to 48 hours. The clot is replaced
Sturge-Weber syndrome. Sturge-Weber syn- with granulation tissue, and bone formation
drome (encephalofacial angiomatosis) is a is well established by 6 to 8 weeks and com-
rare congenital neurocutaneous condition pleted by 6 months. Intact mucosa is noted
characterized by a port-wine stain of the skin, within 2 to 3 weeks. Local factors such as
typically in the ophthalmic and maxillary dis- infection or cemento-osseous dysplasia that
tributions of the trigeminal nerve. Patients leads to bone sclerosis and poor vascularity
present with bluish-purple plaques on the may lead to a poorly healing or nonhealing
buccal mucosa and gingiva on the same side socket, but several systemic conditions must
as the skin lesions. These represent hamarto- also be considered.
matous vascular hyperplasia. Abnormal vessels
in the leptomeninges of the brain and choroid
are the cause of seizures. Diagnosis is made Diabetes mellitus
through clinical examination, angiography, and Patients with poorly controlled type 1 diabetes
imaging. The treatment is symptomatic and mellitus are at risk for poorly healing sockets
includes laser therapy to remove or lighten the because these patients are prone to infections
skin and mucosal lesions, anticonvulsants for and more severe periodontal disease, which in
seizure control, symptomatic and prophylactic itself may impair glycemic control. Conversely,
therapy for headache, and treatments to reduce treatment for severe periodontitis has shown
intraocular pressure. to improve glycemic control. Studies in dia-
betic animals have shown that the formation
Hereditary hemochromatosis. Hereditary of the collagenous framework in the tooth
hemochromatosis results from an inherited extraction socket is inhibited and that aberrant
autosomal-recessive disorder, while hemosid- endothelial activation and impaired angiogenic
erosis may occur from multiple transfusions. response result in delayed socket healing and
Hereditary hemochromatosis is caused by the development of dry socket.42
increased absorption of dietary iron in the gut
leading to iron accumulation in parenchymal
organs and end-organ damage. Iron deposi- Primary bone diseases
tion in the form of hemosiderin and ferritin Paget disease of bone (osteitis deformans) is
leads to skin bronzing, cirrhosis, hypopituita- a disease of unknown etiology characterized
rism, diabetes mellitus, nephrogenic diabetes by hyperactivity of osteoclasts and increased

263
12 Oral Manifestations of Systemic Diseases

bone remodeling that lead to the formation of as sorafenib and sunitinib. Bisphosphonates are
disorganized bone. Paget disease usually occurs potent inhibitors of osteoclast-mediated bone
after the age of 55 years and typically affects resorption and have been used in the manage-
the axial skeleton, especially the pelvis. The ment of multiple myeloma, bone metastases,
skull is involved in about 40% of patients, and and osteoporosis.
involved bones enlarge over time.43 The use of bisphosphonates is associated with
The most common symptom of Paget dis- MRONJ in both osteoporosis (frequency of
ease is bone pain and neurologic signs related 0.1% to 0.2%)45 and cancer therapy (frequency
to pressure on the cranial nerves when their of 3% to 8%).46 The single most important risk
ostia become progressively narrowed from factor for MRONJ is cumulative dose, which is
bone deposition. Enlargement of the maxilla related to duration of therapy47; this is related
and mandible may result in the development in part to the long half-life of 10 years of this
of diastemas of teeth and malocclusion. Early medication. Patients with osteoporosis who are
lesions are radiolucent with coarsening of the prescribed intravenous bisphosphonates gen-
trabecular pattern. Later, cortical thickening erally receive a single annual 5-mg infusion of
develops and radiopacities form, resulting in zoledronic acid, compared with patients with
a cotton wool–like appearance. Patients with myeloma or metastatic cancer to the bones who
Paget disease are at higher risk of developing receive a 4-mg infusion monthly, usually for
osteosarcoma (less than 1% of cases).44 24 months. Other risk factors include dental
The diagnosis is made on the basis of serol- extractions, other dentoalveolar surgery, phys-
ogy (greatly increased bone-specific alkaline iologic trauma, and odontogenic infections,
phosphatase and other bone-turnover markers), although some cases are idiopathic.
histopathology, and radiographic appearance. The American Academy of Oral and Maxil-
Dentists should recognize the symptoms and lofacial Surgeons defines MRONJ on the basis
signs of Paget disease and be aware that a of three criteria: (1) current or prior treatment
dental extraction may be associated with with antiresorptive or antiangiogenic agents,
postextraction bleeding and secondary osteo- (2) exposed bone or bone that can be probed
myelitis. Mild cases of Paget disease do not through an intraoral or extraoral fistula in the
require treatment whereas patients with pain maxillofacial region that has persisted for more
and severe involvement are treated with antire- than 8 weeks, and (3) no history of radiation
sorptive therapy. therapy to the jaws or obvious metastatic
Osteopetrosis is a genetic heterogeneous disease to the jaws.48 Staging and clinical find-
group of heritable diseases of the bone char- ings are presented in Table 12-5. Radiographic
acterized by osteoclastic dysfunction and findings are variable and may include altered
osteosclerosis. Infection, osteonecrosis, and bony trabeculae with mottled osteosclerotic
osteomyelitis may ensue after extractions due changes, bone sequestra and osteolytic changes,
to the reduced blood supply. thickening of the lamina dura and narrowed
periodontal ligament space, and persistent
rarefaction at the site of dental extractions (at
Medication-related osteonecrosis least 6 months after extraction).
of the jaw Patients present with either painful or painless
areas of exposed bone often on the mylohyoid
Medication-related osteonecrosis of the jaw ridge, tori, or nonhealing extraction sockets with
(MRONJ) is a sequela of the use of antiresorptive exposed bone. Some patients may develop ulcers
agents such as bisphosphonates and denosu­ on the tongue from trauma from sharp bone
mab as well as antiangiogenic agents such as edges. Sinus tracts are often seen around the
bevacizumab and other targeted therapies such exposed bone and are a particularly important

264
Poorly Healing or Nonhealing Extraction Sockets and Osteonecrosis

TABLE 12-5  |  MRONJ staging


Stage Clinical presentation

At risk No clinical or radiographic changes consistent with MRONJ in patients who have been treated with either oral or
intravenous bisphosphonates

Stage 0 No clinical evidence of necrotic bone, but radiographic changes and clinical findings consistent with MRONJ

Stage 1 Asymptomatic exposed bone or fistula that probes to bone, with no evidence of infection

Stage 2 Exposed and necrotic bone, or a fistula that probes to bone, with pain, erythema, and infection with or without
purulent drainage

Stage 3 Exposed and necrotic bone or a fistula that probes to bone with pain, erythema, and infection with or without purulent
drainage, and one or more of the following: exposed and necrotic bone extending beyond the region of alveolar bone
(ie, inferior border and ramus in the mandible, maxillary sinus and zygoma in the maxilla) resulting in pathologic
fracture, extraoral fistula, oroantral/oronasal communication, or osteolysis extending to the inferior border of the
mandible or the sinus floor

(Modified from Ruggiero et al.48)

a b

Fig 12-16 |  MRONJ. (a) Nonviable sequestrum on the mylohyoid ridge. (b) Bed of hemorrhagic granulation
tissue after dislodgement (nonsurgical sequestrectomy).

finding in stage 1 disease. Treatment is aimed teeth can be decoronated. However, there is
at pain and infection control, and at every visit no absolute contraindication to extraction.
an attempt should be made to gently dislodge Extraction may be the most appropriate treat-
the bone fragment (nonsurgical sequestrectomy) ment for teeth that are extremely mobile and
(Fig 12-16). Stages 2 and 3 are treated with anti- at risk for aspiration that are within obvious
biotics (amoxicillin with or without clavulanic areas of MRONJ as determined radiographi-
acid, clindamycin, or metronidazole) for a 2- cally, if the patient has continued to have pain
to 4-week or 1- to 2-month course of therapy. and infection in spite of multiple courses of
Chlorhexidine or other antimicrobial oral rinses antibiotic therapy. Contrary to earlier findings,
should be prescribed for all patients at any stage. current data show that surgical resection may
If the patient has had substantial exposure lead to durable remission of disease.49 Preven-
to bisphosphonate therapy, endodontic therapy tion of MRONJ, especially in cancer patients,
is preferable to extraction for nonmobile teeth involves a protocol similar to that for patients
that have good bone support; nonrestorable who are scheduled for head and neck radiation.

265
12 Oral Manifestations of Systemic Diseases

Denosumab (Prolia, Amgen; Xgeva, Amgen) skin or mucosa, tooth mobility, and gingival
is a monoclonal antibody with bone antiresorp- or jaw swelling. Radiographically, the lesions
tive properties that has been cleared by the present as poorly defined radiolucencies. A
US Food and Drug Administration for use in common scenario when such involved mobile
patients with osteoporosis and metastatic can- teeth are extracted is that the socket fails to
cer to the bone as an alternative medication to heal but is instead filled with fleshy soft tissue.52
bisphosphonates. Denosumab binds to a mole-
cule on the osteoclast, inhibiting its activity and
thereby causing inhibition of bone resorption.
As such, its use has also been associated with
Xerostomia and
the development of MRONJ in patients with Hyposalivation
cancer at a frequency of 2% to 3%, similar to
that associated with bisphosphonates.50 Saliva plays a critical role in lubrication, buff-
Prolia is used for treatment of osteoporosis ering, mastication, swallowing, speech, taste,
and is administered at a dose of 60 mg subcu- tooth remineralization, and prevention of oral
taneously once every 6 months, while Xgeva is infection. Therefore, patients with hyposali-
indicated for management of bone metastases vation are at risk of developing erythematous
from solid tumors in adult patients and is admin- areas from friction; experience difficulty with
istered at a dose of 120 mg subcutaneously every eating, swallowing, taste, and speech; and are
4 weeks. Prevention and treatment strategies for at a greater risk of caries and candidiasis.53
denosumab-associated ONJ are the same as for Xerostomia is a common subjective com-
bisphosphonate-associated ONJ.51 plaint of dryness in the mouth. It is only a
Antiangiogenic agents (such as bevacizumab symptom and may or may not be associated
and sunitinib) are prescribed for the man- with reduced saliva in the mouth. Salivary
agement of metastatic cancers and are also gland hypofunction and hyposalivation are the
associated with an increased risk of MRONJ, terms used for objective and measurable reduc-
especially when used together with bisphos- tions in the amount of saliva in the mouth. The
phonates that themselves have antiangiogenic measurable reduction occurs because saliva
properties. Both agents act against blood vessel production and flow are reduced either in nor-
growth factors essential for normal jawbone mally functioning glands or in damaged glands.
remodeling and wound repair and can lead to A simple subjective test for objective dryness
ONJ. Management principles are the same as from hyposalivation that correlates well with
for bisphosphonate-associated ONJ. saliva collection studies involves asking the
following four questions54:

Metastatic tumor to the jaw 1. Do you feel you have less saliva than
Metastases to the oral cavity are rare and normal?
represent approximately 1% of all oral malig- 2. Does your mouth feel dry when eating a
nancies. Two-thirds of metastatic tumors to the meal?
oral cavity occur to the jawbones, and nearly 3. Do you have difficulty swallowing?
80% involve the mandible; one-third occur in 4. Do you need to drink water to swallow dry
the soft tissues, usually involving the gingiva foods?
and sometimes the tongue.52 Cancers of the
breast, lung, prostate, kidney, and colon/rectum A positive answer to all four is strongly asso-
are the most common primary sites. ciated with hyposalivation. A modified Schirmer
Patients with metastasis to the jaw may com- test (normally used in the eyes) also subjectively
plain of pain, paresthesia, or anesthesia of the identifies patients with marked hyposalivation.55

266
Xerostomia and Hyposalivation

Besides feeling that the mouth is dry, patients the salivary and lacrimal glands), while the
may report that they experience sensitivity on secondary form is associated with other auto-
eating foods that are spicy, acidic, or crunchy or immune diseases such as lupus erythematosus
have symptoms of burning. Food may not taste and rheumatoid arthritis. Patients typically
the way it used to, and patients may report a report a dry mouth, dry eyes (keratoconjunc-
metallic or salty taste. In less severe cases, the tivitis sicca), and dry nasal passages.
saliva is frothy, ropey, or thick. In more severe To establish a diagnosis of Sjörgen syndrome,
cases, the mucosa looks dry and erythematous individuals with symptoms and signs suggestive
(especially the tongue) and there is little to no of Sjörgen syndrome need to have at least two
pooling of saliva in the floor of mouth. of the following three objective features: (1)
positive serum anti–SSA/Ro and/or anti–SSB/La
or positive rheumatoid factor and antinuclear
Hyposalivation associated with antibody titer ≥ 1:320; (2) labial salivary gland
normal glands biopsy exhibiting focal lymphocytic sialadenitis
with a focus score ≥ 1 focus/4 mm2; (3) kera-
Hyposalivation associated with normal glands toconjunctivitis sicca with an ocular staining
is by far the most common type of hyposali- score of 3 (assuming that the patient is not
vation. Causative factors include dehydration currently using daily eye drops for glaucoma
(eg, from poor oral intake of fluids, overcon- and has not had corneal surgery or cosmetic
sumption of caffeinated beverages, and/or eyelid surgery in the last 5 years).
smoking), the use of drugs with anticholin- Other possible conditions that may contrib-
ergic activity, the use of multiple medications ute to hyposalivation are IgG4-related disease,
(polypharmacy), and chronic anxiety. Strongly which results in salivary gland enlargement and
xerogenic medications include antidepressants, hyposalivation similar to Sjörgen syndrome,
antipsychotics, diuretics, anticholinergics, anti- chronic GVHD, poorly controlled diabetes, and
hypertensives, anxiolytics, sedatives, NSAIDs, malnutrition.
antihistamines, and opioid analgesic agents. Treatment of xerostomia aims to relieve
symptoms and, in cases of true salivary gland
hypofunction, increase salivary flow. Thera-
Hyposalivation associated with peutic strategies include improved hydration
damaged glands and avoidance of harsh and dehydrating den-
tifrices; avoidance of crunchy hard foods; use
Radiation therapy of the head and neck of saliva substitutes, mucosal lubricants, and
region, especially for head and neck cancer, saliva stimulants such as sugar-free candy or
and Sjögren syndrome occur less commonly gum; and daily fluoride treatment for caries
but result in severe hyposalivation because of prevention. Nonalcoholic oral care products
destruction of salivary glands. Salivary glands such as Biotene products (GlaxoSmithKline)
(especially parotid glands) exposed to radiation and children’s toothpaste are good options.
doses of greater than 60 Gy sustain permanent Medications include pilocarpine (5 to 10 mg
damage with no recovery in salivary hypofunc- three times a day for at least 3 months) and
tion over time. cevimeline (30 mg three times a day for at least
Sjörgen syndrome is a systemic autoim- 3 months). Saline nasal sprays help in lubrica-
mune disorder that mainly affects middle-aged tion and sometimes improve flavor discernment
women. Affected individuals produce autoan- in food.
tibodies that attack and destroy the salivary Damage to the salivary glands with radia-
and other exocrine glands.56 The primary form tion can be somewhat mitigated with the use of
involves mostly the exocrine glands (especially intensity-modified radiation therapy, where the

267
12 Oral Manifestations of Systemic Diseases

radiation is better targeted; with the use of ami- Some patients present not with burning
fostine (a free radical scavenger that protects but with other symptoms, including tingling,
the glands); and with the use of pilocarpine a rough (sandpaper) sensation, a crawling
during radiation. sensation, or taste changes, in which case the
general term of oral dysesthesia may be more
appropriate.
Burning Mouth Syndrome Treatment of BMS is aimed at managing
symptoms, reducing anxiety, and managing
(Oral Dysesthesia Syndrome) underlying psychiatric illnesses. Topical ther-
apies include capsaicin extract used in a rinse,
Primary burning mouth syndrome (BMS) is a clonazepam rinse, and topical anesthetic solu-
chronic pain condition and neuropathy char- tions. Systemic therapy includes alpha lipoic
acterized by a burning dysesthesia mainly acid (300 mg twice a day), clonazepam (0.5
localized to the tip of the tongue and anterior to 2.0 mg per day, usually taken at night), low
dorsum, the lip mucosa, and the anterior hard doses of the tricyclic antidepressants amitripty-
palatal mucosa and is often associated with line and nortriptyline, and gabapentin.61
xerostomia, hyposalivation, and taste and smell Because this is a chronic condition that often
changes.57 lasts for years, nonpharmacologic approaches
The pathophysiology of BMS is not well to management are particularly important to
understood and may be secondary to both reduce dependence on medications and include
biologic and psychologic factors.58 Hematinic stress management/reduction, yoga, exercise,
deficiencies (such as iron and vitamin B12 defi- and cognitive behavioral therapy. Cases that
ciencies) should be ruled out first, although in are refractory to therapy should be referred to
such cases patients usually present with signs a neurologist or pain specialist.
of atrophic glossitis. BMS is often triggered Multitargeted tyrosine kinase inhibitors
by stressful events in a patient’s life and is (such as sunitinib, sorafenib, pazopinib, and
strongly associated with sleep disorders, anx- cabozantinib) are antineoplastic drugs that
iety, depression, psychiatric illness, and signs have been associated with oral dysesthesias
and symptoms of somatization. More serious and palmar-plantar erythrodysesthesia.62,63
conditions associated with BMS include multi- In severe cases, a dose reduction or discon-
ple sclerosis, trigeminal neuralgia, and central tinuation of the medication is recommended.
nervous system lesions. BMS is classified into Symptoms may resolve upon discontinuation
three types: type I, with burning sensation of tyrosine kinase inhibitor therapy.
developing in the late morning, gradually
increasing in severity during the day, and reach-
ing its peak intensity by evening; type II, with
continuous symptoms throughout the day and Conclusion
difficulty falling asleep at night; and type III,
with pain-free periods during the day.59 We have presented eight common oral condi-
Several drug classes have been associ- tions that may also indicate systemic disease.
ated with dysgeusia and dysosmia through Many patients see their dentist on a regular
a multifactorial mechanism; these drugs basis for scaling and prophylaxis, and the dental
include fluoroquinolones, macrolides (clar- hygienist and dentist are therefore in a unique
ithromycin), antimycotics (terbinafine), position to notice subtle changes in the mucosa
angiotensin-converting enzyme inhibitors, pro- or bone that may be the first indication of a sys-
tein kinase inhibitors, proton pump inhibitors, temic disease, be it as common as iron-deficiency
and statins.60 anemia or as life-threatening as leukemia.

268
References

References 18. Lerman MA, Laudenbach J, Marty FM, Baden LR, Treis-
ter NS. Management of oral infections in cancer patients.
Dent Clin North Am 2008;52:129–153.
1. Scully C. Clinical practice. Aphthous ulceration. N Engl 19. Lee FY, Mossad SB, Adal KA. Pulmonary mucormyco-
J Med 2006;355:165–172. sis: The last 30 years. Arch Intern Med 1999; 159:
2. Baccaglini L, Lalla RV, Bruce AJ, et al. Urban legends: 1301–1309.
Recurrent aphthous stomatitis. Oral Dis 2011; 17: 20. Spellberg B, Edwards J Jr, Ibrahim A. Novel perspectives
755–770. on mucormycosis: Pathophysiology, presentation, and
3. Baumgart DC, Sandborn WJ. Crohn’s disease. Lancet management. Clin Microbiol Rev 2005;18:556–569.
2012;380:1590–1605. 21. Chan LS, Ahmed AR, Anhalt GJ, et al. The first interna-
4. Scully C, Hodgson T. Recurrent oral ulceration: tional consensus on mucous membrane pemphigoid:
Aphthous-like ulcers in periodic syndromes. Oral Surg Definition, diagnostic criteria, pathogenic factors, medi-
Oral Med Oral Pathol Oral Radiol Endod 2008; 106: cal treatment, and prognostic indicators. Arch Dermatol
845–852. 2002;138:370–379.
5. Wekell P, Karlsson A, Berg S, Fasth A. Review of auto- 22. Scully C, Mignogna M. Oral mucosal disease: Pemphi-
inflammatory diseases, with a special focus on periodic gus. Br J Oral Maxillofac Surg 2008;46:272–277.
fever, aphthous stomatitis, pharyngitis and cervical 23. Warnakulasuriya S, Johnson NW, van der Waal I.
adenitis syndrome. Acta Paediatr 2016; 105: Nomenclature and classification of potentially malignant
1140–1151. disorders of the oral mucosa. J Oral Pathol Med 2007;
6. Dalvi SR, Yildirim R, Yazici Y. Behcet’s syndrome. Drugs 36:575–580.
2012;72:2223–2241. 24. Mawardi H, Elad S, Correa ME, et al. Oral epithelial
7. Mendes D, Correia M, Barbedo M, et al. Behcet’s dysplasia and squamous cell carcinoma following allo-
disease—A contemporary review. J Autoimmun 2009; geneic hematopoietic stem cell transplantation: Clinical
32:178–188. presentation and treatment outcomes. Bone Marrow
8. Sonis ST. Oral mucositis. Anticancer Drugs 2011; 22: Transplant 2011;46:884–891.
607–612. 25. Al-Hashimi I, Schifter M, Lockhart PB, et al. Oral lichen
9. Shim YJ, Choi JH, Ahn HJ, Kwon JS. Effect of sodium planus and oral lichenoid lesions: Diagnostic and ther-
lauryl sulfate on recurrent aphthous stomatitis: A apeutic considerations. Oral Surg Oral Med Oral Pathol
randomized controlled clinical trial. Oral Dis 2012;18: Oral Radiol Endod 2007;103(suppl):S25.e1–12.
655–660. 26. Schlosser BJ. Lichen planus and lichenoid reactions of
10. Xu F, Schillinger JA, Sternberg MR, et al. Seroprevalence the oral mucosa. Dermatol Ther 2010;23:251–267.
and coinfection with herpes simplex virus type 1 and 27. Au J, Patel D, Campbell JH. Oral lichen planus. Oral
type 2 in the United States, 1988–1994. J Infect Dis Maxillofac Surg Clin North Am 2013;25:93–100.
2002;185:1019–1024. 28. Habash-Bseiso DE, Yale SH, Glurich I, Goldberg JW.
11. Woo SB, Challacombe SJ. Management of recurrent Serologic testing in connective tissue diseases. Clin
oral herpes simplex infections. Oral Surg Oral Med Oral Med Res 2005;3:190–193.
Pathol Oral Radiol Endod 2007;103(suppl):S12.e1–18. 29. Brennan MT, Valerin MA, Napeñas JJ, Lockhart PB.
12. Griffiths PD. Burden of disease associated with human Oral manifestations of patients with lupus erythemato-
cytomegalovirus and prospects for elimination by univer- sus. Dent Clin North Am 2005;49:127–141.
sal immunisation. Lancet Infect Dis 2012;12:790–798. 30. Schubert MM, Correa ME. Oral graft-versus-host
13. Dojcinov SD, Venkataraman G, Raffeld M, Pittaluga S, disease. Dent Clin North Am 2008;52:79–109.
Jaffe ES. EBV positive mucocutaneous ulcer—A study 31. Lalla RV, Patton LL, Dongari-Bagtzoglou A. Oral candi-
of 26 cases associated with various sources of immu- diasis: Pathogenesis, clinical presentation, diagnosis
nosuppression. Am J Surg Pathol 2010;34:405–417. and treatment strategies. J Calif Dent Assoc 2013;41:
14. Williams PM, Conklin RJ. Erythema multiforme: A review 263–268.
and contrast from Stevens-Johnson syndrome/toxic 32. Akpan A, Morgan R. Oral candidiasis. Postgrad Med J
epidermal necrolysis. Dent Clin North Am 2005; 49: 2002;78:455–459.
67–76. 33. Assimakopoulos D, Patrikakos G, Fotika C, Elisaf M.
15. Léauté-Labrèze C, Lamireau T, Chawki D, Maleville J, Benign migratory glossitis or geographic tongue: An
Taïeb A. Diagnosis, classification, and management of enigmatic oral lesion. Am J Med 2002;113:751–755.
erythema multiforme and Stevens-Johnson syndrome. 34. Woo SB. Oral Pathology: A Comprehensive Atlas and
Arch Dis Child 2000;83:347–352. Text. Philadelphia: Elsevier Saunders, 2012:58.
16. Ayangco L, Rogers RS 3rd. Oral manifestations of 35. Armitage GC; Research, Science and Therapy Commit-
erythema multiforme. Dermatol Clin 2003;21:195–205. tee of the American Academy of Periodontology.
17. Creamer D, Walsh SA, Dziewulski P, et al. U.K. guidelines Diagnosis of periodontal diseases. J Periodontol 2003;
for the management of Stevens-Johnson syndrome/ 74:1237–1247.
toxic epidermal necrolysis in adults 2016. Br J Dermatol 36. Casiglia J, Woo SB, Ahmed AR. Oral involvement in
2016;174:1194–1227. autoimmune blistering diseases. Clin Dermatol 2001;
19:737–741.

269
12 Oral Manifestations of Systemic Diseases

37. Holmstrup P, Westergaard J. Necrotizing periodontal 50. Epstein MS, Ephros HD, Epstein JB. Review of current
disease. In: Lindhe J, Lang NP, Karring T (eds). Clinical literature and implications of RANKL inhibitors for oral
Periodontology and Implant Dentistry, ed 5. Ames, IA: health care providers. Oral Surg Oral Med Oral Pathol
Blackwell Munksgaard, 2008:459–475. Oral Radiol 2013;116:e437–442.
38. Kataoka M, Kido J, Shinohara Y, Nagata T. Drug-induced 51. Diz P, López-Cedrún JL, Arenaz J, Scully C.
gingival overgrowth—A review. Biol Pharm Bull 2005; Denosumab-related osteonecrosis of the jaw. J Am Dent
28:1817–1821. Assoc 2012;143:981–984.
39. Seymour RA, Ellis JS, Thomason JM. Risk factors for 52. Hirshberg A, Shnaiderman-Shapiro A, Kaplan I, Berger
drug-induced gingival overgrowth. J Clin Periodontol R. Metastatic tumours to the oral cavity—Pathogenesis
2000;27:217–223. and analysis of 673 cases. Oral Oncol 2008; 44:
40. Li CC, Malik SM, Blaeser BF, et al. Mucosal pigmentation 743–752.
caused by imatinib: Report of three cases. Head Neck 53. Gupta A, Epstein JB, Sroussi H. Hyposalivation in elderly
Pathol 2012;6:290–295. patients. J Can Dent Assoc 2006;72:841–846.
41. Sánchez-Pablo MA, González-García V, del 54. Fox PC, Busch KA, Baum BJ. Subjective reports of
Castillo-Rueda A. Study of total stimulated saliva flow xerostomia and objective measures of salivary gland
and hyperpigmentation in the oral mucosa of patients performance. J Am Dent Assoc 1987;115:581–584.
diagnosed with hereditary hemochromatosis. Series of 55. Chen A, Wai Y, Lee L, Lake S, Woo SB. Using the
25 cases. Med Oral Patol Oral Cir Bucal 2012; 17: modified Schirmer test to measure mouth dryness: A
e45–49. preliminary study. J Am Dent Assoc 2005;136:164–170.
42. McKenna SJ. Dental management of patients with 56. Shiboski SC, Shiboski CH, Seror R, et al. 2016 American
diabetes. Dent Clin North Am 2006;50:591–606. College of Rheumatology/European League Against
43. Colina M, La Corte R, De Leonardis F, Trotta F. Paget’s Rheumatism classification criteria for primary Sjögren’s
disease of bone: A review. Rheumatol Int 2008; 28: syndrome: A consensus and data-driven methodology
1069–1075. involving three international patient cohorts. Arthritis
44. Hansen MF, Seton M, Merchant A. Osteosarcoma in Rheumatol 2017;69:35–45.
Paget’s disease of bone. J Bone Miner Res 2006; 57. Grushka M, Epstein JB, Gorsky M. Burning mouth
21(suppl 2):P58–63. syndrome. Am Fam Physician 2002;65:615–620.
45. Solomon DH, Mercer E, Woo SB, Avorn J, Schneeweiss 58. Klasser GD, Fischer DJ, Epstein JB. Burning mouth
S, Treister N. Defining the epidemiology of syndrome: Recognition, understanding, and manage-
bisphosphonate-associated osteonecrosis of the jaw: ment. Oral Maxillofac Surg Clin North Am 2008; 20:
Prior work and current challenges. Osteoporos Int 2013; 255–271.
24:237–244. 59. Sun A, Wu KM, Wang YP, Lin HP, Chen HM, Chiang
46. Kuhl S, Walter C, Acham S, Pfeffer R, Lambrecht JT. CP. Burning mouth syndrome: A review and update. J
Bisphosphonate-related osteonecrosis of the jaws—A Oral Pathol Med 2013;42:649–655.
review. Oral Oncol 2012;48(10):938–947. 60. Tuccori M, Lapi F, Testi A, et al. Drug-induced taste and
47. Almazrooa SA, Woo SB. Bisphosphonate and smell alterations: A case/non-case evaluation of an
nonbisphosphonate-associated osteonecrosis of the Italian database of spontaneous adverse drug reaction
jaw: A review. J Am Dent Assoc 2009;140:864–875. reporting. Drug Saf 2011;34:849–859.
48. Ruggiero SL, Dodson TB, Fantasia J, et al. American 61. McMillan R, Forssell H, Buchanan JA, Glenny AM,
Association of Oral and Maxillofacial Surgeons position Weldon JC, Zakrzewska JM. Interventions for treating
paper on medication-related osteonecrosis of the burning mouth syndrome. Cochrane Database Syst Rev
jaw—2014 update. J Oral Maxillofac Surg 2014; 72: 2016;11:CD002779.
1938–1956. 62. Lee WJ, Lee JL, Chang SE, et al. Cutaneous adverse
49. Carlson ER, Basile JD. The role of surgical resection in effects in patients treated with the multitargeted kinase
the management of bisphosphonate-related osteone- inhibitors sorafenib and sunitinib. Br J Dermatol 2009;
crosis of the jaws. J Oral Maxillofac Surg 2009; 67: 161:1045–1051.
85–95. 63. Yuan AD, Kurtz SL, Barysauskas CM, Pilotte AP, Wagner
AJ, Treister NS. Oral adverse events in cancer patients
treated with VEGFR-directed multitargeted tyrosine
kinase inhibitors. Oral Oncol 2015;51:1026–1033.

270
CHAPTER 13

Oral Complications in the


Immunocompromised Patient:
The Oncology Prototype
Douglas E. Peterson, dmd, phd, fds rcsed

Oral complications in cancer patients can have • Concurrent/concomitant (synergistic): Com-


considerable adverse impact on both the clini- bination with radiation therapy (eg, head and
cal condition of the cancer patient and the cost neck radiation therapy plus weekly cisplatin)
of cancer care.1,2 This dynamic occurs for a
number of reasons, including the intensity of Although considerable evidence exists about
cancer treatment regimens as well as the extent treatment-based oral toxicity in relation to
of preexisting oral disease, which can evolve tumor type, new frontiers in research are
into acute and/or chronic toxicity. beginning to delineate patient-based factors
However, substantive advances in the field with more precision. This evolving evidence
of oral oncology over the past two decades base continues to more precisely define the
have resulted in reduced incidence and sever- interrelationships between the more classic
ity of selected oral toxicities. Key drivers for modeling of disease (and its treatment) and
this progress include the increasingly effective new knowledge directed to patient-based risk
modeling of molecular science into clinical determinants3,4 (Fig 13-1). This line of research
practice as well as the leading role that scien- represents a promising foundation for future
tists and clinicians from dental medicine have implementation of personalized medicine in
taken in interprofessional research, production which customized oral prevention and man-
of clinical guidelines, and education of health agement interventions are developed for each
professionals. These advances have occurred individual patient.
even as combination cancer therapies for The management of acute myeloid leuke-
advanced stages of oral cancer have become mia is utilized in this chapter as a principal
increasingly prominent: prototype by which the science and its clinical
translation can be applied to clinical decision
• Neoadjuvant (debulk): Sole use prior to sur- making and patient care (Figs 13-2 and 13-3).
gery (eg, 5-fluorouracil, which can cause oral The high-dose induction chemotherapy regi-
mucositis) men that is used to treat patients with acute
• Adjuvant (curative): Use after surgery and myelogenous leukemia is profoundly myelosup-
before radiation therapy (eg. 5-fluorouracil, pressive and typically causes severe ulcerative
which can cause oral mucositis) oral mucositis of at least 2 weeks’ duration.
The interface of profound myelosuppression
with disruption in integrity of the oral mucosal

271
13 Oral Complications in the Immunocompromised Patient: The Oncology Prototype

Patient

Toxicity

Treatment Tumor

Fig 13-1  |  Toxicity of cancer treatment, including oral Fig 13-2 |  Peripheral blood smear from a patient
complications, occurs as a result of patient-based, with newly diagnosed acute myelogenous leukemia
tumor-based, and treatment-based components. in blast crisis.

a b

Fig 13-3 |  Acute leukemia can be considered “naturally metastatic” in that the neoplasm arises within the
white blood cell progenitors produced in the bone marrow. Because of the inherent circulation of these cells,
the disease is widespread by the time of diagnosis. (a) Histopathology based on hepatic biopsy, demonstrating
widespread infiltrate of the blast leukemic cells. (b) Gingival leukemic infiltrate in a patient newly diagnosed with
acute myelogenous leukemia. Extensive gingival engorgement has been caused by the infiltrating leukemic cells.
The resulting ischemia can contribute to the development of tissue necrosis as well as opportunistic infection
such as pseudomembranous candidiasis.

barrier can result in a risk of life-threatening • Management of oral mucosal injury associ-
bacteremia and/or sepsis. ated with targeted cancer therapies. This will
Because of their high clinical importance and illustrate the unique characteristics of these
frequency, this chapter highlights the following: lesions in relation to oral mucositis caused
by conventional cancer treatments.
• Acute toxicities of mucosal injury and their • Contemporary clinical issues associated with
associated pain, as well as oral infection, in medication-related osteonecrosis of the jaw.
the setting of multiple acute sequelae that Discussion of these lesions is presented in
can occur (Table 13-1). By comparison, the context of risk for emergence of this tox-
however, chronic toxicities caused by che- icity in selected oncology patients who are
motherapy are not common; this aspect of receiving bone-stabilizing agents in order to
oncology patient care will thus not be spe- prevent skeletal bone fractures.
cifically addressed in detail.

272
Oral Complications in the Immunocompromised Patient: The Oncology Prototype

TABLE 13-1  |  Oral toxicities in cancer patients

Complication Weighted prevalence

Bisphosphonate-associated All studies: 6.1% (mean)


osteonecrosis  Studies with documented follow-up: 13.3%  
Studies with undocumented follow-up: 0.7%  
Epidemiologic studies: 1.2%  

Dysgeusia  CT only: 56.3% (mean)  


RT only: 66.5% (mean) 
Combined CT and RT: 76% (mean) 

Oral fungal infection  Clinical oral fungal infection (all oral candidiasis):
Pretreatment: 7.5% 
During treatment: 39.1% 
Posttreatment: 32.6% 
Clinical oral candidiasis infection by cancer treatment:
During HNC RT: 37.4% 
During CT: 38% 

Oral viral infection  Patients treated with CT for hematologic malignancies:


Patients with oral ulcerations/sampling oral ulcerations: 49.8% 
Patients sampling oral ulcerations: 33.8% 
Patients sampling independently of the presence of oral ulcerations: 0% 
Patients treated with RT:
Patients with RT only/sampling oral ulcerations: 0% 
Patients with RT and adjunctive CT/sampling oral ulcerations: 43.2% 

Dental disease  Dental caries in patients treated with cancer therapy:


All studies: 28.1% 
CT only: 37.3% 
Post-RT: 24% 
Post-CT and post-RT: 21.4% 
Severe gingivitis in patients undergoing CT: 20.3% 
Dental infection/abscess in patients undergoing CT: 5.8% 

Osteoradionecrosis  Conventional RT: 7.4% 


IMRT: 5.2% 
RT and CT: 6.8% 
Brachytherapy: 5.3% 

Trismus  Conventional RT: 25.4% 


IMRT: 5% 
Combined RT and CT: 30.7% 

Oral pain*  VAS pain level (0–100) in HNC patients: 


Pretreatment: 12/100 
Immediately posttreatment: 33/100 
1 mo posttreatment: 20/100 
EORTC QLQ-C30 pain level (0–100) in HNC patients: 
Pretreatment: 27/100 
3 mo posttreatment: 30/100 
6 mo posttreatment: 23/100 
12 mo posttreatment: 24/100 

273
13 Oral Complications in the Immunocompromised Patient: The Oncology Prototype

TABLE 13-1 (cont)  |  Oral toxicities in cancer patients

Complication Weighted prevalence


Salivary gland hypofunction and All studies: 
xerostomia (in HNC patients by type Pre-RT: 6% 
of RT) During RT: 93% 
1–3 mo post-RT: 74% 
3–6 mo post-RT: 79% 
6–12 mo post-RT: 83% 
1–2 y post-RT: 78% 
> 2 y post-RT: 85% 
Conventional RT: 
Pre-RT: 10% 
During RT: 81% 
1–3 mo post-RT: 71% 
3–6 mo post-RT: 83% 
6–12 mo post-RT: 72% 
1–2 y post-RT: 84% 
> 2 y post-RT: 91% 
IMRT:
Pre-RT: 12% 
During RT: 100% 
1–3 mo post-RT: 89% 
3–6 mo post-RT: 73% 
6–12 mo post-RT: 90% 
1–2 y post-RT: 66% 
> 2 y post-RT: 68% 

*Pain is common in patients with HNCs and is reported by approximately half of patients before cancer therapy, by 81% during
therapy, by 70% at the end of therapy, and by 36% at 6 months posttreatment.
CT, chemotherapy; RT, radiation therapy; HNC, head and neck cancer; IMRT, intensity-modulated radiation therapy; VAS, visual
analog scale; EORTC QLQ-C30, European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire C30.
(Adapted from the National Cancer Institute website.1)

Given the scientific and medical complexity patients whose immune function is attenuated
of cancer and its treatment, the multidisci- as a result of systemic disease. Examples of
plinary oncology team has essential value in these diseases include genetically based or
achieving optimal outcomes while minimizing acquired immunodeficiencies, as well as adverse
side effects of that treatment. As a part of this outcomes of non-neoplastic disease treatment.
team, the dental professional has vital respon-
sibilities, including the following:

• Education of the patient and family members


Acute Oral Toxicities in
• Education of the medical and nursing staff the Myelosuppressed
• Diagnosis and treatment of oral com­pli­
cations
Oncology Patient
• Prevention of oral complications
• Oral health follow-up and maintenance Oral Mucositis
Clinical impact
The interprofessional model as described
for the oncology patient can also provide con- Oral mucositis can have a significant delete-
text for research and clinical management of rious impact on the cancer patient receiving

274
Oral Mucositis

Fig 13-4 |  Oral mucositis in a breast cancer patient


receiving high-dose chemotherapy. Extensive pseu-
domembranous lesions can significantly impair normal
oral function and represent a portal of entry for severe
systemic infection.

Box 13-1  |  Key developments in the field of oral mucositis over the past 20 years2

• Initiation of research studies into pathobiology and clinical trials


• The creation of international organizations of health professionals directed to the mission of supportive care in
cancer
• The first two American Society of Clinical Oncology educational sessions addressing mucositis, held in 2003 and
2004
• Publication of the first evidence-based mucositis guidelines by the Multinational Association of Supportive Care
in Cancer and International Society of Oral Oncology (MASCC/ISOO), which have since been updated twice
• Incorporation of mucositis as a MeSH term by the National Library of Medicine
• The publication of clinical practice guidelines for mucositis by other organizations
• Comprehensive updating of the National Cancer Institute’s Physician Data Query website1 to include a section on
oral complications of chemotherapy and head and neck radiation

high-dose chemotherapy2,4–6 (Fig 13-4). The le- had been viewed by many clinicians and thus
sion can be so painful that oral functions such patients as an inevitable consequence of
as eating, speaking, and swallowing are severe- high-dose cancer therapy. Fortunately, sub-
ly compromised. In cancer patients receiving stantial progress over the past 20 years at the
multicycle chemotherapy regimens, the oral research and clinical levels has set the stage
pain can necessitate subsequent dose delays or for new opportunities for prevention and treat-
reductions to lessen the extent of oral injury ment (Box 13-1).
and discomfort. In neutropenic cancer patients,
ulcerative oral mucositis can be a portal of
entry for systemic infection, resulting in bac- Pathobiology and future research
teremia and/or sepsis. The collective sequelae directions
of the lesion can require extensive use of health
care resources,7 including by some estimates One of the key advances in the last two
approximately US $41,000 in the hematopoi- decades has been the delineation of a contem-
etic stem cell transplant (HSCT) population.8 porary pathobiologic model that illustrates the
Oral mucositis can thus be clinically and eco- complex interactions within and across oral ep-
nomically consequential. ithelial and connective tissues5,6,9 (Fig 13-5). An
Despite the importance of the condition, additional major advance in recent years has
however, until the early 1990s oral mucositis been the scientific and clinical linkage of oral

275
13

276
Oral Complications in the Immunocompromised Patient: The Oncology Prototype

Fig 13-5  |  The conceptual framework for oral mucositis pathobiology consists of five stages, ranging from initial injury within hours of exposure to high-dose cancer
therapy to eventual healing approximately 2 to 4 weeks after cessation of that treatment. Although the illustration depicts an orderly and sequential mechanistic pro-
cess, the course of molecular and cellular events is more likely dysregulated and biologically chaotic. (Reprinted from Sonis5 with permission.)
Oral Mucositis

a b

Fig 13-6  |  Systemic chemotherapy causes architectural and functional disturbances throughout the alimentary
tract, including the intestine. (a) Normal jejunal mucosa in a healthy patient, as imaged by video-capsule endos-
copy. Black arrows, normal villi. (b) Extensive ulceration (white arrow) and hemorrhage (black arrows) of the in-
testine in an HSCT patient approximately 7 days after the last dose of high-dose chemotherapy as a component
of the conditioning regimen. This lesion of the gastrointestinal tract is not typically painful but can be associated
with severe diarrhea, cramping, and compromised absorption of nutrients. (Reprinted from Triantafyllou et al10
with permission.)

mucositis with gastrointestinal mucositis3,4,10,11 typically incorporate multiple considerations,


(Fig 13-6). This paradigm of alimentary tract including the location and staging of the can-
mucositis has provided an essential founda- cer, the age of the patient, and anticipated cure
tion for new research frontiers that collectively rates. Although the chemotherapy utilized in
could permit personalized medicine for each these treatment protocols is not highly myelo-
oncology patient based on a priori prediction suppressive, it can cause oral mucositis that is
of both risk for mucositis and likely response clinically significant in some patients.
to therapeutics (Box 13-2). At present, the US Food and Drug Admin-
Achievement of this goal is important istration has cleared only one nondevice
because the incidence and severity of oral intervention for oral mucositis.12 This agent,
mucositis vary across cancer patient cohorts, palifermin, is a member of the keratinocyte
from approximately 40% of patients receiving growth factor family and is cleared for pre-
standard-dose chemotherapy to approximately vention of oral mucositis in HSCT patients
80% of patients receiving myeloablative con- receiving chemotherapy with or without total
ditioning regimens for HSCT.2 There is thus a body radiation in the autologous transplant
need to implement customized prevention and setting. Despite phase IV studies, the agent has
management for selected, but not all, cancer not demonstrated consistent efficacy in the
patients who are about to begin mucotoxic nonautologous transplant setting.13
cancer treatment. Additional preclinical and clinical develop-
Patients with advanced stages of head and ment of drugs and biologics continues, directed
neck cancer (Fig 13-7) typically receive multi- to key molecular targets and network hubs (see
modality treatment that includes approximately Box 13-2). Notable among many current stud-
two cycles of induction chemotherapy followed ies is work being conducted by investigators
by high-dose head and neck radiation with or at the National Institutes of Health, who are
without weekly concomitant chemotherapy. pursuing the use of tempol for prevention of
Decisions on types of oral cancer treatment oral mucositis14 and gene therapy for treatment

277
13 Oral Complications in the Immunocompromised Patient: The Oncology Prototype

Box 13-2  |  Future research questions: Oral mucositis caused by cancer therapies

Pathobiology
• What is the role of the oral microbiome relative to causation, progression, and/or healing of oral mucositis?
• What are the molecular bases for incidence and severity of pain in relation to degree of oral mucosal injury?
• Why do patients with comparable characteristics, including age, sex, cancer diagnosis, and treatment regimen,
collectively exhibit variability of toxicity expression, including oral mucositis?
• How can delineation of molecular pathogenesis lead to novel models for prediction of incidence, severity, and
response to thera­peutics in individual patients, in contrast to a cohort-wide basis?
• What biologic characteristics contribute to native protection against development of mucositis at selected non–
alimentary tract mucosal sites such as vaginal and conjunctival mucosa?
• What crosstalk occurs between at-risk mucosal and dermal sites for which common etiologies can be
delineated?
• How can systems biology technology be optimally applied to study the regulatory function of gene clusters in
comparison with single gene expression? In addition, how do these interrelated gene cluster functions translate
to studies of symptom burden that are observed in many oncology patients undergoing high-dose cancer
therapies?

Development of pharmacologics, biologics, and devices to manage oral mucositis


• What are the key molecular targets that, if successfully perturbed, are likely to permit achievement of a high
degree of clinical efficacy with the least extent of side effects?
• What is the most efficient preclinical and clinical development strategy to bring new therapeutics to clinical
practice in a cost-effective fashion?

Impact of evidence-based clinical practice guidelines


• What are the most effective ways to ascertain the impact of utilization of evidence-based oral mucositis clinical
practice guide­lines relative to enhanced clinical outcomes and reduced cost of cancer care?

(Reprinted from Peterson et al2 with permission.)

Fig 13-7 |  Advanced stage of squamous cell carci-


noma on the right lateral side of the tongue. Advanced
stages of oral squamous cell carcinoma typically war-
rant multimodality cancer treatment, including surgery
and/or chemotherapy followed by high-dose head and
neck radiation. The chemotherapeutic regimens utilized
are not usually profoundly myelosuppressive, although
they can cause clinically significant oral mucositis.

of chemoradiation-induced oral mucositis.15 of the mucositis trajectory in patients via


This and related lines of biologic research are genetic6,17–21 and imaging-based22 approaches.
being enhanced by the development of novel High-throughput deep sequencing of the oral
technologies such as in vitro tissue engineering microbiome23 is permitting delineation of puta-
models of oral mucosa16 as well as prediction tive pathogens that are not typically cultivable

278
Oral Mucositis

by conventional methods. Systems biology therapy.29 Systematic oral tissue and pain
technology is increasingly being utilized to inte- assessments are then needed throughout the
grate the complex and extensive data sets into period of cancer treatment. A staged approach
cohesive models of pathobiology that translate to pain relief, ranging from topical anesthetics,
to the clinical trajectory.2,24–26 doxepin, and fentanyl through nonopioid and
Thus, basic, translational, and clinical opioid systemic analgesics, can be implemented
research has successfully shifted the historic as needed. It is also essential to monitor side
paradigm of “inevitable consequence” to a effects of the pain therapeutics, particularly
contemporary model for which prediction of with the opioid classes of drugs.
incidence and severity of oral mucositis as well
as response to therapeutics is likely to emerge
over the next few years. These advances will Mucosal injury caused by targeted
likely provide an evidence base on which cancer therapy
comprehensive clinical guidelines for mucosi-
tis management can position the clinician to The use of targeted cancer therapies has escalat-
develop a personalized, customized approach ed in recent years, following their introduction
to prevention and treatment of oral mucositis into clinical oncology practice approximately
for each individual oncology patient. a decade ago.28,30,31 These agents are designed
to selectively inhibit a molecular target that
is abnormal in malignant cells versus cells in
Pain management for patients with normal tissue.32 These agents have a number
oral mucositis of safety and efficacy benefits over convention-
al high-dose chemotherapy, including reduced
The complex proinflammatory and neu- toxicity profiles as well as enhanced cancer pa-
rotransmitter cascade associated with oral tient survivorship in selected cohorts.
mucositis can result in moderate to severe oral These agents have, however, resulted in the
pain that can compromise delivery of cancer emergence of unique oral mucosal lesions,
treatment regimens.27 The pain can escalate such as the mammalian target of rapamycin
mood changes such as anxiety and depres- (mTOR) inhibitors33 (Fig 13-8). These lesions
sion, which in turn can further contribute to mimic recurrent aphthous ulcerations in
the patient’s pain experience. There are likely clinical appearance and in selected patients
multiple molecular mechanisms that contrib- respond well to topical, intralesional, and/or
ute to the degree to which patients report pain systemic corticosteroid therapy.28,34–36 These
severity.28 It is important to note that objective lesions are thus quite different from oral
measures of oral mucosal inflammation (eg, mucositis caused by conventional chemother-
erythema, ulceration) are not wholly homol- apy or head and neck radiation with regard
ogous with patient-based subjective reports to mucosal distribution, clinical appearance,
of pain. and response to steroid management. Further
Thus, both objective and subjective eval- research on the molecular pathobiology of
uation of the pain component is warranted these lesions is needed to enhance understand-
clinically in order to optimize pain manage- ing of their causation as well as the potential
ment. It is essential that an aggressive and for prediction of incidence and severity on a
systematic pain assessment and interven- patient-by-patient basis.
tion strategy be incorporated. The paradigm Unlike high-dose chemotherapy patients,
begins with education of the patient and patients receiving targeted cancer therapies
caregiver regarding oral mucositis and its such as inhibitors of the mTOR are not mye-
consequences prior to initiation of cancer losuppressed. These mTOR inhibitory agents

279
13 Oral Complications in the Immunocompromised Patient: The Oncology Prototype

a b

Fig 13-8  |  Selected mammalian target of rapamycin (mTOR) inhibitors can cause oral mucosal lesions that clin-
ically resemble recurrent aphthous ulceration and often respond to topical, intra­lesional, or systemic corticoste-
roid management. (a) Lesion on the tongue after a patient received three 21-day cycles (63 days) of ridaforolimus.
(b) Inner lips of a patient who developed mTOR inhibitor–associated stomatitis within 10 days after initiation of
treatment with everolimus (10 mg once daily) in combination with figitumumab. (Courtesy of Dr Nathaniel Treister,
Dana-Farber Cancer Institute/Brigham and Women’s Hospital, Boston, Massachusetts.)

are directed to reducing the hyperactivity of • Topical, intralesional, or systemic steroids


mTOR observed in several oncogenic path- may be useful for treatment, in contrast to
ways.30 However, patients receiving selected mucosal injury caused by conventional che-
targeted agents can experience a unique clinical motherapy or head and neck radiation.
expression of mucosal injury not previously • Based upon the current state of the science,
documented prior to the advent of these agents. clinicians cannot typically predict a priori
Key points associated with oral mucosal which patients will or will not develop the
injury caused by mTOR inhibitors are the lesions.
following28:
New research is needed, including risk pre-
• Incidence varies widely (2% to 78%). diction, pathobiology, and the molecular basis
• Grade 3 and grade 4 toxicity occurs in up to of pain (Fig 13-9), in order to enhance preven-
9% of patients. tion at the clinical level in the future.

280
Oral Mucositis

Fig 13-9  |  Integration of the molecular modeling of oral mucosal pain into the current inflammation biology par-
adigm for oral mucositis caused by cancer therapy. Development of novel pain therapeutics targeting key neural
pathways could strategically enhance management of oral mucositis in the future. (Reprinted from Peterson et
al29 with permission.)

281
13 Oral Complications in the Immunocompromised Patient: The Oncology Prototype

Box 13-3  |  Oral complications of hematopoietic stem cell transplantation

Phase I: Preconditioning
• Oral infections: dental caries, endodontic infections, periodontal disease (gingivitis or
periodontitis), and mucosal infections (ie, viral, fungal, or bacterial)
• Gingival leukemic infiltrates
• Metastatic cancer
• Oral bleeding
• Oral ulceration: aphthous ulcers and erythema multiforme
• Temporomandibular dysfunction
Phase II: Conditioning neutropenic phase
• Oropharyngeal mucositis
• Oral infections: mucosal infections (ie, viral, fungal, or bacterial) and periodontal
infections
• Hemorrhage
• Xerostomia
• Taste dysfunction
• Neurotoxicity: dental pain and muscle tremor (eg, jaws or tongue)
• Temporomandibular dysfunction: jaw pain, headache, and joint pain
Phase III: Engraftment hematopoietic recovery
• Oral infections: mucosal infections (ie, viral, fungal, or bacterial)
• Acute graft-versus-host disease (GVHD)
• Xerostomia
• Hemorrhage
• Neurotoxicity: dental pain and muscle tremor (eg, jaws or tongue)
• Temporomandibular dysfunction: jaw pain, headache, and joint pain
• Granulomas/papillomas
Phase IV: Immune reconstitution late posttransplant
• Oral infections: mucosal infections (ie, viral, fungal, or bacterial)
• Chronic GVHD
• Dental and skeletal growth and development alterations (pediatric patients)
• Xerostomia
• Relapse-related oral lesions
• Second malignancies
Phase V: Long-term survival
• Relapse or second malignancies
• Dental and skeletal growth and development alterations

(Adapted from the National Cancer Institute.37 )

Acute and Chronic Graft- framework that includes stem cell rescue
of bone marrow injury that is caused by a
Versus-Host Disease conditioning regimen of high-dose chemother-
apy and/or radiotherapy for treatment of the
The HSCT patient represents a unique cohort malignancy. This rescue is achieved by post-
among immunocompromised patients, with a conditioning infusion of hematopoietic cells
broad scope of acute and chronic oral toxic- that are harvested in advance from one of the
ities that may develop1,37,38 (Box 13-3). This following donor sources:
uniqueness is characterized by a conceptual

282
Infection

• Autologous: Patient’s own hematopoietic in severe cases, particularly if multiorgan


stem cells (human leukocyte antigen [HLA] involvement is present.
identical).
• Syngeneic: Patient’s identical twin or triplet’s
hematopoietic cells (HLA identical).
• Allogeneic: Typically a sibling or donor Infection
relative who is genetically similar based
on matching at the HLA locus. In some The dentition, periodontium, and periradicu-
instances, the donor can be unrelated to the lar sites represent potential sources of acute
patient (HLA identical, haploidentical, or local and systemic infection during myelo-
mismatched). suppression.1 The following preexisting oral
lesions can represent clinically significant risk
Patients who undergo allogeneic HSCT may to the patient during the subsequent period of
develop moderate to severe oral mucositis myelosuppression:
caused by the conditioning regimen.29
In addition, graft-versus-host disease • Advanced and/or symptomatic dental caries
(GVHD) can be clinically significant in these • Periapical pathoses symptomatic within the
patients as well.39,40 For example, at least past 90 days
50% of patients develop chronic GVHD • Severe periodontal disease for which prog-
within about 1 year of the transplant proce- nosis of the dentition is poor
dure.37 Risk for development of this condition • Mucosal lesions secondary to trauma from
is directly influenced by the extent to which prosthetic or orthodontic appliances
there is genetic disparity at the HLA locus with
either a related or unrelated donor. The reac- Many of these acute infectious flare-ups can
tion is in part mediated via alloreactive donor be prevented by stabilization of the oral cavity
lymphocytes that recognize and bind to anti- prior to initiation of the chemotherapy. Unfor-
gens associated with salivary glands and oral tunately, there is no definitive prechemotherapy
mucosa. The resultant cytotoxicity can lead to oral care protocol that clearly delineates risk
irreversible oral tissue injury. The oral mucosal for infectious flare-ups in relation to degree and
lesions associated with chronic GVHD are typ- duration of myelosuppression. For example, a
ically lichenoid in appearance, with an erosive recent study reported that oral health status
or ulcerative component in selected cases. As was not associated with risk of bacteremia
such, they mimic naturally occurring mucosal of potential oral source either at the time of
diseases, such as ulcerative lichen planus, pem- acute myelogenous leukemia consolidation or
phigus, and pemphigoid. Oral chronic GVHD in association with allogeneic transplant.44 Fur-
is considered a risk factor for the development thermore, chronic oral foci of infection present
of potentially malignant and malignant oral prior to, for example, initiation of high-dose
mucosal neoplasms over time. Clinically sig- chemotherapy are not necessarily likely to
nificant salivary hypofunction and xerostomia flare during the subsequent period of intense
can develop as a result of chronic GVHD as myelosuppression.44 Clinicians must therefore
well, with a potentially highly adverse impact incorporate clinical judgment in their preche-
on the dentition, periodontium, and mucosa.37 motherapy decision making. The history of
Considerations for topical management of symptoms within the previous 90 days can
the mucosal lesions include steroids,41 immuno- serve as a useful guide in this regard.
suppressants,42 photobiomodulation,43 and/or The oral assessment should occur as early as
oral psoralen and ultraviolet A therapy.37 Sys- feasible in relation to the upcoming chemother-
temic immunosuppression is usually warranted apy. Clear communication of the oral findings

283
13 Oral Complications in the Immunocompromised Patient: The Oncology Prototype

a b

Fig 13-10  |  Chronic inflammatory periodontal disease represents a potential site for acute infection in the my-
elosuppressed chemotherapy patient. (a) Patient who does not have cancer presenting for periodontal surgical
management of severe chronic periodontal disease. (b) Intraoperative clinical photograph of the patient in part
a. The extensive ulceration provides a direct portal for systemic dissemination of colonizing microflora. In the
chemotherapy patient with profound neutropenia (eg, neutrophil count of less than 100/mm3), this systemic
dissemination can result in morbid or fatal infections.

a b

Fig 13-11  |  (a) Acute periodontal infection in a neutropenic cancer patient with a neutrophil count of less than
500/mm3. Classic inflammatory signs, including erythema and purulence, are not clinically prominent because
the number and function of neutrophils are suppressed. (b) Acute periodontal infection in a chemotherapy pa-
tient with a neutrophil count of less than 1,000/mm3. Classic signs of inflammation are more evident because the
neutrophil response is more robust.

among the oncology team is essential, with a condition. The microbiologic flora has been
focus on the extent of preexisting oral disease historically reported as comprising both peri-
in relation to the potential for acute infectious odontopathic organisms as well as acquired
exacerbation during myelosuppression. pathogens associated with morbidity and mor-
tality in the neutropenic cancer patient.45 When
it does occur, the classic signs of inflammation
Acute periodontal infections may be diminished because of the myelosup-
Chronic periodontal disease can undergo pressed condition of the patient. Culturing
acute infectious exacerbation during neutro- of the attached gingiva, as opposed to direct
penia (Figs 13-10 and 13-11). Acute flare-ups subgingival culturing, may not produce mi-
are infrequent despite the widespread occur- crobial specimens reflective of the causative
rence of the predisposing chronic inflammatory microflora.

284
Infection

a b

Fig 13-12  |  (a) Reactivated HSV is typically a self-limiting clinical infection in the immunocompetent patient.
This patient’s lesion has been present for 8 days and is now in the stages of resolution. (b) Reactivated HSV can
be extensive and fatal in a patient who is immunocompromised following HSCT. The compromised mucosal and
circulating immune defenses result in a rapidly progressing, painful, and sometimes fatal systemic viral infection.
(Part b courtesy of Dr Mark M. Schubert, Fred Hutchinson Cancer Research Center, Seattle, Washington.)

Treatment typically consists of oral rinses immunocompromised patients such as those


with 0.13% chlorhexidine digluconate, irri- undergoing HSCT can be life-threatening (Fig
gation of the periodontal pocket with 3% 13-12). Fortunately, antiviral prophylaxis or
hydrogen peroxide, gentle and supervised treatment with thymidine kinase inhibitors
dental brushing and flossing, and use of sys- such as acyclovir or its derivatives is highly
temic broad-spectrum antibiotics if indicated efficacious. Drug-resistant HSV is an infre-
medically. quent occurrence. The infection can codevelop
in the setting of oral mucositis and/or acute
GVHD.38 This concurrent lesion development
Herpes simplex viral infection not only confounds the diagnostic process
The mucosa can also become primarily or sec- but also intensifies the severity of the mucosal
ondarily infected during this myelosuppression. injury.
Examples of clinical infection with herpes sim- As with all infections in this population, early
plex virus (HSV) and Candida are discussed, diagnosis is key to instituting prompt measures
using the HSCT setting as the prototype. Addi- directed to cure. Viral culturing remains the
tional detailed guidelines for the prevention of gold standard, while other testing such as direct
infectious complications among these patients immunofluorescence, immunoassay, and shell
have been provided by Tomblyn et al.46 vial testing may be useful in generating more
The immunocompromised cancer patient is rapid results.
at risk for either reactivation of latent viruses or The development of antiviral medications
activation of newly acquired viruses.47 Latent such as acyclovir, valacyclovir, and their
HSV, varicella zoster virus, and Epstein-Barr derivatives means that the infections in pre-
virus can be reactivated, while cytomegalovirus chemotherapy and pre-HSCT patients who are
infection can result from either reactivation of seropositive for HSV and varicella zoster virus
latent virus or newly acquired virus. can essentially be prevented. Breakthrough
Unlike the self-limiting recurrent HSV infections, should they occur, are typically
infections in otherwise immunocompetent highly responsive to increased doses of the
individuals, reactivation of latent HSV in antiviral medications, because viral resistance

285
13 Oral Complications in the Immunocompromised Patient: The Oncology Prototype

a b

Fig 13-13  |  Pseudomembranous candidiasis can occur in oncology patients secondary to myelosuppression
and/or compromised salivary defense mechanisms. (a) Clinically documented pseudomembranous candidia-
sis in an allogeneic HSCT patient with chronic GVHD involving the major salivary glands. (b) Cytologic smear
demonstrating the dimorphic candidal organism and hyphae.

Box 13-4  |  Characteristics of candidiasis As with herpetiform infections, the risk and
severity of the human papillomavirus lesion
Risk factors are influenced by the duration and depth of
• Myelosuppression the immunocompromised state. While laser
• Mucosal injury removal or cryotherapy may be implemented,
• Salivary compromise the lesions often regress on immune recovery.
• Antibiotics
• Steroids
• Increased length of hospital stay Candidal infection
Diagnosis
• History Opportunistic infections such as candidiasis
• Assessment of risk factors are common in cancer patients experienc-
• Examination ing chemotherapy-induced myelosuppression,48
• Culture as needed including those patients undergoing HSCT47
Treatment (Fig 13-13). In addition to the patient’s im-
• Nonmedicated oral rinse paired immune response, additional risk
• Topical antifungal (systemic therapy if factors include oral mucositis as well as phar-
indicated) macologic- and/or GVHD-induced salivary
• Removal of dentures
gland hypofunction (Box 13-4). The resultant
compromised salivary flow and composition
(eg, lactoferrin, salivary immunoglobulin A,
histatins, transferrin, and mucins) can lead to
pseudomembranous candidiasis and other op-
is rare. Persistent clinical infection is less likely portunistic infections.
to be due to viral resistance than to less than Diagnosis of candidiasis is typically based on
optimal dosing and/or impaired gastrointesti- history and clinical examination. Smear testing
nal absorption of orally administered agents or culturing can be performed if clinically war-
such as acyclovir. ranted (see Fig 13-13b).
Other nonherpes viral infections such Topical oral antifungal therapy with nystatin
as human papillomavirus can occur in the and clotrimazole can be instituted, although the
immunocompromised HSCT patient as well. yeast infection will likely recur on cessation of

286
Medication-Related Osteonecrosis of the Jaw

a b

Fig 13-14 |  MRONJ. (a) Exposed oral bone involving the lingual aspect of the left mandible. (b) Periapical
radiograph 4 months later demonstrating continuing evidence of an osteolytic process. (Courtesy of Dr Cesar A.
Migliorati, University of Florida College of Dentistry, Gainesville, Florida.)

the medication unless the underlying risk factors Medication-Related


are successfully addressed as well. The follow-
ing regimen of topical therapy (7 to 14 days’ Osteonecrosis of the Jaw
treatment) was recommended by Lerman et al49:
Bone-stabilizing agents have high clinical effi-
• Clotrimazole troche (10 mg), four to five cacy relative to reducing risk of skeletal bone
times per day fractures in patients with metastatic bone
• Nystatin oral suspension (100,000 U/mL), disease. In addition, the agents can exert anti­
five mL, four times per day tumor effects in selected cancer cohorts as
• Nystatin pastilles (200,000 U), four to five well.
times per day However, medication-related osteonecrosis
• Fluconazole solution (eg, 10 mg), swished of the jaw (MRONJ) is a key toxicity associ-
and expectorated, three times per day ated with their use and is unique to the oral
• Amphotericin B, oral suspension (100 mg/ cavity50–52 (Fig 13-14). These lesions can occur
mL), 1 mL, four times per day in cancer patients who have been exposed over
many months to one or more of the following
Systemic antifungal management with agents three classes of bone-stabilizing agents, which
such as fluconazole is typically indicated for are in turn associated with three different
treatment of persistent oral infection as well as molecular mechanisms:
for patients with profound immunosuppression.
In addition to candidal infection, other types • Bisphosphonates, osteoclast inhibitors via
of fungal infection can occur, including lesions mevalonate pathway
caused by Aspergillus, Mucor, and Rhizopus • Denosumab, an inhibitor of the receptor
species. Culturing is essential for diagnosis, activator of nuclear factor κ B (RANK)–
because these lesions may mimic the clinical RANK ligand (RANKL) pathway
appearance of nonyeast toxicities in HSCT • Angiogenesis inhibitors, including vaccular
patients. endothelial growth factor (VEGF) inhibitors

287
13 Oral Complications in the Immunocompromised Patient: The Oncology Prototype

Despite the three uniquely different molecu- Conclusion


lar mechanisms, the incidence as well as clinical
prevention and management of MRONJ in Oral complications can adversely affect the
cancer patients are comparable, regardless of quality of life of myelosuppressed cancer pa-
type of bone-stabilizing agent being utilized. tients. This impact can range from mild and
MRONJ is defined as the unexpected devel- easily tolerated to severe and debilitating. In
opment of necrotic bone anywhere in the oral some cases, the toxicities can decrease the can-
cavity of a patient who has been exposed or cer patient’s chances of survival. In addition to
is currently taking a bone-modifying agent, an the prototypic modeling of oral complications
angiogenesis inhibitor, or both and who has associated with myelosuppression resulting
not received radiation therapy to the head from conventional high-dose chemotherapy,
and neck. MRONJ is persistent and does the increasing use of targeted cancer therapies
not respond within 6 to 8 weeks to standard in recent years has led to the emergence of new
therapy. toxicity profiles. This latter toxicity expression
Current guidelines for prevention and includes a unique form of oral mucosal injury
management are based upon several key prin- for which the clinical phenotype is distinctly
ciples.52 Oral disease should be eliminated different from that of oral mucositis caused
or stabilized prior to initiation of the bone- by conventional chemotherapeutic regimens.
stabilizing therapy, whenever possible. Cancer Treatment of the conditions can be expen-
patients receiving these agents should be evalu- sive, including the cost of prolonged hospital
ated by a dentist based on a systematic timeline stays and supportive care interventions such as
developed in collaboration with the patient’s infection management and nutritional support.
medical oncologist. In addition, daily mainte- Fortunately, substantial and innovative
nance of optimal oral hygiene by the patient is research in recent years has provided new
essential. MRONJ staging is important in order insights into molecular-based causation.
to provide a basis for appropriate level of oral These discoveries are setting the stage for the
management (see Table 12-5). development of novel preventive and ther-
If MRONJ should occur, the lesion should apeutic technologies for future use in the
be managed by health professionals with expe- clinical setting. Researchers and clinicians
rience in managing such lesions. For MRONJ from dental medicine are taking the lead
stages 1 and 2, conservative treatment with role in these scientific advances for the pre-
periodic clinical evaluation of the progress of vention and management of some toxicities,
disease in MRONJ is typically recommended. such as mucositis. In recent years, interdisci-
Use of systemic antibiotics is recommended for plinary collaborations promoted via national
patients with active infection and/or clinical and international organizations of health
paresthesia. Oral rinses with 0.12% or 2.0% professionals have contributed to the trans-
chlorhexidine digluconate two to four times per lation of clinically applicable outcomes into
day should be considered to enhance wound high-quality, evidence-based guidelines for
care. For MRONJ stage 3, more aggressive oncology practice.
surgical intervention may be necessary. Top- The presentation of this oncology paradigm
ical or systemic pain management should be has been designed to highlight these issues in
implemented if clinically appropriate to do so. the context of dental medicine research and
clinical care. Insights and lessons learned from
this modeling may well be applicable to other
patient cohorts in whom disease and/or its
treatment has led to immunocompromise.

288
References

Clinical Considerations: What You Can Take Back to Your Practice


Is there a biologic plausibility for an association The delivery of medically necessary oral health
between acute and/or chronic oral disorders and care prior to and during the period of myelo-
systemic health?  An intact immune response suppression can mitigate selected constituents
is an essential component of maintaining ho- of the oral toxicity profile. An example of this
meostasis that is necessary for oral health. The benefit to patients includes reducing the risk for
oncology patient experiencing chemotherapy- opportunistic mucosal infection in the setting of
induced myelosuppression serves as a key pro- oral mucositis caused by cancer chemotherapy.
totype for how compromised host defenses can
lead to the emergence of clinically significant Is it safe to provide dental care to patients who
oral toxicities. There is thus a profound bio- are immunocompromised?  Clear and evidence-
logic plausibility regarding the association be- based consultation between the dental provider
tween oral health and the immunosuppressed and the oncology team is essential to provide
oncology patient. safe and effective, medically necessary oral care
in these patients. Considerations include po-
Are oral infections independent risk factors for tential risks and benefits associated with each
the development of adverse systemic outcomes specific dental intervention, the patient’s hema-
in the immunocompromised cancer patient?  tologic trajectory including maximum nadir of
Acute oral mucosal infections can occur as a white blood cells, and the degree to which nor-
result of the compromised immune surveillance mal immune function is projected to recover in
and function. Immunosuppression can thus be the future.
viewed as an independent risk factor in these pa-
tients. The infections can be caused by preexist- What should a dental practitioner inform a pa-
ing colonizing microorganisms, newly acquired tient about the association between oral health
pathogens, and/or reactivation of latent viruses. versus oral disease and the development of
systemic outcomes that may arise during peri-
Can treatment of selected oral infections and ods of immunocompromise?  Acute oral com-
related disorders prior to immunocompro- plications arising in myelosuppressed oncolo-
mise reduce the risk for the development of gy patients can cause considerable morbidity
adverse systemic outcomes during the immu- and, in selected cases, can be fatal. The current
nocompromised phase of patient treatment? high-quality basic, translational, and clinical
Similarly, can prompt diagnosis and early research evidence base represents an excellent
treatment of acute oral infection during im- foundation for providing state-of-the-science,
munocompromise reduce risk for subsequent, medically necessary safe and effective dental
more severe oral infection during this phase?  care to these patients.

References
1. National Institutes of Health, National Cancer Institute. 3. Sonis ST, Elting LS, Keefe D, et al. Perspectives on
Oral Complications of Chemotherapy and Head/Neck cancer therapy-induced mucosal injury: Pathogenesis,
Radiation. http://www.cancer.gov/cancertopics/pdq/ measurement, epidemiology, and conse­quences for
supportivecare/oralcomplications/HealthProfessional. patients. Cancer 2004; 100(9 suppl):1995–2025.
Accessed 18 May 2018. 4. Peterson DE, Keefe DM, Sonis ST. New frontiers in
2. Peterson DE, Srivastava R, Lalla RV. Oral mucosal injury mucositis. Am Soc Clin Oncol Educ Book 2012;
in oncology patients: Perspectives on maturation of a 545–551.
field. Oral Dis 2015;21:133–141. 5. Sonis ST. Pathobiology of oral mucositis: Novel insights
and opportunities. J Support Oncol 2007;5(9 suppl 4):
3–11.

289
13 Oral Complications in the Immunocompromised Patient: The Oncology Prototype

6. Al-Dasooqi N, Sonis ST, Bowen JM, et al. Emerging 20. Mougeot JL, Bahrani-Mougeot FK, Lockhart PB, Bren-
evidence on the pathobiology of mucositis. Supportive nan MT. Microarray analyses of oral punch biopsies from
Care Cancer 2013;21:3233–3241. acute myeloid leukemia (AML) patients treated with
7. Carlotto A, Hogsett VL, Maiorini EM, Razulis JG, Sonis chemotherapy. Oral Surg Oral Med Oral Pathol Oral
ST. The economic burden of toxicities associated with Radiol Endod 2011;112:446–452.
cancer treatment: Review of the literature and analysis 21. Mougeot JL, Mougeot FK, Peterson DE, Padilla RJ,
of nausea and vomiting, diarrhoea, oral mucositis and Brennan MT, Lockhart PB. Use of archived biopsy spec-
fatigue. Pharmacoeconomics 2013;31:753–766. imens to study gene expression in oral mucosa from
8. Sonis ST, Oster G, Fuchs H, et al. Oral mucositis and chemotherapy-treated cancer patients. Oral Surg Oral
the clinical and economic outcomes of hematopoietic Med Oral Pathol Oral Radiol 2013;115:630–637.
stem-cell transplantation. J Clin Oncol 2001; 19: 22. Calantog A, Hallajian L, Nabelsi T, et al. A prospective
2201–2205. study to assess in vivo optical coherence tomography
9. Sonis ST. New thoughts on the initiation of mucositis. imaging for early detection of chemotherapy-
Oral Dis 2010;16:597–600. induced oral mucositis. Lasers Surg Med 2013; 45:
10. Triantafyllou K, Dervenoulas J, Tsirigotis P, Ladas SD. 22–27.
The nature of small intestinal mucositis: A video-capsule 23. Diaz PI, Dupuy AK, Abusleme L, et al. Using high
endoscopy study. Support Care Cancer 2008; 16: throughput sequencing to explore the biodiversity in oral
1173–1178. bacterial communities. Mol Oral Microbiol 2012; 27:
11. Yasuda M, Kato S, Yamanaka N, et al. 5-HT3 receptor 182–201.
antagonists ameliorate 5-fluorouracil-induced intestinal 24. Michelson S, Sehgal A, Friedrich C. In silico prediction
mucositis by suppression of apoptosis in murine intes- of clinical efficacy. Curr Opin Biotechnol 2006; 17:
tinal crypt cells. Br J Pharmacol 2013; 168: 666–670.
1388–1400. 25. Molina F, Dehmer M, Perco P, et al. Systems biology:
12. Sonis ST. Efficacy of palifermin (keratinocyte growth Opening new avenues in clinical research. Nephrol Dial
factor-1) in the amelioration of oral mucositis. Core Evid Transplant 2010;25:1015–1018.
2010;4:199–205. 26. Sonis ST, Antin J, Tedaldi MW, Alterovitz G. SNP-based
13. Niscola P, Tendas A, Cupelli L, et al. The prevention of Bayesian networks can predict oral mucositis risk in
oral mucositis in patients with blood cancers: Current autologous stem cell transplant recipients. Oral Dis
concepts and emerging landscapes. Cardiovasc Hema- 2013;19:721–727.
tol Agents Med Chem 2012;10:362–375 27. Elting LS, Keefe DM, Sonis ST, et al. Patient-reported
14. Cotrim AP, Yoshikawa M, Sunshine AN, et al. Pharma- measurements of oral mucositis in head and neck
cological protection from radiation ± cisplatin-induced cancer patients treated with radiotherapy with or without
oral mucositis. Intl J Rad Oncol Biol Phys 2012; 83: chemotherapy: Demonstration of increased frequency,
1284–1290. severity, resistance to palliation, and impact on quality
15. Zheng C, Cotrim AP, Sunshine AN, et al. Prevention of of life. Cancer 2008;113:2704–2713.
radiation-induced oral mucositis after adenoviral 28. Peterson DE, O’Shaughnessy JA, Rugo HS, et al. Oral
vector-mediated transfer of the keratinocyte growth mucosal injury caused by mammalian target of rapa-
factor cDNA to mouse submandibular glands. Clin mycin inhibitors: Emerging perspectives on pathobiology
Cancer Res 2009;15:4641–4648. and impact on clinical practice. Cancer Med 2016; 5:
16. Colley HE, Eves PC, Pinnock A, Thornhill MH, Murdoch 1897–1907.
C. T issue engineered oral mucosa to study 29. National Institutes of Health, National Cancer Institute.
radiotherapy-induced oral mucositis. Int J Rad Biol Oral Mucositis. https: //www.cancer.gov/about-
2013;89:907–914. cancer/treatment/side-effects/mouth-throat/oral-
17. Sonis S, Haddad R, Posner M, et al. Gene expression complications-hp-pdq#link/_337. Accessed 25 May
changes in peripheral blood cells provide insight into 2018.
the biological mechanisms associated with 30. Xie J, Wang X, Proud CG. mTOR inhibitors in cancer
regimen-related toxicities in patients being treated for therapy. F1000Res 2016;5.
head and neck cancers. Oral Oncol 2007;43:289–300. 31. Vigarios E, Epstein JB, Sibaud V. Oral mucosal changes
18. Al-Dasooqi N, Bowen JM, Gibson RJ, Logan RM, induced by anticancer targeted therapies and immune
Stringer AM, Keefe DM. Selection of housekeeping checkpoint inhibitors. Support Care Cancer 2017; 25:
genes for gene expression studies in a rat model of 1713–1739.
irinotecan-induced mucositis. Chemotherapy 2011;57: 32. Hait WN, Hambley TW. Targeted cancer therapeutics.
43–53. Cancer Res 2009;69:1263–1267.
19. Hahn T, Zhelnova E, Sucheston L, et al. A deletion poly- 33. Pilotte AP, Hohos MB, Polson KM, Huftalen TM, Treister
morphism in glutathione-S-transferase mu (GSTM1) N. Managing stomatitis in patients treated with mamma-
and/or theta (GSTT1) is associated with an increased lian target of rapamycin inhibitors. Clin J Oncol Nurs
risk of toxicity after autologous blood and marrow trans- 2011;15:E83–E89.
plantation. Biol Blood Marrow Transplant 2010; 16:
801–808.

290
References

34. Sonis S, Treister N, Chawla S, Demetri G, Haluska F. 43. Epstein JB, Raber-Durlacher JE, Huysmans MC, et al.
Preliminary characterization of oral lesions associated Photobiomodulation therapy alleviates tissue fibroses
with inhibitors of mammalian target of rapamycin in associated with chronic graft-versus-host disease: Two
cancer patients. Cancer 2010;116:210–215. case reports and putative anti-fibrotic roles of TGF-β.
35. Peterson DE, Boers-Doets CB, Bensadoun RJ, Herrst- Photomed Laser Surg 2018;36:92–99.
edt J, ESMO Guidelines Committee. Management of 44. Schuurhuis JM, Span LF, Stokman MA, van Winkelhoff
oral and gastrointestinal mucosal injury: ESMO Clinical AJ, Vissink A, Spijkervet FK. Effect of leaving chronic
Practice Guidelines for diagnosis, treatment, and oral foci untreated on infectious complications during
follow-up. Ann Oncol 2015;26(5 suppl):v139–v151. intensive chemotherapy. Br J Cancer 2016; 114:
36. Chambers MS, Rugo HS, Litton JK, Meiller TF. Stomatitis 972–978.
associated with mammalian target of rapamycin inhibi- 45. Peterson DE, Minah GE, Overholser CD, et al. Microbi-
tion: A review of pathogenesis, prevention, treatment, ology of acute periodontal infection in myelosuppressed
and clinical implications for oral practice in metastatic cancer patients. J Clin Oncol 1987;5:1461–1468.
breast cancer. J Am Dent Assoc 2018;149:291–298. 46. Tomblyn M, Chiller T, Einsele H, et al. Guidelines for
37. National Institutes of Health, National Cancer Institute. preventing infectious complications among hematopoi-
Graft-Versus-Host Disease. https: //www.cancer.gov/ etic cell transplantation recipients: A global perspective.
about-cancer/treatment/side-effects/mouth-throat/ Blood Marrow Transpl 2009;44:453–455.
oral-complications-hp-pdq#link/_271. Accessed 25 47. National Institutes of Health, National Cancer Institute.
May 2018. Infection. https://www.cancer.gov/about-cancer/treat-
38. Schubert MM, Correa MEP, Peterson DE. Oral compli- ment/side-effects/mouth-throat/oral-complications-
cations of hematopoietic cell transplantation. In: Forman hp-pdq#link/_338. Accessed 25 May 2018.
SJ, Negrin RS, Antin JH, Appelbaum FR (eds). Thomas’ 48. Maheronnaghsh M, Tolouei S, Dehghan P, Chadega-
Hematopoietic Cell Transplantation, ed 5. London: nipour M, Yazdi M. Identification of Candida species in
Wiley-Blackwell, 2016:1242–1258. patients with oral lesion undergoing chemotherapy along
39. Martin PJ, Lee SJ, Przepiorka D, et al. National Institutes with minimum inhibitory concentration to fluconazole.
of Health Consensus Development Project on Criteria Adv Biomed Res 2016;5:132.
for Clinical Trials in Chronic Graft-versus-Host Disease: 49. Lerman MA, Laudenbach J, Marty FM, Baden LR, Treis-
VI. The 2014 Clinical Trial Design Working Group Report. ter NS. Management of oral infections in cancer patients.
Biol Blood Marrow Transplant 2015;21:1343–1359. Dent Clin North Am 2008;52:129–153.
40. Carpenter PA, Kitko CL, Elad S, et al. National Institutes 50. Ruggiero SL, Dodson TB, Fantasia J, et al. American
of Health Consensus Development Project on Criteria Association of Oral and Maxillofacial Surgeons position
for Clinical Trials in Chronic Graft-versus-Host Disease: paper on medication-related osteonecrosis of the
V. The 2014 Ancillary Therapy and Supportive Care jaw—2014 update. J Oral Maxillofac Surg 2014; 72:
Working Group Report. Biol Blood Marrow Transplant 1938–1956.
2015;21:1167–1187. 51. Ruggiero SL. Management of medication-related osteo-
41. Zadik Y, Elad S, Shapira A, Shapira MY. Treatment of necrosis of the jaw. Preface. Oral Maxillofac Surg Clin
oral mucosal manifestations of chronic graft-versus-host North Am 2015;27:ix.
disease: Dexamethasone vs. budesonide. Expert Opin 52. Ruggiero SL, Kohn N. Disease stage and mode of ther-
Pharmacother 2017;18:235–242. apy are important determinants of treatment outcomes
42. Reikvam H, Gronningsaeter IS, Mosevoll KA, Lindas R, for medication-related osteonecrosis of the jaw. J Oral
Hatfield K, Bruserud O. Patients with treatment- Maxillofac Surg 2015;73(12 suppl):S94–S100.
requiring chronic graft versus host disease after alloge-
neic stem cell transplantation have altered metabolic
profiles due to the disease and immunosuppressive
therapy: Potential implication for biomarkers. Front
Immunol 2017;8:1979.

291
CHAPTER 14

Oral-Systemic Health:
The Genomic Connection
John R. Shaffer, phd
Robert J. Weyant, ms, dmd, drph

The interdisciplinary field of human genomic answers to these questions are difficult to pre-
science has grown rapidly over the last two dict, in part because of how rapidly the state
decades as key technologic developments have of the science has evolved. The overwhelming
led to landmark projects and a newfound ca- trend in genomic science is one of increasing
pacity to generate vast quantities of biologic complexity, with new genomic discoveries of-
data. In turn, these developments have led to ten revealing that the gap between basic science
the birth of new fields of study and accelerated and clinical application is wider than previ-
the pace of biomedical research. We find our- ously thought. The reality in dentistry is that
selves in a world where whole genomes can genomic science has of yet not contributed in
be quickly and cheaply sequenced. Dynamic substantial ways to direct improvements in
cellular processes such as gene transcription, routine patient care, but this will not always
translation, and protein-protein interactions be so. We are confident that the benefits that
can be assayed; three-dimensional structures of come from the precision medicine initiative will
the genome and protein products can be exper- ultimately contribute in important ways to im-
imentally and computationally modeled; and proved dental care as well.
epigenetic modifications such as DNA meth- As we approach the study of the genetic basis
ylation can be measured and studied, all at of oral disease and its possible linkage with
the scale of the genome. To accommodate the systemic disease, it should be kept in mind that
immensity of the data generated, information almost all diseases arise through some inter-
storage systems have been stretched to the action of environmental (eg, social, chemical),
point where the cost of storing these data may behavioral (eg, smoking, exercise), and biologic
in some cases exceed the cost of simply regen- (eg, genetic) mechanisms. It is important not to
erating the data from a stored biologic sample. limit our thinking about common risks among
And despite the numerous computational and various diseases to only one domain or mech-
bioinformatics tools that have been developed anism. In this chapter we review genetic risk,
to manage, analyze, and interpret these data, but in most cases, genetic risk can be highly
our current capacity to produce genomic data variable based on interactions with environ-
far outstrips our ability to make sense of it. mental and behavioral factors.
How will this “tsunami of data” impact bio- In this chapter we focus on issues of impor-
medical science? How will genomic data be tance to clinical dentists and patient care. We
harnessed to inform clinical care? The specific start with the current state of genomic science

292
State of Genomic Science

by describing the technologies, research meth- have formed the foundation of the field. Among
odologies, and conceptual approaches used to these formative works include the Human
investigate the genetic basis of disease. Next, Genome Project, the International HapMap
we briefly review our understanding of the Project, the 1000 Genomes Project, and the
genomic basis of selected oral diseases and Encyclopedia of DNA Elements (ENCODE).
where genetic research may suggest that oral
and systemic disease share a common patho-
physiology or causal relationship. Finally, we The Human Genome Project
touch on how our growing understanding Launched in 1990 based on plans formed in
of the genetic basis of oral diseases will play the mid-1980s, this international initiative to
an expanding role in clinical dentistry in the sequence the human genome was audacious in
future. scope and difficulty given the technology of the
time. Completion of the first rough draft3,4 of
the human genome sequence was announced
with much fanfare in 2000 by US President Bill
State of Genomic Science Clinton and British Prime Minister Tony Blair.
A more comprehensive (ie, covering 99.7% of
It was not long after the initial research on the euchromatic genomic regions) and greatly
DNA sequencing in the late 1950s that the improved draft of the human genome sequence
genes responsible for many diseases began to was completed in 2004.5 This reference provid-
be identified.1 This early research focused on ed the foundation for seminal discoveries in
identifying monogenic diseases (ie, those de- many areas, including shaping our understand-
rived from mutations in a single gene), and now ing of the composition and structure of the
over 4,000 such disease genes have been identi- human genome itself, advancing our knowledge
fied. Most of these diseases are associated with of genetic variation across human populations,
serious morbidity but also tend to be rare in revealing new insights into human evolution,
the general population. It took several more and greatly forwarding our understanding of
decades for the technology to mature and allow how specific genetic variants impact human
for the study of polygenic (ie, contributions of health and disease. These far-reaching impli-
many genetic variants of small individual ef- cations are all based on the creation of a fairly
fects) disease. simple information resource: the reference se-
The diseases that carry the most social con- quence of approximately 3 billion nucleic acid
sequences due to their high prevalence in the base pairs in the human genome.
general population are polygenic diseases, often
referred to as common complex diseases. The
National Institutes of Health (NIH) describes The International HapMap and
common complex disease as disease thought to 1000 Genomes projects
be caused by many, often hundreds, of genes
in conjunction with behavioral and environ- With the human reference genome complete,
mental factors.2 These diseases tend to be the International HapMap Project6,7 (2002 to
major causes of morbidity and mortality in the 2009) was conceived to map the haplotype
general population and include heart disease, (ie, inheritance of multiple genetic variants to-
certain cancers, stroke, and diabetes, to name gether as a group from a single parent) and
a few. Dental caries and chronic periodontitis correlational structure of the human genome,
also fit into this category. representing important patterns of human ge-
To shed light on the current state of genomic netic variation. Eleven ancestry groups from
science, consider some seminal projects that across the world were represented, including

293
14 Oral-Systemic Health: The Genomic Connection

European Americans, African Americans, the human reference sequence, and, along with
Chinese Americans, Mexican Americans, and some later large-scale sequencing initiatives,
Gujarati Indians from the United States, as well provided the imputation base for many contem-
as people from China, Japan, Nigeria, Kenya, porary GWASs. The major contribution of the
and Italy. Whereas the Human Genome Proj- 1000 Genomes Project was to provide a more
ect provided an initial reference point for the comprehensive catalog of human genetic varia-
genomic sequence, the International HapMap tion than previously known, which has proved
Project provided detailed information about to be a valuable resource for disciplines such
the differences in this sequence between indi- as genomics, bioinformatics, human evolution,
viduals and between populations, and it greatly genetic epidemiology, and pharmacogenomics.
expanded (by many millions) the list of known
polymorphic sites in the human genome.
Comprehensive knowledge of such common ENCODE
human genetic variants was instrumental Another important follow-up to the Human
in informing strategies for conducting unbi- Genome Project is the ENCODE project,10
ased, whole-genome scans to identify variants an ongoing initiative to catalog the function-
associated with human disease, which came to al elements of the genome—that is, all of the
be known as genome-wide association studies non–protein-coding content of the genome
(GWASs). A key advancement provided by the involved in regulating gene activity. DNA ele-
International HapMap Project was its utility ments include gene promoters, transcriptional
as the first “imputation base”—that is, a ref- regulatory sequences such as transcription
erence of the correlation across polymorphic factor binding sites, and regions affecting
sites, which allowed researchers to impute al- chromatin structure and histone modifica-
leles of unobserved genetic loci based on the tions, all of which can be cell type specific.
loci genotyped in an experimental sample. In An array of methods, many based on recent
practice, this allowed researchers to determine advancements in high-throughput sequencing
genotypes for 2.5 to 5 million genetic poly- technologies, have been used to identify these
morphisms based on the data experimentally elements (Fig 14-1). ENCODE and its ancil-
observed by genotyping 200,000 to 500,000 lary projects, as well as other similar initiatives
polymorphisms. In other words, genotyping such as the Function Annotation of the Mam-
projects could leverage the HapMap reference malian Genome (FANTOM)11 and Roadmap
data to collect much higher-resolution genet- Epigenomics projects,12 have provided founda-
ic data based on lower-resolution genotyping. tional insights into the regulatory structure of
Thus, the resource provided by the Interna- the human genome and have provided needed
tional HapMap Project helped make GWASs tools for designing genetic studies and anno-
feasible, allowing for hypothesis-free associ- tating and interpreting their results.
ation scans for genetic variants influencing
human health and disease.
As a successor to the International Hap- Advances in the field
Map Project, with a similar goal of cataloging These seminal projects and others have rap-
human genetic variation, the 1000 Genomes idly reshaped our current understanding of
Project8,9 was launched in 2008 and employed the human genome and its roles in health and
newly developed high-throughput sequencing disease. For example, prior to the Human
technologies to sequence the genomes of over Genome Project, most estimates for the num-
a thousand people across multiple ethnicities. ber of human protein-coding genes ranged
This project yielded deeper understanding of from 100,000 to 200,000, whereas the initial
the genetic variation of humans, helped refine reference sequence suggested this number was

294
State of Genomic Science

Hypersensitive sites CH3


CH3

RNA polymerase

CH3CO

Hi-C DNase-seq ChlP-seq WGBS ENCODE RNA-seq eCLIP


ChlA-PET ATAC-seq –TF RRBS Encyclopedia RAMPAGE Bind-n-Seq
Repli-seq –Histone methyl array –Registry of cREs miRNA-seq
–SCREEN

Genes

Long-range regulatory elements Promoters


(enhancers, repressors/silencers, insulators) Transcripts

Fig 14-1  |  The ENCODE project aims to catalog the functional elements of the human genome. The organi-
zational structure of the genome is depicted from chromosome to double helix. The blue boxes represent the
methods and tools, mostly based on high-throughput sequencing technologies, used to collect multiomics data
on functional elements. Black arrows represent the correspondence between method, target of the assay, and
functional element. (Reprinted from Daryl Leja and Michael Pazin [National Human Genome Research Institute],
Ian Dunham [European Bioinformatics Institute], and the ENCODE Consortium with permission.) Inclusion of
this figure does not imply endorsement by the NIH, the National Human Genome Research Institute, or EN-
CODE for any particular viewpoint expressed in this chapter. Methods: Hi-C, high-throughput chromosome
conformation capture, a method for determining the three-dimensional architecture of chromatin; ChIA-PET,
chromatin interaction analysis by paired-end tag sequencing, a method for determining long-range chromatin
interactions; Repli-seq, a method for determining replication timing of genomic regions; DNase-seq, DNase I
hypersensitivity sites sequencing, a method for determining the location of regulatory regions; ATAC-seq, assay
for transposase-accessible chromatin sequencing, a method for determining chromatin accessibility; ChIP-seq,
chromatin immunoprecipitation sequencing, a method for determining the binding sites for a protein of interest
such as a transcription factor (TF) or for determining histone modifications; WGBS, whole genome bisulfite se-
quencing, a method for determining the DNA methylation status of cytosines across the whole genome; RRBS,
reduced representation bisulfite sequencing, a method for determining the DNA methylation status of cytosines
for regions of the genome (comprising about 1% of the genome) with high CpG content; methyl array, a method
for determining the DNA methylation status of cytosines for a predetermined list of CpG sites (currently about
850,000); cREs, candidate regulatory elements cataloged in the ENCODE encyclopedia; SCREEN, a web inter-
face for searching near-acting cREs derived from ENCODE data; RNA-seq, RNA sequencing, a method for de-
termining gene expression based on the presence and quantity of RNA molecules; RAMPAGE, RNA annotation
and mapping of promoters for analysis of gene expression, a method for determining transcription start sites;
miRNA-seq, microRNA sequencing, a method for determining the presence and quantity of small regulatory
RNA molecules; eCLIP, enhanced crosslinking and immunoprecipitation, a method for determining RNA-binding
protein targets; Bind-n-Seq, a method for determining protein-RNA interactions.

closer to 30,000 to 40,000 protein-coding genes comprising only 1.5% of the genome.
genes. Further work has shown that even this In addition, we now believe that the genome
estimate was high, as there is currently believed contains about 11,000 pseudogenes (ie, DNA
to be approximately 20,000 protein-coding sequences related to and resembling genes but

295
14 Oral-Systemic Health: The Genomic Connection

CH3 CH3 CH3 CH3 CH3


O = C  O = C  O = C  O = C  O = C 
Active
Active
chromatin
chromatin

CH3 CH3 CH3 CH3 CH3 CH3 CH3 CH3 CH3 CH3

Inactive
chromatin

CH3 CH3 CH3 CH3 CH3 CH3 CH3 CH3 CH3 CH3 CH3

Fig 14-2  |  Transcription is regulated, in part, by the state of the chromatin. The nucleosomes comprise DNA
(blue) wrapped around eight histone proteins (orange). Transcription is possible for active (open) chromatin (top),
characterized by unmethylated cytosines (white circles) and acetylated histones. Transcription is impeded for in-
active (silent) chromatin (bottom), characterized by methylated cytosines (red circles) and deacetylated histones.
Histone methylation (not depicted) also differs between active and inactive chromatin.

having lost some functionality) and 30,000 to associated with “switching on” regions of the
60,000 noncoding RNAs (ie, transcribed and genome by activating the chromatin (ie, open-
putatively functional sequences that are not ing the conformation of the DNA so that it is
translated into protein; sometimes referred to physically accessible to the cellular machinery)
as RNA genes). We now know that the rest of to make transcription possible, or conversely,
the genome, which does not code any genes “switching off” regions of the genome by
and had traditionally been regarded as “junk silencing the chromatin (ie, tightly packaging
DNA,” is filled with functional sequences. In the DNA in a closed conformation) to prevent
fact, 80% of the noncoding sequence of the transcription (Fig 14-2).
genome is functional in the sense that it is These insights into the composition and
involved in biochemical activities relevant to structure of the human genome have also
biomedical sciences.13 Many of these sequences reshaped our understanding of the genetic
have importance roles, such as providing the basis of human disease. Whereas genetic dis-
landing sites for regulatory proteins involved ease had traditionally been viewed as the result
in determining when and where genes are of a disruption of a protein-coding gene that
expressed. Other sequences affect the pack- was passed on through families under rela-
aging of the genome within the nucleus of tively simple inheritance models (eg, dominant,
the cell, which is itself a major modality of recessive, sex-linked), we now know that the
gene regulation. For example, epigenetic DNA contributions of genetics to human disease also
modifications, such as histone methylation, his- operates through regulatory variants and epi-
tone acetylation, and DNA methylation, are genetic modifications. Under this perspective,

296
State of Genomic Science

$100,000,000

Moore’s law
$10,000,000
Cost per genome (US $)

$1,000,000

$100,000

$10,000

$1,000
2001 2003 2005 2007 2009 2011 2013 2015 2017

Year

Fig 14-3  |  The cost of sequencing a human genome has decreased substantially over the last 15 years, far
outstripping Moore’s law, originally postulated as a doubling of the number of transistors on a computer mi-
crochip every 2 years; it is shown here as cutting the cost in half every 2 years (orange line). These data were
provided by the National Human Genome Research Institute.14

many common complex diseases, including generate relative short (ie, up to a few hun-
dental caries and periodontitis, have been dred base pairs), partially overlapping, snippets
studied to identify the specific genetic variants of DNA sequence. Billions of these snippets,
contributing to disease susceptibility. In fact, known as reads, must then be assembled into
an important theme that has emerged from one continuous sequence guided by their partial
genomic studies is that the majority (about overlap, which is a computationally intensive
90%) of disease-associated variants are reg- task made possible by concurrent advance-
ulatory in nature rather than protein-coding. ments in computing power. Improvements in
These studies have been made possible through sequencing technology have caused the cost of
the use of high-throughput genotyping and sequencing a whole human genome to plum-
sequencing technologies, many of which were met, from its initial price point of about $100
developed or honed via the seminal genomics million at the end of the Human Genome
projects described earlier. Project to about $1,000 in 2015 (Fig 14-3).
Among the most important genomic technol- This drop in price put clinical applications
ogies of the last decade was the development of next-generation sequencing on the table,
of high-throughput sequencing methods, including applications targeting malignancies,
also known as massively parallel sequencing undiagnosed genetic syndromes, and prenatal
or next-generation sequencing. These terms testing.
are usually intended to refer to any of a few Next-generation sequencing technolo-
separate technologies, which, in contrast to gies have also formed the basis for other
traditional Sanger sequencing developed in the high-throughput assays employed in bio-
1970s, can be used to sequence larger amounts medical research today. For example, RNA
of DNA faster and at much lower cost per sequencing uses next-generation sequencing
nucleotide. Most of these methods are “short to assess gene expression by measuring the
read” sequencing methods, meaning that they presence and quantity of mRNA transcripts

297
14 Oral-Systemic Health: The Genomic Connection

in a biologic sample. Similarly, chromatin common genetic variation in humans obtained


immunoprecipitation (ChIP) sequencing uses through the seminal genomics projects such
next-generation sequencing to assay the physi- as the International HapMap Project and
cal interactions of target proteins with DNA by similar efforts to catalog human genetic vari-
immunoprecipitating the target proteins after ation. These SNP chip technologies, together
crosslinking them to the DNA. The immuno- with public reference data sets necessary for
precipitated DNA regions are then sequenced imputing unmeasured genetic variation, made
to identify the binding sites of the target pro- GWASs possible, which have become the most
teins. Analogous methods have been developed widely used genomic approach and have been
to use next-generation sequencing to identify employed to study the genetics of a great many
DNA methylation states and protein-RNA human traits and diseases; approximately
interactions at the genome scale. Altogether, 15,000 genetic associations have been observed
these technologies have extended the field of to date, as reported in the GWAS Catalog.15
genomics into DNA-adjacent “-omics” are- GWASs have become a preferred approach
nas, such as transcriptomics, regulomics, and for studying human genetics because it is
epigenomics. These next-generation sequenc- hypothesis free. That is, GWASs do not require
ing–based technologies have facilitated the any a priori assumptions about the genetics
collection of vast quantities of data, which has of a trait of interest, such as which genes are
provided opportunities to pursue new scientific involved or how the trait is inherited. Instead,
questions and posed new challenges related to the GWAS approach is to scan the whole
data management, analysis, and interpretation. genome by testing each genetic variant indi-
In fact, based on the degree to which they have vidually for statistical association with a trait
impacted the operation of biomedical research, of interest, with careful interpretation of the
DNA sequencing–based technologies have been results given the fact that many tests were
dubbed “the new microscope” in recognition performed. Typically, GWAS efforts focus on
of their transformative role in allowing us to the contributions of common genetic variants
“see” into the inner workings at the level of occurring at frequencies greater than 1% in the
the tissue and cell. population under study. The goal of GWASs is
In addition to DNA sequencing technologies, to identify genetic variants that are associated
another important advancement in genomics with the trait in order to nominate genes and/
was the development of high-throughput geno- or regulatory elements for further study. To
typing technologies. In contrast to sequencing, date, GWASs have been performed for several
where all nucleotides are observed, genotyping oral health conditions, including dental caries,
technologies target only the genetic variants chronic periodontitis, aggressive periodontitis,
already known to differ between individu- periodontal pathogen colonization, temporo-
als, which are the sites of greatest interest in mandibular disorder, and nonsyndromic
explaining the differences in phenotype, such orofacial clefts. Likewise, GWASs have been
as diseases, among people. Genotyping arrays used to study most systemic human diseases.
have been designed that simultaneously cap-
ture information on 200,000 to 5 million
single nucleotide polymorphisms (SNPs; ie,
differences in the nucleotide base—A, T, C, Genetics and Oral Disease
G—at a given position in the sequence) for low
cost (roughly $50 to $200 per DNA sample). The fact that certain oral diseases covary
Development of these “SNP chips” was made with certain systemic diseases has motivated
possible by advancements in the genotyping research to identify common underlying patho-
technology itself, as well as knowledge of the physiologic mechanisms. Identifying a shared

298
Genetics and Oral Disease

pathophysiology, it is hoped, could lead to a caries scores in addition to affected/unaffected


common treatment and improved efficiencies status.
in patient care, including improved patient out- These efforts were fruitful in identifying
comes and reduced health system costs. Below significantly associated genetic variants. And
we discuss the genetics research related to den- while the majority of GWASs of dental caries
tal caries and periodontal disease. have focused on white populations, the largest
study to date was performed in a US Latino/
Hispanic population,17 identifying a genetic
Genetics of dental caries variant in NAMPT—a gene with roles in mul-
There have been a number of GWASs seek- tiple biologic processes including periodontal
ing to identify the specific variants influencing healing—that is not present in white popula-
susceptibility to decay. These efforts were mo- tions. Altogether, these studies have identified
tivated by the cumulative evidence from prior several genetic variants statistically associ-
twin- and family-based studies going back nine ated with dental caries and have generated a
decades that overwhelmingly demonstrated growing list of genes to follow up in addi-
the heritability of dental caries. Heritabili- tional studies. However, few of these genetic
ty estimates differ across populations, and associations have been robustly replicated in
it is difficult to generalize the exact degree independent samples, which may be due to the
by which caries liability derives from genet- presence of genetic heterogeneity (ie, that the
ic causes. One unresolved issue that concerns variants contributing to dental decay differ
all genetic research, including caries and peri- across populations), environmental differences
odontal disease research, is the concern over across studies that moderate the genetic effect,
missing heritability. This concern relates to the or the low statistical power of these studies to
fact that standard genetic approaches, such as detect genetic variants of small effects. It is also
was used before high-throughput technology likely that some of these genetic associations
was available, often provided large heritability were spurious. Efforts are currently underway
estimates for many (disease) traits. However, by the Gene-Lifestyle Interactions and Dental
the application of subsequent GWASs and oth- Endpoints (GLIDE) Consortium, an interna-
er high-throughput technologies that identified tional collaboration with the goal of leveraging
individual genes associated with the trait fail, existing genetic studies with data on dental phe-
in many cases, to account for a substantial notypes, to combine GWAS results across many
portion of that predicted heritability. Debate independent cohorts via meta-analysis. Simi-
continues on how to interpret this discrepancy. lar meta-analyses have been very fruitful for
Genomic studies of dental caries have typ- other complex diseases; therefore, results from
ically considered the primary dentition in the GLIDE Consortium are likely to further
pediatric samples and the permanent denti- advance our understanding of the genetic archi-
tion in adults separately. The first GWAS of tecture of dental caries. Altogether, GWASs of
dental caries16 simply contrasted the genomes dental caries have demonstrated the polygenic
of caries-affected versus caries-free white chil- nature of dental caries but have not yet led to
dren and did not identify any loci meeting the discoveries that would impact clinical care.
strict statistical threshold of “genome-wide sig-
nificance” (ie, the P value needed to declare a
finding statistically significant given the large Genetics of periodontal disease
burden of multiple comparisons). Later studies By far the most research in the genetics of
looked at dental caries in adult populations and oral diseases has focused on periodontal dis-
expanded the list of dental caries phenotypes in eases. Genetic factors are reported to explain
both children and adults to include quantitative around 50% of the variance in the liability for

299
14 Oral-Systemic Health: The Genomic Connection

periodontitis, and the basis for the heritabili- “doubtful whether the criteria applied could
ty of periodontitis appears to be biologic and be realistically taken to unequivocally identify
not behavioral in nature.18 Periodontal dis- periodontitis.” They also noted a generalized
eases are a family of biofilm-related diseases failure to address the potential for serious con-
that vary by clinical presentation but share an founding, particularly from variables such as
underlying inflammatory basis.19 Van Dyke20 smoking and socioeconomic status. The poten-
describes the disease process as an overgrowth tial for disease misclassification and residual
of commensal organisms creating a “dysbiotic” confounding are likely reasons for the large
microbiome, where the initiation and progres- degree of heterogeneity of results across studies
sion of the disease is largely modulated by the in this area.
host inflammatory response. As Sima and Van Like dental caries, several GWASs have
Dyke21 remind us, a pathogenic biofilm is a investigated the genetic architecture of peri-
necessary but not sufficient cause of periodon- odontitis. One surprising result was that the
tal disease. At a minimum it appears that an initial batch of GWASs of chronic periodonti-
impaired host inflammatory response is also tis, published in 2013 and 2014 and focusing
required for tissue damage to occur, the nature primarily on white populations, did not iden-
of which is under substantial genetic control. tify any associations meeting the statistical
Periodontal medicine, as described by Wil- criteria for genome-wide significance. Only
liams and Offenbacher,22 is the branch of suggestive results were reported. This paucity
periodontology focusing on the relationship of genetic associations was unexpected given
between periodontal disease and systemic the presumed genetic architecture of the dis-
disease. In a review of the history of research ease. Most of the subsequent GWASs using
examining the association of periodontal dis- clinical definitions of chronic periodontal dis-
ease with systemic disease, Monsarrat et al23 ease, which have been undertaken in various
documented 57 systemic conditions that have racial and ethnic populations, have also yielded
been hypothesized as having some linkage with few or no significant associations. However,
periodontal disease. Given that many of the studies looking at innovative phenotypes, such
diseases associated with periodontal disease as periodontal pathogen colonization, or that
are common conditions with high morbidity, cleverly combined gene expression and genetic
the advancement of further research can be association in multiphase study designs, have
quite important from a patient and population been more successful. Likewise, studies of the
health perspective. Hypothesized mechanisms rarer aggressive form of periodontitis have
linking periodontal disease and systemic dis- yielded several interesting genetic associations.
ease consistent with the current evidence have Overall, the genomic studies of periodontitis to
been described as falling into three categories: date have identified several potential genetic
metastatic infection, inflammation, and adap- loci but have not yet generated any knowledge
tive immunity.24 The literature detailing these that has impacted clinical care.
pathophysiologic mechanisms are described
elsewhere in this text. Relevant to genetic
research is the reality that the current litera- GWAS lessons and other
ture suffers from substantial methodologic genome-wide approaches
shortcomings. A particular concern, as with
caries, is the variability across studies in the While genomic studies in the area of oral
case definition or phenotypic description used health have not uncovered any smoking guns,
to determine if individuals have disease. Linden nevertheless, key insights have been obtained.
et al25 said this was a serious concern across In particular, GWASs of many diseases and
studies, concluding in most cases that it was complex phenotypes, including oral conditions,

300
Genetics and Oral Disease

have revealed some big-picture themes about used to perform transcriptome-wide associa-
the role of genetics in complex disease. These tion studies, which investigate tissue-specific
include the observation (which had been hy- gene expression with respect to disease. Like-
pothesized beforehand) that most complex wise, epigenome-wide association studies
human diseases involve the contributions of (also known as methylome-wide association
many genetic variants of small individual ef- studies) can be performed to investigate the
fect sizes. A consequence of this is that most connection between tissue-specific patterns
complex diseases, like dental caries and peri- of DNA methylation and disease. Few such
odontitis, can only be poorly predicted based studies have been performed in the area of
on our current knowledge of their genetic ar- oral health, although some pioneering work
chitecture. Moreover, this observation casts includes an epigenome-wide association studies
some doubt on the ultimate utility of targeting by Langevin et al, who used DNA methylation
individual genetic variants via pharmaceuticals patterns to develop a classifier that could accu-
or gene therapies, as individual variants may rately predict oral and pharyngeal carcinoma.26
only contribute a small amount to total genet- In summary, the science of genomics has
ic liability. It also reflects a need for genomic come a long way in just a few decades, fueled
studies to move past examining single variants by technologic advancements facilitating swift
individually and instead to consider regulato- and economical collection of huge masses of
ry networks operating within the cell. Other data. Despite the advancements pioneered in
themes that have emerged from the mountain the seminal genomics projects, the field still
of GWASs performed to date include the obser- has unmet needs in terms of understanding and
vation that disease-associated genetic variants interpreting these data. For most clinical areas,
often differ in frequency across ancestry (ie, including oral disease, genomic information
racial and ethnic) groups and that some genes/ has yet to be successfully translated to clinical
genetic variants are involved in multiple sep- practice. However, with the amassing GWASs
arate diseases, a concept known in traditional and other genomic studies, our understanding
genetics as pleiotropy. In fact, pleiotropy is so of the genetic basis of human disease contin-
commonplace that study designs leveraging ues to grow. Emergent themes from the field of
giant data warehouses of electronic medical re- genomics suggests that as our understanding
cords have flipped the GWAS upside down by of the genetic architecture of disease grows, a
investigating the association of a single genetic genomic connection between oral and systemic
variant of interest with hundreds to thousands diseases is likely to be revealed.
of different medical conditions, an approach
that has been dubbed the phenome-wide asso-
ciation study (PWAS or PhenWAS). Evidence of the genomic
In addition to GWASs and PWASs, which connection between oral and
focus primarily on common genetic variants,
other genomic study designs have also been
systemic disease
developed and deployed to understand human Recent results from the GLIDE Consortium
disease. For example, whole-exome sequenc- provide some of the most systematic and salient
ing studies are used to identify the coding evidence to date of the genomic connection
variants associated with disease, including between oral and systemic disease.27 Statisti-
the rare variants that are not well captured in cal methods (ie, linkage disequilibrium score
GWASs. Similarly, whole-genome sequencing regressions) were used to contrast GWAS re-
studies can be used to identify all variants, sults for dental caries and periodontitis with
regulatory and coding, rare and common, that those of various traits and diseases reported
contribute to disease. RNA sequencing can be in separate studies. This approach allowed the

301
14 Oral-Systemic Health: The Genomic Connection

genetic correlations between dental outcomes Paradigms for the shared genetics
and a number of other human diseases and
of oral and systemic disease
traits to be quantified. In other words, separate
genomic studies were compared to estimate the An important point to appreciate is that many
proportion of shared genetics between oral and of the genomic studies of human disease are ob-
nonoral health measures. In total, genetic cor- servational in nature rather than experimental.
relations were estimated for both dental caries Such research leads to statistical associations,
and periodontitis with dozens of health-related and, as the axiom states, association is not
traits. For dental caries, significant shared ge- causation. That said, the constitutional ge-
netic etiology was observed with smoking nome of a person is determined at fertilization
habits; measures of obesity; lipid levels; dia- and remains unchanged thereafter (ignoring
betes; autoimmune diseases such as systemic somatic mutations). Therefore, studies of the
lupus erythematosus and rheumatoid arthri- constitutional genome are unaffected by the
tis; psychiatric conditions such as depression, problem of epidemiologic confounding due to
anorexia nervosa, and bipolar disorder; and environmental exposures. Namely, a risk fac-
educational attainment. Weaker statistical ev- tor such as smoking, for example, does not
idence was observed for the shared genetics of confound the relationship between a genetic
dental caries with Alzheimer’s disease, atten- variant and a disease, such as periodontitis,
tion deficit hyperactivity disorder, asthma, and because a person’s genotype is not affected by
celiac disease. For periodontal disease, signifi- smoking (or any other external factor) even
cant shared genetic etiology was observed with though smoking is a risk factor for the dis-
smoking habits, depression, measures of obesi- ease. This detail can be helpful in interpreting
ty, psychiatric disorders, subjective well-being, genomic studies. However, even though ge-
and educational attainment. Weaker statistical netic associations with disease are robust to
evidence of shared heritability was observed confounding by environmental factors, the
for periodontitis and multiple sclerosis, atten- associations themselves often do not provide
tion deficit hyperactivity disorder, diabetes, and much insight into the mechanisms, biologic
lipid levels.27 or otherwise, through which they impact dis-
These results indicate that the genetic etiol- ease. Follow-up studies, both observational and
ogies of both dental caries and periodontitis experimental, are needed to glean knowledge
are shared with a variety of systemic diseases, about these mechanisms. Likewise, when con-
plus tobacco use and education. However, sidering the genomic connection between oral
the genetic correlations observed from these and systemic disease, the mechanisms through
genomic studies only provide a global indicator which the comorbidities are coupled can be
of the degree to which genetic determinants challenging to determine. We will consider a
are shared between oral and systemic diseases. few paradigms (Fig 14-4) while acknowledging
These estimates of genetic correlation do not that other connections are also possible.
indicate which genetic variants specifically are The simplest paradigm for a genomic con-
involved, nor do they point to the biologic nection between oral disease and systemic
mechanisms linking the genetic etiologies of disease is pleiotropy—the single gene connec-
oral and systemic disease. Additional research tion. Traditionally, pleiotropy was taken to
will be needed to identify these genetic variants mean that a genetic variant in a gene simultane-
and elucidate these mechanisms. Like other ously affected two or more distinct phenotypes.
comorbidities, we expect that the shared genet- We see this phenomenon in a number of genetic
ics of oral and systemic disease may operate syndromes with both oral and systemic mani-
through a variety of mechanistic paradigms. festations (see Table 14-1 for a non-exhaustive
set of examples). In these syndromes, disruption

302
Genetics and Oral Disease

D1
Pleiotropy: Single gene
G
connection
D2

D1
G E
Gene-environment D2
correlation

G D1 E D2

Shared molecular D1
basis of complex
disease
D2

Mendelian ?
G D1 D2
randomization
(instrumental
variables) E

Fig 14-4  |  Paradigms for the shared genetics of oral and systemic disease. The node diagrams illustrate theo-
retical modalities by which two diseases can share a genetic basis. G, gene; E, environmental factor(s); D1 and
D2, oral or systemic disease or trait; arrows indicate causal relationships.

TABLE 14-1  |  Examples of single-gene syndromes with oral and systemic symptoms

Syndrome Gene(s) Oral manifestations Systemic manifestations

Waardenburg type 1 PAX3 Cleft lip and/or palate, tooth enamel Hearing loss, musculoskeletal anomalies,
malformations, and tooth agenesis hirschsprung disease, and other features

Jalili CNNM4 Amelogeneis imperfecta, dental caries, dentin Cone-rod dystrophy (vision loss, light
dysplasia sensitivity, poor color vision)

Marfan FBN1 Orthodontic anomalies, high palate, crowded Over 30 symptoms of skeletal,
teeth, fragility of the temporomandibular joint, cardiovascular, and nervous systems, lung,
periodontal disease and eye

Crouzon FGFR2 High palate, malocclusion, tooth agenesis, Craniosynostosis, pronounced facial features,
crowded teeth hearing loss

Rothmund-Thomson RECQL4 Tooth agenesis, microdontia, tooth dysplasia Skin manifestations such as sun-sensitive
rash, poikiloderma, telagiectasias, skeletal
defects, cataracts, risk of malignant disease

303
14 Oral-Systemic Health: The Genomic Connection

of the proper functioning of a key gene results oral lesions on the tongue and cheek. Recent
in signs and symptoms across separate tissues work28 has shown that common protein-coding
or systems. These examples show that del- variants in the keratin genes associated with
eterious genetic variants can simultaneously pachyonychia congenita are also associated
impact oral health and other biologic systems. with increased susceptibility to dental caries
They also suggest the plausibility that other in unaffected individuals. A similar single gene
not-quite-so-deleterious genetic variants may connection has been observed for the hair ker-
simultaneously impact risk for chronic oral and atin gene KRT75. Variants in this gene cause
systemic diseases. a mild hair disorder leading to skin inflamma-
Though not due to a single gene, several tion after shaving known as pseudofolliculitis
aneuploidies (chromosomal disorders) includ- barbae; recent work has shown that variants
ing trisomies of chromosomes 9p, 12p, 13, in KRT75 also cause increased susceptibility
18, 21, and 22, exhibit craniofacial and oral to dental caries and reduced enamel hard-
manifestations in addition to other systemic ness as well as subclinical enamel defects.29
manifestations. The same is also true for dele- These examples illustrate that variation in a
tion syndromes of chromosomes 13q and 18q, gene can impact chronic oral disease, such
as well as aneuploidies of the sex chromo- as dental caries, while also impacting other
somes (ie, multiple X [XXX+], Turner [X0], tissues/systems. Given that dental caries and
Klinefelter [XXY], and XXY syndromes). Due periodontitis are prevalent and exhibit a wide
to the fact that these syndromes are caused by range of disease severity in the general popu-
chromosomal anomalies encompassing large lation, it is plausible that many single genes
amounts of genomic content, it is likely that connecting these oral diseases to other systemic
distinct symptoms are due to the gain/loss of maladies have gone unnoticed. Indeed, Offen-
different genes across the affected genomic bacher et al30 suggested that variation in disease
regions. That is, the gain/loss of genetic mate- progression among patients could indicate sub-
rial responsible for the oral manifestations may classifications of disease based on genetic and
differ from those causing other symptoms. Still, microbial-inflammatory signatures.
these chromosomal disorders demonstrate the Another paradigm through which genom-
shared genomic origins of both oral and sys- ics may act to connect oral and systemic dis-
temic maladies under a fairly simple paradigm ease is gene-environment correlation. This
involving a single genetic aberration. occurs when an exposure to an environ-
Other examples following the single gene mental condition is dependent on an indi-
connection paradigm are ectodermal dyspla- vidual’s genotype. For example, genetic
sias, a group of related disorders affecting variants in the nicotinic receptor gene clus-
the ectodermal tissues such as teeth, salivary ter CHRNA5-CHRNA3-CHRNA4 have
glands, hair, nails, and sweat glands. Many of well-established effects on nicotine depen-
these, such as Hay-Wells syndrome, Naegeli dence and smoking behavior. Smoking, in turn,
syndrome, and hypohidrotic ectodermal dys- impacts risk for numerous diseases, including
plasia, result in overt tooth dysplasias alongside periodontal disease. Therefore, periodontal
other clinical features. Recent studies have also disease shares a genomic origin with cardio-
observed the connection between ectodermal vascular disease, various cancers, and other
dysplasias and chronic oral conditions, such as smoking-related diseases by way of the genetic
dental caries. For example, mutations in keratin basis for nicotine dependence/smoking behav-
genes KRT6A and KRT6B cause the condition ior. Knowledge of the genetic basis for an envi-
pachyonychia congenita, which is character- ronmental exposure is not always of particular
ized mainly by hyperkeratosis lesions on the interest. In instances where the environmental
hands and feet, nail dysplasia, and white plaque exposure is known, the genomic connection

304
Genetics and Oral Disease

takes a back seat to the exposure itself. That that increase risk of caries. Thus, asthma and
is, smoking per se is of greater interest in con- dental caries share a genetic underpinning in
necting periodontitis to other smoking-related that asthma-associated genetic variants lead
diseases than is the genetic underpinnings to caries-associated environmental exposures.
of smoking status. Indeed, knowledge of the Knowledge of such environmental exposures
genetic underpinnings of the exposure do not could provide an actionable point to intervene,
add much, from a clinical perspective, in man- breaking the connection between diseases.
aging disease beyond what can be obtained Another proposed gene-environmental cor-
by asking about the exposure itself. In some relation involving oral and systemic traits is
cases, knowledge of a genetic predilection prior the connection between anxiety disorders, pain
to key environmental exposures may be help- experience, and oral health. Dental fear, which
ful for gauging future risk of disease. Where is anxiety or apprehension regarding receiving
smoking status may turn out to be important dental treatment, is related to other anxiety
clinically is in risk estimation. As an example, disorders and has shared heritability with fear
Divaris et al31 in a GWAS of the genetic basis of pain.32 Specific genetic variants, including
of chronic periodontitis showed that the esti- variants in MCR1 (the “redhead” gene), are
mate of the heritability of severe chronic peri- associated with both dental fear and fear of
odontitis from all SNPs increased from 18% to pain.33 (This observation fits with dental lore
52% when they included an interaction term that red-headed individuals may experience
between the genetic data and smoking. pain differently.) Genetic variants predis-
The gene-environment correlation para- posing to anxiety disorders or affecting pain
digm may be of greater interest in situations experience or pain-blocker metabolism may
where the environmental exposure connect- contribute to dental fear, which in turn leads to
ing oral and systemic disease is theorized or avoidance behavior. Patients who avoid dental
unknown. For example, several large studies treatment experience poor oral health. Thus,
have reported a correlation between childhood oral health conditions may share a genetic basis
asthma and dental caries, but the reason for with anxiety and pain phenotypes; the genomic
this connection is not clear. Moreover, unpub- connection is due to the gene-environmental
lished family-based studies have indicated a correlation between anxiety and pain percep-
shared heritability of these two diseases, and tion–related genetic variants and avoidance of
genetic associations have been reported for dental care. Incidentally, poor oral health may
variants in the mucin-coding gene MUC7 with lead to painful symptoms, thereby exacerbat-
both asthma and dental caries. Nevertheless, ing the problem of dental fear and avoidance.
the exact mechanism by which these two dis- Knowledge of such gene-environment correla-
eases are linked at the genetic level remains tions may be useful in breaking the cycle of
unknown. One potential explanation for the avoidance. That is, treating the anxiety could
genomic connection between dental caries improve oral health.
and asthma is through the gene-environment In summary, genetics may play a key role
correlation with antiasthmatic medications, in risk of exposures that impact oral health,
such as albuterol. The proposed scenario is and these exposures may simultaneously affect
that asthma-associated variants contribute to systemic disease. This genomic connection,
asthma, which in turn leads to antiasthmatic which is mediated by an environment, may be
medication exposure. Albuterol, for exam- less interesting from a clinical perspective than
ple, alters the oral microbiome and can cause the exposure itself. However, genomic connec-
dry mouth, both of which may contribute tions following this paradigm may be helpful in
to increased dental caries. Other antiasth- identifying environmental exposures that could
matic medications may include sweeteners serve as targets for disease-prevention strategies.

305
14 Oral-Systemic Health: The Genomic Connection

Another paradigm for the oral-systemic measures. More work will be necessary to tease
health genomic connection is through the out the specifics of these genetic underpinnings.
shared molecular basis of complex diseases. Genetics and “omics” data resources, combined
A hallmark of complex disease is that numer- with bioinformatics and computational tools,
ous biologic processes may impact etiology may provide a framework for understanding
and predilection. Diseases may partly share the overlap in the genetic architectures of oral
signaling pathways and molecular networks, and systemic diseases, complementing and
and genetic variants impacting these biologic informing experimental work in model organ-
processes may impact both diseases. Genomic isms and in vitro.
studies hold potential for uncovering such In addition to explaining the connection
shared genetics, although to date little research between oral and systemic disease, genetics
has been conducted in this area linking oral can be leveraged in order to assess the causal
diseases to systemic diseases. The most studied relationships between oral and systemic dis-
relationship is the possible genomic connection ease using Mendelian randomization. This
for periodontal disease and cardiovascular dis- approach simply employs the statistical method
ease. Twin studies suggest a shared heritability of instrumental variables where genetic vari-
of cardiovascular and periodontal diseases. A ants serve as the instrument. One application
recent review34 of the periodontal and cardio- of the method has been to use genetics to test
vascular GWAS literature indicated that though the causal relationship of adiposity and peri-
few periodontitis-associated genetic variants odontitis. A genetic risk score, which combined
have been mapped, to date, three out of four multiple well-established genetic variants asso-
loci associated with periodontal disease are ciated with body mass index, was used as an
also associated with cardiovascular disease. instrumental variable to test whether adipos-
These preliminary results support the notion ity caused periodontal disease. The combined
that cardiovascular and periodontal diseases results of this analysis across many large stud-
involve some common underlying biology ies showed that adiposity was not a causal
influenced by genetic variation. risk factor for periodontitis.35 This conclusion
The paucity of evidence for a shared molecu- is consistent with a PWAS showing pleiotro-
lar basis of complex oral and systemic diseases pic associations between obesity and chronic
reflects the current state of the field in that few periodontitis.36
attempts to consider this area of investigation Altogether, oral and systemic disease can
have been made to date. Unlike most epidemi- share a genomic connection through multi-
ologic studies, genomic studies can use separate ple different paradigms. Understanding these
samples to investigate the shared heritability. connections could lead to novel therapies,
Using genomics as the link, researchers can improved risk prediction, and improved patient
estimate the shared heritability of two dis- outcomes.
eases using two separate samples, one having
data on each disease. A major limitation is that
these methods require large samples, preferably
10,000 or more individuals, and presently there
Genetics and Clinical
are few such samples in which to study the Practice
genetics of oral diseases. As described earlier,
recent efforts by the GLIDE Consortium have The full benefit from genomic research will
achieved such large sample sizes by combining only be realized when that knowledge is
multiple independent studies26 and provided brought into routine clinical practice. Although
some of the first evidence for the shared her- we are not yet to the point where actionable ge-
itability of oral diseases and general health netic information is available for use in general

306
Genetics and Clinical Practice

dental settings, that day will eventually arrive. predict caries and periodontal disease rely
Through its translational research programs, primarily on measures of social, behavioral,
the NIH is leading the way to ensure that the and clinical parameters. Caries risk assessment
substantial resources it is investing in genetic (CRA) tools include caries management by risk
research will pay practical dividends in im- assessment (CAMBRA)40 and the American
proved patient outcomes. One of the principal Dental Association41 risk assessment forms.
mechanisms by which NIH supports trans- The predictive ability of CRAs tend to lack
lational research is through its Clinical and proper validation through longitudinal stud-
Translational Science Awards program, initiat- ies and thus have unknown predictive value.
ed in 2006. NIH defines translational research Another problem that limits the predictive
as follows: ability of CRAs is their failure to incorporate
genetic information in their risk model. As
Translational research includes two areas noted previously, the heritability estimates for
of translation. One is the process of apply- both caries and periodontal disease suggest that
ing discoveries generated during research roughly 50% of the variation in disease liability
in the laboratory, and in preclinical stud- comes from genetic risk factors. But a valid
ies, to the development of trials and studies algorithmic approach to caries risk prediction
in humans. The second area of translation is still some ways off.
concerns research aimed at enhancing the Periodontal risk predictors include the peri-
adoption of best practices in the commu- odontal risk calculator (PRC)42 and periodontal
nity. Cost-effectiveness of prevention and risk assessment (PRA) model.43 Both risk pre-
treatment strategies is also an important part dictors rely on clinically measured disease
of translational science.37 parameters such as bone loss, pocket depth,
and bleeding and tend to suffer from similar
The goal of this program, as described by shortcomings as the CRAs. The PRA, however,
Reis et al,38 is to “catalyze the transformation of in its original version and through subsequent
biomedical research at a national level, speed- modifications has endeavored to incorporate
ing the discovery and development of therapies, genetic information, specifically by consider-
fostering collaboration, engaging communities, ation of interleukin (IL) 1 gene polymorphisms.
and training succeeding generations of clinical There have been some efforts to advocate for
and translational researchers.” Over the now the use of single gene markers such as IL-1
more than 12 years of this program’s operation, or IL-6 as the basis for treatment planning
it has fostered major advances in genomic sci- and insurance reimbursement for periodontal
ence and is helping to ensure that genetics will disease, but the clinical value of such a test
become central to the future of clinical practice, has been questioned.44 These single-gene tests
including clinical dental practice. were developed based on science that has not
The two areas where translational genomic been replicated. In fact, large meta-analysis
science is currently having a large impact on has proven that the effects of these variants, if
clinical practice is through improved disease any, are very small—much too small to be of
prediction and through genomic targeting of clinical utility.
drugs in ways that optimize patient outcomes.39 Oral cancers, given their high incidence,
Accurate prediction of the risk for future dis- particularly in developing countries, and sub-
ease development allows at-risk patients to stantial morbidity and mortality attributed to
be appropriately provided with prevention frequent late-stage diagnosis, would benefit
interventions while sparing the inconvenience from early risk predictors. Although environ-
and cost of such interventions for patients that mental and behavioral risk factors (eg, tobacco,
would not derive benefit. Presently, efforts to alcohol) are considered the major determinants

307
14 Oral-Systemic Health: The Genomic Connection

of oral cancer disease liability, genetic and discuss genetic information with their patients.
epigenetic risks are under investigation45 and Storing and managing genetic data will require
may provide some additional early diagnostic changes in the electronic health records used
benefit. Unfortunately, the current microarray by dentists, as no electronic health records
technology is too expensive for broad-based today are equipped to store or make available
screening, particularly in low-income countries, genetic information. Medicine has responded
where the disease has its highest incidence.46 to this challenge by deploying automated
Pharmacogenomics is a rapidly growing decisions-support software that incorporates
area. The US Food and Drug Administration genetic information and is designed to improve
now provides a framework for pharmaceuti- diagnosis and treatment response. Moreover,
cal companies on their requirements around such stored information requires careful han-
genetic data submission. The requirements are dling as it carries substantial risk for patient
based on the intended use and data availability. harm. If its confidentiality is not well guarded,
Presently there are several hundred drugs that it could impact patient insurability and
are required to provide product labeling text employability. Finally, the cost-benefit ratio of
regarding genetic indications or contraindica- genetic testing will need to be fully documented
tions for their use. Kornman and Polverini47 if insurance companies are to consider this as
reviewed the current state of the use of genetics part of a dental benefit plan.
to stratify patients in ways that could improve The advantages of genetic research for the
prevention and treatment of oral diseases. routine practice of dentistry will continue to
As stated earlier, this review highlights that grow and benefit dentists and patients. But
methodologic issues—including disease mis- this coming wave of new genetic knowledge
classification, residual confounding, and small requires that we start preparing for it now.
sample size—prevent any clear guidance on
actionable genetic traits that would be of value
to clinicians.
Methodologic shortcomings notwithstand- References
ing, given the rapid advances in genomic
science, it seems reasonable to assume that 1. Ankeny RA. Sequencing the genome from nematode
to human: Changing methods, changing science.
some clinically actionable genetic informa- Endeavour 2003: 27: 87–92.
tion will be available for use in routine dental 2. National Institutes of Health. Fact Sheet: Genetics of
practice in the not-too-distant future. If this Common Complex Disease. https: //report.nih.gov/
information is to be fully exploited for patient NIHfactsheets/Pdfs/GeneticsofCommonComplexDis-
ease(NHGRI).pdf. Accessed 29 May 2018.
benefits, the implication is that this will neces- 3. Lander ES, Linton LM, Birren B, et al; International
sitate important changes in how we train Human Genome Sequencing Consortium. Initial
dentists, finance dental care, deploy relevant sequencing and analysis of the human genome. Nature
2001; 409:860–921.
technology, and ethically treat patients.
4. Venter JC, Adams MD, Myers EW, et al. The sequence
At present there is a perceived inability of the human genome. Science 2001;291:1304–1351.
for clinicians to understand and effectively 5. International Human Genome Sequencing Consortium.
implement genetic or pharmacogenetic infor- Finishing the euchromatic sequence of the human
genome. Nature 2004;431:931–945.
mation in routine patient care.48 If dentistry is
6. International HapMap Consortium. A haplotype
to respond to this, it will require substantial map of the human genome. Nature 2005; 437:
changes to dental school curricula and con- 1299–1320.
tinuing education offerings. This training will 7. International HapMap Consortium; Frazer KA, Ballinger
DG, Cox DR, et al. A second generation human haplo-
need to allow dentists to develop a sophisti-
type map of over 3.1 million SNPs. Nature 2007; 449:
cated understanding of complex genetic data 851–861.
and to have the skills necessary to meaningfully

308
References

8. 1000 Genomes Project Consortium; Abecasis GR, 25. Linden GJ, Lyons A, Scannapieco FA. Periodontal
Altshuler D, Auton A, et al. A map of human genome systemic associations: Review of the evidence. J Clin
variation from population-scale sequencing. Nature Periodontol 2013;40(suppl 14):S8–S19.
2010;467:1061–1073. 26. Langevin SM, Elliot M, Butler RA, et al. CpG island
9. 1000 Genomes Project Consortium; Abecasis GR, methylation profile in non-invasive oral rinse samples is
Auton A, Brooks LD, et al. An integrated map of genetic predictive of oral and pharyngeal carcinoma. Clin
variation from 1,092 human genomes. Nature 2012; Epigenetics 2015;7:125.
491:56–65. 27. Shungin D, GLIDE Consortium Investigators. Shared
10. ENCODE Project Consortium; Birney E, Stamatoyan- genetic contributions between oral and general health
nopoulos JA, Dutta A, et al. Identification and analysis using GWAS in 506,667 adults. J Dent Res 2018;
of functional elements in 1% of the human genome by 97(special issue):1717.
the ENCODE pilot project. Nature 2007;44:799–816. 28. Duverger O, Carlson JC, Karacz CM. Genetic variants
11. FANTOM Consortium and the RIKEN PMI and CLST in pachyonychia congenita-associated keratins increase
(DGT); Forrest AR, Kawaji H, Rehli M, et al. A promoter- susceptibility to tooth decay. PLoS Genet 2018; 14:
level mammalian expression atlas. Nature 2014;507: e1007168.
462–470. 29. Duverger O, Ohara T, Shaffer JR, et al. Hair keratin
12. Bernstein BE, Stamatoyannopoulos JA, Costello JF, et mutations in tooth enamel increase dental decay risk.
al. The NIH Roadmap Epigenomics Mapping Consor- J Clin Investig 2014;124:5219–5224.
tium. Nature Biotechnol 2010;28,1045–1048. 30. Offenbacher S, Divaris K, Barros SP, et al. Genome-wide
13. Germain PL, Ratti E, Boem F. Junk or functional DNA? association study of biologically informed periodontal
ENCODE and the function controversy. Biol Philos 2014; complex traits offers novel insights into the genetic basis
29:807–808. of periodontal disease. Hum Mol Genet 2016; 25:
14. Wetterstrand KA. DNA Sequencing Costs: Data from 2113–2129.
the NHGRI Genome Sequencing Program (GSP). www. 31. Divaris D, Monda KL, North KE, et al. Exploring the
genome.gov/sequencingcostsdata. Accessed 24 genetic basis of chronic periodontitis: A genome-wide
August 2018. association study. Hum Mol Genet 2013; 22:
15. MacArthur J, Bowler E, Cerezo M, et al. The new 2312–2324.
NHGRI-EBI Catalog of published genome-wide asso- 32. Randall CL, McNeil DW, Shaffer JR, Crout RJ, Weyant
ciation studies (GWAS Catalog). Nucleic Acids Res RJ, Marazita ML. Fear of pain mediates the association
2017;45(database issue):D896–D901. between MC1R genotype and dental fear. J Dent Res
16. Shaffer JR, Wang X, Feingold E, et al. Genome-wide 2016;95:1132–1137.
association scan for childhood caries implicates novel 33. Randall CL, Shaffer JR, McNeil DW, Crout RJ, Weyant
genes. J Dent Res 2011;90:1457–1462. RJ, Marazita ML. Toward a genetic understanding of
17. Morrison J, Laurie CC, Marazita ML, et al. Genome-wide dental fear: Evidence of heritability. Community Dent
association study of dental caries in the Hispanic Oral Epidemiol 2016;45:66–73.
Communities Health Study/Study of Latinos (HCHS/ 34. Aarabi G, Zeller T , Seedorf H, et al. Genetic susceptibility
SOL). Hum Mol Genet 2016;25:807–816. contributing to periodontal and cardiovascular disease.
18. Michalowicz BS, Diehl SR, Gunsolley JC, et al. Evidence J Dent Res 2017;96:610–617.
of a substantial genetic basis for risk of adult periodon- 35. Shungin D, Corneilis MC, Divaris K, et al. Using genetics
titis. J Periodontol 2000;71:1699–1707. to test the causal relationship of total adiposity and
19. Armitage GC. Development of a classification system periodontitis: Mendelian randomization analyses in the
for periodontal diseases and conditions. Ann Periodontol Gene-Lifestyle Interactions and Dental Endpoints
1999;4:1–6. (GLIDE) Consortium. Int J Epidemiol 2015;44:638–650.
20. Van Dyke TE. Pro-resolving mediators in the regulation 36. Cronin RM, Field JR, Bradfork Y, et al. Phenome-wide
of periodontal disease. Mol Aspects Med 2017; 58: association studies demonstrating pleiotropy of genetic
21e36. variants within FTO with and without adjustment for
21. Sima C, Van Dyke TE. Therapeutic targets for manage- body mass index. Front Genet 2014;5;5:250.
ment of periodontitis and diabetes. Curr Pharm Des 37. National Institutes of Health. Institutional Clinical and
2016;22:2216–2237. Translational Science Award (U54) [Announcement
22. Williams RC, Offenbacher S. Periodontal medicine: The RFA-RM-07-002]. http: //grants.nih.gov/grants/guide/
emergence of a new branch of periodontology. Peri- rfa-files/RFA-RM-07-007.html. Accessed 29 May 2018.
odontol 2000 2000;23:9–12. 38. Reis SE, Berglund L, Bernard GR, et al. Reengineering
23. Monsarrat P, Blaizot A, Kémoun P, et al. Clinical research the national clinical and translational research enterprise:
activity in periodontal medicine: A systematic mapping The strategic plan of the national clinical and translational
of trial registers. J Clin Periodontol 2016;43:390–400. science awards consortium. Acad Med 2010; 85:
24. Van Dyke TE, van Winkelhoff AJ. Infection and inflam- 463–469.
matory mechanisms. J Periodontol 2013; 84(4 suppl): 39. Larson EA, Wilke RA. The integration of genomics in
S1–S7. primary care. Am J Med 2015;128:1251.e1–5.

309
14 Oral-Systemic Health: The Genomic Connection

40. Chaffee BW, Cheng J, Featherstone JD. Baseline caries 45. D’Souza W, Saranath D. Clinical implications of epigen-
risk assessment as a predictor of caries incidence. J etic regulation in oral cancer. Oral Oncology 2015;51:
Dent 2015;43:518–524. 1061–1068
41. American Dental Association. Caries Risk Assessment 46. Liu Y, Li Y, Fu Y, et al. Quantitative prediction of oral
Form (Age 0–6). https: //www.ada.org/~/media/ADA/ cancer risk in patients with oral leukoplakia. Oncotarget
Member%20Center/FIles/topics_caries_under6.pdf ? 2017;11;8:46057–46064.
la=en. Accessed 29 May 2018. 47. Kornman KS, Polverini PJ. Clinical application of genet-
42. Page RC, Martin J, Krall EA, Mancl L, Garcia R. Longi- ics to guide prevention and treatment of oral diseases.
tudinal validation of risk calculator for periodontal Clin Genet 2014;86:44–49.
disease. J Clin Periodontol 2003;30:819–827. 48. Selkirk CG, Weissman SM, Anderson A, Hulick PJ.
43. Lang NP, Tonetti MS. Periodontal risk assessment (PRA) Physicians’ preparedness for integration of genomic
for patients in supportive periodontal therapy (SPT). Oral and pharmacogenetic testing into practice within a major
Health Prev Dent 2003;1:7–16. healthcare system. Genet Test Mol Biomarkers 2013;
44. Diehl SR, Kuo F, Hart TC. Interleukin 1 genetic tests 17:219–225.
provide no support for reduction of preventive dental
care. J Am Dent Assoc 2015;46:164–173.

310
CHAPTER 15

Common Risk Factors:


The Link Between Oral and
Systemic Disease
David M. Williams, bds, msc, phd, frcpath, fds rcs (engl)

There is currently enormous interest in the role in advocating for the inclusion of oral
associations between oral diseases, particular- disease in wider strategies for the prevention
ly dental caries and periodontal disease, and of the major NCDs that are the major kill-
the major noncommunicable diseases (NCDs) ers of the 21st century. Not only will such an
such as cardiovascular disease (CVD), diabe- approach have the potential to reduce the bur-
tes, stroke, and cancer.1 The literature is replete den of NCDs in general, but it will lead to the
with reports of the association between these more effective prevention of oral diseases and
NCDs and oral diseases, but despite extensive the more cost-effective use of scarce financial
research, the evidence for a causal relation- and human resources in their treatment. This
ship between oral diseases such as periodontal shift substantially expands the strategies and
disease and systemic disease—particularly resources available to address oral disease and
CVD—is weak. By contrast, it is well estab- should lead to a breakdown in the unnecessary
lished that the major oral diseases and NCDs silos that have kept oral health in a separate cat-
share the same social determinants and com- egory from general health. Such a transition is
mon risk factors. Particularly important among also imperative because current strategies that
the latter are tobacco, alcohol, and dietary sug- deal with the two most common oral diseases,
ar intake. It is imperative that all oral health periodontal disease and caries, are focused
care professionals (OHCPs) understand the im- overwhelmingly on treatment rather than on
portance of these associations because it is in prevention and oral health promotion. These
large measure based on this evidence that the approaches are too costly to remain viable,3–5
World Health Organization (WHO) has en- and the limitations of the traditional surgical
couraged an integrated approach to chronic model of oral health care demonstrate the need
disease prevention.2 to change to an approach for the prevention
This chapter considers the evidence that oral of oral diseases that will involve integrating
and systemic disease share the same determi- efforts with all health professionals to achieve
nants and common, modifiable risk factors and optimal oral and general health.3
makes the case that OHCPs have a significant

311
15 Common Risk Factors: The Link Between Oral and Systemic Disease

The Challenge of the Global on 291 diseases—including oral diseases—in


21 regions. In a systematic analysis of the
Oral Disease Burden and global burden of oral diseases within the wider
the Growing Problem of Oral GBD study, Marcenes et al14 reported that in
2010 oral diseases affected 3.9 billion people
Health Inequality worldwide. Untreated caries in the permanent
dentition was the most prevalent condition
Current approaches to improve oral health are in the entire GBD study (Table 15-1), with a
relatively ineffective yet very costly, and high- global prevalence of 35%. Additionally, severe
income countries are facing a “dental crisis.”6 periodontitis in adults and untreated caries in
Oral and dental diseases afflict almost every- primary teeth were the 6th and 10th most prev-
one and leave a legacy of substantial morbidity alent conditions, respectively. The authors also
and functional deficits among older adults.7 Al- observed a 20.8% increase in the total burden
though the oral health of children and young of oral disease between 1990 and 2010, attrib-
adults has improved significantly in recent utable principally to population growth.
decades,8 with falls in the prevalence of both Not only is the prevalence of oral disease
dental caries and periodontal disease, these high in relation to all other diseases, but there
falls mask persistent inequalities. It has been are very marked inequalities in the distribution
estimated that only 3% of the variation in car- of all major oral diseases, including oral cancer.
ies levels in 12-year-olds can be attributed to Figure 15-1 shows the global distribution of
dental services and that 65% is explicable on dental caries, where it can be seen that there is
the basis of socioeconomic factors.9 More re- a gradient of prevalence with higher levels of
cent studies have confirmed the importance of disease in low- and middle-income countries.
social determinants and environmental factors, Similar gradients in disease prevalence are also
as exemplified by the work of Elani et al,10 who seen for periodontal disease and oral cancer.15
showed that in the United States and Canada, In addition to being highly prevalent, oral
the overall prevalence of edentulism and untreat- diseases have a significant negative impact on
ed dental caries in adults reduced significantly people’s quality of life. In children, they are the
between 1970 to 1972 and 2007 to 2008. How- most common causes of pain that disturb sleep
ever, a persistent oral health inequality gradient and retard growth. Caries is also responsible
remained that was attributable to place of birth, for poor learning and school performance. In
educational attainment, and level of income. In adults and older people, dental pain, suffering,
the United States, the oral health of older adults, and discomfort severely restrict dietary intake
currently one of the fastest-growing segments of and social functioning.6
the population, “is in a state of decay.”11 From a global perspective, oral disease pre­
From a global perspective, a more disturbing sents two major challenges: (1) how to address
and challenging picture of oral disease emerges, the existing burden of disease and (2) how to
particularly in low- and middle-income coun- implement effective prevention that will lead
tries, with a major and growing overall bur- to sustainable improvements in oral health and
den of oral disease accompanied by marked a reduction in oral health inequality. For the
inequalities both within and between countries. first of these challenges, a restorative approach
Indeed, oral diseases constitute a “neglected is simply unsustainable. Yee and Sheiham5
epidemic” in these countries, and rates are have calculated that to restore one tooth per
increasing.12,13 The only comprehensive effort 6- to 18-year-old child would cost between
to date to estimate global population health by US $1,618 and US $3,513 per 1,000 children,
cause is the Global Burden of Diseases, Injuries, which is unaffordable on a global scale. Figure
and Risk Factors (GBD) Study, which reported 15-2 is a map of the ratio between the burden

312
The Challenge of the Global Oral Disease Burden and the Growing Problem of Oral Health Inequality

TABLE 15-1  |  Global prevalence of oral conditions in 2010 by sex


Overall Men Women
Rank Condition n° % n° % n° %

1 Untreated caries of 2,431,636 35.29 1,194,051 34.37 1,237,585 36.23


permanent teeth

2 Tension-type headache 1,431,067 20.77 655,937 18.88 775,131 22.69

3 Migraine 1,012,944 14.70 371,072 10.68 641,873 18.79

4 Fungal skin disease 985,457 14.30 516,167 14.86 469,291 13.74

5 Other skin and 803,597 11.66 417,129 12.01 386,468 11.32


subcutaneous diseases

6 Severe periodontitis 743,187 10.79 378,407 10.89 364,780 10.68

7 Mild hearing loss 724,689 10.52 386,147 11.11 338,543 9.91

8 Acne vulgaris 646,488 9.38 311,349 8.96 335,140 9.81

9 Low back pain 632,045 9.17 334,793 9.64 297,252 8.7

10 Untreated caries of 621,507 9.02 352,085 10.13 269,421 7.89


primary teeth

36 Severe tooth loss 158,284 2.3 67,264 1.94 91,020 2.66

°Numbers of cases reported in thousands. (Adapted from Marcenes et al14 with permission.)

ICELAND
TOOTH DECAY
WORLDWIDE
SWEDEN FINLAND
NORWAY

ESTONIA Average number


LATVIA
LITHUANIA of decayed (D), missing (M),
DENMARK
IRELAND
UK and filled (F) teeth (T)
NETH. BELARUS
GERMANY
POLAND
in 12-year-olds
BELGIUM UKRAINE RUSSI A
C A N A D A
LUX.
CZECH
REP. SLOVAKIA latest available data
MOLDOVA
FRANCE SWITZ. AUSTRIA HUNGARY
SLOV. ROMANIA
1994–2014
CROATIA B-H
BULGARIA
PORTUGAL
MONT. more than 3.5 high
ALBANIA FYROM
MONGOLIA
SPAIN
ITALY 2.6 – 3.5 moderate
U S A GREECE
UZBEK. JA PAN 1.2 – 2.5 low
TURKEY SOUTH
KOREA

MALTA
CYPRUS SYRIA C H I N A 0.0 – 1.1 very low
LEBANON IRAQ
MOROCCO TUNISIA ISRAEL I RA N
JORDAN
KUWAIT
no data
PAKIS TAN BHUTAN
MEXICO BAHAMAS
L I B YA BAHRAIN
NEPAL
CAYMAN IS. CUBA DOMINICAN QATAR Macau SAR
EGYPT UAE
REP.
BANGLADESH HK SAR
JAMAICA PUERTO RICO SAUDI ARABIA INDIA KIRIB ATI
HAITI ANGUILLA LAOS
BELIZE OMAN MYANMAR
ANTIGUA & BARBUDA VIET NAM TUVALU
GUATEMALA ST KITTS & NEVIS
HONDURAS DOMINICA
SENEGAL NIGER SUDAN ERITREA YEMEN
THAILAND TOKELAU
EL SALVADOR ST LUCIA PHILIPPINES
NICARAGUA GRENADA GAMBIA
BARBADOS BURKINA
FASO CAMBODIA
COSTA RICA TRINIDAD & TOBAGO SAMOA
B ENI N

NIGERIA
GHANA

VENEZUELA C‘TE
PANAMA GUYANA SOUTH ETHIOPIA SRI LANKA
DíIVOIRE
SURINAME SUDAN BRUNEI VANUATU
COLOMBIA
MALAYS I A FIJI
UGANDA
COOK ISLANDS
SINGAPORE
ECUADOR KENYA NIUE
GABON
TONGA
SEYCHELLES
PAPUA
PERU TANZANIA I N D O N E S I A NEW
GUINEA FRENCH POLYNESIA
BRAZIL
SOLOMON
ISLANDS

BOLIVIA
NAMIBIA
PARAGUAY MOZAMBIQUE
CHILE

SWAZILAND AUSTRALI A
SOUTH
AFRICA
URUGUAY

NEW
ZEALAND

From The
From The Challenge
ChallengeofofOral
OralDisease
Disease—A callforforglobal
– A call globalaction
actionbybyFDI
FDIWorld
WorldDental
DentalFederation.
Federation.
Maps and
Maps and graphics
graphics©©Myriad
MyriadEditions
Editions2015
2015

Fig 15-1  |  Global inequalities in the prevalence of dental caries. (Reprinted from FDI World Dental Federation15
with permission.)

313
15 Common Risk Factors: The Link Between Oral and Systemic Disease

ICELAND THE BURDEN OF


NORWAY
SWEDEN FINLAND
DISEASE/PROVIDER RATIO
ESTONIA The ratio between burden of
LATVIA
LITHUANIA
oral disease in DALYs
DENMARK
IRELAND
UK and number of oral health
NETH.
GERMANY
POLAND
BELARUS
personnel per country
BELGIUM UKRAINE
CZECH
SLOVAKIA
LUX.
REP.
HUNGARY
MOLDOVA
RUSSI A
highest ratio 500 or more
FRANCE SWITZ. AUSTRIA
SLOV. ROMANIA
CROATIA SERBIA 100 – 499
BULGARIA
MONT. KOSOVO
PORTUGALANDORRA
CA NADA FYROM 20 – 99
SPAIN
ITALY KAZAKHSTAN
GREECE MONGOLIA 2 – 19
U S A
AZERBAIJAN
UZBEK. KYRGYZSTAN JA PAN lowest ratio 2 or less
TURKEY ARMENIA TURKMEN.
TAJIKIS TAN
CYPRUS SYRIA
no data
MALTA
CHIN A
LEBANON IRAQ AFGHANISTAN
MOROCCO TUNISIA ISRAEL I R AN
JORDAN
KUWAIT PAKIS TAN BHUTAN
BAHAMAS NEPAL
ALGERIA LIBYA BAHRAIN QATAR
MEXICO CUBA
EGYPT UAE MARSHALL ISLANDS
DOMINICAN
JAMAICA REP. BANGLADESH
HAITI SAUDI ARABIA LAOS
BELIZE CAPE IN DI A MYANMAR KIRIB ATI
ANTIGUA & BARBUDA VERDE MAURITANIA MALI
OMAN
GUATEMALA HONDURAS DOMINICA NIGER SUDAN THAILAND
EL SALVADOR SENEGAL CHAD ERITREA YEMEN
NICARAGUA GRENADA GAMBIA PHILIPPINES
BARBADOS BURKINA
FASO DJIBOUTI CAMBODIA
TRINIDAD & TOBAGO GUINEA-
COSTA RICA
BISSAU GUINEA

B ENI N
SAMOA
GHANA
VENEZUELA NIGERIA
TOGO
PANAMA GUYANA CÔTE ETHIOPIA
SIERRA LEONE D’IVOIRE CENTRAL SRI LANKA
SURINAME AFRICAN REP. BRUNEI
COLOMBIA LIBERIA VANUATU
MALDIVES
EQUATORIAL CAMEROON SOMALIA MALAYSIA FIJI
GUINEA UGANDA
KENYA SINGAPORE
ECUADOR GABON
SÃO TOME DEM. REP. RWANDA
& PRINCIPE CONGO OF CONGO BURUNDI SEYCHELLES TONGA
PAPUA
PERU TANZANIA I N D O N E S I A NEW
GUINEA SOLOMON
BR A Z I L COMOROS ISLANDS
ANGOLA EAST TIMOR

ZAMBIA

BOLIVIA MAURITIUS
ZIMBABWE MADAGASCAR
NAMIBIA
BOTSWANA
PARAGUAY MOZAMBIQUE
CHILE
SWAZILAND AUSTRALI A
LESOTHO
URUGUAY

ARGENTINA

NEW
ZEALAND
From The Challenge of Oral Disease – A call for global action by FDI World Dental Federation.
From The Challenge of Oral Disease—A call for global action by FDI World Dental Federation.
Maps and graphics © Myriad Editions 2015
Maps and graphics © Myriad Editions 2015

Fig 15-2  |  Inequalities in the global distribution of oral health personnel: The burden of disease/provider ratio.
DALYs, disability-adjusted life years. (Reprinted from FDI World Dental Federation15 with permission.)

of disease in disability-adjusted life years and responsible. The understanding exists to


the number of oral health personnel by coun- prevent a very large proportion of oral dis-
try. This shows very clearly that in low- and eases, and community-based prevention
middle-income countries there is a major dis- generally is cost-saving compared with
proportion between the availability of personnel a treatment-focused approach, particularly
and the burden of disease. From this it is clear for communities and individuals at high risk
that a traditional high-income country model for disease.
of dental care, such as that seen in the United
States and Europe, is completely inappropriate It is clear that current approaches to pre-
to address the challenge of poor oral health. vention have not been effective at a global
Even if it were appropriate, financial constraints level, and it is imperative to understand the
in low- and middle-income countries mean that reasons for this and develop strategies that
there is no realistic prospect of addressing this could deliver effective disease prevention
human resource challenge in the short term. and sustainable improvements in oral health
This means that if the current burden of oral worldwide. In this regard, there are important
disease is to be managed effectively, particularly lessons to be learned from addressing the asso-
in low- and middle-income countries, very dif- ciations between oral and systemic disease and
ferent models of care will be required. understanding whether the approaches pro-
In a critique of the US dental care system, posed to reduce the global burden of NCDs
Tomar and Cohen16 summed up the situation could also result in improvements in global
worldwide thus: oral health.
However, it is not enough for oral health
A system focused primarily on treatment of professionals simply to observe the associations
disease in individuals is not economically between oral and systemic disease. As Garcia
sustainable, socially desirable, or ethically and Tabak17 have so presciently stated:

314
Intervening to Improve Oral Health: The Common Risk Factor Approach

Fig 15-3 |  Downstream to up-


National and/or local policy initiatives stream: Options for oral disease
Legislation/regulation prevention. (Reprinted from Watt19
Fiscal measures
with permission.)
Upstream healthy
public policy Healthy settings–HPS
Community development
Training other professional groups
Media campaigns
School dental health education
Chairside dental health education
Downstream health Clinical prevention
education and
clinical prevention

Tackling global oral health inequalities will global health agenda that includes for exam-
require creativity, diligence and a strong ple, tobacco and alcohol control, clean water
commitment to partner with the many play- and sanitation, maternal and child health,
ers involved in global health. These include and health systems research.
research institutions, institutions of higher
education, private and public organizations, The evidence presented in this section sets
professional associations, health officials, out the scale of the oral disease challenge that
corporations and foundations that promote all countries face, and it is clear from this that
health. The oral health community must ‘be the burden of disease falls disproportionately
at the table’ with these and other partners, on those countries least able to respond. The
be willing to frame oral health needs in the sections that follow consider strategies to
context of overall health, and develop clearly reduce both the overall burden of disease and
articulated objectives driven by strong evi- the unjust inequalities in oral health.
dence of the burden posed by oral diseases
and conditions. […] The oral health com-
munity needs to become more integrated
with the broader family of global health. Intervening to Improve Oral
As a discipline, oral health has evolved Health: The Common Risk
from the one-dimensional understanding of
health and disease to a more comprehensive Factor Approach
view. Individual risk and susceptibility to
oral diseases can no longer be understood The failure of most approaches to improve
in isolation, but as part of a complex set of oral health and reduce inequalities in oral
influences that include individual, family, health were cogently summed up by Kwan
community or neighborhood characteristics, and Petersen18:
and health system factors. This broad con-
ceptual framework of oral health facilitates Measures that focus on downstream factors
interactions with other health professions (Fig 15-3) only, such as lifestyle and behav-
and with other sectors including education ioral influences, may have limited success in
and social services. Framing oral health as a reducing oral health inequities. … Such ap-
vital part of overall health will enable the in- proaches may be counterproductive as they
tegration of oral health as part of a broader are often ineffective and costly and fail to

315
15 Common Risk Factors: The Link Between Oral and Systemic Disease

Fig 15-4 |  The social determinants of health.


, (Reprinted from Whitehead and Dahlgren20 with
permission.)
Poverty

Work Water

Unemployment
Sanitation

Tobacco Alcohol
Nutrition
Housing

Sugar Diet

Education Health care

address the wider social determinants that decayed, missing, and filled teeth with age
cause people to get ill in the first place. Peo- result from dental caries caused by sugars and
ple in more privileged social positions tend to periodontal disease associated with inadequate
benefit from interventions more than those oral hygiene and smoking.21–24 Consequently,
in disadvantaged groups. Hence, inappropri- the priorities for oral disease prevention should
ate interventions can widen inequities. It is include controlling dietary sugar intake and
necessary to address the root causes, tackling tobacco use and making healthy choices the
social determinants and the environment. easier choices.25 These shared risk factors (Fig
15-5) provide the conceptual basis for the com-
The social determinants of health are the cir- mon risk factor approach (CRFA),26 which is
cumstances into which people are born, grow, one of the most important concepts for oral
live, work, and age (Fig 15-4). These circum- disease prevention. At the same time, it paves
stances, which largely determine the behaviors the way for the close integration of oral health
people adopt and the choices they make, are into strategies addressing NCDs.
in turn shaped by a wider set of forces: eco- The key concept underlying the CRFA is that
nomics, social policies, education, politics, controlling a small number of important clus-
and many more. The unequal distribution tered risk factors should have a major impact on
of all these determining factors accounts for a number of common chronic diseases, includ-
the persisting and growing global differences ing oral conditions, at a lower cost than narrow
in health status and disease burden.20 These disease-specific approaches.26–28 Interventions
inequalities in general and in oral health within to prevent NCDs on a population-wide basis
and between populations pose significant chal- are not only achievable but also cost-effective.29
lenges for policy­makers and those in public The WHO Global Strategy for Control of
health.15 NCDs stated: “We know what works, we know
A major reason that oral diseases remain what it costs, and we know that all countries
a global problem is the absence of rational, are at risk. We have an Action Plan to avert
sustained, interlinked, multitargeted policies millions of premature deaths and help promote
directed at the proximal causes of dental caries a better quality of life for millions more.” 29
and periodontal disease and the determinants Emphasis should therefore be on directing
of those causes. Increases in the numbers of actions at shared behavioral risks common

316
Intervening to Improve Oral Health: The Common Risk Factor Approach

Tooth decay
Unhealthy
Tobacco
diet use
Periodontal disease

Stress Oral trauma Alcohol

Diabetes

Obesity

Lack of control Cancers


Lack of exercise

Cardiovascular disease

Respiratory disease

Poor hygiene From The Challenge of Oral Disease—A call for global action by FDI World Dental Federation. Injuries
Maps and graphics © Myriad Editions 2015

Fig 15-5  |  Common risk factors and their importance for oral health. (Reprinted from FDI World Dental Feder-
ation15 with permission.)

to NCDs.30 Such an approach would expand water fluoridation, and supporting food and
the roles of oral health professionals and nutrition standards for school meals and other
strengthen collaborative linkages with other food and drink sold in schools. Examples of
professional cadres.31,32 local interventions include encouraging schools
Another reason why oral diseases remain to become part of the Health Promoting
a major global problem is the dependence Schools Network,37 encouraging commu-
on individualized “high-risk” approaches to nity action groups to engage in oral health
prevention and health education.19,33,34 These projects, and supporting the development of
approaches divert limited resources away from local infant feeding policies that include oral
upstream healthy public policies and blame health messages19 (see Fig 15-3). Downstream
the victim35; they produce a lifestyle approach interventions operate at the individual level
to health policy instead of a social policy and include such measures as chairside oral
approach to healthy lifestyles and overlook health education and tertiary prevention.19
the way behaviors are generated and struc- These latter interventions are of limited effec-
turally maintained.36 Upstream interventions tiveness and are costly,36 so a shift is required
are those directed at the proximal causes of away from downstream efforts toward a more
disease and operate at national and local lev- appropriate whole-population upstream public
els. National-level actions include encouraging health approach to health policy. In general,
stronger legislation on food labeling and health dentists principally work downstream or mid-
claims on products, supporting legislation on stream, but they do not address the upstream

317
15 Common Risk Factors: The Link Between Oral and Systemic Disease

From The Challenge of Oral Disease—A call for global action by FDI World Dental Federation.
Maps and graphics © Myriad Editions 2015

Fig 15-6  |  The global impact of NCDs. (Reprinted from FDI World Dental Federation15 with permission.)

determinants of oral disease. More effort of 56 million deaths worldwide, with nearly
needs to be focused on the upstream forces 75% of these NCD-related deaths occurring
that cause disease in the first place. Refocus- in low- and middle-income countries. They are
ing efforts upstream requires a healthy public now the major cause of premature death, with
policy approach that involves national and the overwhelming majority of these early deaths
local initiatives, legislation, fiscal measures, and occurring in low and middle-income countries
community development to increase healthy (Fig 15-6). In 2013, the WHO asserted:
settings.
Most of these premature deaths from NCDs
are largely preventable by enabling health
Systemic Disease: The systems to respond more effectively and eq-
uitably to the health-care needs of people
Growing Challenge of NCDs with NCDs, and influencing public policies
in sectors outside health that tackle shared
While deaths due to infectious diseases are de- risk factors—namely tobacco use, unhealthy
clining, NCDs are increasing and have now diet, physical inactivity, and the harmful use
become the leading cause of death and disabil- of alcohol.29
ity worldwide. The most significant of these in
terms of prevalence and severity are cancers, In 2014, the WHO stated that “the human,
CVD, chronic respiratory diseases, and diabe- social and economic consequences of NCDs
tes.1 In 2012, they accounted for 38 million out are felt by all countries but are particularly

318
The WHO Global Action Plan for the Prevention and Control of NCDs 2013 to 2020

devastating in poor and vulnerable popula- The WHO Global Action


tions. Reducing the global burden of NCDs
is an overriding priority and a necessary con- Plan for the Prevention and
dition for sustainable development.”1 A key Control of NCDs 2013 to 2020
entry point for the inclusion of oral disease
within the context of the wider agenda for the
prevention and control of NCDs arose from The stated goal of the WHO Global Action
a 2011 meeting of the General Assembly of Plan for the Prevention and Control of NCDs
the United Nations.38 Addressing the threat 2013–2020 published in 2013 is the following:
of NCDs, the United Nations recognized that
they “undermine social and economic devel- To reduce the preventable and avoidable bur-
opment throughout the world, and threaten den of morbidity, mortality and disability
the achievement of internationally agreed due to noncommunicable diseases by means
development goals.”38 The role of common of multisectoral collaboration and coopera-
risk factors and social determinants in the tion at national, regional and global levels,
etiology of NCDs was acknowledged, and so that populations reach the highest attain-
the high-level declaration that followed the able standards of health and productivity at
meeting also stated “that renal, oral and eye every age and those diseases are no longer
diseases pose a major health burden for many a barrier to well-being or socioeconomic
countries and that these diseases share com- development.29
mon risk factors and can benefit from common
responses to non-communicable diseases.”38 The key objectives of the action plan are
This single sentence in the declaration was a to raise the priority accorded to prevention
highly significant recognition at the highest and control of NCDs in global, regional, and
policy level of the need for an NCD strategy national agendas; strengthen national capacity,
that includes oral disease. leadership, governance, multisectoral action,
The integration of oral and general health and partnerships to accelerate country response
should be the cornerstone of policy approaches for the prevention and control on NCDs; reduce
to improve prevention and control of oral dis- modifiable risk factors for NCDs and under-
eases, as was acknowledged in the Oral Health lying social determinants through creation of
Action Plan adopted by the 60th World Health health-promoting environments; strengthen
Assembly in 2007.2 This plan emphasizes “the and orient health systems to address the preven-
intrinsic link between oral health, general tion and control on NCDs and the underlying
health and quality of life” and identifies “the social determinants through people-centered
need to incorporate programmes for promo- primary health care and universal health cover-
tion of oral health and prevention of oral age; promote and support national capacity for
diseases into programmes for the integrated high-quality research and development for the
prevention and treatment of chronic diseases.”2 prevention and control on NCDs; and monitor
In the same document, the Ministers of Health trends and determinants of NCDs and evaluate
call for the creation of innovative workforce progress in their prevention.29 A high priority
models to integrate essential oral health care for all oral health professionals is to ensure that
into primary health care.2 This is also one of the all NCD action plans also address the global
key strategies set out in FDI’s Vision 20203 and burden of oral disease. This issue is addressed
is developed further later in this chapter (see in more detail later in this chapter (see “Oral
“Taking Action on Common Risk Factors and Health in All Policies”).
the Social Determinants of Health”).

319
15 Common Risk Factors: The Link Between Oral and Systemic Disease

The Associations Between for heart disease and stroke are unhealthy diet,
physical inactivity, tobacco use, and harmful
Oral Diseases and NCDs: use of alcohol.46 This is the basis for the WHO
Making the Case for an recommendation for the cessation of tobacco
use, reduction of salt in the diet, consump-
Integrated Strategy tion of fruits and vegetables, regular physical
activity, and avoiding harmful use of alcohol
The importance of tobacco use, unhealthy to reduce the risk of CVD.
diet, physical inactivity, and the harmful use The relationship between diabetes and oral
of alcohol as common risk factors for CVD, diseases, particularly periodontal disease, has
cancers, respiratory disease, and diabetes29 has been the subject of intense research. For exam-
already been discussed (see “Systemic Disease: ple, there is good evidence from meta-analyses
The Growing Challenge of NCDs”), and the that type 2 diabetes is a risk factor for peri-
role of these same risk factors in the etiology odontal disease.47,48 Furthermore, a separate
of oral diseases is also the basis of the CRFA meta-analysis has shown that while diabetic
discussed previously (see “Intervening to Im- patients had the same prevalence of periodon-
prove Oral Health: The Common Risk Factor tal disease as nondiabetic patients, this was of
Approach”). Thus, it is not surprising that there significantly higher severity in the former.49
are associations of varying strength between Over the last 20 years, consistent and robust
oral diseases and NCDs, as shown in Fig 15-5. evidence has emerged that severe periodontitis
For example, there is high-level evidence that adversely affects glycemic control in patients
individuals with periodontitis have a signifi- with diabetes and glycemia in patients without
cantly higher risk of various other problems, diabetes. In diabetic patients, there is a direct
including CVD, diabetes mellitus, respirato- and dose-dependent relationship between
ry disease, and preterm delivery of low–birth periodontitis severity and diabetes complica-
weight babies,39 but the evidence for these as- tions. Emerging evidence supports an increased
sociations is of variable strength. There is also risk for diabetes onset in patients with severe
intense debate in the literature as to whether periodontitis.50 While there is some evidence
these associations are indicative of causal re- suggesting that periodontal disease adversely
lationships between oral and systemic disease. affects diabetes outcomes, further longitudi-
This matter is addressed in detail in other chap- nal studies are needed to test this assertion.
ters in this publication. However, it is reasonable to conclude on the
There is good evidence from epidemiologic basis of current evidence that patients should
studies for a moderate, positive association be encouraged to adopt a healthy lifestyle to
between periodontal diseases and CVD,40 reduce the risk of developing diabetes, one of
and evidence from systematic reviews with the complications of which is severe periodon-
meta-analyses indicates that patients with tal disease. The presence of severe periodontal
chronic periodontal disease are at increased disease should alert the oral health practitioner
risk of developing coronary heart disease41 to the possibility that the patient may have
and carotid atherosclerosis.42 This has led to undiagnosed diabetes. To this end, Chapple et
speculation about plausible biologic mecha- al50 have recommended that, given the current
nisms to implicate periodontal disease in the evidence, it is timely to provide guidelines for
etiology of CVD.43 However, in spite of intense periodontal care in diabetic patients for medical
research, there is insufficient evidence for a and dental professionals as well as recom-
causal relationship between periodontal disease mendations for patients and the public. These
and CVD.44,45 By contrast, it is well established guidelines include the specific recommendation
that the most important behavioral risk factors that patients who present without a diabetes

320
Taking Action on Common Risk Factors and the Social Determinants of Health: An Integrated Strategy for Oral Health

diagnosis but with obvious risk factors for type countries since 1990. Rapid decreases have
2 diabetes and signs of periodontitis should be occurred in some countries. For example, five
informed about their risk for having diabetes, countries (Israel, Denmark, Norway, South
assessed using a chairside hemoglobin A1c test, Korea, and the United Kingdom) had at least
and/or referred to a physician for appropriate a 65% decrease in age-standardized death
diagnostic testing and follow-up care. rates for ischemic heart disease. Many other
In addition to the associations discussed countries have had decreases of 40% to 65%.53
previously, there is also evidence for associa- These decreases coincide with the introduction
tions of varying strength between periodontitis of policies to reduce exposure to risk factors for
and caries with respiratory disease, particu- CVD, specifically the cessation of tobacco use,
larly chronic obstructive pulmonary disease reduction of salt in the diet, increased consump-
(COPD).51 In light of the foregoing discussion tion of fruits and vegetables, regular physical
about periodontal disease, it is important to activity, and avoiding harmful use of alcohol.
note that the most significant risk factor for the The key conclusions arising from this sec-
development of COPD is cigarette smoking,52 tion are that actions to reduce the burden of
with case-control studies also pointing to an NCDs—including oral diseases—should (1)
increased risk associated with passive smoking. address the common risk factors and the social
To summarize the evidence presented in determinants of health, (2) be implemented
this section, there are associations of varying early in the life course, and (3) be directed
strength between oral disease and NCDs, partic- at upstream interventions operating at the
ularly diabetes and CVD. There is also evidence level of national policies, and they need to be
for the importance of CRFA in these associa- integrated.
tions, to which should be added the importance
of tobacco and alcohol use as risk factors for
oropharyngeal cancer and other body-site can-
cers.15 The effects of common risk factors and Taking Action on Common
the social determinants of health are cumulative
over the life course, so many of the reported
Risk Factors and the Social
associations occur in middle-aged and older Determinants of Health: An
adults as the prevalence of NCDs increases. Integrated Strategy for Oral
The long latent period between initial expo-
sure and the development of frank clinical Health
disease means that intervention studies are
difficult to conduct, but there is good reason A new model for oral health promotion is need-
to believe that upstream interventions directed ed.3,54 The traditional “vertical” approach to
at reducing the effects of common risk factors public health, where individual diseases are ad-
are effective. The most notable example is the dressed in isolation, should be replaced by the
success of actions directed at reducing pre- “horizontal” approach that addresses a num-
mature mortality due to CVD. The aging and ber of diseases at the same time and uses the
growth of populations has led to an increase CRFA.55 Intersectoral action is necessary at all
in the total number of cardiovascular deaths, stages of policy formulation and implementa-
which accounted for almost one-third of all tion because the major determinants of chronic
deaths globally in 2013. However, while the disease lie outside the health sector. Hence, pol-
overall number of deaths has risen between icies and plans should focus on the common
1990 and 2013, age-standardized death rates risk factors and cut across specific diseases.
for cardiovascular and circulatory diseases have They should be population-wide, with indi-
fallen in high-income and many middle-income vidual interventions being integrated with the

321
15 Common Risk Factors: The Link Between Oral and Systemic Disease

Box 15-1  |  Taking action on oral health equity: Things the oral health team should do

1. All members of the oral health team should acquire a thorough understanding of the importance that social
determinants play in oral as well as general health. They should have a thorough understanding of how the con-
ditions in which people are born, live, work, and age can affect their health and how they can act to tackle these.
2. Dentists and the oral health team should engage in partnership with communities to help them better understand
and tackle the social, economic, and environmental factors that determine oral health and increase inequalities.
3. Dentists and the oral health team should engage with colleagues such as primary health care professionals in
the development of cross-sectoral partnerships so that oral health promotion strategies become incorporated
into all strategies for health.
4. Dentists should become advocates for health, particularly oral health, with their patients and the wider community.
This should include an emphasis on acting as enablers, helping to make healthy choices the easier choices and
empowering people to take control of their own lives and health.

(Based on Williams et al.58 )

population approaches.56 If behaviors are to is required. Some of the principles that must
change, the environment that predisposes people underpin action include the following:
to adopt health-compromising behaviors must
also be addressed. Health promotion to improve • Tailoring the response to the level of oral
social and physical environments supportive of and general health need
health is pivotal to improving health and should • Building on community assets and
redress the balance of influences away from pre- strengthening family competence to
scription to make healthier choices easier and self-manage health, including oral health
facilitate decision-making skills. This includes • Emphasizing the early years and early
combatting the upstream influences of those intervention
who produce and profit from ill health and • A family focus
points to a requirement for greater regulation • A personalized approach to delivering
of the food, drink, and tobacco industries. services
Integrated policy action across individual,
community, and societal intervention levels, All primary health care professionals should
as well as across the life course, are needed tackle the needs of families in the context of
to tackle complex health problems such as the environment and their experience. There is
obesity, CVD, and caries effectively and to a fundamental need to integrate initiatives to
reduce health inequalities.57 While it is clear improve oral health with more general inter-
that physicians and other health professionals ventions to support good physical and mental
have a role in promoting health equity, dental health. Primary care is the first point of con-
teams are in an important position to actively tact with the health service and is the setting
engage in promoting oral health equity, both in which most care—both general and oral—
for their patients and the wider community58 is provided. Oral health teams, collaborating
(Box 15-1). with primary care teams, have the largely
Given the close links between oral health unexploited potential to be important advo-
and other indicators such as family income cates, enablers, and mediators for oral health.
and educational attainment of children and Because the risk factors for oral and general
parents, a whole-systems approach to improv- health are the same, such activities will also
ing oral health in the context of general health promote good general health.

322
Oral Health Professionals as Advocates for Oral Health

Primary medical care is increasingly appre- are often highly invested in the status quo and
ciating the pivotal importance of social resistant to change. Moreover, adding oral
determinants in influencing health status and health concerns to already overtaxed public
health outcomes. If oral health care is to be health and primary care systems requires a
properly integrated with health care in general, redistribution of resources that needs to be
it is essential that all members of the oral health driven by a sustained political will. The exam-
team understand the importance of the social ple of the US legislative battles to achieve
determinants of oral health and integrate their parity for mental health services gives reason
activities with other groups. for optimism. As Hernandez and Uggen63 have
shown, important policy changes that include
integration of services and the redistribution
of scarce resources can be achieved, even when
Oral Health in All Policies organized resistance to policy change is wide-
spread and neoliberal ideology predominates.
Because oral diseases share social determinants, But policy change depends on a well-organized
common risk factors, and biologic pathways and consistent advocacy message from various
with other NCDs, oral health belongs in a stakeholders who have the stability to sustain
“Health in All Policies” (HiAP) approach.59 This their messaging throughout a policy-change
integrative approach will require high-level pol- process that may take decades to achieve.
icy initiatives that lead to an “Oral Health in All
Policies” (OHiAP) concept. The WHO Consti-
tution60 acknowledges that “enjoyment of the
highest attainable standard of health is one of
Oral Health Professionals as
the fundamental rights of every human being,” Advocates for Oral Health
and the WHO Adelaide Statement on Health in
All Policies61 advocated for an HiAP approach Oral health professionals have opportunities
as an important strategy to advance this human to become strong advocates for upstream pol-
right. The acknowledgment of oral health as a icy changes that can reduce health-harming
serious threat to overall health and well-being social conditions and close the inequality
and understanding that many oral disease share gap in disease by working collectively at the
common risk factors and biologic pathways community and national levels through their
with other NCDs legitimizes the inclusion of professional dental organizations. This is not
oral health in an HiAP approach and could a role generally embraced by oral health pro-
lead to new intersectoral partnerships and, as fessionals, but national dental organizations
suggested by Meier et al,62 shift the focus in have it within their power to make their voic-
oral health from one of technical interventions es heard and become advocates for policies
toward one based on social justice and consid- that will address these inequalities effectively.
eration of the social determinants of health. This Leaders within the dental profession need to
shift substantially expands the strategies and improve understanding among their members
resources available to address oral disease and of the central role that social determinants of
will lead to a breakdown in the artificial silos health play in creating these persistent inequal-
that have for too long maintained oral health in ities in oral disease. By shifting the policy focus
a separate category from general health policy. toward common risk factor reduction and up-
Achieving parity in policy and full integra- stream interventions, the dental profession can
tion of oral health necessitates substantial significantly expand their alliances with other
modifications of national (dental and non- chronic disease stakeholders and leverage that
dental) health care delivery systems, which collective influence as they seek to reduce sugar

323
15 Common Risk Factors: The Link Between Oral and Systemic Disease

Box 15-2  |  Recommendations on integrating oral health into the NCD agenda

• Improve access to health services and ensure more supportive social conditions for disadvantaged groups to
reduce social inequities.
• Adopt both an HiAP and OHiAP approach to minimize and manage risks to oral health, general health, and health
equity arising from policies in other sectors.
• Implement cost-effective evidence-based population-wide oral health promotion measures as a way of protecting
overall health and well-being.
• Strengthen interprofessional collaboration between oral health and other health professionals to improve prevention
and management of comorbidities, for example through shared health records.
• Include oral health in curricula for other health care professionals and ensure that oral health care professional
education addresses associated diseases and interdisciplinary care.
• In low and middle-income countries, integrate oral and NCD care into current programs, such as those for HIV/
AIDS, to improve collaboration and capitalize on existing systems.
• Integrate oral and NCD care into broader efforts to achieve universal health coverage.
• Include oral health and NCD workforce planning in overall planning for human resources for health.
• Strengthen health care professional education and collaboration to ensure that oral disease and NCD risks are
appropriately considered in maternal and pediatric care.
• Implement community-based initiatives, such as school education programs, to promote healthy behaviors from
an early age.
• Ensure healthy environments for children, for example through banning sugar-sweetened beverages and unhealthy
snacks in schools and ensuring that healthy food is available.
• Systematically include oral disease and NCD surveillance in epidemiologic monitoring, including surveillance of
common modifiable risk factors.
• Promote research into effective interventions for oral health and NCDs, focusing on what works in the area of
social and behavioral interventions to tackle the common modifiable risk factors.
• Integrate oral health perspectives into national NCD action plans and other relevant NCD governance mechanisms.
• Fully integrate oral health into sustainable development goals strategies and monitoring frameworks.

(Based on the policy brief from the FDI World Dental Federation and the NCD Alliance.64 )

consumption, increase access to healthy diets, civil society organizations in more than 170
reduce tobacco and excessive alcohol consump- countries with the mission to combat the NCD
tion, and address the other risk factors that epidemic by putting health at the center of all
contribute to inequalities in oral and gener- policies. Strategic partners include the United
al health. Advocating for common risk factor Nations, WHO, and governments, and the
reduction and upstream interventions will al- long-term goals of the organization are aligned
low dentistry to take part in championing the with the WHO Global Action Plan for the Pre-
value of health equity, create clarity of goals, vention and Control of NCDs 2013–2030.28
and further integrate oral health into general The FDI World Dental Federation has taken a
health at all levels. lead among dental organizations in joining the
Consistent with the principle of integration NCD Alliance, and the most tangible output
that runs throughout this chapter, if the oral from this partnership has been the publica-
health community is to be effective in its advo- tion of a joint FDI/NCD Alliance policy brief
cacy efforts, then it will be essential to develop titled “Accelerating Action on Oral Health and
strategic partnerships with those engaged in NCDs: Achieving an Integrated Response.”64
action against NCDs in general. Arguably the This policy brief makes a number of practical
most effective organization engaged in this recommendations for action (Box 15-2) that
regard is the NCD Alliance. Founded in 2009, are consistent with the goals discussed else-
the NCD Alliance is a network uniting 2,000 where in this chapter.

324
Conclusion

The Care Team: into primary health care.66 Furthermore, in


their role as clinical service providers, they need
Interprofessional Education to adapt the chronic care model for manage-
and Collaboration ment of oral disease67 to that used for other
chronic systemic diseases. Thus, there needs
to be widespread implementation of health
The current training, service provision, and dis- promotion efforts that improve the ability of
tribution of dental care teams responsible for patients to manage their own health.
the delivery of the majority of clinical dental Full implementation of the provision of
services do not adequately address the needs of oral health services throughout the primary
most populations.6 Therefore, a change is re- care system will require changes in the over-
quired, and a first step is to change the existing all care delivery infrastructure. The chronic
education model for oral health professionals care model also emphasizes the provision of
and give equal emphasis to preventive services clinical services that are evidence-based and
and risk reduction through use of a chronic cost-effective. As stated earlier, a new model
disease management approach. Risk reduction for oral health promotion is needed3,54—one
through such a medical management ap- that considers oral health as an integral part
proach to chronic diseases such as caries and of general health, addresses the needs and
periodontal disease should provide immediate demands of populations, and includes an
improvements in patient outcomes while reduc- integrated upstream public health approach,
ing costs. However, this will only happen if it is moving health from an individual lifestyle
accompanied by changes in the way dentists and choice model to one that tackles the social
other oral health professionals are remunerated. determinants of chronic diseases.
We need to move away from procedure-based
reimbursement and toward value-based
models that reward improvements in individu-
al- and population-level health status. Conclusion
A second step is the expansion and integra-
tion of the dental care delivery system into The challenge that the burden of oral disease
the broader general medical care infrastruc- presents is immense and will not be met ef-
ture. Incorporating oral health services into fectively by the dental profession continuing
primary health care will improve access and to work in a dental silo. It will require closer
provide better coordination of overall health engagement with the other health professions,
care. To facilitate the necessary conceptual engagement with—and advocacy on behalf
change requires acknowledgement by schools of—civil society, and a high degree of leader-
educating non–oral health care professionals ship. The leaders within the dental profession
that oral health is integral to overall health. must increase understanding among their
Their curricula should change in line with members of the central role that the social de-
WHO recommendations on “Transforming terminants of health play in creating persistent
and Scaling Up Health Professionals’ Educa- inequalities in oral disease. Advocating for re-
tion and Training,”65 with the goal of creating ductions in common risk factors will allow
an “Oral Health in All Training” approach dentistry to take part in championing the val-
across all health professional education. ue of health equity, create clarity of goals, and
Oral health professionals can improve their integrate oral health into monitoring of general
ability to contribute to the overall health care health. National dental organizations should
mission by playing a stronger role in overall advocate for policies that will address inequal-
health management through greater integration ities in oral and general health effectively. They

325
15 Common Risk Factors: The Link Between Oral and Systemic Disease

should advocate for much greater emphasis on 2. Agenda item 12.9. Oral health: Action plan for promotion
and integrated disease prevention [Proceedings of the
controlling dietary sugars and tobacco use, re-
Sixtieth World Health Assembly, 14–23 May 2007,
ducing excessive alcohol consumption, and Geneva]. Geneva: World Health Organization, 2007.
making healthy choices the easy choices. 3. Glick M, Monteiro da Silva O, Seeberger GK, et al. FDI
Current approaches to the control of oral Vision 2020: Shaping the future of oral health. Int Dent
J 2011;62:278–291.
disease are of limited effectiveness and econom- 4. Statistics and Information Department, Ministry of
ically unsustainable. Oral diseases share the Health, Labour and Welfare Department. Estimates of
same common risk factors and social determi- National Medical Care Expenditure [in Japanese]. http://
nants as the major NCDs. Thus, to reduce the www.mhlw.go.jp/toukei/saikin/hw/k-iryohi/08/index.
html. Accessed 31 May 2018.
growing global threat to society of poor oral 5. Yee R, Sheiham A. The burden of restorative dental
health, the focus must shift toward a greater treatment for children in third world countries. Int Dent
emphasis on health promotion and the inte- J 2002; 52:1–9.
6. Sheiham A, Williams DM, Weyant RJ, et al. Billions with
gration of oral health with general health in
oral disease: A global health crisis—A call to action. J
all strategies. The horizontal HiAP approach Am Dent Assoc 2015;146:861–864.
that addresses a number of diseases at the same 7. Petersen PE, Bourgeois D, Ogawa H, Estupinan-Day
time and uses the CRFA should be widened to S, Ndiaye C. The global burden of oral diseases and
risks to oral health. Bull World Health Organ 2005;83:
include oral diseases.
661–669.
Acting alone, the dental profession is 8. Petersen PE. The World Oral Health Report 2003:
unlikely to be effective in achieving good oral Continuous improvement of oral health in the 21st
health outcomes. Hence there is a need for a century—The approach of the WHO Global Oral Health
Programme. Community Dent Oral Epidemiol 2003;
“multi-sectoral approach to oral health, linking
31(suppl 1):3–23.
the health sector with sectors of society such 9. Nadanovsky P, Sheiham A. Relative contribution of
as education, agriculture, trade, environment, dental services to the changes in caries levels of
energy, social welfare, housing and others that 12-year-old children in 18 industrialized countries in the
1970s and early 1980s. Community Dent Oral Epidemiol
impact on oral health.”68 1995;23:331–339.
10. Elani HW, Harper S, Allison PJ, Bedos C. Kaufman JS.
Socio-economic inequalities and oral health in Canada
and the United States. J Dent Res 2012;91:865–870.
Acknowledgments 11. Oral Health America. State of Decay: Are Older Amer-
icans Coming of Age Without Oral Healthcare? http://
origin-qps.onstreammedia.com/origin/multivu_archive
My views on the response of the dental pro- / PRNA / ENR / FX-20131008-MM93259.pdf. Accessed
31 May 2018.
fession to the global challenge of oral disease
12. Bagramian RA, Garcia-Godoy, Volpe AR. The global
have evolved during numerous discussions with increase in dental caries. A pending public health crisis.
the late Professor Aubrey Sheiham, my mentor Am J Dent 2009;22:3–8.
and friend over many years. I also gratefully ac- 13. Benzian H. The neglect of global oral health: Symptoms
and solutions [thesis]. Amsterdam: University of Amster-
knowledge the contribution to development of
dam, 2014. http://dare.uva.nl/record/462712. Accessed
the principles set out in sections 8 and 9 by my 31 May 2018.
coauthors Sheiham A, Williams DM, Weyant 14. Marcenes W, Kassebaum NJ, Bernabé E, et al. Global
RJ, et al.6 burden of oral conditions in 1990-2010: A systematic
analysis. J Dent Res 2013;92:592–597.
15. The Challenge of Oral Disease. A Call for Global Action.
The Oral Health Atlas, ed 2. Geneva: FDI World Dental
Federation, 2015.
References 16. Tomar SL, Cohen LK. Attributes of an ideal oral health
care system. J Public Health Dent 2010; 70(suppl 1):
S6–S14.
1. Global Status Report on Noncommunicable Diseases
17. Garcia I, Tabak LA. Global oral health inequalities: The
2014. Geneva: World Health Organization, 2014.
view from a research funder. Adv Dent Res 2011;23:
207–210.

326
References

18. Kwan S, Petersen PE. Oral health: Equity and social 37. Health Promoting Schools: A Framework for Action.
determinants. In: Blas E, Sivasankara Kurup A (eds). Geneva: World Health Organization, 2009.
Equity, Social Determinants and Public Health 38. United Nations. Political Declaration of the High-Level
Programmes. Geneva: World Health Organization, Meeting of the General Assembly on the Prevention and
2010:159–176. Control of Non-communicable Diseases. http: //www.
19. Watt RG. From victim blaming to upstream action: Tack- who.int/nmh/events/un_ncd_summit2011/political_
ling the social determinants of oral health inequalities. declaration_en.pdf. Accessed 1 June 2018.
Community Dent Oral Epidemiol 2007;35:1–11. 39. Azarpazhooh A, Tenenbaum HC. Separating fact from
20. Whitehead M, Dahlgren G. What can be done about fiction: Use of high-level evidence from research synthe-
inequalities in health? Lancet 1991;338:1059–1063. ses to identify diseases and disorders associated with
21. Moynihan P, Kelly S. Effect on caries of restricting sugars periodontal disease. J Can Dent Assoc 2012;78:c25.
intake: Systematic review to inform WHO guidelines. J 40. Kebschull M, Demmer RT, Papapanou PN. “Gum bug,
Dent Res 2014;93:8–18. leave my heart alone!”—Epidemiologic and mechanistic
22. Thomson WM, Sheiham A, Spencer AJ. Sociobehavioral evidence linking periodontal infections and atheroscle-
aspects of periodontal disease. Periodontol 2000 2012; rosis. J Dent Res 2010;89:879–902.
60:54–63. 41. Bahekar AA, Singh S, Saha S, Molnar J, Arora R. The
23. Watt RG, Petersen PE. Periodontal health through public prevalence and incidence of coronary heart disease is
health—The case for oral health promotion. Periodontol significantly increased in periodontitis: A meta-analysis.
2000 2012;60:147–155. Am Heart J 2007;54:830–837.
24. Bergström J. Tobacco smoking and chronic destructive 42. Zeng XT, Leng WD, Lam YY et al. Periodontal disease
periodontal disease. Odontology 2004;92:1–8. and carotid atherosclerosis: A meta-analysis of 17,330
25. Milio N. Making healthy public policy; Developing the participants. Int J Cardiol 2016;203:1044–1051.
science of art: An ecological framework for policy stud- 43. Papanou PN, Trevisan M. Periodontitis and atheroscle-
ies. Health Promot 1987;2:236–274. rotic vascular disease: What we know and why it is
26. Sheiham A, Watt RG. The common risk factor approach: important. J Am Dent Assoc 2012;143:826–828.
A rational basis for promoting oral health. Community 44. Lockhart PB, Bolger AF, Papapanou PN, et al. Peri-
Dent Oral Epidemiol 2000;28:399–406. odontal disease and atherosclerotic vascular disease:
27. Watt RG, Sheiham A. Integrating the common risk factor Does the evidence support an independent associa-
approach into a social determinants framework. tion?: A scientific statement from the American Heart
Community Dent Oral Epidemiol 2012;40:289–296. Association. Circulation 2012;125:2520–2544.
28. Global Status Report on Noncommunicable Diseases 45. Dietrich T, Sharma P, Walter C, Weston P, Beck J. The
2010. Geneva: World Health Organization, 2010. epidemiological evidence behind the association
29. Global Action Plan for the Prevention and Control of between periodontitis and incident atherosclerotic
NCDs 2013–2020. Geneva: World Health Organization, cardiovascular disease. J Periodontol 2013; 84(4 suppl):
2013. S70–S84.
30. Beaglehole R, Bonita R, Horton R, et al. Priority actions 46. World Health Organization. Cardiovascular Diseases
for the non-communicable diseases crisis. Lancet 2011; (CVDs). http: //www.who.int/mediacentre/factsheets/
377:1438–1447. fs317/en/. Accessed 31 May 2018.
31. Frenk J, Chen L, Bhutta ZA, et al. Health professionals 47. Chávarry NG, Vettore MV, Sansone C, Sheiham A. The
for a new century: Transforming education to strengthen relationship between diabetes mellitus and destructive
health systems in an interdependent world. Lancet periodontal disease: A meta-analysis. Oral Health Prev
2010;376:1923–1958. Dent 2009;7:107–127.
32. FDI World Dental Federation. FDI policy statement on 48. Borgnakke WS, Ylostalo PV, Taylor GW, Genco RJ.
non-communicable diseases. Adopted by the FDI Effect of periodontal disease on diabetes: Systematic
General Assembly: 30 August 2013—Istanbul, Turkey. review of epidemiologic observational evidence. J Peri-
Int Dent J 2013;63:285–286. odontol 2013;84(4 suppl):S135–S152.
33. Bachelor P, Sheiham A. The limitations of a ‘high-risk’ 49. Khader YS, Dauod AS, El-Qaderi SS, Alkafajei A,
approach for the prevention of dental caries. Community Batayha WQ. Periodontal status of diabetics compared
Dent Oral Epidemiol 2002;30:302–312. with nondiabetics: A meta-analysis. J Diabetes Compli-
34. Harper S. Rose’s strategy of preventive medicine. Geof- cations 2006;20:59–68.
frey Rose with commentary by Kay-Tee Khaw and 50. Chapple IL, Genco R, working group 2 of the joint EFP/
Michael Marmot. Int J Epidemiol 2008;38:1743–1745. AAP workshop. Diabetes and periodontal diseases:
35. Crawford R. You are dangerous to your health: The Consensus report of the Joint EFP/AAP Workshop on
ideology and politics of victim blaming. Int J Health Serv Periodontitis and Systemic Diseases. J Periodontol
1977;7:663–680. 2013;84(4 suppl):S106–S112.
36. McKinlay JB. Paradigmatic obstacles to improving the 51. Manger D, Walshaw M, Fitzgerald R, et al. Evidence
health of populations—Implications for health policy. summary: The relationship between oral health and
Salud Publica Mex 1998;40:369–379. pulmonary disease. Br Dent J 2017;222:527–533.

327
15 Common Risk Factors: The Link Between Oral and Systemic Disease

52. Devereux G. ABC of chronic obstructive pulmonary 60. World Health Organization. Constitution of the World
disease: Definition, epidemiology and risk factors. BMJ Health Organization. http://www.who.int/governance/
2006;322:1142–1144. eb/who_constitution_en.pdf. Accessed 1 August 2018.
53. GBD 2013 Mortality and Causes of Death Collaborators. 61. World Health Organization. Adelaide Statement on
Global, regional, and national age-sex specific all-cause Health in All Policies: Moving Towards a Shared Gover-
and cause-specific mortality for 240 causes of death, nance for Health and Well-Being. http: //www.who.int/
1990–2013: A systematic analysis for the Global Burden social_determinants/hiap_statement_who_sa_final.pdf.
of Disease Study 2013. Lancet 2015;385:117–171. Accessed 1 June 2018.
54. Glick M, Monteiro da Silva O, Seeberger GK, et al. FDI 62. Meier BM, Brodish PH, Koivusalo M. Human rights
Vision 2020: Shaping the future of oral health. Int Dent provide justification for the Health in All Policies
J 2012;62:278–291. approach. http: www.hhrjournal.org2013 06 10 human -
55. Frenk J. Bridging the divide: Global lessons from rights-provide-justification-for-the-health-in-all-policies-
evidence-based health policy in Mexico. Lancet 2006; approach. Accessed 1 June 2018.
368:954–961. 63. Hernandez EM, Uggen C. Institutions, politics, and
56. Epping-Jordan JE, Galea G, Tukuitonga C, Beaglehole mental health parity. Soc Ment Health 2012;2(3).
R. Preventing chronic diseases: Taking stepwise action. 64. FDI World Dental Federation, NCD Alliance. Accelerating
Lancet 2005;366:1667–1671. Action on Oral Health and NCDs: Achieving an Inte-
57. Bambra CL, Hillier FC, Moore HJ, Summerbell CD. grated Response. http://www.fdiworlddental.org/sites/
Tackling inequalities in obesity: A protocol for a system- default/files/media/resources/ncda_fdi-policy_
atic review of the effectiveness of public health brief_oral_health_ncds.pdf. Accessed 1 June 2018.
interventions at reducing socioeconomic inequalities in 65. Transforming and Scaling Up Health Professionals’
obesity amongst children. Syst Rev 2012;1:16. Education and Training: World Health Organization
58. Williams DM, Sheiham A, Watt RG. Dentists and oral Guidelines 2013. Geneva: World Health Organization,
health teams. In: Allen M, Allen J, Hogarth S, Marmot 2013.
M (eds). Working for Health Equity: The Role of Health 66. Glick M. Our health: Do we get what we pay for? J Am
Professionals. London: UCL Institute of Health Equity, Dent Assoc 2013;144:1218–1220.
2013:93–94. 67. Coleman K, Austin BT, Brach C, Wagner EH. Evidence
59. Ståhl T, Wismar M, Ollila E, Lahtinen E, Leppo K. Health on the chronic care model in the new millennium. Health
in All Policies: Prospects and Potentials. Helsinki: Minis- Aff (Millwood) 2009;28:75–85.
try of Social Affairs and Health, 2006. 68. Mossey PA, Petersen PE. Budapest Declaration:
IADR-GOHIRA®. J Dent Res 2014; 93(7 suppl):
120S–121S.

328
CHAPTER 16

Antibiotic Prophylaxis for


Patients at Risk for Infective
Endocarditis
Peter B. Lockhart, dds, fds rcsed, fds rcps

The ancient notion of an association between the efficacy for prevention of IE has increased
oral and systemic disease has had a reviv- greatly in recent years.
al in the past 25 years, although these more
recent claims have not been confirmed by
clinical studies.1 However, scientific evidence
connecting oral bacterial pathogens and infec- Antibiotic Prophylaxis
tive endocarditis (IE) is undisputed, and the to Prevent Distant Site
strength of the association between invasive
procedures and IE has a long history. Around Infections
1940, with the advent of the first antibiotics,
it was proposed that people at risk for IE (eg, Antibiotic prophylaxis recommendations for
history of rheumatic fever) might benefit from invasive dental procedures can be broken
the use of antibiotics prior to invasive proce- into two types: primary and secondary. Pri-
dures. In 1955, the American Heart Association mary prophylaxis involves giving antibiotics
(AHA) issued their first recommendations for during a procedure (ie, intraoperative) to pre-
antibiotic prophylaxis,2 which included ad- vent infection at the operative site. For some
ministration of an intramuscular injection 30 surgical procedures, such as placement of a
minutes before the procedure. In a revised AHA prosthetic joint or an aortic graft, it has been
recommendation in 1957, oral antibiotics four shown that primary prophylaxis reduces the
times per day for 2 days before, on the day of, incidence of infection at the site of surgery.
and for 2 days after the procedure were added Secondary antibiotic prophylaxis is given pri-
to the intramuscular dose. The trend since then, or to an invasive procedure in order to reduce
however, has been a reduction in the number a procedure-related bacteremia and thereby
of days of prophylaxis as well as the dose, to prevent the development of an infection at a
eventually eliminate the intramuscular dose in distant site; the best-known example is IE. It is
1990. These recommendations have evolved unclear if or to what extent secondary prophy-
to the present time, and the controversy about laxis prevents distant site infections.

329
Antibiotic Prophylaxis for Patients at Risk for Infective Endocarditis

80–

70– 67.2%

60–
Percent of respondents

50–

40– 36.9%
30.3%
30–
22.4% 22.1%
20– 13.8%
10–

0–
Mitral valve Congenital Rheumatic Aortic Bicuspid Other
prolapse heart heart or mitral valve
conditions* disease stenosis disease

*Such as ventricular septal defect, atrial septal defect, and hypertrophic cardiomyopathy

Fig 16-1  |  Cardiac conditions for which some patients continue to receive antibiotic prophylaxis although this
is no longer recommended by the 2007 AHA guidelines.4

What Do We Know About dermatologic, and other invasive procedures


may also cause a bacteremia of pathogenic
the Issues Surrounding species known to cause IE.
Antibiotic Prophylaxis? • Antibiotics given an hour prior to an inva-
sive dental procedure will likely reduce but
not eliminate the number of circulating oral
The following section lists what we know, what viridans group streptococci, and it is believed
we think we know, and what we need to know that this reduces the risk of IE.7
about antibiotic prophylaxis: • Antibiotic prophylaxis, both primary and
secondary, is being used for many patient
• Antibiotic prophylaxis compliance is poor populations in dental practice for reasons
for patients and clinicians in part because that lack a scientific basis.
of confusion and a lack of understanding • Antibiotic prophylaxis may be cost-effective
concerning the AHA recommendations as in certain situations, but only where it is suf-
well as a wide diversity of opinion among ficiently effective in preventing disease and
physician specialty groups about the need where the costs of developing the disease
for prophylaxis prior to dental procedures3–6 outweigh the costs of preventing it.8 This
(Fig 16-1). This confusion also arises because may be true in the case of antibiotic pro-
of conflicting evidence in the literature over phylaxis for IE in those patients at high risk9
time and a lack of definitive answers for the but not in other situations.
issue of efficacy. • Recent data suggest that a single dose of
• There is a long-standing assumption that amoxicillin prior to a dental procedure is
dental procedures are the main risk factor safe with regard to drug reactions,10 but clin-
for IE despite evidence that gastrointestinal, damycin may put patients at significant risk.

330
What Is Infective Endocarditis?

Fig 16-2  |  IE of the aortic valve. Note the vegeta-


tions on the valve leaflets (arrows).

• We need to know if single-dose use of anti- valves. There are about 40,000 to 50,000
biotics is contributing to the problem of new cases in the United States each year.11
drug-resistant organisms. The mechanism for these infections has sev-
• We need to know which dental procedures eral steps. Briefly, a platelet plug forms on a
put patients at risk. damaged heart valve and bacteria from the
• The risk of bacteremia following dental bloodstream lodge in this platelet plug, which
extractions and other manipulations of grows to become a vegetation (Fig 16-2). Vege-
the gingival tissues comes from bacter­ tations may obstruct the normal flow of blood
emia studies, but the incidence and nature through the valve by perforating or obstructing
of bacteremia from necrotic pulp tissue is valve closure and may allow the backflow of
unknown. Although often referred to, there blood through the valve. Both mean the heart
are few data on bacteremia during chewing becomes inefficient in pumping blood around
food or flossing teeth. the body, resulting in heart failure. In a fur-
• We need a definitive epidemiologic study to ther complication, fragments of vegetations
determine if antibiotic prophylaxis prior to may break off and travel as infected emboli to
dental procedures reduces the risk for IE. distant sites in the body, including the brain,
• We need to know if those with poor oral where they can cause a stroke. For these rea-
hygiene are really at significantly greater risk sons, patients with IE require urgent hospital
of developing IE as a result of daily activities admission and high-dose intravenous antibi-
compared to similar individuals with good otics for up to 6 weeks. Complications are
oral hygiene. common, and the prognosis includes a mortal-
ity rate of 15% to 20% during hospitalization,
and 40% to 50% will require valve repair or
What Is Infective replacement.11 Upward of 25% of patients will
die within the first year following the infection.
Endocarditis? These patients are also at risk for IE in the fu-
ture, which carries a poor prognosis, and they
IE is an uncommon if not rare infection of are at risk for developing heart failure and ad-
the heart myocardium, particularly the heart ditional long-term morbidity.

331
16 Antibiotic Prophylaxis for Patients at Risk for Infective Endocarditis

Types of IE (ie, morbidity and mortality) and the risk of


acquiring IE. The moderate-risk group is made
There are two general types of IE: community- up of people with a history of rheumatic fever
acquired and medically related. Community- or mitral valve prolapse, for example; in com-
acquired IE occurs in people with a heart valve parison, the high-risk group involves four
defect who acquire this infection from a bac- categories of cardiac patients, those with (1)
teremia from a skin or mucosal surface either artificial heart valves, (2) a prior history of IE,
during an invasive procedure or routine daily (3) certain congenital heart defects, and (4) a
activities (eg, tooth brushing). These infections previous heart transplant who now have a val-
are often caused by the viridans streptococci vular defect. The moderate-risk group includes
group of bacteria, some of which are very com- about 90% of patients at risk for IE, and the
monly found in the mouth. This is the group remaining 10% are considered high risk.8
of people who are targeted for antibiotic pro- The 2007 AHA recommendations are
phylaxis prior to invasive dental procedures. generally well accepted by both dentists and
Medically related IE occurs in people who physicians involved in the care of patients with
have a medical condition or device that puts cardiac conditions. These recommendations,
them at risk. This includes people on renal however, do not cover all scenarios of patients
dialysis, with prosthetic heart valves, involved who might be considered for antibiotic pro-
with injection drug use, or with chronic der- phylaxis. For example, there are patients in the
matologic conditions. These medically related moderate-risk group with additional medical
cases are not believed to be caused by bacteria problems (eg, immunosuppression) who may
from the mouth, and therefore they are not be at increased risk either for and/or from IE.
targeted for antibiotic prophylaxis prior to There may need to be a discussion with the
dental procedures. patient’s primary care physician or cardiologist,
but it is important beforehand to have a clear
understanding of the current AHA recommen-
dations and of the information needed from the
The AHA Recommendations physician. Otherwise, these recommendations
provide for the vast majority of the situations
The AHA prophylaxis regimen in the mid- to that arise in community dental practice.
late 1950s was aggressive and difficult for den-
tists to accomplish. Since that time, the dose
and the duration of antibiotic prophylaxis has Changes in the current 2007 AHA
been reduced to the current recommendation recommendations
of just one dose 30 to 60 minutes prior to the
dental procedure. More than 60 years later, sur- The 2007 AHA recommendations underwent
veys suggest that the AHA recommendations significant changes from the prior 1997 rec-
are the most recognized of any publication, ommendations. These changes included the
with a 98% awareness among dentists in the following:
United States.4
It should be remembered that these AHA • An increased focus on the existing science
recommendations focus on two major groups in general.
of people at risk: those in the “moderate risk” • The moderate-risk group was dropped from
group and those in the “high risk” group.12 antibiotic coverage, leaving the high-risk
The determination of patients that are in one group, which has only approximately 10%
or the other of these two groups is based on of all people at risk for IE.
a consensus of both the risks from getting IE

332
The AHA Recommendations

Box 16-1  |  Medical conditions and devices reported to put patients at increased risk from a bacteremia
following invasive dental procedures

• Cardiac: • Hereditary hemorrhagic telangiectasia


–– Prosthetic heart valves • Immunosuppression from drugs or disease
–– Congenital defects (some) • Indwelling catheters
–– Previous IE • Nondental implants: deep brain stimulator, breast,
–– Heart transplants (some) penile, and others
–– Native heart valve disease • Prosthetic joints
–– Pacemakers and defibrillators • Shunts: cerebral spinal fluid, renal dialysis, and others
–– Coronary stents • Systemic lupus erythematosus
• Asplenism • Transplants: solid organs, bone marrow, and others
• Autoimmune disease • Type 1 diabetes mellitus
• Debilitated patients • Vascular grafts

• An increased emphasis on the frequency of • The recommendation to consider an antibac-


bacteremia due to routine daily events (eg, terial mouthrinse prior to a dental procedure
tooth brushing) and a stronger suggestion was dropped due to conflicting data.14
that poor oral hygiene and gingival disease • The reference to bleeding as an indicator of
may play a role in the incidence of naturally risk for bacteremia was dropped for lack of
occurring bacteremia and therefore the risk data to show that “visible bleeding during
for community-acquired IE. the dental procedure is a reliable predictor
• Prior AHA recommendations included a of bacteremia.”13
table listing dental procedures that might
put patients at risk for bacteremia and IE. The reasoning behind some of these changes
The 2007 recommendations dropped this is an increasing appreciation for the likelihood
list because of the lack of evidence base and that community-acquired IE from oral bacterial
replaced it with a single sentence to guide species is far more likely to result from frequent
dentists on procedures most likely to cause bacteremia from daily activities such as tooth
a bacteremia: “All dental procedures that brushing than from dental office procedures.
involve manipulation of gingival tissue or the Therefore, the maintenance of optimal oral
periapical region of teeth or perforation of health and hygiene may be far more impor­tant
the oral mucosa.”13 There was concern that as prevention than prophylactic antibiotics.
by eliminating the list of dental procedures AHA recommendations for cardiac con-
the new definition in the 2007 recommen- ditions have been adopted for a variety of
dations would not cover all procedures that noncardiac patient populations as well, partic-
might result in a bacteremia. A comparison ularly in patients with prosthetic joints. Other
of more than 21 million claims for payments examples include patients with a variety of
to dentists in a large provider network sug- shunts, central line catheters, and transplanted
gested that there would be no difference in organs. In fact, in the dental literature there
the dental procedures covered in the 1997 are suggestions or recommendations for over
versus the 2007 recommendations. 25 different patient populations to receive sec-
ondary antibiotic prophylaxis prior to dental
procedures, usually opinion-based8 (Box 16-1).

333
16 Antibiotic Prophylaxis for Patients at Risk for Infective Endocarditis

However, there is no proof of efficacy for any What Do Bacteremia Studies


of these purported indications for antibiotic
prophylaxis. In addition, there are other com- Tell Us?
mon uses for secondary antibiotic prophylaxis
that have little or no support from the scientific There have been many studies of bacteremia
literature. Examples include use of secondary following various dental procedures. Most
antibiotic prophylaxis prior to invasive dental focus on surrogate measures of risk for IE,
procedures to prevent local infection (eg, third most commonly the incidence, duration, na-
molar removal), to resolve failure of local anes- ture (bacterial species), and magnitude of
thesia due to inflammation in the region, for bacteremia. It seems clear that bacteremia
extractions following radiotherapy to the jaws, may arise from any procedure that involves
and for toothache without signs or symptoms gingival tissues adjacent to the teeth. A wide
of infection.15 range of incidence has been reported follow-
ing single- and multiple-tooth extractions,
scaling, endodontic procedures, and other pro-
cedures.14,16–21 However, it is not known if there
Why Is There a Focus on is a clinically significant difference between the
Dental Procedures for People bacteremia caused by various invasive dental
procedures or the degree to which they put pa-
at Risk for IE? tients at risk for IE.
A large randomized study comparing tooth
Distant site infections from oral bacterial spe- brushing with single-tooth extraction reported
cies are rare, but people with specific cardiac that the incidence and duration of bacteremia
conditions are at risk for IE. The gingival sulcus from tooth brushing is not greatly different
and periodontal pocket have a wide variety of from that of an extraction done with antibiotic
both pathogenic and nonpathogenic bacteria prophylaxis7 (Fig 16-3). This raises the question
within them. It has long been assumed that the of the relative risk between naturally occur-
gingival crevicular tissue adjacent to the tooth ring bacteremia from daily activities such as
surface becomes ulcerated in the presence of tooth brushing and invasive procedures in the
inflammation from plaque and calculus. The dental office. Given that individuals have far
scientific evidence for this is weak, but it is gen- more frequent bacteremia from tooth brushing
erally regarded that this greatly increases the (and perhaps other oral hygiene measures and
likelihood that a wide variety of bacteria may chewing food) than they would ever have from
enter the inflamed gingival tissues and capil- dental office procedures, we may need to focus
lary blood vessels around the teeth following more on preventing bacteremia by improving
any manipulation and that these bacterial spe- oral hygiene and eliminating dental disease.
cies may travel to and lodge themselves onto This is especially important because we are
damaged heart valves. Some of the bacterial currently employing antibiotics for only the
species in the dental plaque are of particular approximately 10% of people who are at high
concern with regard to IE, such as some of risk for IE. Improving oral hygiene is likely
the viridans streptococci group (Streptococcus to be an important prevention measure for all
mitis, Streptococcus oralis, Streptococcus mu- people at increased risk of IE (both high risk
tans, and Streptococcus salivarius). Much of and moderate risk) and is the only prevention
what is known about dental procedures and method available for those at moderate risk
oral disease as well as IE comes from bacte- for IE who are not currently recommended for
remia studies. antibiotic prophylaxis.

334
Support For and Against Antibiotic Prophylaxis

60

50
Positive cultures (% ) 40

30

20

10 Extraction +
placebo
0 Extraction +
0 1.5 5 20 40 60 amoxicillin
Tooth
Minutes after extraction or brushing brushing

Fig 16-3  |  Size and duration of bacteremia following single-tooth extraction versus tooth
brushing.7 Note that tooth extraction with a placebo had the largest and longest duration
of bacteremia but that tooth brushing can also have a bacteremia duration of up to 1 hour.

Support For and Against • Antibiotic prophylaxis has been the primary
if not the only method for prevention of IE.
Antibiotic Prophylaxis • Reports that upward of 35% of cases of
community-acquired IE come from oral
The support for antibiotic prophylaxis comes bacterial species.
from a variety of sources. The original source • Viridans group streptococci were highly
could be considered a remnant of the focal susceptible to antibiotics at the time of the
infection theory. In 1955, the intention for original AHA recommendations.
prophylaxis was to reduce the incidence of IE, • Bacteremia with IE-related bacterial species
which was fatal for virtually everyone with the frequently occurs during dental procedures.22
disease in the preantibiotic era. The emergence • There is an increasing number of cardiac and
of antibiotics in the early 1940s, along with noncardiac devices that put people at risk for
early animal studies where prophylaxis was IE, and these people have high morbidity and
shown to be effective, resulted in the opinion mortality from this disease.
that patients might benefit by having antibiotic • It is believed that the antibiotic will kill the
prophylaxis before invasive procedures. This bacteria both in the bloodstream before they
support for prophylaxis also comes from the can attach to a heart valve and after they
following sources: attach to a vegetation.
• A large epidemiologic study in the United
• Over 60 years of AHA recommendations Kingdom reported a benefit from the use of
that advise prophylaxis and long-established antibiotic prophylaxis, and its use may be
practice patterns that are deeply entrenched cost-effective as well.9,23
with the belief that prophylaxis works.

335
16 Antibiotic Prophylaxis for Patients at Risk for Infective Endocarditis

• Data from a study of 3 million prescriptions IE, particularly in those at high risk.9 There
for antibiotic prophylaxis in the United are a variety of other costs associated with
Kingdom focused on single-dose amoxicillin, this use of antibiotics, such as the cost asso-
and the data suggest that amoxicillin is safe ciated with a single dose of amoxicillin for
for this purpose because the drug reaction cardiac patients,8 and they increase greatly
rate is very low.10 However, clindamycin, a if used for the other noncardiac patient
backup drug for amoxicillin, has a higher populations mentioned previously (see Box
incidence of drug reactions than anticipated, 16-1).
and the study reported cases of Clostridium • Much of the confusion surrounding antibi-
difficile and death following administration otic prophylaxis stems from the conflicting
for dental prophylaxis. literature over the years, which is often based
on expert opinion and case reports. For
There has been increasing support in the example, there have been well over 200 case
last two to three decades to reduce the use of reports suggesting that a dental procedure
prophylactic antibiotics in general and for IE caused a case of IE, but none are documented
specifically. This support comes from the fol- well enough to show even a strong associ-
lowing sources: ation.25 Most of the studies that purport to
show a causal relationship have flawed meth-
• There are no well-documented cases of IE odologies, including small sample sizes and
from a dental procedure, although it is likely follow-ups that are too short.
that this does occur. • Finally, there is often an exaggerated tem-
• Bacteremia studies rely on surrogate mea- poral relationship between the time of the
sures of risk for IE. dental procedure and the onset of IE. IE is
• In the absence of a randomized controlled generally thought to occur within 1 to 6
trial, there is no proof of efficacy for the use weeks from the time of the bacteremia, but
of antibiotic prophylaxis, and the epidemi- this time frame can extend out to more than
ology studies often have conflicting findings. 3 months in unusual cases, usually those
• There are many well-documented cases of IE caused by oral viridans group streptococci
where prophylaxis failed to work. and other less pathogenic bacterial species.
• Even if prophylaxis is effective, a huge num-
ber of antibiotic doses may be required to
prevent a small number of cases of IE.
• There is long-standing concern about drug What Do Current Data Tell Us
reactions from antibiotics, especially for About the Prevention of IE by
clindamycin.
• There is an increasing concern across the Prophylactic Antibiotics?
world about the development of drug resis-
tance and more virulent strains of bacterial Since the original 1955 AHA recommenda-
species and a conviction that we must elim- tions, there has been increasing concern that
inate any unnecessary use of antibiotics.10,24 the benefit of antibiotic prophylaxis prior to
• There are concerns about the cost- invasive procedures has been exaggerated, and
effectiveness of antibiotic prophylaxis. The there is a strong need to get definitive data con-
2007 AHA recommendations suggest that cerning the extent to which prophylaxis might
this practice may not be cost-effective, but a prevent some of these cases. There is much lit-
large study in the United Kingdom reported erature on the subject, but there are no studies
that it may be cost-effective for preventing robust enough to provide a definitive answer

336
Do Oral Hygiene and Periodontal Diseases Matter for Patients at Risk for IE?

to this question. Of interest is the opportunity Do Oral Hygiene and


that arose when a guidelines committee in the
United Kingdom decided in 2008 to stop anti- Periodontal Diseases Matter
biotic prophylaxis entirely.26 A study published for Patients at Risk for IE?
in 2011 suggested that although the prescribing
of antibiotics fell significantly soon after 2008,
there was no change in the incidence of IE.27 The connection between oral hygiene and
However, another study by the same group of gingival disease and the risk for IE was first
researchers with a longer follow-up period re- suggested well over a hundred years ago,32
ported a significant increase in IE after 2008, and it is well documented that oral bacte-
suggesting that antibiotic prophylaxis may re- rial species are responsible for many cases
duce the risk of IE following dental procedures, of community-acquired IE. It has long been
especially for the AHA moderate-risk group assumed that dental plaque and calculus and
of patients.23 There has been much discussion the resulting inflammation and periodontal
concerning this study and the importance of pocket formation cause ulceration of the mu-
determining any change in the incidence of IE cosal lining of the gingiva adjacent to the teeth
in the United States since the 2007 AHA rec- (Figs 16-4 and 16-5). This allows bacterial
ommendations. There have been several such species to enter the bloodstream with manip-
studies published since 200728–30; the signifi- ulation of the gingiva during dental procedures
cance of these papers has been covered in a and daily activities (eg, tooth brushing).
recent review of this topic.31 A study of tooth brushing and single-tooth
What is clear is that most of these studies extraction showed an eight-fold increased risk
failed to give strong evidence one way or the of bacteremia from IE-related bacterial spe-
other because they (1) are too small in size and cies.33 This study found that bleeding following
therefore underpowered, (2) have too short a brushing, mean plaque scores, and mean cal-
time period for follow-up after 2007, (3) do not culus scores were highly associated with the
have data on the microbiology of these infec- risk for bacteremia during tooth brushing. This
tions that would implicate the oral cavity, or work also suggests that the incidence of bacter­
(4) do not have antibiotic prescribing data to emia in patients with heavy calculus is similar
compare with a change in the incidence of IE. to that of a single-tooth extraction. However,
There is an ongoing effort to get these data it is important to note that these measures of
and compare them with a likely decrease in oral hygiene and periodontal diseases and the
the use of prophylactic antibiotics since 2007. resultant bacteremia are only surrogate mea-
At least one study suggests that oral disease sures of the risk for IE.
significantly increases the risk for a bacteremia Given that there is ample evidence that den-
both from tooth brushing as well as a dental tal procedures and tooth brushing often cause
extraction,7 and there is a large ongoing study bacteremia and because there is evidence that
underway to determine if poor oral hygiene oral hygiene and disease increase the risk of
and periodontal disease are more common in bacteremia, it is entirely possible that this puts
patients with IE as opposed to people who are some people at greater risk for IE33 (see Fig
simply at risk for IE. In the meantime, there is 16-5). Ongoing investigations are attempting
an increased understanding of the likely greater to determine the strength of the association
risk for community-acquired IE from poor oral between oral hygiene and disease and IE. The
hygiene and periodontal disease. impact of demonstrating a strong association
would do the following:

337
16 Antibiotic Prophylaxis for Patients at Risk for Infective Endocarditis

Fig 16-4 |  Comparison of oral hygiene and the periodontium in healthy (left side) and diseased (right side)
states.7

Poor oral hygiene Gingivitis Advanced periodontal


disease

Ulcerated
sulcus/pocket
Bacteremia
with IE bacterial
species

Evidence-
Infective endocarditis based
May reduce
risk for* Assumed
Poor oral hygiene No gingivitis/
Unknown
and periodontal periodontal
diseases Treatment and prevention diseases

*It is unknown if resolving poor oral hygiene and periodontal disease will reduce the risk for IE.

Fig 16-5 |  Associations between oral hygiene and periodontal disease and
IE.

338
What Can We Expect from Current and Future Research Studies?

• Add greatly to our understanding of risk incidence of IE after stopping prophylaxis


factors for IE. altogether have been discussed at great length
• Provide the opportunity to reduce the among the authorities in this field, and we
incidence and the associated morbidity, mor- await future studies that will either confirm
tality, and cost for community-acquired IE. or revise these findings.23 There are studies
• Give patients the opportunity to prevent this currently underway in the United States that
disastrous infection by maintaining good should help greatly to answer the following
oral hygiene and avoiding periodontal dis- key questions:
ease and decrease the likelihood of IE from
naturally occurring activities such as tooth 1. Has there been an increase, a decrease, or
brushing. no change in the incidence of IE as a result
• Provide policymakers with specific informa- of the 2007 AHA recommendations, when
tion for future recommendations concerning the moderate-risk group was no longer rec-
the importance of oral hygiene and prevent- ommended for prophylaxis? This should
ing gingival disease. help to resolve the fundamental question
• Over time, reduce the need for dental concerning the efficacy of antibiotic pro-
procedures that might require antibiotic phylaxis to prevent IE.
prophylaxis. 2. What is the strength of the association
• Provide a rationale for reimbursement for between indexes of oral hygiene and peri-
medically necessary dental care for patients odontal diseases for cases of IE that are
at risk for IE. caused by oral versus nonoral bacterial
species?
3. Are we using antibiotic prophylaxis for
the most appropriate cardiac patient pop-
What Can We Expect from ulations and for the appropriate dental
Current and Future Research procedures?
4. How well do dentists understand the cur-
Studies? rent AHA recommendations and the lack
of scientific data to support the use of pri-
Although there have been attempts to fund a mary and secondary antibiotic prophylaxis
large study that would give a more definitive in other patient populations?
answer to this long-standing question, there
has never been a prospective randomized con- Studies published in the last decade are
trolled trial of antibiotic prophylaxis. Reasons bringing clarity to some of the other more sig-
for this include (1) logistics and study design is- nificant unanswered questions with regard to
sues that would require over 100,000 high-risk this interface between medicine and dentistry.
patients in a multi-center, multiyear study In the meantime, given the lack of proof of
that might cost in excess of $60 million, and efficacy for antibiotic prophylaxis, guidelines
(2) there are ethical and legal issues, such as committees are charged with sorting out con-
randomizing patients at high risk for IE to a flicts and confusion with the literature and
placebo. Because a randomized controlled tri- creating recommendations on the risks and
al of prophylaxis is highly unlikely, an answer benefits of this practice. Until these studies have
to the question of causality will require one been completed, current data suggest that all
or more large, well-designed epidemiologic patients at risk for IE may benefit by eliminat-
studies. ing oral disease and maintaining good hygiene.
The results of the study done in the United Once answers to these questions are available,
Kingdom that reported an increase in the we will want to know if people with specific

339
16 Antibiotic Prophylaxis for Patients at Risk for Infective Endocarditis

cardiac conditions are at risk from poor oral 4. Lockhart PB, Hanson NB, Ristic H, Menezes AR,
Baddour L. Acceptance among and impact on dental
hygiene and periodontal diseases. If so, does
practitioners and patients of American Heart Association
treatment and prevention of caries and peri- recommendations for antibiotic prophylaxis. J Am Dent
odontal disease decrease the incidence of IE in Assoc 2013;144:1030–1035.
people at risk? 5. Lockhart PB, Brennan MT, Fox PC, Norton HJ, Jernigan
DB, Strausbaugh LJ. Decision-making on the use of
It is also important to understand that there antimicrobial prophylaxis for dental procedures: A survey
are no official guidelines concerning the use of infectious disease consultants and review. Clin Infect
of primary or secondary antibiotic prophy- Dis 2002;34:1621–1626.
laxis in dental practice for any other patient 6. Lockhart PB. Antibiotic prophylaxis for dental proce-
dures: Are we drilling in the wrong direction? Circulation
population with the exception of patients 2012;126:11–12.
with prosthetic joints, a highly controversial 7. Lockhart PB, Brennan MT, Sasser HC, Fox PC, Paster
issue given the lack of association between the BJ, Bahrani-Mougeot FK. Bacteremia associated with
toothbrushing and dental extraction. Circulation 2008;
nature of bacterial species typically found in
117:3118–3125.
infected prosthetic joints by comparison with 8. Lockhart PB, Loven B, Brennan MT, Fox PC. The
the strong association for patients with IE. This evidence base for the efficacy of antibiotic prophylaxis
is an important distinction for several reasons, in dental practice. J Am Dent Assoc 2007;138:458–474.
9. Franklin M, Wailoo A, Dayer MJ, et al. The cost-
including the increasing concern about the
effectiveness of antibiotic prophylaxis for patients at risk
abuse of antibiotics and the global problem of of infective endocarditis. Circulation 2016; 134:
drug-resistant strains of bacteria. 1568–1578.
Following a period of decades during which 10. Thornhill MH, Dayer MJ, Prendergast B, Baddour LM,
Jones S, Lockhart PB. Incidence and nature of adverse
there were relatively minor changes in the gen-
reactions to antibiotics used as endocarditis prophylaxis.
eral thinking about antibiotic prophylaxis for J Antimicrob Chemother 2015;70:2382–2388.
dental procedures, the last 15 years of research 11. Cahill TJ, Baddour LM, Habib G, et al. Challenges in
have brought about significant changes in infective endocarditis. J Am Coll Cardiol 2017; 69:
325–344.
our understanding of the people at risk for 12. Wilson W, Taubert KA, Gewitz M, et al. Prevention of
this disease and those factors that put them infective endocarditis: Guidelines from the American
at increased risk. The increased number of Heart Association: A guideline from the American Heart
well-designed studies should result in better Association Rheumatic Fever, Endocarditis, and Kawa-
saki Disease Committee, Council on Cardiovascular
prevention protocols for cardiac patients and Disease in the Young, and the Council on Clinical Cardi-
other patient populations for whom there has ology, Council on Cardiovascular Surgery and
been well over 100 years of debate. Anesthesia, and the Quality of Care and Outcomes
Research Interdisciplinary Working Group. Circulation
2007;116:1736–1754.
13. Wilson W, Taubert KA, Gewitz M, et al. Prevention of
infective endocarditis: Guidelines from the American
References Heart Association: A guideline from the American Heart
Association Rheumatic Fever, Endocarditis and Kawa-
1. Lockhart PB, Bolger AF, Papapanou PN, et al. Peri- saki Disease Committee, Council on Cardiovascular
odontal disease and atherosclerotic vascular disease: Disease in the Young, and the Council on Clinical Cardi-
Does the evidence support an independent associa- ology, Council on Cardiovascular Surgery and
tion?: A scientific statement from the American Heart Anesthesia, and the Quality of Care and Outcomes
Association. Circulation 2012;125:2520–2544. Research Interdisciplinary Working Group. J Am Dent
2. Jones TD, Baumgartner L, Bellows MT, et al. Prevention Assoc 2007;138:739–760.
of rheumatic fever and bacterial endocarditis through 14. Lockhart PB. An analysis of bacteremias during dental
control of streptococcal infections. Circulation 1955; 11: extractions: A double-blind, placebo-controlled study
317–320. of chlorhexidine. Arch Intern Med 1996;156:513–520.
3. Dayer MJ, Chambers JB, Prendergast B, Sandoe JA, 15. Runyon MS, Brennan MT, Batts JJ, et al. Efficacy of
Thornhill MH. NICE guidance on antibiotic prophylaxis penicillin for dental pain without overt infection. Acad
to prevent infective endocarditis: A survey of clinicians’ Emerg Med 2004;11:1268–1271.
attitudes. QJM 2013;106:237–243.

340
References

16. Lockhart PB, Brennan MT, Kent ML, Norton HJ, Weinrib 25. Durack DT, Kaplan EL, Bisno AL. Apparent failures of
DA. Impact of amoxicillin prophylaxis on the incidence, endocarditis prophylaxis. Analysis of 52 cases submitted
nature, and duration of bacteremia in children after intu- to a national registry. JAMA 1983;250:2318–2322.
bation and dental procedures. Circulation 2004; 109: 26. Centre for Clinical Practice at NICE. Prophylaxis Against
2878–2884. Infective Endocarditis: Antimicrobial Prophylaxis Against
17. Baltch AL, Schaffer C, Hammer MC, et al. Bacteremia Infective Endocarditis in Adults and Children Undergoing
following dental cleaning in patients with and without Interventional Procedures. London: National Institute
penicillin prophylaxis. Am Heart J 1982; 104: for Health and Clinical Excellence, 2008.
1335–1339. 27. Thornhill MH, Dayer MJ, Forde JM, et al. Impact of the
18. Debelian GJ, Olsen I, Tronstad L. Profiling of Propioni- NICE guideline recommending cessation of antibiotic
bacterium acnes recovered from root canal and blood prophylaxis for prevention of infective endocarditis:
during and after endodontic treatment. Endod Dent Before and after study. BMJ 2011;342:d2392.
Traumatol 1992;8:248–254. 28. Pant S, Patel NJ, Deshmukh A, et al. Trends in infective
19. Giglio JA, Rowland RW, Dalton HP, Laskin DM. Suture endocarditis incidence, microbiology, and valve replace-
removal-induced bacteremia: A possible endocarditis ment in the United States from 2000 to 2011. J Am Coll
risk. J Am Dent Assoc 1992;123:65–70. Cardiol 2015;65:2070–2076.
20. Erverdi N, Biren S, Kadir T, Acar A. Investigation of 29. Mackie AS, Liu W, Savu A, Marelli AJ, Kaul P. Infective
bacteremia following orthodontic debanding. Angle endocarditis hospitalizations before and after the 2007
Orthod 2000;70:11–14. American Heart Association Prophylaxis Guidelines. Can
21. Heimdahl A, Hall G, Hedberg M, et al. Detection and J Cardiol 2016;32:942–948.
quantitation by lysis-filtration of bacteremia after different 30. van den Brink FS, Swaans MJ, Hoogendijk MG, et al.
oral surgical procedures. J Clin Microbiol 1990; 28: Increased incidence of infective endocarditis after the
2205–2209. 2009 European Society of Cardiology guideline update:
22. Lockhart PB, Brennan MT, Sasser H, Noll J, Coleman A nationwide study in the Netherlands. Eur Heart J Qual
S, Ashar J, et al. Tooth extraction and tooth brushing Care Clin Outcomes 2017;3:141–147.
both produce bacteremia of endocarditis-related patho- 31. Thornhill MH, Dayer M, Lockhart PB, Prendergast B.
gens. J Am Coll Cardiol 2007;49(9 suppl A):301A. Antibiotic prophylaxis of infective endocarditis. Curr
23. Dayer MJ, Jones S, Prendergast B, Baddour LM, Lock- Infect Dis Rep 2017;19:9.
hart PB, Thornhill MH. Incidence of infective endocarditis 32. Horder TJ. Infective endocarditis: With an analysis of
in England, 2000–13: A secular trend, interrupted 150 cases and with special reference to the chronic
time-series analysis. Lancet 2015;385:1219–1228. form of the disease. QJM 1909;2:289–324.
24. Centers for Disease Control and Prevention. Antibiotic 33. Lockhart PB, Brennan MT, Thornhill M, et al. Poor oral
Resistance Threats in the United States. Atlanta: U.S. hygiene as a risk factor for infective endocarditis-
Department of Health and Human Services, 2013. related bacteremia. J Am Dent Assoc 2009; 140:
1238–1244.

341
CHAPTER 17

Oral-Systemic Connection:
The Salivaomics and
Exosomics Connection
Taichiro Nonaka, dds, phd
Karolina Elżbieta Kaczor-Urbanowicz, dmd, phd, msc, msl
David T.W. Wong, dmd, dmsc

The oral-systemic connection is further creden- minor glands produce a liter of whole saliva dai-
tialed in the development and genesis of salivary ly per individual (see Fig 17-1a). The source of
biomarkers for systemic diseases (Fig 17-1). The the saliva fluid and its omics constituents (pro-
three major pairs of salivary glands (parotid, teome, genome, and transcriptome) are from
submandibular, and sublingual) together with systemic circulation where passive, active, and

Capillaries

  1   2     3    4

    6
Blood    7

   8

   9

Capillaries

Trigeminal nerve (V) Na+

Facial nerve (VII)     5 K+

Parotid gland

Blood
Sublingual gland
a Submandibular gland b Saliva

Fig 17-1  |  (a) Major salivary glands: parotid, submandibular, and sublingual. (b) Transport of circulatory mole-
cules and fluid into salivary glands: (1) ultrafiltration; (2) active transport or passive diffusion across the cell mem-
brane; (3) simple diffusion through cell membrane pores; (4) transepithelial movement of water along a sodium
chloride (NaCl) gradient via channel proteins; (5) creation of hypotonic salivary solution via ductal Na+ reabsorp-
tion; (6) acinar cell membrane; (7) cell membrane pore; (8) intercellular space; (9) acinar cell. K+, potassium ion.

342
Salivaomics

secretory mechanisms are in place to transfer the molecular constituency of oral fluids.
fluid and omics targets from the capillary bed This suggests that circulating biomarkers of
(see Fig 17-1b). This systemic-oral passaging disease absorbed by the salivary glands can
of omics constituents embodies a compelling alter the biochemical composition of salivary
rationale for systemic-oral communication. Yet secretions. Consequently, oral fluids contain
the mechanistic underpinnings and scientific molecular information capable of communi-
data are only beginning to emerge. This chapter cating an individual’s current state of health.
focuses on the systemic-oral connection from This constituted the theoretical rationale for
a biomarkers development perspective using oral-systemic communication and connection
basic, translational, and clinical studies to un- for biomarker development.
derpin the mechanistic rationale.

Salivaomics
Saliva
Salivaomics was coined in 2008 to reflect the
Human saliva is a clear, slightly acidic (pH rapid development of knowledge about the
6.0 to 7.0) heterogeneous biofluid composed “omics” constituents in saliva.5 The terms
of water (99%), proteins (0.3%), and inorganic salivary proteome, extracellular RNA (exR-
substances (0.2%).1 On average, individual sal- NA), microRNA (miRNA), metabolome, and
ivation can range from 0.3 to 0.7 mL of saliva microbiome have since entered the scientific
per minute,2 producing approximately one liter lexicon.5 The metabolome is the complete set
daily. Saliva is multifunctional, not only serving of small molecular metabolites found within
to facilitate digestion, swallowing, tasting, and a biologic sample (carbohydrate, lipid, amino
tissue lubrication but also acting as a protective acid, nucleic acid, hormones, and other signal-
barrier against pathogens. Saliva is generated ing molecules). Translationally, these are the
within the salivary glands by acinus cells, col- diagnostic alphabets of saliva, and their discov-
lected in small ducts, and subsequently released ery poised saliva for translational and clinical
into the oral cavity.3 There are three major and applications, including personalized medicine
numerous minor saliva-producing glands lo- and dentistry. These salivaomics constituents,
cated in and around the mouth and throat their discovery, annotations, and documenta-
(see Fig 17-1). Each gland is entirely innervat- tions are assembled at the Salivary Proteome
ed autonomically, subject to parasympathetic Knowledge Base (Fig 17-2).
and sympathetic stimulation, and exocrine in The defining of salivaomics constituents pre-
function. The three major glands—the parotid, sented tools that can permit, for the first time,
submandibular, and sublingual glands—con- development of salivary biomarkers de novo
tribute approximately 90% of total saliva, for the detection of oral and systemic diseases.
while the minor glands—the labial, buccal, lin- The outcomes of these studies represent trans-
gual, and palatal glands—supply the remainder. lational evidence of oral-systemic connection.
Each salivary gland is highly permeable Key examples are highlighted below.
and enveloped by capillaries (see Fig 17-1),
allowing free exchange of blood-based mol-
ecules into saliva-producing acinus cells. Salivary proteomics
Blood-derived molecules can enter salivary Comprehensive analysis of the salivary
tissues via trans­ cellular (eg, passive and proteome is critical for appreciating its full diag-
active transport) or paracellular (eg, extra- nostic potential. A consortium of three research
cellular ultrafiltration) routes4 that influence groups from the Scripps Research Institute; the

343
17 Oral-Systemic Connection: The Salivaomics and Exosomics Connection

Salivaomics Knowledge Base


Scientific
Saliva ontology
foundations

Proteome

Transcriptome
Point-of-care
technology

microRNA

Metabolome

Saliva diagnostic Genome


atlas

Fig 17-2  |  Salivaomics Knowledge Base. An assembly of the omics constituent databases, publications, and
point-of-care technologies (www.skb.ucla.edu).

University of California, San Francisco; and the salivary proteins appear to be more suscepti-
University of California, Los Angeles compiled ble to degradation.10 Esser et al11 reported that
a comprehensive catalog of the salivary pro- salivary protein can be degraded rapidly even
teome of healthy individuals, identifying 1,166 during saliva collection and handling, which
proteins in parotid and submandibular/sublin- may hamper the downstream experiments and
gual gland ductal saliva.6 The data set from this application. We have established the methods
study has been deposited into the Saliva Pro- to stabilize the salivary protein using protease
teome Knowledge Base for public access.7 inhibitors, enabling saliva samples to be stored
Bandhakavi et al made a significant contri- up to 2 weeks without significant degradation.12
bution to expand our understanding of the The first attempt at cancer detection using
salivary proteome by using three-dimensional salivary protein was made by Hoerman et al
peptide fractionation and generated the larg- more than 50 years ago, in which they showed
est whole saliva proteome data set, including that patients with prostate cancer exhibited
2,340 proteins that are involved in a variety elevated acid phosphatase enzymatic activity
of biologic functions in the oral cavity.8 Com- in parotid saliva.13 There has been a marked
parative analysis of human saliva and plasma advancement in protein analytical technologies
proteome showed that distributions of salivary combined with bioinformatics, creating a new
proteins are enhanced in two gene ontology revolution in salivary proteomics. Currently,
categories (metabolic and catabolic processes) high-throughput mass spectrometry is the
compared with plasma, suggesting that saliva core technology for salivary protein identifi-
may be advantageous over plasma, especially cation, and salivary proteins were identified
for less abundant proteins involved in these as potential biomarkers for the detection and
biologic processes.9 Unlike the serum protein, monitoring of cancer (Table 17-1).14–29

344
Salivaomics

TABLE 17-1  |  Salivary protein biomarkers for cancers

Cancer Sample Salivary protein biomarker Ref


Breast cancer Whole saliva EGF 14
C-erbB-2 15
CA15-3, C-erbB-2 15
VEGF, EGF, CEA 16
CA6 17
LRP 18

Squamous cell Whole saliva A1BG, CFB 19


carcinoma M2BP, MRP14, CD59, CAT, PFN 20
FGB, S100, TF, IGHG, CFL1 21
ADA 22
IL-8, M2BP, IL-1β 23
Salivary extracellular vesicles A2M, HPa, MUC5B, LGALS3BP, IGHA1, PIP, PKM1/M2, GAPDH 24

Gastric cancer Whole saliva 1472.78 Da, 2936.49 Da, 6556.81 Da, 7081.17 Da 25
CSTB, TPI1, DMBT1, CALML3, IGH, IL-1RA 26

Lung cancer Whole saliva HP, AZGP1, CALPR 27


Salivary extracellular vesicles Annexin A1, A2, A3, A5, A6, A11; NPRL2, CEACAM1, 28
HIST1H4A, MUC1, PROM1, TNFαIP3

Ovarian cancer Whole saliva CA125 29


EGF, epidermal growth factor; VEGF, vascular endothelial growth factor; CEA, carcinoembryonic antigen; LRP, lung resistance protein; CFB, complement
factor B; FGB, beta fibrin; S100, S100 calcium-binding protein; TF, transferrin; IGHG, immunoglobulin heavy chain constant region gamma; CFL1, cofilin-1;
ADA, adenosine deaminase activity; IL-8, interleukin-8; IL-1β, interleukin-1β; A2M, α 2-macroglobulin; HPa, haptoglobin α chain; MUC5B, mucin-5B;
LGALS3BP, galectin-3-binding protein; IGHA1, immunoglobulin α1 chain C region; PIP, prolactin-inducible protein; PKM1/M2, pyruvate kinase isozymes
M1/M2; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; NPRL2, nitrogen permease regulator 2-like protein; CEACAM1, carcinoembryonic antigen-
related cell adhesion model 1; HIST1H4A, histone H4; MUC1, mucin 1; PROM1, prominin-1; TNFαIP3, tumor necrosis factor α –induced protein 3.

Breast cancer under the curve [AUC] = 0.84). Furthermore,


we identified CA6 and psoriasin as potential
Breast cancer is a well-studied cancer by salivary salivary biomarkers for breast cancer and per-
proteomics. Significantly elevated epidermal formed validation using an independent cohort.
growth factor (EGF),14 C-erbB-2, and CA15-315 The results showed a significant difference in
were identified in saliva of breast cancer pa- salivary CA6 between breast cancer patients and
tients compared to noncancer groups. Positive healthy controls (P = .0017).17 Recently, Wood
correlation was found between serum and sa- and Streckfus analyzed the concentrations of
liva CA15-3 level, supporting the potential of lung resistance protein (LRP) in saliva of Stage I
saliva as a diagnostic body fluid for breast can- breast cancer patients.18 The results showed that
cer. We analyzed vascular endothelial growth the saliva of the patients had significantly higher
factor (VEGF), EGF, and carcinoembryonic an- levels of LRP compared to healthy controls, sug-
tigen (CEA) in saliva from breast cancer patients gesting it is a novel biomarker for breast cancer.
to evaluate the predictive power of each protein
individually and in combination.16 The logistic
regression model revealed that the best combi- Squamous cell carcinoma
nation was salivary VEGF and EGF, with 83% Our prior salivary proteomic study discov-
sensitivity and 74% specificity, respectively (area ered five candidate markers (M2BP, MRP14,

345
17 Oral-Systemic Connection: The Salivaomics and Exosomics Connection

CD59, catalase, and profilin) associated with is suggested to improve the sensitivity for the
oral squamous cell carcinoma (OSCC).20 A lo- diagnosis of gastric cancer.30 Despite enormous
gistic regression model including five markers effort in serum biomarker discovery, there are
achieved 90% sensitivity and 83% specificity only a few studies related to salivary protein
with a cross-validation prediction accuracy rate biomarkers for detection of gastric cancer. Wu
of 85% (AUC = 0.93). Furthermore, by using et al identified four salivary proteins (1472.78
two additional independent cohorts, we have Da, 2936.49 Da, 6556.81 Da, and 7081.17 Da)
validated the previously identified three salivary by mass spectrometry and found that these pro-
protein markers for oral cancer (inter­leukin-8 teins were significantly different between gastric
[IL-8], M2BP, and IL-1β) and found IL-8 and cancer and normal groups.25 Recently, we iden-
M2BP to be significantly different (P < .02) tified and quantified 519 proteins in the saliva
between cancer and control groups.23 Ohshiro of gastric cancer patients using mass spectrom-
et al analyzed whole saliva from three head etry with tandem mass tag, among which 48
and neck SCC (HNSCC) patients using liquid showed a significant differential expression pro-
chromatography–mass spectrometry (LC-MS/ file between cancer patients and controls.26 Five
MS).19 Among 34 proteins detected in sali- proteins (CSTB, TPI1, DMBT1, CALML3, IGH,
va from NHSCC patients, α1-B glycoprotein and IL-1RA) were selected for initial verifica-
(A1BG) and complement factor B (CFB) were tion by enzyme-linked immunosorbent assay
identified as unique proteins in cancer. Dow- (ELISA) and found to be downregulated in the
ling et al analyzed whole saliva samples from saliva of gastric cancer patients. The logistic re-
HNSCC patients using two-dimensional dif- gression model using three biomarkers (CSTB,
ference gel electrophoresis (DIGE) analysis TPI1, and DMBT1) in the prevalidation sample
and subsequent mass spectrometry.21 The re- set resulted in 85% sensitivity and 80% specific-
sult identified five proteins: Beta fibrin (FGB), ity (AUC = 0.93). These findings provided the
S100 calcium-binding protein (S100), transfer- proof of concept of salivary protein biomarkers
rin (TF), immunoglobulin heavy chain constant for the noninvasive detection of gastric cancer.
region gamma (IGHG), and cofilin-1 (CFL1)
were increased in the saliva from HNSCC pa-
tients compared to the control group. Rai et al Lung cancer
assessed adenosine deaminase activity (ADA) We investigated salivary proteomic biomark-
in saliva of tongue cancer and found signifi- ers in lung cancer patients and found that
cant differences between cancer and control the levels of three proteins (HP, AZGP1, and
groups, suggesting that ADA levels might be CALPR) were significantly higher in lung can-
useful as a diagnostic tool for early detection cer patients than in healthy controls. A logistic
of tongue cancer.22 regression model including the three proteins
achieved 88.5% sensitivity and 92.3% speci-
ficity (AUC = 0.90), suggesting that salivary
Gastric cancer protein biomarkers have the potential for de-
Much effort has been made to develop serum tection of lung cancer.27
biomarkers for gastric cancer, and over 2,000
studies were reported in the past decade. The
most commonly used diagnostic biomarkers Ovarian cancer
are CEA and CA19-9. However, these bio- Chen et al analyzed CA125 in saliva samples
markers are not widely acknowledged in early from ovarian cancer patients and showed sig-
detection of cancer because of their limited sen- nificant differences in salivary CA125 levels
sitivity (< 21%), and the combination use of between the malignant and benign groups,
CEA, CA19-9, CA125, and α-fetoprotein (AFP) with 81.3% sensitivity.29 There was a linear

346
Salivaomics

correlation between salivary and serum CA125 suggested that salivary mRNA can be a poten-
levels, suggesting the promising utility of saliva tial biomarker for oral cancer detection and
for monitoring ovarian cancer. Using a sali- opened a new avenue for salivary biomarker
va exRNA approach, we have discovered and discovery for other human cancers.
validated that five salivary exRNA biomarkers We found four salivary transcriptomic
(AGPAT1, B2M, BASP2, IER3, and IL-1β) can biomarkers (KRAS, MBD3L2, ACRV1, and
significantly discriminate ovarian cancer pa- DPM1) to be associated with resectable pan-
tients (n = 21) from healthy controls (n = 35), creatic cancer.39 A logistic regression model
yielding a receiver operating characteristic plot demonstrated that the combination of four
AUC value of 0.909, with 85.7% sensitivity markers could differentiate cancer patients
and 91.4% specificity.31 from control subjects with 90% sensitivity and
95% specificity (AUC = 0.971). The utility of
salivary mRNAs was further demonstrated for
Salivary transcriptomics the detection of breast cancer,17 ovarian can-
A transcriptome is the complete set of RNA cer,31 and lung cancer.40 We profiled salivary
molecules, including mRNA, miRNA, piwi- transcriptomes and proteomes of breast cancer
interacting RNA (piRNA), and other small patients.17 Preclinical validation demonstrated
RNAs such as transfer RNA (tRNA) and ribo- that the combination of eight transcriptomic
somal RNA (rRNA). Salivary transcriptomics biomarkers (CSTA, TPT1, IGF2BPI, GRM1,
has emerged as a powerful approach for ex- GRIK1, H6PD, MDM4, S100A8) and one
ploring salivary biomarkers.32 Although the proteomic biomarker (CA6) could discriminate
genome is the same in all host cells, different between breast cancer patients and controls
cells and body fluids show different patterns with 83% sensitivity and 97% specificity. Tran-
of RNA composition; therefore, transcriptom- scriptomic analysis of saliva in ovarian cancer
ic analysis can provide valuable information identified 4 upregulated and 16 downregulated
about disease states. In 2004, the human sali- genes.31 The logistic regression model demon-
vary transcriptome was first discovered in our strated that the combination of five biomarkers
laboratory using expression microarrays.33 We (AGPAT1, B2M, BASP1, IER3, IL-1β) showed
characterized the salivary transcriptome as a 85.7% sensitivity and 91.4% specificity. Fur-
highly fragmented coding and noncoding gene thermore, we found significant differences
transcript derived from host and oral microbio- in salivary mRNA between patients with
ta.34,35 We also developed a method that allows lung cancer and healthy controls.40 A logis-
stable and direct saliva transcriptomic analysis tic regression model using the combination
without further processing.36 of five markers (CCNI, EGFR, FGF19, FRS2,
We analyzed the salivary transcriptome in and GREB1) could differentiate lung cancer
cancer patients in attempts to improve early patients from controls with 93.75% sensitivity
diagnosis. Our prior transcriptomic studies dis- and 82.81% specificity (AUC = 0.925). Hence,
covered seven mRNA biomarkers (IL-8, SAT, salivary exRNA biomarkers could be useful for
IL-1β, OAZ1, H3F3A, DUSP, S100P) in the screening for cancers.
saliva of OSCC patients.37,38 A logistic regres- Detection of miRNA from body fluids is
sion model including four markers (IL-8, SAT, becoming increasingly important in liquid
IL-1β, and OAZ1) achieved a prediction accu- biopsy. Many studies have shown that miRNAs
racy rate of 81%. We further validated these are frequently dysregulated in cancer.41 There-
oral cancer biomarkers using two additional fore, the changes in miRNA expression
independent cohorts and demonstrated that the patterns often correlate with disease and can
AUC for prediction of OSCC ranges from 0.74 be promising diagnostic biomarkers for cancer.
to 0.86 across the five cohorts.23 These findings Importantly, salivary miRNAs are more stable

347
17 Oral-Systemic Connection: The Salivaomics and Exosomics Connection

and discriminatory than salivary mRNA; thus, standardized exRNA protocols, and other use-
circulating miRNAs are attractive potential ful tools and technologies generated by funded
biomarkers. In 2009, we discovered miRNA projects. Investigators from all five initiatives
in saliva and characterized it as a possible will form the ERCC with goals to discover
biomarker for oral cancer detection.42 Two fundamental biologic principles about the
miRNAs (miR-125a and miR-200a) were sig- mechanisms of exRNA biogenesis, to develop
nificantly reduced in the saliva of oral cancer an exRNA catalog in normal human body
patients compared to the control group. Fur- fluids, and to investigate the use of exRNAs
thermore, in the validation study for salivary as therapeutic molecules or biomarkers of
gland tumors, we demonstrated the combina- disease. The NIH Common Fund ERCC initia-
tion of four miRNAs with 95% specificity and tive included saliva extracellular RNA in both
69% sensitivity (AUC = 0.90).43 biomarker development and reference catalog
More recently, we reported piRNA in development, firmly credentialing the salivary
saliva as an emerging potential biomarker exRNA scientifically and translationally. Figure
for cancers.44 We conducted high-throughput 17-3 is an insert from “exRNA Atlas” depict-
sequencing of piRNA in human saliva from ing contributions of RNA-sequencing data sets
healthy individuals, showing comparable from various bodily fluids. Saliva’s contribu-
expression patterns and levels compared to tion stands at 248 small RNA libraries (http://
that in other body fluids. These findings opened exrna-atlas.org).
doors for further investigation of salivary
piRNA as a cancer biomarker.
It should be mentioned that while RNA has
been demonstrated in saliva since 2004, its Saliva-Exosomics
presence and authenticity has been challenged.
A defining moment in the field of exRNA came
Discovery of salivary exosomes
in 2012 when the NIH Common Fund funded Extracellular vesicles (EVs) are classified into
“The NIH Extracellular RNA Communication three subgroups (exosomes, microvesicles, and
Consortium (ERCC)” to develop a commu- apoptotic bodies) based on their size and bio-
nity of investigators to address the critical genetic pathways.45 Exosomes are intraluminal
issues and to generate fundamental scientific vesicles (ILVs) that are formed within multive-
discoveries and innovative tools and technol- sicular bodies (MVBs) and released upon fusion
ogies in five key initiatives: (1) defining the of the MVBs with the cell membrane. Over 30
fundamental principles of exRNA biogenesis, years ago, two independent groups observed
distribution, uptake, and function as well as the that MVBs in reticulocytes released small ves-
development of the molecular tools, technol- icles into the extracellular space.46 Then, the
ogies, and imaging modalities to enable these term exosomes was coined in 1987 by John-
studies; (2) generating a reference catalog of stone et al to describe that vesicles shed from
exRNAs present in the body fluids of normal cultured cells retained enzymatic activity as a
healthy individuals that would facilitate disease remnant from the parent cells.47 It was later
diagnosis and therapeutic interventions; (3) determined that exosomes are EVs of endoso-
identifying exRNA biomarkers of disease; (4) mal origin that are not degraded by lysosomes,
demonstrating the clinical utility of exRNAs noting that exosome secretion is a way to ex-
as therapeutic agents and developing the scal- crete unnecessary products from the cells.48
able technologies required for these studies; With the discovery that exosomes contain
and (5) developing the Data Management and RNA, exosomes acquired substantial interest
Resource Repository (DMRR), a community as mediators of cell-to-cell communication.48
resource to provide access to exRNA data, Salivary exosomes were first described in 2008

348
Saliva-Exosomics

Fig 17-3  |  exRNA Atlas.


Biofluid
Amniotic fluid (10)
Urine (293)
Bile (17)

Sputum (15) Bronchoalveolar lavage


fluid (10)

Cerebrospinal fluid
Serum (376) (615)

Seminal fluid (8) Conditioned media (22)

Ovarian follicle fluid (10)


Saliva (256)

Plasma (4,118)

when Ogawa et al found that 30- to 130-nm of novel biomarkers in cancer. We coined
vesicles were present in human whole saliva.49 the term saliva-exosomics to describe the
The stability of exosomes in the circulation and next-generation salivaomics that studies sal-
body fluids has made exosomes attractive as ivary exosomes through the application and
potential biomarkers. Typically, exosomes are integration of advanced “-omics” technologies
defined as vesicles ranging from 30 to 100 nm to better delineate their specific functions and
in size and 1.13 to 1.19 g/mL in density and for use as a source of noninvasive biomarkers
are isolated through density gradient or sucrose for disease diagnosis.52
cushion by ultracentrifugation at 100,000 g.
However, the term exosome is now often used
in a less restrictive manner than Johnstone’s Structure of salivary exosomes
original definition.50 For the nomenclature Exosome characterization typically includes
and definition of extracellular vesicles, see the morphologic analysis. Due to their nanoscale
websites of the International Society for Ex- size, morphologic analysis of exosomes had
tracellular Vesicles (http://www.isev.org), the solely been limited to electron microsco-
American Society for Exosomes and Microves­ py (EM).53 While EM has been a standard
icles (http://www.asemv.org), and Vesiclepedia technique for exosome characterization, this
(http://microvesicles.org). technique may not provide a representative
Human saliva is an ideal fluid with distinct view of the exosomes due to the harsh sample
advantages for oral cancer detection.23 The processing such as fixation. Indeed, transmis-
isolation of exosomes from saliva has been sion EM showed a cup-shaped appearance,
optimized, and the use of this small but highly while cryo-EM showed the round shape of
informative fraction may reduce the overall the exosomes.54 We first applied atomic force
complexity of saliva as a result of contribu- microscopy (AFM) and field emission scan-
tion from local and systemic sources.51 For ning electron microscopy (FESEM) to assess
these reasons, the study of vesicles secreted by the native salivary exosome structure and sub-
cancer cells via exocytosis into saliva will be structural organization unresolvable in EM.55,56
a valuable clinical approach for the detection We found the distinct substructure of single

349
17 Oral-Systemic Connection: The Salivaomics and Exosomics Connection

isolated 70- to 100-nm human salivary exo- Contents of salivary exosomes


somes in the form of trilobed structures and
demonstrated their reversible elastic nano- In general, exosomes carry a unique cargo of
mechanical properties. High-resolution AFM proteins and nucleic acids that can be relative-
images correlated well with the exosome struc- ly distinct from those of the cell of origin. The
tures obtained from FESEM, where low-force most commonly detected proteins are mem-
imaging resulted in round-shaped exosomes, brane transport and fusion proteins such as
suggesting exosomes have spherical morphol- tetraspanins (eg, CD63, CD9, and CD81), heat
ogy unless outside force is exerted on them. shock proteins (eg, Hsp70 and Hsp90), Rab
In addition, AFM phase-contrast images of GTPases, major histocompatibility complexes
salivary exosomes indicated a heterogeneous (MHC class I and II), and endosomal proteins
surface, which was attributable to the presence (eg, Alix and Tsg101) that are involved in the
of proteins in the highly dense lipid membrane, biogenesis of exosomes from MVBs.60 Other
consistent with previous proteomic analysis of proteins found on exosomes include signaling,
salivary exosomes.57 cytoskeletal, metabolic, and carrier proteins. Of
Salivary exosomes are naturally occurring note, MHC class I is ubiquitously expressed on
bioparticles, parts of which are secreted from all exosomes, while MHC class II is confined
the surfaces of oral epithelial cells into saliva. to exosomes derived from antigen-presenting
Because exosomes released by normal and cells, including dendritic cells, macrophages,
tumor cells have been suggested to differ in and B cells.61 Proteomic studies on mammali-
both functional and structural properties,58 oral an EVs have yielded extensive catalogs of the
cancer–derived exosomes in saliva have great proteins found in various types of EVs isolated
potential as biomarkers for cancer detection. from cells, tissue, or body fluids. The databas-
To elucidate the structural differences between es are publicly available at Vesiclepedia45 and
the salivary exosomes derived from healthy ExoCarta.62 Both databases include data on
subjects and oral cancer patients, Sharma et proteins, nucleic acids, lipids, and the constit-
al investigated morphologic characteristics at uents isolated from salivary exosomes.
single-vesicle level using high-resolution AFM.59 The first comprehensive proteomic analysis
AFM imaging displayed irregular morphologies of human salivary exosomes was performed by
and higher intervesicular aggregation in cancer Gonzalez-Begne et al using a multidimensional
exosomes than normal exosomes. Quantita- protein identification technology (MudPIT).57
tive analysis revealed that the size and CD63 The analysis identified 491 proteins in the exo-
surface density were significantly increased in some fraction of human parotid saliva, and
cancer exosomes (98.3 ± 4.6 nm) compared to Gene Ontology analysis found that cytosolic
normal exosomes (67.4 ± 2.9 nm) (P < .05). proteins comprise the largest category of the
The structural and morphologic aberrations proteins. Comparison between the parotid
in the exosomes indicated that these exosomes salivary exosomes (491 proteins) and the
are at least in part cancer-derived products that previously identified global parotid saliva pro-
were directly shed into saliva. Interestingly, teome (914 proteins6) showed a 23% overlap
MVBs were identified in oral cancer salivary between the parotid salivary exosomes and the
exosome fractions. These multivesicular struc- parotid saliva, whereas 20% were unique to
tures revealed the presence of ruptures and the parotid exosomes and 57% were unique
elongated nanofilaments around the lumen of to the parotid saliva. Ogawa et al revealed the
these MVBs, suggesting these to be the sites presence of two different types of vesicles (exo-
for the release of the exosomes as well as the somes I and II) with different mean diameter
filamentous extension of nucleic acids. (I: 83.5 nm, II: 40.5 nm) and protein constit-
uents.63 A total of 101 and 154 proteins were

350
Saliva-Exosomics

identified in exosomes I and II, respectively, sequencing (RNA-Seq) using human cell-free
and 68 proteins including common markers whole saliva and found that the most abun-
(CD63, Alix, Tsg101 and Hsp70) were found dant types of small RNAs were piRNA (7.5%),
overlapped between the two groups. Approx- miRNA (6.0%), and snoRNA (0.02%), con-
imately 40% of the proteins identified were sistent with prior study. Exosomes offer
extracellular (eg, immunoglobulin chain) or protection from RNase, thereby inhibiting
secretory proteins (eg, serum albumin), indi- RNAs from degradation and allowing them
cating that saliva contains vesicles originating to be taken up by the cells with subsequent
from circulating lymphocytes and intravascular putative effects on gene expression in the target
fluid. The presence of DPP IV (CD26) in the cells.66 Indeed, prior studies have shown that
vesicles and its enzymatic activity were also salivary exosomal RNAs are stable and can
demonstrated by showing DPP IV–dependent be taken up and translated by recipient cells,
degradation of substrates (substance P and indicating that these RNAs are biologically
GIP), suggesting that the vesicles were biolog- functional. Further saliva-exosomics studies
ically active. will provide important insight regarding the
We performed the proteomic profiling of mechanisms that control the epithelial cell
salivary microvesicles (MVs) with an average homeostasis in the oral cavity.
diameter of 100 to 1,000 nm to investigate a
different type of EVs in saliva.64 LC-MS/MS
combined with gel electrophoresis identified Salivary exosomes as biomarkers
63 proteins in salivary MVs. Among these, 35 for cancer
proteins were exclusively identified in MVs by
comparing parotid exosomes proteome, sug- A breakthrough in the field of EVs was made
gesting that salivary MVs contained their own in 2008 when it was discovered that glioblas-
unique proteins. The most striking finding was toma MVs carry cancer-specific mutant mRNA
that saliva triggers factor VII–mediated coagu- (EGFRvIII).67 Strikingly, the tumor-specific
lation of human plasma. Berckmans et al tested EGFRvIII was detected in the serum MVs
the ability of saliva to induce clot formation from glioblastoma patients, suggesting
of autologous plasma and found that sali- that tumor-derived MVs may serve as cancer
vary exosomes shortened the clotting time of biomarkers. Since this discovery, cancer-derived
exosomes-depleted plasma. Western blot anal- exosomes have been recognized as an im-
ysis and transmission EM with immunogold portant diagnostic tool. A series of “-omics”
labeling identified tissue factor, the initiator of approaches has revealed that exosomal
coagulation activation in salivary exosomes. miRNA, DNA, and protein signatures may
Moreover, the salivary exosomes-induced also serve as biomarkers to aid diagnosis. Se-
shortening of the clotting time was completely rum exosomal miR-21 and miR-141 levels
abolished in the presence of antifactor VII. are significantly increased in the patients of
These results indicated that saliva facilitates esophageal SCC and prostate cancer, respec-
hemostasis as one of the first steps in the pro- tively.68 Mutated KRAS and p53 DNA were
cess of wound healing. detected by whole genome sequencing in the se-
Salivary exosomes are also known to contain rum exosomes of pancreatic cancer patients,69
mRNA55 and small RNA65 including miRNA,65 and the membrane-anchored exosomal pro-
piRNA, small nucleolar RNA (snoRNA), and tein Glypican-1 (GPC-1) was identified as a
other small RNAs (rRNA and tRNA).44 Inter- promising biomarker of pancreatic cancer.70
estingly, piRNA appears to be found at higher These findings indicate that serum exosomes
abundance in exosomes compared to whole in cancer patients have great potential for can-
saliva.44 We conducted high-throughput RNA cer diagnosis.

351
17 Oral-Systemic Connection: The Salivaomics and Exosomics Connection

PBS Tumor (WT) Tumor (exosome secretion inhibited)

Control group (healthy) Tumor group #1 Tumor group #2

Control group Tumor group #1

Saliva Salivary gland Serum Saliva Salivary gland Serum Tumor

(Panel 1) ESTABLISHMENT OF TUMOR-SPECIFIC SALIVARY TRANSCRIPTOMIC BIOMARKER PROFILE


Daf2
mRNA
p < .0001
15

ΔCt (GAPDH)
10
Discovery of Verification/validation of 5
tumor-specific tumor-specific
salivary transcriptomic salivary transcriptomic 0
Microarray profile profile
–5

er
hy

nc
alt

Ca
He
qPCR

(Panel 2) DETERMINE ROLE OF TUMOR-DERIVED EXOSOMES IN TUMOR-SPECIFIC SALIVARY TRANSCRIPTOMIC BIOMARKERS


Daf2
p = .656
20
Control group #1 (PBS) Tumor group #1 (GFP) Tumor group #2 (DN-Rab11-GFP)
ΔCt (GAPDH) 10 p = .001
Determine whether established tumor-
specific salivary transcriptomic profile 0
is altered with the inhibition of exosome
secretion at the tumor source –10

FP
hy

-G
GF
Saliva Saliva Saliva –20
alt

11
He

ab
-R
DN
qPCR

Fig 17-4  |  Mouse pancreatic cancer model demonstrating that tumor-derived exosomes are responsible for
the transport of tumor-specific RNA biomarkers from the organ of pathology to salivary glands and saliva.71

We have focused our efforts on under- salivary transcriptome were the result of the
standing the mechanisms and roles of salivary changes in cancer-derived exosomes. This study
exosomes in cancer. To provide proof of demonstrated that cancer-derived mRNAs are
concept for the potential utility of salivary the cargo of exosomes and reach the salivary
exosomes for cancer detection, we developed gland via circulation, providing a mechanistic
a pancreatic cancer mouse model in which link between discriminatory salivary biomark-
a mouse pancreatic cancer cell line (Panc02) ers and distal tumor.
was orthotopically injected into the pancreas To further our understanding of the link
of syngeneic mice.71 We investigated the role between salivary exosomes and distal tumor,
of pancreatic cancer–derived exosomes in sal- we generated a xenograft lung cancer mouse
ivary biomarker development by inhibiting the model in which H460 human lung cancer cells
exosome biogenesis in Panc02 cells (Fig 17-4). that stably express hCD63-GFP were ortho­
Stable transfection of the dominant negative topically injected into immunocompromised
form of the GTPase Rab11 (DN-Rab11) effec- mice.72 We identified human GAPDH mRNA
tively inhibited the biogenesis of exosomes in in hCD63+GFP+ exosome-like MVs in mouse
Panc02 cells and resulted in the ablation of saliva, indicating that exosome-like MVs carry
discriminatory salivary biomarker signatures tumor cell–specific mRNA and travel to the cir-
between tumor-bearing mice and control mice, culation and reach the saliva where they have
suggesting that the observed changes in the a potential role as tumor biomarkers.

352
Acknowledgment

Our most recent animal study demon- (PROM1), and tumor necrosis factor α–induced
strated a role of salivary exosomes in immune protein 3 (TNFα IP3). These works thus open
surveillance.73 To investigate an immunoregu- up promising new lines of research that may
latory effect of salivary exosomes, saliva from lead to the identification of new classes of can-
Panc02-bearing mice was orally administered cer biomarkers. Further functional studies of
to non–tumor-bearing control mice. Expres- salivary exosomes will provide new clues to its
sion levels of NK activation markers CD69 and mechanism of action and simultaneously raise
NKG2D was significantly decreased by garage fundamental questions about the coregulation
feeding of tumor saliva, while these effects were of serum and salivary exosomes on the progres-
ablated by DN-Rab11–mediated inhibition of sion of cancer.
exosome biogenesis in Panc02 cells. These
animal studies supported our hypothesis that
cancer-derived exosomes provide a rationale
for the development of salivary biomarkers Future Perspectives
that are applicable to distal tumor.
Winck et al performed a proteomic analy- In the past decade, salivaomics studies have
sis of salivary EVs from OSCC patients and revealed the utility of saliva in identifying the
healthy controls, and a total of 381 proteins presence of diseases. Much progress has been
were identified in the EVs from the two groups made in understanding the characteristics of
by mass spectrometry.24 Among the pro- saliva, with significant advances on how the
teins identified in salivary EVs, eight proteins salivary constituents relate to their biomark-
including α 2-macroglobulin (A2M), hapto- ers and functions. In addition, a growing body
globin α chain (HPa), mucin-5B (MUC5B), of saliva-exosomics study is highlighting the
galectin-3-binding protein (LGALS3BP), Ig α1 role of disease-derived exosomes in saliva.
chain C region (IGHA1), prolactin-inducible The unique properties of exosomes in saliva,
protein (PIP), pyruvate kinase isozymes M1/M2 which originate from organelles and then mi-
(PKM1/M2), and GAPDH were differentially grate into saliva, are attracting the attention of
expressed between the two groups (ANOVA, scientists as these could be used for diagnostic
P < .05). Gene Ontology analysis showed that biomarkers, potential surrogate markers for
the salivary EVs proteome in OSCC patients other physical conditions, or novel immune
was enriched in proteins related to molecular regulatory systems through the gastrointes-
transport and cellular growth and prolifera- tinal tract. These basic, translational, and
tion, suggesting that the property of salivary clinical research advancements of salivaomics
EVs may reflect the tumor of origin. Sun et al and saliva-exosomics support and credential
analyzed the proteome of salivary EVs isolated the mechanistic underpinnings of oral-systemic
from lung cancer patients and identified 113 connection and communication (Fig 17-5).
and 95 proteins in cancer patients and healthy
controls, respectively.28 Among the total 113
proteins identified in the cancer group, 63 pro-
teins were exclusively detected in this group. Acknowledgment
The literature survey showed that 12 proteins
are lung cancer–related biomarkers, including This work was supported by Public Health Ser-
annexin family members (annexin A1, A2, vice (PHS) grants from the National Institutes
A3, A5, A6, A11), nitrogen permease regula- of Health (NIH)—UH3 TR000923 and R90
tor 2-like protein (NPRL2), CEA-related cell DE022734—and the Ronnie Dios Stand Up
adhesion molecule 1 (CEACAM1), histone H4 and Shout Cancer Research Fund.
(HIST1H4A), mucin 1 (MUC1), prominin-1

353
17 Oral-Systemic Connection: The Salivaomics and Exosomics Connection

Salivary gland
Serous acinar cells Salivary gland

Acinus-derived exosomes
Cancer-derived Exosomes
exosomes

Endocytosis
Striated duct
Membra
ne fusio Cancer-derived exosomes
n

Pancreatic cancer Mucous acinar cells

Fig 17-5  |  Oral-systemic connection: The salivaomics and saliva-exosomics connection.

Disclosures 4. Jusko WJ, Milsap RL. Pharmacokinetic principles of


drug distribution in saliva. Ann N Y Acad Sci 1993;694:
36–47.
David Wong is cofounder of RNAmeTRIX 5. Wong DT. Salivaomics. J Am Dent Assoc 2012;143(10
Inc, a molecular diagnostic company. He suppl):19S–24S.
holds equity in RNAmeTRIX and serves as a 6. Denny P, Hagen FK, Hardt M, et al. The proteomes of
human parotid and submandibular/sublingual gland
company Director and Scientific Advisor. The salivas collected as the ductal secretions. J Proteome
University of California also holds equity in Res 2008;7:1994–2006.
RNAmeTRIX. Intellectual property that David 7. Ai J, Smith B, Wong DT. Saliva Ontology: An ontology-
Wong invented and which was patented by the based framework for a salivaomics knowledge base.
BMC Bioinformatics 2010;11:302.
University of California has been licensed to 8. Bandhakavi S, Stone MD, Onsongo G, Van Riper SK,
RNAmeTRIX. Dr Wong is also a consultant Griffin TJ. A dynamic range compression and
for GlaxoSmithKline, Wrigley, EZLife Bio, three-dimensional peptide fractionation analysis platform
expands proteome coverage and the diagnostic potential
and Colgate-Palmolive.
of whole saliva. J Proteome Res 2009;8:5590–5600.
9. Yan W, Apweiler R, Balgley BM, et al. Systematic
comparison of the human saliva and plasma proteomes.
Proteomics Clin Appl 2009;3:116–134.
References 10. Helmerhorst EJ, Oppenheim FG. Saliva: A dynamic
proteome. J Dent Res 2007;86:680–693.
1. Van Nieuw Amerongen A, Bolscher JG, Veerman EC. 11. Esser D, Alvarez-Llamas G, de Vries MP, Weening D,
Salivary proteins: Protective and diagnostic value in Vonk RJ, Roelofsen H. Sample stability and protein
cariology? Caries Res 2004;38:247–253. composition of saliva: Implications for its use as a diag-
2. Edgar WM. Saliva and dental health. Clinical implications nostic fluid. Biomark Insights 2008; 3:25–27.
of saliva: Report of a consensus meeting. Br Dent J 12. Xiao H, Wong DT. Method development for proteome
1990;169:96–98. stabilization in human saliva. Anal Chim Acta 2012;722:
3. Tiwari M. Science behind human saliva. J Nat Sci Biol 63–69.
Med 2011;2:53–58.

354
References

13. Hoerman KC, Chauncey HH, Herrold RD. Parotid saliva 28. Sun Y, Xia Z, Shang Z, et al. Facile preparation of salivary
acid phosphatase in prostatic cancer. Cancer 1959; 12: extracellular vesicles for cancer proteomics. Sci Rep
359–363. 2016;6:24669.
14. Navarro MA, Mesía R, Díez-Gibert O, Rueda A, Ojeda 29. Chen DX, Schwartz PE, Li FQ. Saliva and serum CA
B, Alonso MC. Epidermal growth factor in plasma and 125 assays for detecting malignant ovarian tumors.
saliva of patients with active breast cancer and breast Obstet Gynecol 1990;75:701–704.
cancer patients in follow-up compared with healthy 30. He CZ, Zhang KH, Li Q, Liu XH, Hong Y, Lv NH.
women. Breast Cancer Res Treat 1997;42:83–86. Combined use of AFP, CEA, CA125 and CAI9-9
15. Streckfus C, Bigler L, Tucci M, Thigpen JT. A preliminary improves the sensitivity for the diagnosis of gastric
study of CA15-3, c-erbB-2, epidermal growth factor cancer. BMC Gastroenterol 2013;13:87.
receptor, cathepsin-D, and p53 in saliva among women 31. Lee YH, Kim JH, Zhou H, Kim BW, Wong DT. Salivary
with breast carcinoma. Cancer Invest 2000;18:101–109. transcriptomic biomarkers for detection of ovarian
16. Brooks MN, Wang J, Li Y, Zhang R, Elashoff D, Wong cancer: For serous papillary adenocarcinoma. J Mol
DT. Salivary protein factors are elevated in breast cancer Med (Berl) 2012;90:427–434.
patients. Mol Med Rep 2008;1375–1378. 32. Park NJ, Li Y, Yu T, Brinkman BM, Wong DT. Charac-
17. Zhang L, Xiao H, Karlan S, et al. Discovery and preclinical terization of RNA in saliva. Clin Chem 2006;52:988–994.
validation of salivary transcriptomic and proteomic 33. Li Y, Zhou X, St John MA, Wong DT. RNA profiling of
biomarkers for the non-invasive detection of breast cell-free saliva using microarray technology. J Dent Res
cancer. PLoS One 2010;5:e15573. 2004;83:199–203.
18. Wood N, Streckfus CF. The expression of lung resistance 34. Park NJ, Zhou X, Yu T, et al. Characterization of salivary
protein in saliva: A novel prognostic indicator protein for RNA by cDNA library analysis. Arch Oral Biol 2007;52:
carcinoma of the breast. Cancer Invest 2015; 33: 30–35.
510–515. 35. Spielmann N, Ilsley D, Gu J, et al. The human salivary
19. Ohshiro K, Rosenthal DI, Koomen JM, et al. Pre-analytic RNA transcriptome revealed by massively parallel
saliva processing affect proteomic results and biomarker sequencing. Clin Chem 2012;58:1314–1321.
screening of head and neck squamous carcinoma. Int 36. Lee YH, Zhou H, Reiss JK, et al. Direct saliva transcrip-
J Oncol 2007;30:743–749. tome analysis. Clin Chem 2011;57:1295–1302.
20. Hu S, Arellano M, Boontheung P, et al. Salivary proteom- 37. Li Y, Elashoff D, Oh M, et al. Serum circulating human
ics for oral cancer biomarker discovery. Clin Cancer Res mRNA profiling and its utility for oral cancer detection.
2008;14:6246–6252. J Clin Oncol 2006;24:1754–1760.
21. Dowling P, Wormald R, Meleady P, Henry M, Curran A, 38. Li Y, St John MA, Zhou X, et al. Salivary transcriptome
Clynes M. Analysis of the saliva proteome from patients diagnostics for oral cancer detection. Clin Cancer Res
with head and neck squamous cell carcinoma reveals 2004;10:8442–8450.
differences in abundance levels of proteins associated 39. Zhang L, Farrell JJ, Zhou H, et al. Salivary transcriptomic
with tumour progression and metastasis. J Proteomics biomarkers for detection of resectable pancreatic
2008;71:168–175. cancer. Gastroenterology 2010;138:949–957.
22. Rai B, Kaur J, Jacobs R, Anand SC. Adenosine deam- 40. Zhang L, Xiao H, Zhou H, et al. Development of tran-
inase in saliva as a diagnostic marker of squamous cell scriptomic biomarker signature in human saliva to detect
carcinoma of tongue. Clin Oral Investig 2011; 15: lung cancer. Cell Mol Life Sci 2012;69:3341–3350.
347–349. 41. Lin S, Gregory RI. MicroRNA biogenesis pathways in
23. Elashoff D, Zhou H, Reiss J, et al. Prevalidation of salivary cancer. Nat Rev Cancer 2015;15:321–333.
biomarkers for oral cancer detection. Cancer Epidemiol 42. Park NJ, Zhou H, Elashoff D, et al. Salivary microRNA:
Biomarkers Prev 2012;21:664–672. Discovery, characterization, and clinical utility for oral
24. Winck FV, Prado Ribeiro AC, Ramos Domingues R, et cancer detection. Clin Cancer Res 2009; 15:
al. Insights into immune responses in oral cancer 5473–5477.
through proteomic analysis of saliva and salivary extra- 43. Matse JH, Yoshizawa J, Wang X, et al. Discovery and
cellular vesicles. Sci Rep 2015;5:16305. prevalidation of salivary extracellular microRNA biomark-
25. Wu ZZ, Wang JG, Zhang XL. Diagnostic model of saliva ers panel for the noninvasive detection of benign and
protein finger print analysis of patients with gastric malignant parotid gland tumors. Clin Cancer Res 2013;
cancer. World J Gastroenterol 2009;15:865–870. 19:3032–3038.
26. Xiao H, Zhang Y, Kim Y, et al. Differential proteomic 44. Bahn JH, Zhang Q, Li F, et al. The landscape of micro­
analysis of human saliva using tandem mass tags quan- RNA, piwi-interacting RNA, and circular RNA in human
tification for gastric cancer detection. Sci Rep 2016;6: saliva. Clin Chem 2015;61:221–230.
22165. 45. Kalra H, Simpson RJ, Ji H, et al. Vesiclepedia: A
27. Xiao H, Zhang L, Zhou H, Lee JM, Garon EB, Wong DT. compendium for extracellular vesicles with continuous
Proteomic analysis of human saliva from lung cancer community annotation. PLoS Biol 2012;10:e1001450.
patients using two-dimensional difference gel electro-
phoresis and mass spectrometry. Mol Cell Proteomics
2012;11:M111.012112.

355
17 Oral-Systemic Connection: The Salivaomics and Exosomics Connection

46. Pan BT, Johnstone RM. Fate of the transferrin receptor 60. Witwer KW, Buzás EI, Bemis LT, et al. Standardization
during maturation of sheep reticulocytes in vitro: Selec- of sample collection, isolation and analysis methods in
tive externalization of the receptor. Cell 1983; 33: extracellular vesicle research. J Extracell Vesicles 2013;2.
967–978. 61. Colombo M, Moita C, van Niel G, et al. Analysis of
47. Johnstone RM, Adam M, Hammond JR, Orr L, Turbide ESCRT functions in exosome biogenesis, composition
C. Vesicle formation during reticulocyte maturation. and secretion highlights the heterogeneity of extracellular
Association of plasma membrane activities with released vesicles. J Cell Sci 2013;126:5553–5565.
vesicles (exosomes). J Biol Chem 1987; 262: 62. Simpson RJ, Kalra H, Mathivanan S. ExoCarta as a
9412–9420. resource for exosomal research. J Extracell Vesicles
48. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, 2012;1.
Lötvall JO. Exosome-mediated transfer of mRNAs and 63. Ogawa Y, Miura Y, Harazono A, et al. Proteomic analysis
microRNAs is a novel mechanism of genetic exchange of two types of exosomes in human whole saliva. Biol
between cells. Nat Cell Biol 2007;9:654–659. Pharm Bull 2011;34:13–23.
49. Ogawa Y, Kanai-Azuma M, Akimoto Y, Kawakami H, 64. Xiao H, Wong DT. Proteomic analysis of microvesicles
Yanoshita R. Exosome-like vesicles with dipeptidyl pepti- in human saliva by gel electrophoresis with liquid
dase IV in human saliva. Biol Pharm Bull 2008; 31: chromatography-mass spectrometry. Anal Chim Acta
1059–1062. 2012;723:61–67.
50. Gould SJ, Raposo G. As we wait: Coping with an imper- 65. Lässer C, Alikhani VS, Ekström K, et al. Human saliva,
fect nomenclature for extracellular vesicles. J Extracell plasma and breast milk exosomes contain RNA: Uptake
Vesicles 2013;2. by macrophages. J Transl Med 2011;9:9.
51. Al-Tarawneh SK, Border MB, Dibble CF, Bencharit S. 66. Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki
D e fi n i n g s a l i v a r y b i o m a r k e r s u s i n g m a s s Y, Ochiya T. Secretory mechanisms and intercellular
spectrometry-based proteomics: A systematic review. transfer of microRNAs in living cells. J Biol Chem 2010;
OMICS 2011;15:353–361. 285:17442–17452.
52. Nonaka T, Wong DTW. Saliva-exosomics in cancer: 67. Skog J, Würdinger T, van Rijn S, et al. Glioblastoma
Molecular characterization of cancer-derived exosomes microvesicles transport RNA and proteins that promote
in saliva. Enzymes 2017;42:125–151. tumour growth and provide diagnostic biomarkers. Nat
53. Raposo G, Nijman HW, Stoorvogel W, et al. B lympho- Cell Biol 2008;10:1470–1476.
cytes secrete antigen-presenting vesicles. J Exp Med 68. Tanaka Y, Kamohara H, Kinoshita K, et al. Clinical impact
1996;183:1161–1172. of serum exosomal microRNA-21 as a clinical biomarker
54. Conde-Vancells J, Rodriguez-Suarez E, Embade N, et in human esophageal squamous cell carcinoma. Cancer
al. Characterization and comprehensive proteome profil- 2013;119:1159–1167.
ing of exosomes secreted by hepatocytes. J Proteome 69. Kahlert C, Melo SA, Protopopov A, et al. Identification
Res 2008;7:5157–5166. of double-stranded genomic DNA spanning all chromo-
55. Palanisamy V, Deshpande AM, Sharma S, et al. Nano- somes with mutated KRAS and p53 DNA in the serum
characterization of exosomal-like micro vesicles exosomes of patients with pancreatic cancer. J Biol
enriched with mRNAs in human saliva. Presented at the Chem 2014;289:3869–3875.
38th Meeting of the American Association for Dental 70. Melo SA, Luecke LB, Kahlert C, et al. Glypican-1 iden-
Research, Miami, 1–4 April 2009. tifies cancer exosomes and detects early pancreatic
56. Sharma S, Rasool HI, Palanisamy V, et al. Structural- cancer. Nature 2015;523:177–182.
mechanical characterization of nanoparticle exosomes 71. Lau C, Kim Y, Chia D, et al. Role of pancreatic
in human saliva, using correlative AFM, FESEM, and cancer-derived exosomes in salivary biomarker devel-
force spectroscopy. ACS Nano 2010;4:1921–1926. opment. J Biol Chem 2013;288:26888–26897.
57. Gonzalez-Begne M, Lu B, Han X, et al. Proteomic anal- 72. Yang J, Wei F, Schafer C, Wong DT. Detection of tumor
ysis of human parotid gland exosomes by cell-specific mRNA and protein in exosome-like microve-
multidimensional protein identification technology sicles from blood and saliva. PLoS One 2014; 9:
(MudPIT). J Proteome Res 2009;8:1304–1314. e110641.
58. Taylor DD, Lyons KS, Gerçel-Taylor C. Shed membrane 73. Katsiougiannis S, Chia D, Kim Y, Singh RP, Wong DT.
fragment-associated markers for endometrial and ovar- Saliva exosomes from pancreatic tumor-bearing mice
ian cancers. Gynecol Oncol 2002;84:443–448. modulate NK cell phenotype and antitumor cytotoxicity.
59. Sharma S, Gillespie BM, Palanisamy V, Gimzewski JK. FASEB J 2017;31:998–1010.
Quantitative nanostructural and single-molecule force
spectroscopy biomolecular analysis of human-saliva-
derived exosomes. Langmuir 2011;27:14394–14400.

356
CHAPTER 18

The Economic Impact of


Periodontal Inflammation
Michael C. Alfano, dmd, phd

This chapter appears at the end of this sec- even partial or contributory causality, is estab-
ond edition of The Oral-Systemic Health lished, the dimensions of economic impact
Connection because it is logical to consider expand substantially.
the economic effects of a biologic process, a What is a contributory cause of a disease?
disease or set of diseases, or their various treat- Bale et al2 define it as follows: “A contributory
ments after the basic aspects of the processes cause does not require that all those who possess
involved are clearly understood. Thus, I will the contributory cause experience the disease,
not endeavor to repeat the explanations of nor does it require that all those who are free
specific biologic processes, treatments, and out- of the contributory cause be free of the disease.
comes addressed earlier in this text. However, It also means the contributory cause may not
the various biologic processes and interactions be necessary to experience the disease.” We can
have been studied for several decades, more clarify this explanation by using the relation-
than 10,000 papers have been written on these ship between smoking and lung cancer. A person
topics, and tens of millions of research dollars can smoke and never get lung cancer; another
have been expended, all in the search for great- person who has never smoked can get lung
er clarity in our understanding of the precise cancer. But in the majority of the population
interactions of oral and systemic diseases.1 (80% to 90%, according to the American Lung
While not every paper has shown a clear Association5), smoking becomes the cause—the
relationship between oral infection, especially contributory cause—of their lung cancer.
periodontal disease, and systemic health, the vast I deliberately selected lung cancer as an
majority of the published literature supports a example because while there is no doubt that
claim that is now virtually universally accepted: smoking is a contributory cause of lung can-
Periodontal infection is associated with a num- cer, no randomized, double-blind, controlled
ber of noncommunicable systemic inflammatory trial of smoking and lung cancer has ever
diseases. The goal of this chapter is to take that been performed. This is important because
agreement and move the discussion from the most researchers today insist it is impossible
association of periodontal disease with a num- to say that periodontal disease is a contrib-
ber of inflammatory-based systemic diseases to utory cause of noncommunicable systemic
the role of periodontal disease as a contributory inflammatory diseases such as cardiovascular
cause of several inflammatory-based noncom- disease (CVD) or diabetes until a large, ran-
municable systemic diseases.2–4 Once causality, domized, well-controlled clinical trial confirms

357
18 The Economic Impact of Periodontal Inflammation

it. I submit that such a large, expensive, and periodontal treatment versus those who did not
difficult-to-control study will never be done. was also measured; the following systemic con-
Indeed, based on what we have learned from ditions and associated savings were reported:
the private insurance sector on this topic, which $2,840 annual savings with type 2 diabetes,
is described later in this chapter, I further submit $5,681 annual savings with cerebrovascular
that such a large prospective study might be disease, and $1,090 annual savings with coro-
unethical. Thus, by insisting on a study that will nary artery disease.
likely never be performed, the research com- One criticism of this type of retrospective
munity is potentially denying society a huge study is that the subjects in the dental treatment
benefit from improved periodontal care driven groups are simply more compliant with their
not only by an interest in oral health but also by health care regimens. However, one of the cited
a desire to mitigate the effects of life-threatening studies, conducted by United Healthcare, spe-
diseases such as diabetes and heart disease. cifically controlled for this compliance effect,
This benefit, as shown by the insurance stud- and it showed the opposite effect of what might
ies described in the next section, consists of a be expected.8 Those individuals who received
significantly lower chance of hospitalization periodontal care and were not compliant with
and complications from inflammatory-based their medical treatment regimens had much
systemic diseases and a potential annual savings higher annual savings. Specifically, those who
of billions of dollars in the United States alone. received periodontal care and were not com-
It is time to consider whether it continues to be pliant with medical recommendations saved
ethical to deny direct causal inference between $1,849 per year compared to savings of $264
periodontal disease and systemic disease. per year by those who received periodontal
care and were compliant with medical recom-
mendations. In short, both groups who had
The Insurance Studies: the benefit of periodontal care showed medical
savings, with the noncompliant group show-
A Framework for Action ing much greater savings. While this study is
not dispositive in proving periodontal care
Several large, retrospective analyses of insurance reduces total health care cost, it is very sup-
data have consistently shown that individuals portive of this relationship and mitigates the
who receive periodontal therapy have signifi- most obvious objection to such retrospective
cantly lowered costs of health care, driven study designs. More definitive proof of the ben-
primarily by fewer hospitalizations and emer- efits of periodontal care in reducing total health
gency department visits.6–9 For example, Mosen care costs will be described in the next section.
et al6 reported significantly better hemoglobin All three studies cited earlier7–9 were sup-
A1c (HbA1c) control, a 44% reduction in hospital ported by the dental insurance industry and
admissions, and a 38% reduction in emergen- conducted on proprietary databases. Some
cy department visits in a population of diabetic would devalue the importance of two of them,
patients who received regular dental care com- though published in peer-reviewed journals,
pared with a matched set of diabetic patients because of the sponsorship of the research. This
who did not receive regular dental care. Using makes the work of the team at the American
data from United Concordia, Jeffcoat et al7 Dental Association (ADA) Health Policy Insti-
reported a strikingly similar reduction in hos- tute led by Marko Vujicic more meaningful.
pitalizations of 39.4% in a study of patients In this study, Nasseh et al9 used a commer-
with diabetes who received periodontal care cially available database from Truven Health
versus those who did not. The annual reduc- MarketScan Research to explore the effect of
tion in treatment costs for those who received periodontal treatment on the health care costs

358
The Economic Savvy of Dental Insurers

associated with newly diagnosed type 2 diabe- grow to 20% of eligible individuals seeking
tes. One might suspect that in cases of newly care by year 10. Thus, over a 10-year period,
diagnosed diabetes, some of the effects of the periodontal care would cost $7.2 billion. How-
diabetic process may not yet have taken hold, ever, the projected savings, driven primarily by
potentially minimizing any salutary effect of fewer hospitalizations for the systemic condi-
periodontal treatment. However, the authors tions during this same 10-year period, would
reported total health care savings in the peri- be $70.7 billion, for a net savings of $63.5
odontal treatment group of $1,799, a figure billion over 10 years. A year-by-year analysis
that is remarkably similar to what others have showed that the program generated net savings
reported, even though the study was conducted of $500 million in year 1, growing to a net sav-
on a different group of individuals at a differ- ings of $12.2 billion dollars by year 10. If more
ent time using different methodologies. people accessed the program, the savings would
In an analysis of the earlier studies plus an go up, and Avalere estimated that these positive
additional Cigna study,10 Chávez et al11 esti- effects would continue over the long term.
mated the potential savings nationwide for the The remarkable similarity of the reports from
pool of Medicare patients. Their results were six different insurance studies, completed at six
remarkable: When they assessed the total sav- different times by six different investigators on
ings available if all Medicare patients with a six different populations using somewhat differ-
history of stroke, congestive heart failure, or ent methods and definitions, makes a compelling
diabetes received periodontal care each year, the case that the minimization of oral inflammation
annual savings would be $19.0 billion based through periodontal care improves certain sys-
on the Cigna study, $18.8 billion based on the temic diseases so that total health care costs
United Concordia data, and $20.4 billion based are dramatically lowered. However, before we
on the United Healthcare dataset. Given the dif- can complete the case that periodontal care is
ferent approaches, timelines, investigators, and a contributory cause of systemic disease, some
populations used to generate these calculations, prospective data is needed. The next section
the similarities in the numbers are amazing. reports on such prospective data. However, the
While the analysis by Chávez et al11 is proprietary nature of the next level of support
impressive, it assumes that everyone who is requires the reader to accept the actions of the
eligible for periodontal care will receive it, and insurance sector based on verbal reports. The
this is not at all likely. Therefore, we should prospective data have not been published, most
look to another analysis, completed by the likely in an effort to retain competitive advan-
respected consulting firm Avalere.12 The Avalere tage in the marketplace.
study took data from three of the studies dis-
cussed earlier (Cigna,10 United Concordia,7 and
United Healthcare8) and added data generated
by Aetna to develop a model to determine the
The Economic Savvy of
impact of periodontal care on the cost of the Dental Insurers
Medicare program if periodontal care was
made available through Medicare to anyone Unencumbered by traditional academ-
with type 2 diabetes, CVD, or stroke. They esti- ic norms, which would have required
mated the cost of periodontal care at $825 for large, well-controlled clinical trials of the im-
initial treatment (scaling and root planing) and pact of periodontal care on systemic disease,
$250 for maintenance visits every 6 months the insurance industry acted on its retrospec-
thereafter. In addition, they assumed that only tive assessments of periodontal therapy on
5% of eligible individuals would seek the care the costs of treating systemic disease. To be
in year 1 and that this number would only clear, the industry did not act in a vacuum

359
18 The Economic Impact of Periodontal Inflammation

based on its own studies alone; it consult- As I am best able to determine, these mar-
ed the decades-long research enterprise that keting actions took several shapes. First, the
showed numerous epidemiologic associations, dental insurers began to share their data with
highly plausible biologic mechanisms, smaller large corporations to convince them of the out-
clinical trials, and surrogate endpoint analyses, size value of dental insurance in terms of its
each of which, on balance, supported the con- ability to save money in other areas of health
cept that oral inflammation is a contributory care. This argument resonates with corpora-
cause of systemic diseases that are also driven tions because most of them are self-insured,
by inflammatory processes. So how exactly did and any savings derived from the addition of
the insurance industry respond? periodontal services to the benefits mix delivers
To answer this question requires indulgence savings directly to the bottom line by reducing
on the part of the reader, because the infor- health care payments for employees who suffer
mation on which I base this discussion comes from one of the inflammatory-based systemic
from personal conversations with insurance diseases. Second, those dental insurers that
industry executives and direct observation of were a subsidiary of a larger health insurance
presentations made by industry representatives company began to work out reimbursement
to the Centers for Medicare and Medicaid arrangements from the general health side of
Services (CMS) and the Congressional Budget the parent company for cost benefits delivered
Office (CBO). In addition, due to the com- by the dental company to the general health
petitive nature of the insurance business and of the insured population, to be shared back
because the data presented can represent a with the dental company in some appropriate
proprietary advantage to one company over ratio. I am told that such funds only changed
another, I feel obliged to protect the identities hands after those insured individuals who suf-
of the various presenters. Indeed, they came fer from one of the noncommunicable diseases
to the table in generous common cause with actually received the requisite periodontal care.
several not-for-profit organizations, including Indeed, it is reported that the dental insurers
the Santa Fe Group, Oral Health America, would actually recruit the affected insured indi-
Pacific Dental Services Foundation, and the viduals for periodontal care and would add
Center for Medicare Advocacy, in an effort to enticements such as waiving copayments and
shed light on their actions vis-à-vis oral and deductibles to enhance the chances that the
systemic health, so that the huge federal pro- affected people would seek periodontal care.
grams of Medicaid and Medicare might benefit Finally, some dental insurers are reported to
from their real-world experiences. In sum, these have taken these various processes to the next
individuals acted for the public good, and they level by offering their dental/periodontal insur-
and the insurance industry per se should be ance services to general health insurers that did
commended for this action. not have a dental insurer in their portfolios of
Disclaimers aside, we can now answer the companies. These arrangements presumably
question: How exactly did the insurance indus- worked through contracts in which the gen-
try respond? The industry responded by acting eral health care savings were shared via some
on its retrospective studies and marketing its appropriate formula between the two different
periodontal insurance services not only for insurers.
their inherent benefits to oral health, which This chapter could have been written
include tooth retention, fresh breath, improved without the disclosures on insurance indus-
mastication, and social confidence, but also try practices included in this section. Indeed,
for the benefits to systemic health that include while everything to my knowledge about these
markedly lower costs for the management of practices is legal, and although nothing was
diseases such as diabetes and heart disease. told to me or observed by me in confidence,

360
Challenge to the Dental Profession: Moving from Association to Causation

I take some risk in dispensing with the norms as the gold standard of evidence required prior
of referencing for this section of the chapter to approving a new drug or device, accepting
even through such means as named personal a surgical technique, or changing a therapeutic
communications. However, this information paradigm. Indeed, the well-executed clinical tri-
was included because it is important that al creates a sort of safe space for researchers and
individuals from the private insurance sector thought leaders, and in general it has served so-
continue to be forthcoming with their data and ciety very well to protect against sham products
practices so that the public insurance sector can or outright scams through the years. However,
derive the same benefits. Even more important there are many scenarios of clinical trials not
is that the research, practice, and educational serving society well. The best examples come
communities know of these private insurance from drug development, where side effects often
practices because they constitute real-world, go unnoticed until a given drug is delivered not
prospective confirmation that the effects of hundreds or even thousands of times but tens of
periodontal care to mitigate certain systemic thousands of times; or perhaps the drug is not
diseases are, in fact, valid. given for weeks or months but for years. This
Some of the dental insurers involved have does not mean that clinical trials are of no use
been acting in the way described earlier for but that they must be accompanied by continu-
more than a decade. If the effect of periodon- ing surveillance as in the phase IV approach or
tal care to reduce total health care costs is in real-world scenarios in people with comorbid
not real, their reimbursement practices would disease as in the N of 1 approach.
have collapsed under their own weight because Further assault on the value of the clinical
they would have failed economically a long trial can come in the form of the evidence-based
time ago. In sum, I consider the six insurance review system, which can devalue scores if not
studies summarized in the prior section and hundreds of studies in a single analysis. Indeed,
the prospective actions of the private insur- while the evidence-based approach has done
ance industry described in this section to be wonders to enhance critical albeit retrospective
equal to or better than the proof that may thinking on clinical design, we are regularly
have been provided by a well-controlled, large confronted with the need to rethink norms of
clinical trial. In a very significant way, these clinical care because the evidence is weak. I
real-world insurance studies and the current reiterate that the discipline of evidence-based
insurance company actions are the equivalent reviews has been a good thing, but I fear that
of a phase IV trial or an N of 1 clinical trial, as we are abusing the process and are in danger
eloquently defined by Curro et al.13 Finally, this of approaching a situation in which no study
information provides the basis for the challenge is good enough to retrospectively pass muster.
to the dental research, education, and practice Indeed, when was the last time you read an
communities as described in the next section. evidence-based review that concluded that the
evidence is strong and consistent? Continuing
along the current path will have the insidious
effect of further debasing science in the minds
Challenge to the Dental of the public, resulting in a situation in which
Profession: Moving from political opinion, folklore, religion, and such
are placed on an equal plane with science
Association to Causation (eg, creationism versus evolution). While not
directly related to the purpose of this chapter,
I cut my teeth in clinical research, both as an ac- I urge all who are engaged in evidence-based
ademic researcher and as an industry executive, reviews to set their parameters and choose their
on the value of the well-controlled clinical trial words carefully lest the very purpose for which

361
18 The Economic Impact of Periodontal Inflammation

the evidence-based process was created is mar- Santa Fe Group Position Statement
ginalized, with the unintended consequence of on Oral-Systemic Interactions
demeaning all scientific data on all subjects.
Finally, as noted previously, although there After decades of research and thousands of
never was a well-controlled human trial on scientific papers, the relationships between
the effects of smoking on lung cancer, it is oral health, especially periodontal health,
virtually universally accepted that smoking is and systemic health are well known. More-
a contributory cause of lung cancer and that over, during the past ten years, data analysis
smoking affects 80% to 90% of all lung can- by health economists, and public statements
cers. How did it become acceptable to blame and actions by several large, private dental
smoking for lung cancer? When did the data insurers have identified additional benefits of
become compelling enough to move the dia- oral health by revealing that insured individ-
logue from association to causation? Was it uals who receive treatment for periodontal
the report of the Surgeon General in 1964, as disease show fewer hospitalizations and re-
referenced in chapter 1? Was it the work of duced cost of care for a number of systemic
Hammond and Auerbach on smoking beagles diseases including diabetes, cardiovascular
in early 1970?14 Or was it merely the constant disease, and stroke. Therefore, the Santa
repetition of all of the observational data? I do Fe Group has concluded that sufficient ev-
not know when the relationship tipped from idence now exists that periodontal disease
associative to causal, but it did tip, and it never is a contributory cause to certain systemic
went back, despite the efforts of the Tobacco diseases, and the public should benefit from
Institute and the cigarette industry to malign this knowledge. Specifically, Medicare, Med-
the causal relationship. icaid, and other public and private health
I submit that it is past time for the oral insurance programs should incorporate
health research community to tip the dialogue oral health benefits as a component of com-
on the relationship between oral and systemic prehensive health insurance. These health
health from associative to causal. By clinging benefits will not only improve oral health
to a messianic belief that we cannot speak of for its own sake, including speech, masti-
causation until the large clinical trial is deliv- cation and social acceptance, but will also
ered, we are denying society the opportunity produce substantial economic benefits and
to benefit fully from the decrease in hospital- total health improvement for the public.
izations and emergency department visits that
results from periodontal therapy. In particular, This Santa Fe Group statement has been
we are denying the large number of individuals published in two of the most widely read den-
who are on public health insurance to benefit tal journals in the United States, The Journal
in the way those insured by private entities are of the American Dental Association3 and the
benefitting. As noted previously, when society Compendium of Continuing Education in
loses the indirect systemic benefits of periodon- Dentistry4; moreover, it has been presented to
tal care, it also loses the direct benefits of oral senior leadership of the CMS and CBO. The
care, including tooth retention, fresh breath, Santa Fe Group statement has been used to
improved mastication, and the enhanced social solidify the importance of oral health to total
acceptance derived therefrom. health, to galvanize support for oral health
Arguably no entity has done more to tip the from numerous not-for-profit organizations in
discussion from association to causation than the health care segment, to motivate the global
the Santa Fe Group. This group has published periodontal research community to be more
a statement on the relationships of oral and forthcoming, and to endeavor to convince the
systemic health,3,4 which reads as follows: federal and state governments that it is foolish

362
Reordering Priorities in Medicare and Medicaid Policy

to try to save money by cutting dental care you will, or even a cosmetic service that comes
benefits. in near last in any priority-setting exercise for
To date, the Santa Fe Group statement has allocation of research dollars. We see this atti-
generated great interest and surprisingly lit- tude play out almost every day, as many states
tle open criticism. While there is much work choose not to exercise their right to provide
to do, the dental, medical, and public health dental care under federal Medicaid rules. We
communities—hopefully soon joined by crit- see it when states with public dental insurance,
ical governmental agencies—are poised to even progressive states like California, decide
promote the concept that periodontal disease to eliminate dental care first as soon as there is
is a contributory cause to many systemic health a financial crisis. We even see it in the fact that
problems. Will you join this effort? Will you dental infection is the only bodily infection that
accept the substantial real-world experience Medicare does not routinely cover.
described in the prior section? Or will you stay This historic separation has been made
in the safe place of waiting for the large clinical worse by the dental profession’s aversion to
trial that will probably never come? participating in public insurance programs.
What are the risks of stating that the rela- Practicing dentists seem to perceive the rules
tionship between oral and systemic health is and guidelines that accompany programs like
causal as opposed to merely associative? Are Medicaid and Medicare as an unwieldy intru-
we asking people to take new drugs? No. Are sion on their independence. This attitude was in
we suggesting a different form of periodontal play even before the rise of Medicaid services,
therapy? No. Are there unintended conse- with its flawed dental reimbursement model,
quences of periodontal care? Very few (eg, gave dentists good reason to resist the expan-
root sensitivity, rare anesthetic reactions) are sion of such programs. Indeed, dentists were
reported. Even if the thesis presented herein opposed to Medicare when it was founded in
should be disproved—and I submit that it will 1965, well before they began to have difficult
not—the outcome of periodontal care will still experiences with Medicaid reimbursement.
be positive. People will have better masticatory Moreover, the ADA has had a resolution on
function, less chance of painful inflammatory its dockets since 1993 to expand Medicare to
flare-ups, fresher breath, less tooth loss, better cover all medically necessary dental care, but
speech, and enhanced confidence in social set- it has been unwilling or unable to advance this
tings. Stated another way, there is no significant cause for the past 25 years.
downside to delivering periodontal care, and The resistance to these federal health pro-
the potential benefits, direct and indirect, in grams is actually more principled than practical
terms of total health and in terms of finance or self-serving. Nevertheless, any principles
are enormous. that oppose federal involvement in health care
delivery must be measured against the socie-
tal failure that occurs when low-income indiv
iduals are unable to afford health care. To be
Reordering Priorities in sure, one-off events such as Missions of Mercy
Medicare and Medicaid and even Give Kids a Smile are worthy gestures
of the charitable nature of many in the dental
Policy profession, but they do little to assist a person
with an off-cycle, painful dental health prob-
One of the many idiosyncrasies of the historic lem. The dental profession cannot simply resist
separation of the medical and dental profes- efforts to improve health care access; it needs
sions is the belief that dental care is not really to bring viable plans to the table to sustain the
health care. It is an elective service—a luxury, if dignity of the profession.

363
18 The Economic Impact of Periodontal Inflammation

One potentially viable plan for a routine den- benefit. The primary global benefit in the Jones
tal benefit within Medicare has arisen as a joint approach has a specific focus to “prevent pain,
effort of the Santa Fe Group, the DentaQuest inflammation, and infection,” which was
Foundation, and Oral Health America. A key designed specifically to ensure that all partici-
element of this plan is to parallel reimburse- pants in the Medicare oral health benefit have
ment practices of the private insurance sector optimal ability to minimize the effects of oral
in the belief that it makes no sense to proffer a inflammation on systemic health.15
poorly financed plan for dental coverage that The Jones approach applies to a broad den-
no clinical provider is interested in accepting. tal benefit for all Medicare recipients, so it is
Great credit is due to Judy Jones and her coau- what could be called a net coster. In contrast, a
thors and colleagues for identifying the scope plan like that proposed in the Avalere report,12
of the need for oral health care in America’s whereby only selected periodontal services are
aged population and for framing out a realistic offered only to Medicare recipients who have a
approach to a dental benefit in Medicare.11,15–19 diagnosis of diabetes, heart disease, or stroke,
The work of Dr Jones and her colleagues is a net saver. Let’s explore the economic dif-
to add a dental care benefit to Medicare will ferences in these approaches.
be referred to as the Jones approach. While The Jones approach to a Medicare dental
it is not the only approach, it is the one on benefit is biphasic. The first phase, Level 1, con-
which the most information is published, and sists of the core global benefit with a primary
it appears to be the broadest approach in that goal of preventing pain, inflammation, and
34 collaborators from more than two dozen infection. It therefore includes diagnostic, pre-
agencies and institutions participated in some ventive, nonsurgical periodontal therapy and
way.15 Other efforts worthy of mention are the nonelective oral surgery, reimbursed at 70% of
joint effort to expand the definition of medi- usual, customary, and reasonable fees with no
cally necessary dental care, driven primarily by patient copayments to increase participation.
the Dental Lifeline Network and the Center for The estimated cost for this benefit is $32.01
Medicare Advocacy; the economic argument per member per month (PMPM). Level 2 ben-
articulated by Avalere12 and used by a large efits under the Jones approach would include
coalition of health and social service orga- “restorative, removable, fixed, endodontic
nizations to foster change; and the effort of and selected implant (ie, two implants under a
the ADA facilitated through the work of PwC lower complete denture) as well as a spending
Consulting, about which little is yet publically cap ($1,500).”15 Level 2 benefits would cost
known. $31.58 PMPM in addition to the costs of the
The Jones approach incorporates several Level 1 benefits. The core global benefit would
important principles, many of which were cost $16.85 billion, not including any projected
articulated in the Santa Fe Group sympo- savings (eg, $12.20 billion in year 10) as esti-
sium on this topic.20 These principles include mated by the Avalere analysis described later
the incorporation of the dental benefit for in this section. Thus, even assuming the full
all participants in Medicare; that any dental benefit of cost reductions from reduced hos-
benefit should be incorporated into Medicare pitalizations and emergency room visits due
Part B (physician, outpatient hospital, home to the periodontal care provided, because the
health, and other services) and not developed global benefit proposed by Jones is available
as a separate benefit; that providers should be to all seniors, it is a net coster of $4.65 bil-
reimbursed at rates that are comparable to lion in year 10 ($16.85 billion minus $12.20
private dental insurance; and that a primary, billion). These net costs can be mitigated (or
global benefit should be provided to all par- not) based on additional premiums paid by
ticipants, along with an optional second-level Medicare recipients as in the current system.

364
Reordering Priorities in Medicare and Medicaid Policy

The initiative of the Dental Lifeline Net- heart disease, and stroke), Avalere estimated
work and the Center for Medicare Advocacy the savings that could be provided if a basic
(DLN-CMA initiative) seeks expanded dental dental benefit consisting of nonsurgical peri-
care that is medically necessary for Medicare odontal therapy was added to Medicare. Their
recipients under the currently authorized summary of the analysis was as follows:
Medicare legislation. This initiative basically
argues that the CMS already has the authority We estimate providing a periodontal disease
to expand the definition of medically necessary treatment benefit will produce a savings of
dental care by simple administrative action. At $63.5 billion over the period of 2016–2025
present, Medicare does pay for a very limited and should continue long term. This savings
amount of dental care, such as eliminating reflects new costs of approximately $7.2 bil-
oral infection in Medicare patients who are lion from covering periodontal treatment for
undergoing organ transplants. This initiative Medicare beneficiaries with one of the three
did not estimate the costs or the specific nature target chronic conditions. This new spending
of the expanded dental benefits that might be will be offset by an estimated $70.7 billion
provided, so the economic impact cannot be reduction in Medicare spending, largely
discussed at this time. Moreover, the petition to related to fewer hospitalizations and emer-
the CMS remains under review by the agency gency room visits.
many months after it was submitted.
The Avalere analysis12 is not actually an This analysis was actually quite conservative.
initiative but rather a report on which other For example, it assumed only a 5% uptake of
initiatives are based, in full or in part. For the new periodontal benefit in the first year,
example, the Jones approach acknowledges that growing to a 20% utilization of the benefit in
the assessment by Avalere would substantially year 10. Yet the financial outcome was a net
reduce the total cost of the Medicare dental positive beginning in the first year with $500
benefits in her proposal. Like the DLN-CMA million in savings and grew to a net benefit
initiative, the Avalere analysis limits benefits to of $12.2 billion in year 10. While some might
certain Medicare recipients with medical needs. argue that the Avalere analysis would only ben-
However, the Avalere analysis only provides efit a small portion of the senior population, the
dental benefits to Medicare recipients with number of people with periodontal disease and
three specific medical conditions (ie, diabetes, one of the three systemic conditions is actually
heart disease, and stroke), while the DLN-CMA quite large. Indeed, in follow-up to a meeting
initiative would presumably provide dental with the CBO, it was estimated that a scenario
benefits to any Medicare recipient with a med- like the one proposed by Avalere would have
ically necessary dental treatment requirement. the potential to benefit 19 million people.
By providing more limited dental benefits (eg, This is more people than have benefited from
diagnosis and nonsurgical periodontal care) either the Children’s Health Insurance Program
only to a more limited population , the Avalere (CHIP) or the Affordable Care Act (ACA) insur-
analysis easily has the best outcome from a ance pools. Therefore, efforts to characterize
purely economic perspective. Simply, it is a net the Avalere analysis as small in scope are inac-
cost saver, and a substantial one at that. curate. Moreover, the Avalere analysis based
Using data on the effect of periodontal care the dental benefit on costs of $825 for initial
on the number of hospitalizations and emer- treatment and $250 for biannual maintenance
gency department visits from the insurance visits. Thus, the program delivers more than
studies described earlier and data on Medicare two-thirds of a typical, private insurance dental
costs and the numbers of Medicare recipients benefit in year 1 ($1,500 per year) and one-third
who have the three signature diseases (diabetes, of a private dental benefit in subsequent years.

365
18 The Economic Impact of Periodontal Inflammation

The preventive techniques learned by seeking the public sector insured parallels that from the
the periodontal benefit will likely improve the private sector insured would provide substantial
patient’s other oral health problems. In addi- evidence to prompt the federal government to
tion, patients could elect to have other oral act on expanded oral care coverage.
health problems treated out-of-pocket once Finally, and perhaps most importantly, a
they develop a level of comfort with the den- large coalition of organizations guided by
tist who provides the basic periodontal service. Eric Berger, a principal in Liberty Partners in
Washington, DC, has developed a compelling
community statement on this issue.21 This
statement, which is supported by 70 signifi-
What Comes Next? cant organizations from the medical, dental,
social sciences, and patient advocacy sectors,
At the time of this writing, a number of col- is reprinted here:
laborative efforts are underway to affect “the
economic impact of periodontal inflammation” Community Statement on Medicare
as this chapter is titled. First, more effort must Coverage for Medically Necessary Oral
be applied to educate the CMS and Congress. I and Dental Health Therapies
have had the privilege of participating in three
meetings with the CMS on this topic and one The undersigned organizations are proud to
meeting with the CBO. At each meeting, the join in support of Medicare coverage for med-
CMS participants, who sometimes represented ically necessary oral/dental health therapies.
the highest levels in the agency, were attentive It is well established that chronic dis-
and interested in the subject. In fact, it is not a eases disproportionately impact Medicare
stretch to say that some of them were amazed beneficiaries and impose a substantial cost
by the data set. That said, the administration on the federal government. It is also well
has changed, as has the leadership of the CMS, established that untreated oral microbial in-
and new efforts will be required to rekindle the fections are closely linked to a wide range
interest in the enormous savings that could ac- of costly chronic conditions, including di-
crue to the Medicaid and Medicare programs abetes, heart disease, dementia, and stroke.
if the agency were to act to expand dental care In addition, oral diseases have been docu-
services, especially periodontal services. mented by researchers and medical specialty
Furthermore, as I write this, the CBO is societies as precluding, delaying, and even
reported to be scoring the cost/savings of a new jeopardizing medical treatments such as or-
dental benefit in Medicare. This benefit would gan and stem cell transplantation, heart valve
be similar to the one in the Avalere analysis repair or replacement, cancer chemothera-
and has the potential, for example, to cover pies, placement of orthopedic prostheses,
most of the costs of the CHIP program, which and management of autoimmune diseases.
Congress is desperate to approve. By the time Despite these factors, most Medicare ben-
you read this, we will know if this effort was eficiaries do not currently receive oral/dental
in fact successful. care even when medically necessary for the
To enhance the likelihood that Congress or the treatment of Medicare-covered diseases. In
CMS will act to add a dental benefit to Medicare, fact, Medicare coverage extends to the treat-
the Santa Fe Group is funding a new program ment of all microbial infections except for
to assess whether the Medicaid population in those relating to the teeth and periodontium.
New York State benefits from dental care in the There is simply no medical justification for
same way that those in the private insured pop- this exclusion, especially in light of the broad
ulation benefit. Confirmation that the data from agreement among medical specialists that such

366
What Comes Next?

care is integral to the medical management of dontists; American College of Rheumatology;


numerous diseases and medical conditions. American Dental Association; American Den-
Moreover, the lack of medically necessary oral/ tal Education Association; American Dental
dental care heightens the risk of costly medical Hygienists’ Association; American Diabetes
complications, increasing the financial burden Association; American Head and Neck Soci-
on Medicare, beneficiaries, and taxpayers. ety; American Kidney Fund; American Liver
At least six major insurance carriers Foundation; American Nurses Association;
offering dental plans provide enhanced peri- American Parkinson’s Disease Association;
odontal and preventive coverage to targeted American Psychiatric Association; American
enrollees with conditions such as diabetes, Public Health Association; American Society
heart disease, stroke, head/neck cancers, for Radiation Oncology; American Society of
and transplants. According to some reports, Clinical Oncology; American Society of Trans-
such coverage has realized important bene- plant Surgeons; American Thoracic Society;
fits, including markedly lower hospitalization Arthritis Foundation; Association of Dental
and emergency department admission rates Support Organizations; Association of State
as well as substantial cost reductions. On a and Territorial Dental Directors; California
further note, veterans getting care through Dental Association; Catholic Health Associa-
the Veterans Health Administration receive tion of the United States; Center for Medicare
medically adjunctive oral/dental treatment Advocacy; Children’s Dental Health Project;
in many instances when a dental diagnosis Crohn’s and Colitis Foundation of America;
affects their medical prognosis. These are Dental Lifeline Network; Dental Trade Alli-
all important steps forward, and medically ance; Eating Disorders Coalition; Epilepsy
necessary oral/dental healthcare including Foundation; Families USA; Head and Neck
periodontal treatment should be provided Cancer Alliance; Justice in Aging; Leukemia
in traditional Medicare as well. and Lymphoma Society; Lupus Foundation
The Medicare program and all its benefi- of America; Medicare Rights Center; Mental
ciaries should not be without the vital clinical Health America; National Alliance on Mental
and fiscal benefits of coverage for medically Illness; National Association of Area Agen-
necessary oral/dental health therapies. Given cies on Aging; National Association of Com-
the significant potential to improve health munity Health Centers; National Association
outcomes and reduce program costs, we urge of Dental Plans; National Council for Behav-
Congress and the Administration to explore ioral Health; National Kidney Foundation;
options for extending such evidence-based National Multiple Sclerosis Society; Nation-
coverage for all Medicare beneficiaries. al Network for Oral Health Access; Nation-
al Osteoporosis Foundation; National Rural
Signed by: AARP; Acuity Specialists; Health Association; National Stroke Asso-
American Academy of Maxillofacial Prosthet- ciation; Oral Health America; Pacific Den-
ics; American Academy of Periodontology; tal Services Foundation; Parkinson’s Founda-
American Association for Dental Research; tion; PEW Dental Campaign; Renal Physicians
American Association of Clinical Endocri- Association; Santa Fe Group; School-Based
nologists; American Association of Hip and Health Alliance; Society for Transplant So-
Knee Surgeons; American Autoimmune Re- cial Workers; Support for Persons with Oral,
lated Diseases Association; American Col- Head, and Neck Cancer; The Gerontological
lege of Emergency Physicians; American Col- Society of America; The Michael J. Fox Foun-
lege of Gastroenterology; American College dation; The Society for Thoracic Surgeons
of Physicians; American College of Pros­tho­

367
18 The Economic Impact of Periodontal Inflammation

This important statement, signed by an 9. Nasseh K, Vujicic M, Glick M. The relationship between
periodontal interventions and healthcare costs and utili-
almost unprecedented number of organizations
zation: Evidence from an integrated dental, medical,
from across the health care spectrum, will be and pharmacy commercial claims database. Health
used in the coming year in an effort to generate Econ 2017;26:519–527.
an administrative solution that leverages the 10. Cigna. Improved Health and Lower Medical Costs: Why
Good Dental Care Is Important. https://www.cigna.com/
power of periodontal care to reduce hospitaliza- assets/docs/life-wall-library/Whygooddentalcareisim-
tions and emergency room utilization. Coupled portant_whitepaper.pdf. Accessed 30 December 2017.
with many other efforts by these organizations 11. Chávez EM, Calvo JM, Jones JA. Oral Health and Older
and other groups, there is reason for some opti- Americans: A Santa Fe Group White Paper. http: //
santafegroup.org/wp-content/uploads/2017/
mism even in a political environment that is 06/Oral-Health-and-Older-Americans-A-Santa-Fe-
toxic, partisan, and struggling for resources. By Group-White-Paper.pdf. Published June 2017.
the time Dr Glick publishes the third edition Accessed 30 December 2017.
12. Avalere Health. Evaluation of Cost Savings Associated
of this book, I hope we will be able to describe
with Periodontal Disease Treatment Benefit. http: //
a wonderful success story of improved health pdsfoundation.org/downloads/Avalere_Health_Esti-
complemented by favorable economics. mated_Impact_of_Medicare_Periodontal_Coverage.
pdf. Accessed 30 December 2017.
13. Curro FA, Robbins DA, Naftolin F, Grill AC, Vena D, Terra-
cio L. Person-centric clinical trials: Defining the N-of-1

References clinical trial utilizing a practice-based translational


network. Clin Investig (Lond) 2015;5:145–159.
14. Hammond EC, Auerbach O, Kirman D, Garfunkel L.
1. Lamster IB. Oral conditions and systemic health. Effects of cigarette smoking on dogs. 1. Design of exper-
Presented at the Santa Fe Group Salon: Expanding Oral iment, mortality, and findings in lung parenchyma. Arch
Healthcare for America’s Seniors, Arlington, VA, 28–30 Environ Health 1970;21:740–753.
September 2016. https://www.youtube.com/ 15. Jones JA, Monopoli M. Designing a new payment model
watch?v=WE9-Agum1Y8. Accessed 30 December for oral care in seniors. Compend Contin Educ Dent
2017. 2017;38:616–624.
2. Bale BF, Doneen AL, Vigerust DJ. High-risk periodontal 16. Chávez EM, Hendre A. Clinical care for an aging popu-
pathogens contribute to the pathogenesis of atheroscle- lation. Compend Contin Educ Dent 2017;38:595–602.
rosis. Postgrad Med J 2017;93:215–220. 17. Calvo JM, Chávez EM, Jones JA. Financial roadblocks
3. Slavkin HC, Santa Fe Group. A national imperative: Oral to oral health for older adults. Generations 2016;40:
health services in Medicare [editorial]. J Am Dent Assoc 85–89.
2017;148:281–283. 18. Chávez EM, Calvo JM, Jones JA. Dental homes for
4. Alfano M. The seniors are coming! [editorial]. Compend older Americans: The Santa Fe Group call for removal
Contin Educ Dent 2017;38:586–588. of the dental exclusion in Medicare. Am J Public Health
5. American Lung Association. Health Effects of Smoking. 2017;107(suppl 1):S41–S43.
http: //www.lung.org/stop-smoking/smoking- 19. Chávez EM, Calvo JM, Jones JA. The Santa Fe Group
facts/health-effects-of-smoking.html. Accessed on 30 strategy: How Medicare can integrate health and oral
December 2017. care for older Americans. Generations 2016;40:94–99.
6. Mosen DM, Pihlstrom DJ, Snyder JJ, Shuster E. Assess- 20. Wilentz JB, Kleinman DV, Alfano MC. Meeting commen-
ing the association between receipt of dental care, tary: Santa Fe Group Salon Expanding Oral Healthcare
diabetes control measures and health care utilization. for America’s Seniors, September 28–30, 2016. J Am
J Am Dent Assoc 2012;143:20–30. Dent Assoc 2017;148:272–274.
7. Jeffcoat MK, Jeffcoat RL, Gladowski PA, Bramson JB, 21. Community Statement on Medicare Coverage for Med-
Blum JJ. Impact of periodontal therapy on general health: ically Necessary Oral and Dental Health Therapies. https:
Evidence from insurance data for five systemic condi- //cdn.nof.org/wp-content/uploads/2017/10/
tions. Am J Prev Med 2014;47:166–174. Medicare-Community-Statement.pdf. Ac­cessed 30
8. United Healthcare. Medical Dental Integration Study: December 2017.
March 2013. http: //www.unitedhealthgroup.com/~/
media/UHG/PDF/2013/UHC-Medical-Dental-
Integration-Study.ashx?la=en. Accessed 30 December
2017.

368
Index
Page numbers followed by “f” denote figures; cost effectiveness of, 330, 336
“t” tables; and “b” boxes. distant site infection prevention with, 329, 334
for infective endocarditis
American Heart Association recommendations,
329–330, 332–336
A current data regarding, 336–337
drug resistance concerns, 336
ACA. See Affordable Care Act.
history of, 329
ACTH. See Adrenocorticotropic hormone.
research studies about, 339–340
Actinomyces naeslundii, 183
support for and against, 335–336
Activated partial thromboplastin time, 187
issues regarding, 330–331
Activities of daily living, 109
primary, 329–330, 340
Acute monocytic leukemia, 259, 259f
secondary, 329–330, 340
Acute myelogenous leukemia, 271, 272f
Antibiotics
Acute myocardial infarction, 3
bacteria control using, 77
Acute-phase proteins, 89
prophylactic uses of. See Antibiotic prophylaxis.
Acute-phase response, 171–172
Antifungal agents, for candidiasis, 256
ACVD. See Atherosclerotic cardiovascular disease.
Anti-inflammation, 97
Acyclovir, 250t
Antimalarial agents, 261
Addison disease, 262
Antimicrobial oral hygiene treatment, in pneumonia,
Adenotonsillar hypertrophy, 122
202–203
Adipocytes, 93, 106, 108
Antiviral medications, 250t, 285
Adipocytokines, 115
Anxiety disorders, 305
Adipokines, 93
Aortic valve, 331f
Adiponectin, 93, 108
AP. See Aspiration pneumonia.
Adipose tissue, 125
Aphthous-like ulcers, 244–246
Adiposity, 106, 306
Apical periodontitis, 51
Adjuvant therapy, 271
aPTT. See Activated partial thromboplastin time.
ADLs. See Activities of daily living.
Arachidonic acid, 89
Adolescent pregnancy, 224
Aristotle, 4
Adrenocorticotropic hormone, 262
Aspergillosis, 251
Advanced glycation end products, 93
Aspergillus flavus, 251
Affordable Care Act, 366
Aspergillus fumigatus, 251
AFM. See Atomic force microscopy.
Aspiration, 63–64
AGEs. See Advanced glycation end products.
Aspiration pneumonia, 194–195, 197, 204
Aggregatibacter actinomycetemcomitans, 51, 58, 63,
Associations
65, 70, 97, 237
Bradford Hill criteria for, 5–6, 6b, 12
AIDS-associated linear gingivitis, 258
causation and, 361–363
Albuterol, 305
cause-and-effect interpretation of, 13
Alleles, 18
in randomized controlled trials, 34
Allogeneic transplantation, 280, 282
α level, 26–27 Asymptomatic viral shedding, 75
Atherogenesis, 65f, 95
Alternate hypothesis, 25
Atherosclerosis
Alzheimer’s disease, 68–69
description of, 72–73, 95
Amalgam tattoo, 260
oral infections as contributing factor, 164–165
American Dental Association Health Policy Institute,
periodontal pathogens and, 166
359
Atherosclerotic cardiovascular disease
American Heart Association antibiotic prophylaxis
at-risk population, 172–173
recommendations, 329–330, 332–336
endothelial dysfunction and, 174
Amoxicillin, 330, 336
epidemiology of, 169
Amphotericin B, 286
intervention trials, 169–174
Amyloid-beta, 68
mechanisms of, 165–168
Angiogenesis, obesity and, 108–109
periodontal intervention effects on, 173–174
Angular cheilitis, 256
periodontitis and, 169, 175–176
Anthropometry, 102
risk factors for, 139, 169
Antiangiogenic agents, 266
Atherosclerotic plaque, 63
Antibiotic prophylaxis
Atherothrombogenesis, 168f
cardiac conditions requiring, 330f
Atomic force microscopy, 349–350
compliance with, 330

369
I Index

Attribution, 11 Body fluids, microbiome in, 41


Autologous transplantation, 280 Body mass index, 102–103, 104t, 109–110, 114,
Avalere, 359, 364–365 119–120, 157f
ACVD. See Atherosclerotic cardiovascular disease. Body weight, masticatory efficiency and, 121
Bone diseases, primary, 263–264
Bradford Hill criteria, 5–6, 6b, 12–13, 14f–15f
B Breast cancer, 345, 345t
Burket, Lester W., 3
Bacteremia
Burning mouth syndrome, 268
infective endocarditis and, 331, 333b, 334, 335f
oral hygiene as risk for, 337
pathogenesis of, 66–67
periodontal status and, 200 C
tooth brushing and, 67, 177, 200, 332–334, 335f, CA15-3, 345
337 CA19-9, 346
transient, 89 CA125, 346
Bacteria. See also specific bacteria. CAL. See Clinical attachment loss.
core set of, 46–47 CAMBRA. See Caries management by risk
fungi and, 61 assessment.
gram-negative, 42–43 Campylobacter rectus, 217–218
gram-positive, 42–43 CAMs. See Cell adhesion molecules.
interspecies interactions, 60f Cancer. See also Squamous cell carcinoma.
in intracellular spaces, 41 breast, 345, 345t
modifiable changes to decrease, 61–63 colorectal, 236
in oral plaque, 54f deaths caused by, 231
on orthodontic appliances, 52–53 early detection of, 237–238
in peri-implant space, 40 Fusobacterium nucleatum and, 233–234
periodontal breakdown and, 39–40 gastric, 346
periodontal therapy effects on, 61 genetic susceptibility to, 232–233
at phylum level, 46, 47f–48f head and neck, 234–235
on removable dentures, 53 lung, 232, 235–236, 346, 357
in saliva, 49–50, 58f, 62f MiRNA as salivary biomarker for, 347
in subgingival plaque, 39, 56, 65 obesity as risk factor for, 232
supragingival, 51f–52f oral infections and, 238
Bacterial DNA, 70 ovarian, 346–347
Bacteroides gingivalis, 45 pancreatic, 234, 236–237, 352f
Base pair, 43 periodontal disease and
Behavioral Risk Factor Surveillance System, 149 biologic plausibility for, 233–234
Behçet disease, 244–245 observational studies regarding, 234–237
Benzocaine, 243t shared risk factors for, 231–233
Bergeyella, 216 prevention of, 237–238
Betamethasone, 243t salivary biomarkers for
Betel nut chewing, 61 description of, 345t, 345–348
Bidirectional causal relationship, 4 salivary exosome as, 351–353
Binary outcome event, 10 salivary proteomics applied to, 344–347
Biologic plausibility smoking as risk factor for, 232
for cancer and periodontal disease, 233–234 type 2 diabetes as risk factor for, 232
for chronic kidney disease and periodontal disease, Cancer treatment
183–184 options for, 272, 273f
for end-stage renal disease and periodontal disease, oral toxicities associated with
183–184 graft-versus-host disease, 254, 280, 282b,
for oral infection correlation 282–283
with adverse pregnancy outcomes, 214–219, 216f mucositis, 275f–278f, 275–280
with systemic health, 289 overview of, 273t–274t
Biomarkers Candida albicans, 53, 67
cancer, salivary exosomes as, 351–353 Candidal infection, 285–286, 286f
clinical outcomes and, 18 Candidiasis
Bisphosphonates-induced osteonecrosis of the jaw. See characteristics of, 255–256
Medication-related osteonecrosis of the jaw. erythematous, 255
Bite marks, 70 hyperplastic, 255
Blastomycosis, 251 pseudomembranous, 255, 255f, 286f, 286t
Blinding, 33 CAP. See Community-acquired pneumonia.
Blood pigmentations, 262–263 Capsaicin, 268
BMI. See Body mass index. Carcinoembryonic antigen, 345–346
BMS. See Burning mouth syndrome. Cardiovascular disease
Body fat atherosclerotic. See Atherosclerotic cardiovascular
distribution of, 114 disease.
measurements of, 102, 114 deaths caused by, 231

370
Index

diabetes mellitus as risk factor for, 137–138 Chloroquine, 261


diet and, 105 Chorioamnionitis, 70
inflammatory elements of, 95 CHRNA5-CHRNA3-CHRNA4, 304
oral infections and, 164–177 Chromatin, 296f
periodontitis and, 94–96 Chromatin immunoprecipitation sequencing, 298
risk factors for, 94, 320 Chromosomes, 43, 304
Caries management by risk assessment, 307 Chronic kidney disease
Caries risk assessment, 307 antibiotic therapy in, 187
Cariogenic bacteria, 70 definition of, 181
Carney complex, 262 dental management in, 186–188
Case reports, 36 end-stage renal disease progression of, 187
Case series, 36 estimated glomerular filtration rate in, 181
Case-control studies, 35–36 inflammatory burden in, 185
Cauliflower structure, 50, 55f periodontal therapy in
Causal relationships nonsurgical, 186
awareness of, 1 outcomes of, 186
bidirectional, 4 response of, 185–186
cyclic, 4 systemic outcomes of, 186
direct, 4 periodontitis in patients with
experimental studies for identifying, 11 biologic plausibility for, 183–184
hallmarks of, 2 conceptual model of, 184f
indirect, 4 epidemiology of, 181–182
necessary conditions for, 6–8, 7f, 8b–9b prevalence of, 182, 183t
reciprocal, 4 prevalence of, 181
reverse, 4 risk factors for, 182
sufficient conditions for, 6–8, 7f, 8b–9b, 15–16 summary of, 188
Causation. See also Cause. uremia in, 183–184
definition of, 3–5 Chronic obstructive pulmonary disease
frameworks for. See Causation frameworks. characteristics of, 195
instrumental variables and, 17f, 17–18 classification of, 196
Mendelian randomization and, 17f, 18–19, 19f description of, 164
reasons for caring about, 1–2 oral hygiene in, 203
studies on, 361–363 periodontal health in, 204
theories of, 4 periodontitis and, 201, 321
words that imply, 3 prevalence of, 195
Causation frameworks smoking as risk factor for, 195, 201, 321
Bradford Hill criteria, 5–6, 6b, 12–13, 14f–15f CI. See Confidence intervals.
contributory cause, 8 Cicatricial pemphigoid, 251
counterfactual, 10–11, 16, 18 CKD. See Chronic kidney disease.
description of, 5 CLASI-FISH. See Combinatorial labeling and spectral
Henle-Koch postulates, 5, 5b imaging fluorescence in situ hybridization.
necessary and sufficient conditions, 6–8, 7f, 8b–9b Clinical attachment loss, 141, 143, 185
probabilistic, 11 Clinical importance
sufficient-component, 8–9, 10f, 16 confidence interval and, 28–32
Cause. See also Causation. definition of, 24
contributory, 8 effect size and, 31
definition of, 3–5 evaluation of, 28–32
sufficient-component model of, 8–9, 10f, 16 minimal clinically important differences and, 24,
CBO. See Congressional Budget Office. 28, 31
CC chemokine receptor type 2, 107 Clinical outcomes, biomarkers and, 18
CEA. See Carcinoembryonic antigen. Clinical pathway programs, 150
Celiac disease. See Gluten-sensitive enteropathy. Clinical significance, 32
Cell adhesion molecules, 108 Clinical trials, 362
Cell-to-cell signaling, 77 Clobetasol, 243t
Centers for Medicare and Medicaid Services, 361 Clotrimazole, 256, 286
Cevimeline, 268 CMS. See Centers for Medicare and Medicaid
Chemokines, 88, 96 Services.
Chemotherapy, 275f–278f, 275–280 CMV. See Cytomegalovirus.
Children Coated tongue, 256f, 256–257
obesity in, 104, 119 Cohabitation, 59
physical activity in, 106 Cohort studies, 35, 37
pneumonia in, 194 Colony-forming units, 108
Children’s Health Insurance Program (CHIP), 365 Colorectal cancer, 236
Chlamydia pneumoniae, 184 Combinatorial labeling and spectral imaging
Chlorhexidine mouthwash fluorescence in situ hybridization, 50
for candidal infections, 256 Commensal-turned pathogen, 42
for medication-related osteonecrosis of the jaw, 287 Common complex diseases, 293
for oral hygiene in pneumonia patients, 202

371
I Index

Common risk factors approach, 316, 317f, 321–323, sugar-sweetened beverages as cause of, 118–119
326 systemic diseases and, 302
Community-acquired infective endocarditis, 332, 337 tooth loss secondary to, 148
Community-acquired pneumonia, 195 Dental examination, 149–150
Concurrent therapy, 271 Dental fear, 305
Confidence intervals, 28–32, 30t Dental health
Confounders, 4, 12, 14b–15b, 18, 32 acute myocardial infarction and, 3
Confounding, 33, 170b Medicare coverage for therapies, 366–367
Congressional Budget Office, 360, 366 Dental home concept, 223
Consolidation of Standards for Reporting Trials Dental implants, in diabetes mellitus, 40–41
(CONSORT) guidelines, 34 Dental Lifeline Network and the Center for Medicare
Continuous positive airway pressure, 122 Advocacy, 364
Contributory cause, 8 Dental plaque, gingivitis caused by, 257
Control group, 33 Descriptive studies, 36
COPD. See Chronic obstructive pulmonary disease. Desired effect size, 31
Corncob structures, 50, 55f Desquamative gingivitis, 251–252, 252f, 258
Coronary artery disease, 13 Destructive membranous periodontal disease, 260
Corticosteroids, for desquamative gingivitis, 251 Deterministic cause framework, 11
Corynebacterium, 54f–55f Dexamethasone, 243t
Counterfactual cause framework, 10–11, 16, 18 Diabetes mellitus
Counterfactual outcome, 10 burden of, 136–138
Cowden syndrome, 260 cancer risks associated with, 232
Coxsackievirus, 247 case definitions of, 137–138
CPAP. See Continuous positive airway pressure. complications of, 138
CRA. See Caries risk assessment. cost savings in, 151
C-reactive protein costs of, 138
coronary heart disease and, 18 criteria for testing for, 158b
description of, 89 definition of, 136–137
obesity and, 106, 115 dental examination referrals, 149–150
periodontal treatment effects on, 174 dental implants in, 40–41
periodontitis and, 94–95, 166t–167t diagnostic criteria for, 137t
preterm birth and, correlation between, 215, 219 economic burden of, 138
Crohn disease, 244 gestational, 137–138
Cross syndrome, 260 global prevalence of, 137
Cross-sectional studies, 35–36 glycemic control in, 143–144, 264
CRP. See C-reactive protein. guidelines for, 151–152, 152b–155b
CVD. See Cardiovascular disease. hemoglobin A1C test for, 93
Cyclic causal relationship, 4 hyperglycemia associated with, 137
Cyclic neutropenia, 245 interprofessional collaboration for, 150–152
Cyclooxygenases, 89 lifestyle changes for, 147
Cyclosporine-induced gingival hyperplasia, 259, 259f obesity and, 111
Cytokines office screening for, 146–148
in atherosclerotic plaque, 94–95 oral diseases and, 320
in inflammation, 88–89, 108, 215 periodontal disease and, association between
in periodontal infection, 218–219 description of, 138–139, 320
in periodontitis, 200 evidence for, 140–150
Cytomegalovirus, 68, 247 guidelines for, 151–152, 152b–155b
health care costs, 358–359
mechanisms linking, 139–140, 140f
D shared risk markers for, 139, 150
summary of, 152–155
Denosumab
periodontitis and, 92–94, 152b
medication-related osteonecrosis of the jaw caused
poorly healing extraction sockets in, 263
by, 266
prevalence of, 136–137
osteoporosis treated with, 266
risk assessment form for, 156f–157f
Dental care, in immunosuppressed patients, 289
screening for, 146–148
Dental care teams, 324
tooth loss and, 140–141, 148
Dental caries
type 2, 137, 139, 156f–157f, 232
in cancer therapy patients, 273t
Diabetic nephropathy, 182
caries management by risk assessment, 307
Diabetic neuropathy, 148–149
genetics of, 299, 302
Diabetic retinopathy, 138
genome-wide association studies of, 299–300
Diet
global prevalence of, 312, 313f
cardiovascular disease and, 105
metabolic syndrome and, 116
obesity and, 105
obesity and, 116–119
tooth loss effects on, 148
prevalence of, 312
Direct causal relationship, 4
risk assessment, 307
Disability, 109
socioeconomic factors, 312

372
Index

Discoid lupus erythematosus, 254–255 Estrogens, 261


Disease(s). See also specific disease. Evidence-based review system, 361–362
cardiovascular. See Cardiovascular disease. Exosomes, salivary
contributory causes of, 357 as cancer biomarkers, 351–353
oral. See Oral diseases. contents of, 350–351
periodontal. See Periodontal disease. definition of, 349
systemic. See Systemic diseases. discovery of, 349
Distant site infections, 329, 334 mRNA in, 351
DLN-CMA. See Dental Lifeline Network and the structure of, 349–350
Center for Medicare Advocacy. summary of, 353
DNA mutations, 233 Experimental trials, 32–35
Docosahexaenoic acid, 90 ExRNA, 348
Dose-dependent relations, 11 ExRNA Atlas, 348f
Double blinding, 33 External validity, 33
Drug resistance, 336 Extracellular DNA, 59
Drug-induced gingival hyperplasia, 259, 259f Extracellular RNA, 343
Drug-induced oral pigmentation, 261 Extracellular RNA Communication Consortium, 348
Drug-induced oral ulcers, 245–246 Extraction sockets, 263–264
Drug-induced osteonecrosis of the jaw. See
Medication-related osteonecrosis of the jaw.
Dual energy x-ray absorptiometry, 120
Dyclonine hydrochloride, 243t
F
Fallacy of affirming the consequent, 4
Dysbiosis, 46–47
False negative, 26, 27f
Dysgeusia, 273t
False positive, 26, 27f
Famciclovir, 250t
E FANTOM. See Function Annotation of the
Mammalian Genome.
Ecchymoses, 262–263 FDI World Dental Federation, 324, 325
Edema, 86 FESEM. See Field emission scanning electron
Effect modifiers, 12, 14b–15b microscopy.
Effect size, 31 Fibromatosis gingivae, 260
Eicosapentaenoic acid, 90 Fibronectin, 200
Emphysema, 201 Fibrous cap, 94
ENCODE project, 294, 295f Field emission scanning electron microscopy, 349
Endothelial cells, 108 Filifactor alocis, 65
Endothelial dysfunction, 174 FISH. See Fluorescence In Situ Hybridization.
Endotoxins, 71 Fluconazole, 256, 286
End-stage renal disease Fluocinolone, 243t
antibiotic therapy in, 187–188 Fluorescence In Situ Hybridization, 50, 51f–52f, 56f
chronic kidney disease progression to, 187 Focal infection theory, 3, 87
description of, 182 Focus of infection, 2
inflammatory burden in, 185 Function Annotation of the Mammalian Genome, 294
periodontal therapy in Fungal infections, 250–251, 273t
nonsurgical, 186 Fungal microbiome, 49
outcomes of, 186 Fungemia, 67
response of, 185–186 Fungi, 61, 68
systemic outcomes of, 186 Fusobacterium, 216–217
periodontitis in patients with Fusobacterium nucleatum
biologic plausibility for, 183–184 antibiotics for, 65
epidemiology of, 182 cancer and, 233–234, 236
prevalence of, 182, 183t as commensal-turned pathogen, 42
summary of, 188 description of, 65
uremia in, 183–184 illustration of, 54f
Enterococcus faecalis, 56 in peri-implantitis, 51
Epidemiologic research, 32 in pneumonia, 198
Epidemiologic studies, 18 in pregnancy, 97
Epigenetics, 296
Epstein-Barr virus, 75–76, 247
Epulis, 213, 214f
ERCC. See Extracellular RNA Communication
G
Galen, 2
Consortium.
Gastric cancer, 346
Erythema areata migrans. See Migratory glossitis.
Gastritis, 76
Erythema multiforme, 248–250
Gastrointestinal mucositis, 277
Erythematous candidiasis, 255
GDM. See Gestational diabetes mellitus.
Escherichia coli, 56
Geisinger system, 110
ESRD. See End-stage renal disease.
Gene, 43
Estimated glomerular filtration rate, 181, 187

373
I Index

Gene-Lifestyle Interactions and Dental Endpoints Gram-negative bacteria, 42–43


Consortium, 299, 301, 306 Gram-positive bacteria, 42–43
Genetic variant, 18 Granuloma, pregnancy, 213, 214f
Genetics Graphite, 260
cancer risks associated with, 232–233 GWASs. See Genome-wide association studies.
clinical practice and, 306–308
data storage issues, 308
of dental caries, 299
oral diseases and, 298–306
H
Haemophilus influenzae, 195
of periodontal disease, 299–300
Hairy tongue, 256f, 256–257
Genome, 43–44
Hand, foot, and mouth disease, 247
Genome-wide association studies
HapMap Project, 293–294
of dental caries, 299–300
Hashimoto thyroiditis, 254
description of, 26, 232, 294, 298
Hay-Wells syndrome, 304
findings of, 300–301
HCAP. See Health care–associated pneumonia.
Genomic science
Head and neck squamous cell carcinoma, 277, 278f,
advances in, 294–298
346
ENCODE project, 294, 295f
Health care–associated pneumonia, 194
Human Genome Project, 43, 293–294
Health in All Policies, 323, 326
International HapMap Project, 293–294
Health Promoting Schools Network, 317
1000 Genomes project, 293–294
Health promotion, 322
oral disease and systemic disease connections,
Heart disease, 231
301–306
Heavy metal pigmentation, 261
overview of, 292–293
Hedgehog, 54f
Genotyping, 298
HeLa genome, 66f
Geographic tongue. See Migratory glossitis.
Helicobacter pylori, 52, 76–77, 233
Gestational diabetes mellitus, 137–138
Hematopoietic stem cell transplantation
Gingiva
autologous, 280
acute leukemia involvement of, 272f
graft-versus-host disease secondary to, 254, 280,
bleeding of, obesity and, 113
282, 282b
hyperplasia of
nonherpes viral infection after, 285
acute leukemia as cause of, 259f, 259–260
oral complications of, 282b
drug-induced, 259, 259f
oral mucositis secondary to, 275
hereditary causes of, 260
Hemoglobin A1C , 93, 144, 147
pathophysiology of, 258–259
Henle-Koch postulates, 5, 5b
plaque-induced, 259
Hereditary gingival fibromatosis, 260
syndromic causes of, 260
Hereditary hemochromatosis, 263
pregnancy-related inflammation of, 213
Herpangina, 247
Gingival crevicular fluid
Herpes simplex virus
inflammation and, 115, 334
antiviral agents for, 250t
oral microbiome in, 61
clinical presentation of, 248t
tumor necrosis factor α levels in, 112
description of, 75
Gingivitis. See also Periodontal disease.
in myelosuppressed patients, 284–285, 285f
Alzheimer’s disease and, 68
nerve pathways, 68
description of, 39
oral ulcers caused by, 246–247, 249
desquamative, 251–252, 252f, 258
recurrent, 249f
HIV/AIDS-associated linear, 258
HiAP. See Health in All Policies.
ligneous, 260
High-throughput assays, 297
necrotizing ulcerative, 258
High-throughput mass spectrometry, 344
plaque-induced, 257
High-throughput RNA sequencing, 351
plasma cell, 258
High-throughput sequencing, 348
pregnancy, 213
Hippocrates, 2, 86
Glaucoma, 69
Histoplasmosis, 250–251
Glioblastoma microvesicles, 351
HIV
Global Burden of Diseases, Injuries, and Risk Factors,
linear gingivitis associated with, 258
312
oral complications in, 245
Gluten-sensitive enteropathy, 244
HLA-B51, 244
Glycated hemoglobin, 52
Homeostasis, 90
Glycemic control
HOMINGS, 57
in diabetes mellitus, 263
Hospital-acquired pneumonia, 194–196, 206
nonsurgical periodontal treatment effects on,
Host-parasite interaction, 90
144–146, 145t
HSCT. See Hematopoietic stem cell transplantation.
periodontitis effects on, 143–144, 320
HSV. See Herpes simplex virus.
Glycemic status, 146
Human coronary artery endothelial cells, 64
Glypican-1, 351
Human genome, 43–44, 295f, 297f
Gonadal steroids, 212
Human Genome Project, 43, 293–294
Graft-versus-host disease, 254, 280, 282b, 282–283
Human herpesvirus 1, 68

374
Index

Human herpesvirus 2, 68 oral hygiene in


Human herpesvirus 4, 75 description of, 334
Human herpesvirus 5, 247, 248t gingival disease and, 337–339, 338f
Human immunodeficiency virus. See HIV. oral infection and, 3
Human microbiome, 44–45 oral pathogens and, 329
Human Microbiome Project, 44–45, 45f, 77 time of onset, 336
Human Oral Microbiome Database, 45–46 types of, 332
Human papillomavirus, 66, 66f, 73–75, 235 vegetations, 331, 331f
Hunter, William, 87 Inflammation
Hydroxyeicosatetraenoic acids, 89 adiponectin in, 108
Hyperglycemia in chronic kidney disease, 185
in diabetes mellitus, 137 cytokines associated with, 108
hyposalivation caused by, 62 description of, 88–90
oral manifestations of, 148–149 in end-stage renal disease, 185
periodontal disease and, 139 macrophages in, 107
periodontal tissue affected by, 140 mediators of, in obesity, 115–116
periodontitis and, 139–141 in obesity, 106–108, 107f, 115–116
tooth eruption affected by, 140 periodontal. See Periodontal inflammation.
tooth loss affected by, 140 periodontal disease and, 97
Hyperplastic candidiasis, 255 in periodontitis, 139
Hypohidrotic ectodermal dysplasia, 304 in pneumonia, 194
Hyposalivation, 62, 266–268 systemic actions of, 92f
Hypothesis testing, 26–28, 27f Information bias, 33
Hypothetical condition, 10 Innate immunity, 88
Hypoxia, 109 Instrumental variables, 17f, 17–18
Hypoxia-inducible factor-1, 109 Insulin resistance, 112, 116
Insulin-like growth factor 2, 218
Insurance industry
I economic savvy of, 359–361
studies by, 358–359
IARC. See International Agency for Research on
Integrated oral health practice patient visit, 124f
Cancer.
Interferon- τ, 96
Immunoglobulin G, 216–217
Interleukin-1, 307
Immunoglobulin M, 216–217
Interleukin-2, 218
Immunosuppressed patients
Interleukin-6, 106, 108, 115, 214, 307
dental care for, 289
Interleukin-10, 218
oral complications in
Interleukin 1ß, 88, 97
herpes simplex viral infection, 284–285, 285f
International Agency for Research on Cancer, 232
infections, 283–286, 289
International HapMap Project, 293–294
mucositis, 275f–278f, 275–280
Interpersonal travel, 63
summary of, 288
Interprofessional education and collaboration,
oral health in, 289
324–325
Immunosuppressive agents, 243t
Intrauterine growth restriction, 211
Incidence, 35
IVs. See Instrumental variables.
Indirect causal relationship, 4
Infection. See also Oral infections.
in Alzheimer’s disease, 68
definition of, 42 J
diabetes mellitus as cause of, 263 Jaw
in immunosuppressed patients, 283–286 medication-related osteonecrosis of. See
in myelosuppressed patients, 283–286 Medication-related osteonecrosis of the jaw.
pathogen and, 42 metastatic tumor to, 266
Infective endocarditis Jones approach, 364
antibiotic prophylaxis for Junk DNA, 296
American Heart Association recommendations,
329–330, 332–336
current data regarding, 336–337
drug resistance concerns, 336
K
Keystone pathogen, 42
history of, 329
KRT75, 302
research studies about, 339–340
KRT6A, 302
support for and against, 335–336
KRT6B, 302
of aortic valve, 331f
bacteremia and, 334
community-acquired, 332, 337
definition of, 331
L
dental procedures, 334 Lateral gene transfer, 66
medically related, 332 Laugier-Hunziker syndrome, 262

375
I Index

Leptin Metagenomics, 43
adipocyte secretion of, 108 Metastatic tumors, to jaw, 266
in obesity, 115 Methotrexate, 246
skeletal growth affected by, 121 Methylome-wide association studies, 301
Leukemia Microbiome. See also Oral microbiome.
acute monocytic, 259, 259f in body fluids, 41
acute myelogenous, 271, 272f body site, 78f
gingival hyperplasia secondary to, 259f, 259–260 definition of, 44
Leukoplakia, 253, 253f description of, 44–45
Life expectancy, 180 functions of, 44
Ligneous gingivitis, 260 fungal, 49
Lipopolysaccharides, 71–72, 88, 107, 218 in saliva, 56–58, 57f
Lipoxins sampling sites for, 45f
A4, 98t smoking effects on, 41, 57, 61–62
description of, 90 Microvesicles, salivary, 351
Lipoxygenases, 89 Migratory glossitis, 257, 257f
Literature Mild periodontitis, 136t
assessment of, 24 Miller, Willoughby Dayton, 2
oral infection–systemic health link in, 25f, 357 Minimal clinically important differences, 24, 28, 31
Low birth weight, 211, 212b MiRNA, 347, 351
Low-density lipoproteins, 94 Mitral valve prolapse, 332
Lung cancer, 232, 235–236, 346, 357 Moderate periodontitis, 136t
Lung resistance protein, 345 MODY. See Maturity-onset diabetes of the young.
Lupus erythematosus, 254–255 Monogenic diseases, 293
Lymphonodular pharyngitis, 247 MRONJ. See Medication-related osteonecrosis of the
jaw.
mTOR inhibitors. See Mammalian target of
M rapamycin inhibitors.
MUC7, 305
Macrophages, 107, 109, 139
Mucopeptide, 43
MAGIC syndrome, 245
Mucormycoses, 250–251
Mammalian target of rapamycin inhibitors
Mucositis, oral, 245, 246f, 275f–278f, 275–280
description of, 246, 279–280, 280f
Mucous membrane pemphigoid, 251, 252f
stomatitis associated with, 246
Multidimensional protein identification technology
Maresin-1, 98t
(MudPIT), 350
Masking, 33
Murein, 43
Massively parallel sequencing, 297
Murray-Puretic-Drescher syndrome, 260
Masticatory efficiency, 121–122
Mycobacterium tuberculosis, 184
Matrix metalloproteinases, 95, 97
Mycobiome, 49
Maturity-onset diabetes of the young, 137
Myelosuppressed patients, oral complications in
McCune-Albright syndrome, 262
herpes simplex viral infection, 284–285, 285f
MCID. See Minimal clinically important differences.
infections, 283–286
Medicaid, 360, 363–366
mucositis, 275f–278f, 275–280
Medically necessary dental care, 364–367
Myocardial infarction, 3
Medically related infective endocarditis, 332
Medicare, 360, 363–366
Medication-induced oral pigmentation, 261
Medication-related osteonecrosis of the jaw N
antiangiogenic agents as cause of, 266 Naegeli syndrome, 304
bisphosphonates as cause of, 264–266 NAMPT, 299
clinical presentation of, 264–265, 265f, 265t National Cholesterol Education Program Adult
definition of, 262, 287 Treatment Panel III, 110
denosumab as cause of, 266 National dental organizations, 326
description of, 264–266, 273t National Health and Nutrition Examination Survey.
extractions in, 265 See NHANES.
illustration of, 265f, 287f National Institutes of Health, 293, 307
management of, 287–288 NCD Alliance, 324, 325b
prevention of, 265–266, 287 NCEP/ATPIII. See National Cholesterol Education
Prolia as cause of, 266 Program Adult Treatment Panel III.
staging criteria for, 288b Necessary and sufficient condition cause framework,
Melanin, 261–262 6–8, 7f, 8b–9b, 15–16
Melanotic macules, 262 Necessary conditions, 6–8, 7f, 8b–9b, 15–16
Mendelian randomization, 17f, 18–19, 19f Necrotizing ulcerative gingivitis, 259
Mendel’s law of segregation, 18 Neoadjuvant therapy, 27`
Metabolic syndrome Neonatal disorders
dental caries and, 116 low birth weight, 211, 212b
obesity and, 110–111 preterm birth, 210–211, 211f, 212b, 215, 219
periodontal disease risks, 116 Net coster, 364

376
Index

Neurofibromatosis, 262 inflammatory mediators, 115–116


Neuroinflammation, 68 longitudinal studies of, 113–114
Neutropenia, 245 Mendelian randomization on, 17f, 19
Neutrophil extracellular traps, 77 treatment effects, 114–115
Neutrophils, 77 periodontitis and, 111–113, 112f, 123
Next-generation sequencing, 297 physical activity for prevention of, 106
NHANES, 120, 135, 141, 182, 192 prevalence of, 102, 103f, 104–105, 151
Nifedipine-induced gingival hyperplasia, 259, 259f skeletal maturation in, 121
Nikolsky sign, 251 sleep apnea associated with, 111, 122–123
95% confidence interval, 28–29 sugar-sweetened beverages as cause of, 105–106,
No effect, 29 118–119
Noncommunicable diseases summary of, 123–124
description of, 311, 314 tooth eruption and movement affected by, 119–121
global impact of, 318f, 318–319 tooth loss and, 121
interventions to prevent, 316 treatment of, 124
oral diseases and, 320–321 upper airway structure affected by, 122
World Health Organization and Observational studies
Global Action Plan for the Prevention and case-control studies, 35–36
Control of Noncommunicable Diseases cohort studies, 35, 37
2013–2020, 316, 319 cross-sectional studies, 35–36
statement by, 318–319 on dental health–acute myocardial infarction link,
Nonhealing extraction sockets, 263–264 3
Not-for-profit organizations, 360 description of, 1–2, 35
Nuclear factor κ B, 88 randomized controlled trials versus, 1–2
Null hypothesis, 25–27 Obstructive sleep apnea, 122–123
Null value, 29, 31 Odds ratio, 34, 36
Nutrition, tooth loss effects on, 148. See also Diet. OHiAP. See Oral Health in All Policies.
Nystatin, 286 Older adults
periodontal disease in, 180
pneumonia in, 198–199
O 1000 Genomes project, 293–294
Operational taxonomic units, 47, 48f
OAT. See Oral appliance therapy.
Oral appliance therapy, 122–123
Obesity
Oral cancer, 91, 307–308
angiogenesis and, 108–109
Oral cavity metastases, 266
assessment of, 102–104
Oral diseases
body mass index, 102–103, 104t, 109–110, 114,
challenges for, 312
157f
contributory causes of, 357
cancer risks associated with, 232
diabetes mellitus and, 320
in children, 104, 119
downstream interventions for, 317
chronic disease associated with, 110
genetics of, 298–306
clinical considerations regarding, 125
global burden of, 312–315, 313f
consequences of, 109–111
global prevalence of, 313f
definition of, 102
high risk approaches to, 317
dental caries and, 116–119
noncommunicable diseases and, 320–321
diabetes mellitus and, 111
prevention of, 315f, 316
diet and, 105
quality of life affected by, 312
disability caused by, 109
social determinants involved in, 323
disorders associated with, 102, 123
systemic diseases and
economic impact of, 110
common risk factors between, 311, 316, 317f,
ethnicity and, 104
321–323, 326
etiology of, 105–106
genomic connection between, 301–306, 303f
genetic predisposition, 105
shared molecular basis between, 306
gingival bleeding and, 113
summary of, 325–326
global prevalence of, 103f, 104–105
upstream interventions for, 317
inflammation in, 106–108, 107f
Oral dysesthesia syndrome, 268
insulin resistance in, 112, 116
Oral epithelial cells, 56
masticatory efficiency and, 121–122
Oral health
measures of, 113
common risk factors approach for, 316, 317f,
metabolic syndrome and, 110–111
321–323
morbidity and mortality of, 109–110
general health and, 319
obstructive sleep apnea associated with, 122–123
global
oral infections and, 125
inequalities in, 312–315, 314f
pathophysiology of, 106–109
interventions to improve, 315–318
periodontal disease and
in immunosuppressed patients, 289
conceptual analysis of, 123
integrated strategy for, 321–323
cross-sectional clinical studies of, 111–113
oral health professionals as advocates for, 323–324
description of, 111, 123

377
I Index

in pregnant adolescents, 224 future travels, 77–78


social determinants of, 316, 316f, 323 Helicobacter pylori, 76–77
Oral health care professionals herpes simplex virus, 75
description of, 311 human papillomavirus, 73–75
health management involvement by, 324 human transfer of, 69–71
oral health advocacy by, 323–324 indirect transfer of, 70–71
Oral Health in All Policies, 323 interpersonal methods, 69–71
Oral hygiene intrapersonal methods, 63–69
antimicrobial, in pneumonia, 202–203 nerve pathways, 68
in chronic obstructive pulmonary disease, 203 pacifier transfer of, 71
gingival disease and, 337 salivary transfer, 70
in infective endocarditis, 334 travel options, 63–71
periodontal disease and, 338f travelers, 71–77
Oral infections. See also Infection. Oral mucositis, 245, 246f, 275f–278f, 275–280
adverse pregnancy outcomes caused by, 226 Oral pain, 273t, 279
atherosclerosis and, 164–165 Oral sepsis, 87
cancer and, 238 Oral squamous cell carcinoma, 345, 353
cardiovascular disease and, 164–177 Oral ulcers
fungal, 273t aphthous-like ulcers, 243–245
in immunosuppressed patients, 289 deep fungal infections as cause of, 250–251
obesity and, 125 description of, 242
pulmonary diseases and, 205 drug-induced, 245–246
systemic diseases and, 165 erythema multiforme, 248–250
tumor growth and, 238 immunosuppressive agents for, 243t
viral, 273t recurrent aphthous stomatitis, 242–243, 244f
Oral infection–systemic health link. See also Oral- traumatic, 242
systemic connections. vesiculobullous disease as cause of, 251–253,
benefits of establishing, 20 252f–253f
Bradford Hill criteria applied to, 12–13, 14f–15f viral infections as cause of, 246–247, 248t, 250t
counterfactual cause framework applied to, 16 Oral-systemic connections. See also Oral infection–
history of, 2–3 systemic health link.
literature published regarding, 25f, 357 inflammation in. See Inflammation.
necessary and sufficient condition cause framework overview of, 86–88
applied to, 15–16 summary of, 99
position statements on, 362–363 Oropharyngeal cancer, 321
probabilistic framework applied to, 16 Orthodontic appliances, 52–53
research on, 2 Osteitis deformans. See Paget disease of bone.
salivaomics, 354f. See also Salivaomics. Osteonecrosis of the jaw, medication-related. See
salivary biomarkers, 342 Medication-related osteonecrosis of the jaw.
salivary-exosomics, 349–353, 354f Osteopetrosis, 264
sufficient-component cause framework applied to, Osteoporosis, 266
16 Osteoradionecrosis, 273t
Oral lichen planus, 253–255, 254f Ovarian cancer, 346–347
Oral microbiome Overweight, 104
core, 46–50
definition of, 38
distribution of, 50–59
dynamic nature of, 59–63
P
P value, 26–28, 28t, 30t
geographic diversity, 58–59
Paget disease of bone, 263–264
human, 45–46
Pain, oral
intraoral diversity of, 50–56
management of, 279
modifiable changes to, 61–63
topical medications for, 243t
periodontal pockets, 50
Palifermin, 277
phylotypes in, 47f
Panc02 cells, 352, 352f
tooth surfaces, 50
Pancreatic cancer, 234, 236–237, 352f
traveling
Parotid gland, 342f, 342–343
air transfer of, 71
Pathogen, 42
Alzheimer’s disease, 68–69
Pemphigus vulgaris, 252, 252f
antibiotics for, 77
Peptidoglycan, 43
aspiration, 63–64
Periapical periodontitis, 50–51
atherosclerosis, 72–73
Peri-implant space, 40
bloody entryways, 67
Peri-implantitis, 40, 51, 52f
bloody travel vehicles, 67
Periodic fever, aphthous stomatitis, pharyngitis, and
cell invasion, 64–66
adenitis syndrome, 245
destinations of, 63
Periodontal disease. See also Gingivitis; Periodontitis.
direct transfer, 66, 69–70
burden of, 135–136
fungi, 68
cancer and biologic plausibility for, 233–234

378
Index

observational studies regarding, 234–237 during pregnancy, 222–224


shared risk factors for, 231–233 preterm birth affected by, 221t
characteristics of, 92 Periodontitis. See also Periodontal disease.
description of, 90–91 Alzheimer’s disease and, 68
destructive membranous, 260 animal studies of, 98t
diabetes mellitus and, association between atherosclerotic cardiovascular disease and, 169,
description of, 138–139, 146, 321 175–176
evidence for, 140–150 atherothrombogenesis and, 168f
guidelines for, 151–152, 152b–155b bacteria that cause, 46, 53f
health care costs, 359–360 cardiovascular disease and, 94–96
mechanisms linking, 139–140, 140f case definitions of, 135, 136t
shared risk markers for, 139, 150 chronic obstructive pulmonary disease and, 201,
summary of, 152–155 321
genetic component of, 232 contributory factors for, 193
genetics of, 299–300 costs associated with, 193
historical descriptions of, 3 C-reactive protein and, 94–95, 166t–167t
host-parasite interaction, 90 cytokine levels in, 200
hyperglycemia and, 139 definition of, 135, 170b
inflammation and, 97 description of, 39, 75, 90
lung cancer and, 235–236 diabetes mellitus and, 92–94, 152b
in myelosuppressed patients, 284f etiology of, 92
obesity and genetic causes of, 303
conceptual analysis of, 123 glycemic control affected by, 143–144, 321
cross-sectional clinical studies of, 111–113 hyperglycemia and, 139–141
description of, 111, 123 inflammation in, 139
inflammatory mediators, 115–116 mild, 136t
longitudinal studies of, 113–114 moderate, 136t
Mendelian randomization on, 17f, 19 obesity and, 111–113, 112f, 123
treatment effects, 114–115 periapical, 50–51
treatment effects of, 114–115 pneumonia and, 197–201
in older adults, 180 pregnancy and, 96–97, 219–222
oral hygiene and, 338f prevalence of, 135–136, 136f, 180, 193
smoking as risk factor for, 112, 234 pulmonary diseases and
systemic conditions and, 92–97 epidemiologic association between, 196–197
treatment of mechanisms of, 197–201, 204
delivery of, 171–172 summary of, 204–206
obesity effects on, 114–115 risk factors for, 169, 176, 193
Periodontal infections, 283–284, 284f risk indicators for, 40
Periodontal inflammation. See also Inflammation. severe, 136t
adverse pregnancy outcomes associated with, 216f systemic diseases and, 170b
description of, 91, 95 tooth loss secondary to, 148
economic impact of, 357–368 tumor necrosis factor α levels in, 115
Periodontal medicine, 180, 300 waist circumference and, 113, 114f
Periodontal pathogenic bacteria, 39–40 Periodontitis and vascular events, 173
Periodontal pockets, 50, 135 Periopathogens, 166–167
Periodontal risk assessment model, 307 Peritonitis, 98t
Periodontal risk calculator, 307 Peroxisome proliferator-activated receptor gamma,
Periodontal risk predictors, 307 108–109
Periodontal therapy Petechiae, 262–263
atherosclerotic cardiovascular disease affected by, Peutz-Jeghers syndrome, 262
173–174 PGN. See Progranulin.
in chronic kidney disease Pharmacogenomics, 308
nonsurgical, 186 Phenome-wide association study, 301
outcomes of, 186 Phenytoin-induced gingival hyperplasia, 259
response of, 185–186 Physical activity, for obesity prevention, 106
systemic outcomes of, 186 Pigmented lesions, 260–263
C-reactive protein concentrations affected by, 174t Pilocarpine, 267
description of, 61, 124 Piwi-interacting RNA, 347–348, 351
in end-stage renal disease Placebo group, 33
nonsurgical, 186 Placental growth factor, 218
outcomes of, 186 Plaque
response of, 185–186 gingival hyperplasia caused by, 259
systemic outcomes of, 186 gingivitis caused by, 257
insurance studies regarding, 358–359 subgingival. See Subgingival plaque.
nonsurgical, glycemic control affected by, 144–146, supragingival, 55f
145t Plasma cell gingivitis, 258
periodontal pathogen dissemination during, 200 Platelet aggregation, 166

379
I Index

Platelet/endothelial cell adhesion molecules, 108 Probing pocket depth, 141, 143, 185, 196
Pleiotropy, 301–302, 303f Progranulin, 115
Pneumonia Proinflammatory cytokines, 89, 95
antimicrobial oral hygiene treatment in, 202–203 Prolia, 266
aspiration, 194–195, 197, 204 Prostaglandin E 2 , 89, 215
childhood, 194 Protectin D1, 98t
community-acquired, 195 Pseudogenes, 295
definition of, 194 Pseudomembranous candidiasis, 255, 255f, 286f, 286t
health care–associated, 194 Pseudomonas aeruginosa, 198–199
hospital-acquired, 194–196, 206 Psoralen and ultraviolet A therapy, 283
inflammation in, 194 Puberty, 119
in older adults, 198–199 Pulmonary aspiration, 63
pathogens that cause, 195, 198–200 Pulmonary diseases
periodontal health in, 204 chronic obstructive pulmonary disease. See Chronic
periodontitis and, 197–201 obstructive pulmonary disease.
ventilator-associated, 194, 198, 202–203, 206 clinical considerations regarding, 205
Polymorphonuclear leukocytes, 88, 93 definition of, 196
Poorly healing extraction sockets, 263–264 overview of, 193
Pores of Kohn, 195 periodontitis and
Porphyromonas gingivalis epidemiologic association between, 196–197
atherosclerosis and, 165–167 mechanisms of, 197–201, 204
bacteremia and, 67 summary of, 204–206
cell invasion/penetration by, 41, 64 pneumonia. See Pneumonia.
classification of, 45, 46b PWAS. See Phenome-wide association study.
illustration of, 46f Pyogenic granuloma, 213
nerve transport by, 68 Pyostomatitis vegetans, 244
pancreatic cancer and, 234
in peri-implantitis, 51
periodontitis caused by, 95, 198
in pregnancy, 97, 214
R
Radiation therapy
salivary transfer of, 70
hyposalivation caused by, 267
sleep pattern disturbances caused by, 69
salivary gland damage caused by, 267–268
Potential outcome, 10
Randomized controlled trials
PPD. See Probing pocket depth.
advantages of, 32–33
PRA model. See Periodontal risk assessment model.
association measures in, 34
PRC. See Periodontal risk calculator.
blinding in, 33
Preadipocytes, 108
control group in, 33
Precision medicine, 39
of dental treatment during pregnancy, 223–225
Prediabetes, 137t, 137–138, 146–147, 155
description of, 1
Preeclampsia, 211
disadvantages of, 33–34
Pregnancy
ethical concerns for, 34, 37
adolescent, 224
external validity issues, 33
adverse outcomes, oral infection correlation with
hallmark of, 32
biologic plausibility of, 214–219, 216f
limitations of, 1–2
description of, 210–211, 226
Mendelian randomization versus, 19f
indirect pathway of, 218–219
observational studies versus, 1–2
periodontal inflammation, 216f
randomization in, 32
gingival inflammation during, 213
statistical significance of, 31
granulomas during, 213, 214f
RCTs. See Randomized controlled trials.
oral infections during, 210, 226
Receptor activator of NFκ B ligand, 88
periodontal health during, 210, 226
Reciprocal causal relationship, 4
periodontal therapy during, 222–224
Recurrent aphthous stomatitis, 242–243, 244f
periodontal tissues affected by, 211–214
Relative risk, 34–35
periodontitis and, 96–97, 219–222
Removable dentures, 53
preterm birth, 210–211, 211f, 212b, 215, 219
Renal diseases
sex steroids during, 212–213
chronic. See Chronic kidney disease.
Pregnancy gingivitis, 213
end-stage. See End-stage renal disease.
Preterm birth
overview of, 181–182
C-reactive protein and, 215, 219
Renin-angiotensin blockers, 182
description of, 210–211, 211f, 212b
Research
periodontal treatment effects on, 221t
hypothesis-driven, 25
Prevotella intermedia, 198, 214
purpose of, 25
Prevotella nigrescens, 214
Resolution, of inflammation, 97
Primary antibiotic prophylaxis, 329–330, 340
Resolvin D1, 98t
Primary bone diseases, 263–264
Resolvin D2, 98t
Primary medical care, 322
Resolvin E1, 98t
Probabilistic cause framework, 11, 16
Retrospective cohort study, 35

380
Index

Retrospective studies, 358 Skeletal-dental age, 120


Reverse causal relationship, 4 Sleep apnea, 111, 122–123
Rheumatic fever, 332 Small nucleolar RNA, 351
Rheumatoid arthritis Smoking
historical descriptions of, 2–3 cancer risks associated with, 232
leptin in, 108 chronic obstructive pulmonary disease caused by,
Ribosomal RNA, 39 195, 201, 321
Risk factors lung cancer and, 357
for atherosclerotic cardiovascular disease, 169 microbiome affected by, 41, 57, 61–62
for periodontitis, 169, 176 periodontal disease and, 112, 234
RNA genes, 296 “SNP chips,” 298
RNA sequencing, 301 Social determinants of health, 316, 316f, 323
Rutherford syndrome, 260 Sodium lauryl sulfate, 247
Spurious correlations, 11–12
Spurious relationships, 11–12
S Squamous cell carcinoma. See also Cancer.
head and neck, chemotherapy for, 277, 278f
Saliva
salivary proteomics and, 345–346
bacteria in, 49–50, 58f, 62f, 197
Staphylococcus aureus, 47, 195, 198–199
cancer biomarkers in, 345t, 345–348
Statistical analyses, 2
composition of, 343
Statistical significance testing
extracellular vesicles, 353 α level for, 26–27
factor VII–mediated coagulation triggered by, 351
confidence intervals for, 30t
functions of, 266, 343
definition of, 25–26
hyposalivation, 266–268
hypothesis testing for, 26–28, 27f
microbiome in, 56–58, 57f–58f
P value for, 26–28, 28t, 30t
oral microbe transfer via, 70
Stevens-Johnson syndrome, 249–250
xerostomia, 266–268, 274t
Streptococcus pneumoniae, 195
Salivaomics
Streptococcus salivarius, 183
definition of, 343
Streptococcus viridans, 332, 334
salivary proteomics, 343–347
Stromal-vascular component, 106
schematic diagram of, 354f
Study designs
Salivary exosomes
classification of, 32, 33f
as cancer biomarkers, 351–353
description of, 32
contents of, 350–351
evidential interpretations from, 34f
definition of, 349
experimental trials, 32–35
discovery of, 349
randomized controlled trial. See Randomized
mRNA in, 351
controlled trials.
structure of, 349–350
Sturge-Weber syndrome, 263
summary of, 353
Subgingival plaque
Salivary glands
bacteria in, 39, 56, 65
anatomy of, 342f, 343
microbiomes in, 50
hypofunction of, 274t
Sublingual gland, 342f, 342–343
radiation therapy–induced damage to, 267–268
Submandibular gland, 342f, 342–343
Salivary microvesicles, 351
Sufficient conditions, 6–8, 7f, 8b–9b, 15–16
Salivary Proteome Knowledge Base, 343, 344f
Sufficient-component cause framework, 8–9, 10f, 16
Salivary proteomics
Sugar-sweetened beverages, 105–106, 118–119
breast cancer applications of, 345
Supragingival bacteria, 51f–52f
description of, 343–345
Supragingival plaque, 55f
gastric cancer applications of, 346
Susceptibility, 11
lung cancer applications of, 346
Syngeneic transplantation, 280
ovarian cancer applications of, 346–347
Systemic diseases. See also specific disease.
squamous cell carcinoma applications of, 345–346
dental caries and, 302
Salivary transcriptomics, 347–349
oral conditions caused by
Salivary-exosomics, 349–353, 354f
candidiasis, 255f, 255–256
Sanger sequencing, 297
coated/hairy tongue, 256f, 256–257
Santa Fe Group position statement, on oral-systemic
diffuse gingival hyperplasia, 258–260
interactions, 362–363
gingivitis. See Gingivitis.
Scaling and root planing, 115, 144, 200
hyposalivation, 266–268
Secondary antibiotic prophylaxis, 329–330, 340
leukoplakia, 253, 253f
Selection bias, 33
medication-related osteonecrosis of the jaw. See
Severe early childhood caries, 116–117
Medication-related osteonecrosis of the
Severe periodontitis, 136t
jaw.
Sex steroids, 212–213
metastatic tumors, 266
Signature innate cytokines, 88
migratory glossitis, 257, 257f
Sink drain, 71
oral lichen planus, 253–255, 254f
16S rRNA gene, 43–44
Sjögren syndrome, 267

381
I Index

oral ulcers. See Oral ulcers. Trismus, 273t


overview of, 242–243 Tumor necrosis factor- α , 89, 93, 96, 106, 108, 112
pigmented lesions, 260–263 Tumor necrosis factor α-induced protein 3, 353
poorly healing or nonhealing extraction sockets, Type I error, 26, 27f
263–264 Type II error, 26, 27f
white/red lesions, 241t, 253–257 Tyrosine kinase inhibitors, for burning mouth
xerostomia, 266–268 syndrome, 268
oral diseases and. See also Oral infection–systemic
health link.
common risk factors between, 311, 316, 317f,
321–323, 326
U
Ulcers. See Oral ulcers.
genomic connection between, 301–306, 303f
Umbrella reviews, 144
shared molecular basis between, 306
Unfractionated heparin, 187
summary of, 325–326
Uremia, 183–184
oral infections and, 165
Uveitis, 69
periodontal disease and, 92–97
periodontitis and, 170b
salivary biomarkers for, 342
Systemic lupus erythematosus, 254–255
V
Valacyclovir, 250t
VAP. See Ventilator-associated pneumonia.
T Varicella-zoster virus, 247
Vascular endothelial growth factor, 345
T cells, 95–96, 107
Vascular lesions, 263
Tacrolimus, 243t
Vegetations, 331, 331f
Tacrolimus solution, 243t
Veillonella species, 72
Tannerella forsythia, 51
Ventilator-associated pneumonia, 194, 198, 202–203,
Targeted therapies, 279
206
Telangiectatic granuloma, 213
Vesiculobullous disease, 251–253, 252f–253f
Temporality, 2
Viral infections, oral ulcers caused by, 246–247, 248t,
Tetracycline, 261
250t
Th1 cells, 108
Visceral adiposity, 106
Th2 cells, 108
Viscous lidocaine, 243t
Tiffeneau-Pinelli index, 196
Vitamin D deficiency, 184
Toll-like receptors, 88
Tooth brushing
bacteremia after, 67, 177, 200, 332–334, 335f, 337
in diabetes mellitus patients, 154b
W
infective endocarditis and, 337, 339 Waist circumference, 104, 113, 114f
oral microbiome altered by, 61 Waist-height ratio, 104
Tooth eruption and movement Waist-hip ratio, 104
hyperglycemia and, 140 White/red lesions
obesity effects on, 119–121 leukoplakia, 253, 253f
Tooth extractions, 337 oral lichen planus, 253–255, 254f
Tooth loss Whole-exome sequencing, 301
causes of, 121 World Health Organization (WHO)
dental caries as cause of, 148 Global Action Plan for the Prevention and Control
diabetes mellitus and, 140–141 of Noncommunicable Diseases 2013–2020,
dietary effects of, 148 316, 319
hyperglycemia and, 140 noncommunicable diseases statement by, 318–319
nutritional effects of, 148
Toxic epidermal necrolysis, 249
Transcription, 296f X
Transcriptome, 347 Xerostomia, 148–149, 266–268, 274t
Transfer RNA, 347
Translational research, 307
Traumatic ulcers, 242
Traveling oral microbiome. See Oral microbiome,
Z
traveling. Z variable, 17f, 17–18
Triamcinolone, 243t Zimmerman-Laband syndrome, 260

382

You might also like