Salvatore L. Ruggiero, DMD, MD, Clinical
Professor, Division of Oral and Maxillofacial
Surgery, Stony Brook School of Dental
Medicine, Hofstra North Shore-LIJ School
of Medicine, New York Center for Orthognathic
and Maxillofacial Surgery, Lake Success, N.Y.
Thomas B. Dodson, DMD, MPH, Professor
and Chair, University of Washington School of
Dentistry, Department of Oral and Maxillofacial
Surgery, Seattle, Wash.
Tara Aghaloo, DDS, MD, PhD, Professor,
Oral and Maxillofacial Surgery, Assistant Dean
for Clinical Research, UCLA School of Dentistry,
Los Angeles, Calif.
Eric R. Carlson, DMD, MD, EdM, Professor
and Kelly L. Krahwinkel Endowed Chairman,
Department of Oral and Maxillofacial Surgery,
University of Tennessee Graduate School of
Medicine, Knoxville, Tenn.
Brent B. Ward, DDS, MD, Chalmers J. Lyons
Professor of Oral and Maxillofacial Surgery,
Associate Professor of Dentistry, Chair of the
Department of Oral and Maxillofacial Surgery/
Hospital Dentistry in the School of Dentistry
and Associate Professor of Surgery for the
Medical School, University of Michigan
Hospital, Ann Arbor, Mich.
Deepak Kademani, DMD, MD, Chief of
Sta North Memorial Health, Fellowship
Director, Oral/Head and Neck Oncologic
and Reconstructive Surgery Attending Surgeon,
North Memorial Health and the University of
Minnesota. Private practice, Minnesota Oral
and Facial Surgery and Minnesota Head and
Neck Surgery, Minneapolis, Minn.
PAGE 1 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Abstract
Strategies for management of patients with, or at risk for,
medication-related osteonecrosis of the jaw (MRONJ) –
formerly referred to as bisphosphonate-related
osteonecrosis of the jaw (BRONJ) – were set forth
in the American Association of Oral and Maxillofacial
Surgeons (AAOMS) position papers in 2007, 2009
and 2014. The position papers were developed by a
committee appointed by the AAOMS Board of Trustees
and comprising clinicians with extensive experience in
caring for these patients, as well as clinical and basic
science researchers. The knowledge base and experience
in addressing MRONJ continues to evolve and expand,
necessitating modications and renements to the previous
position papers. Three members of the AAOMS
Committee on Oral, Head, and Neck Oncologic and
Reconstructive Surgery (COHNORS) and three authors
of the 2014 position paper were appointed to serve as a
working group to analyze the current literature and revise
the guidance as indicated to reect current knowledge in
this eld. This update contains revisions to diagnosis and
management strategies and highlights the current research
status. AAOMS maintains that it is vitally important for
this information to be disseminated to other relevant
healthcare professionals and organizations.
Introduction
Medications prescribed for dental and medical conditions
have potential side eects that warrant a risk-benet
discussion. Where therapeutic margins are wide and
complications are readily corrected, decisions are
implemented in a straightforward fashion. Where
therapeutic margins are wide but complications are
signicant, deciding to proceed with pharmacologic
treatment becomes more challenging. In most cases of
MRONJ, local therapies can be successful. The fact that
more complex treatment is required for a few patients
should not impact decision-making for all other patients
with osteonecrosis of the jaw. The medications associated
with MRONJ have proven to be safe and eective in
clinical trials and postmarketing analyses for most
Position Paper
American Association of Oral and Maxillofacial Surgeons
Medication-Related Osteonecrosis of the Jaw – 2022 Update
patients and should continue as a mainstay therapy when
indicated. Communicating the risks of MRONJ to patients
and providers is critical to ensure appropriate medical
management for the primary disease.
Undoubtedly, risk proles may change as new medications
come to market. In addition, our understanding of disease
pathophysiology, risk modiers and treatment strategies
will continue to evolve. It is of the utmost importance
that clinicians base their patient treatment decisions on
currently available scientic evidence.
Strategies for management of patients at risk for or with
MRONJ were set forth in AAOMS Position Papers in
2007,
1
2009
2
and 2014.
3
These position papers were
developed by a committee appointed by the AAOMS
Board of Trustees and comprised of clinicians with
extensive experience in caring for these patients as well as
clinical and basic science researchers. The knowledge base
and experience in addressing MRONJ continues to evolve
and expand, necessitating modications and renements to
the previously published position papers. A working group
comprised of three members of the AAOMS Committee
on Oral, Head, and Neck Oncologic and Reconstructive
Surgery and three authors of the 2014 paper convened
remotely in the fall of 2020 to appraise the current
literature and revise the guidelines as indicated to reect
the current knowledge in this eld. This update contains
revisions to the pathogenesis and management strategies
and highlights the current research status. AAOMS
maintains it is vitally important for this information to be
disseminated to other relevant healthcare professionals and
organizations.
Purpose
The purpose of this position paper is to provide updates
regarding:
1. Risk estimates of developing MRONJ.
2. Comparisons of the risks and benets of medications
related to osteonecrosis of the jaw in order to facilitate
medical decision-making for the treating physician,
dentist, dental specialist and patient with the
establishment of algorithms.
3. Guidance to clinicians regarding:
a. the dierential diagnosis of MRONJ in patients
with a history of exposure to antiresorptive
medications.
b. MRONJ prevention measures and management
strategies for patients with MRONJ based on the
disease stage.
PAGE 2 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Medications
Bisphosphonates (BPs) are antiresorptive medications
that are eective in managing cancer-related conditions,
including hypercalcemia of malignancy, spinal cord
compression and pathologic fractures (skeletal-related
events [SREs]) associated with bone metastases in the
context of solid tumors (such as breast, prostate and lung
cancers) and multiple myeloma.
4, 5, 6, 7, 8, 9, 10, 11, 12, 13
While
the potential for BPs to improve cancer-specic survival
remains controversial, these medications have had a
signicant positive eect on the quality of life for patients
with advanced cancer involving the skeleton and reducing
or preventing skeletal-related events.
Bisphosphonates also are used for the prevention of
osteoporosis-related fractures (fragility fractures) in
patients with osteoporosis and osteopenia.
14, 15, 16
BPs
administered orally – including alendronate (Fosamax),
risedronate (Actonel) or parenterally (zoledronic acid
[Reclast]), and ibandronate (Boniva) – can result in
a signicant reduction in vertebral and nonvertebral
fractures for patients with osteoporosis.
17, 18, 19, 20
Bisphosphonate therapy also is indicated for other
metabolic bone diseases such as Paget’s disease of bone
and osteogenesis imperfecta.
21, 22, 23
However, clinical trials
have not demonstrated the ecacy of bisphosphonate
therapy in the management of brous dysplasia.
24
Denosumab (DMB), a receptor activator of nuclear factor
kappa-B ligand (RANK-L), is an antiresorptive agent
that exists as a fully humanized antibody against RANK
ligand and inhibits osteoclast function and associated bone
resorption. When denosumab (Prolia) is administered
subcutaneously every six months, there is a signicant
reduction in the risk of vertebral, nonvertebral and hip
fractures in osteoporotic patients.
25, 26, 27, 28
Denosumab
(Xgeva) also is eective in reducing SREs related
to metastatic bone disease from solid tumors when
administered monthly.
29, 30, 31
RANK ligand inhibitors also have proven ecacy in
the treatment of giant cell tumors of bone and brous
dysplasia.
32, 33, 34, 35, 36
In contrast to BPs, RANK-L
Position Paper
PAGE 3 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
inhibitors do not bind to bone, and their eects on bone
remodeling are mostly diminished within six months of
treatment cessation.
Romosozumab is a new monoclonal antibody used
for fracture prevention in osteoporotic women.
Romosozumab, administered subcutaneously, works via
the Wnt pathway by binding to and inhibiting sclerostin,
resulting in increased bone formation and decreased bone
resorption.
37
MRONJ Case Denition
MRONJ should be distinguished from other forms of
osteonecrosis (ONJ) conditions and identied by history
and clinical exam. The clinical criteria required to establish
a diagnosis of MRONJ have remained unchanged from the
previous position paper.
3
The case denition of MRONJ includes all the following
elements:
1. Current or previous treatment with antiresorptive
therapy alone or in combination with immune
modulators or antiangiogenic medications.
2. Exposed bone or bone that can be probed through an
intraoral or extraoral stula(e) in the maxillofacial
region that has persisted for more than eight weeks.
3. No history of radiation therapy to the jaws or
metastatic disease to the jaws.
Staging
A staging system for MRONJ was introduced in the
2009 AAOMS position paper and then modied in the
2014 position paper to characterize more accurately all
aspects of the clinical presentation of MRONJ. Since
these modications, the AAOMS staging system has
continued to be a straightforward and relevant system to
properly stratify these patients. It has been adopted by
several professional societies and research organizations.
The staging system facilitates the creation of rational
treatment guidelines and guides data collection to assess
the prognosis and outcomes for MRONJ patients. While
AAOMS recognizes that dierent classication systems
are being used by other organizations,
38
the Association
considers the AAOMS system to be a useful and widely
implemented assessment tool guiding clinicians involved
in the care of MRONJ patients. AAOMS remains
concerned that overemphasizing variable radiographic
features often attributed to MRONJ may overestimate
the true disease frequency by including false positives
in the numerator (e.g., cases with radiographic ndings
suggestive of MRONJ), but these patients do not t the
criteria for the diagnosis of MRONJ. In the orthopedic
literature, the usefulness of a Stage 0 category has been
established for staging avascular necrosis (AVN) of the
femoral head when there is a suspicion of AVN in a patient
at risk, but the diagnostic information is not conclusive.
39
AAOMS believes the Stage 0 category for MRONJ is
analogous in principle and does account for the wide-
ranging radiographic presentation of MRONJ that exists
prior to overt bone exposure. Therefore, AAOMS has
decided to maintain the current classication system with
no modications.
Patients at-Risk
No apparent necrotic bone in asymptomatic patients who
have been treated with IV or oral antiresorptive therapy.
Stage 0 (Nonexposed Bone Variant)
Patients with no clinical evidence of necrotic bone but
who present with nonspecic symptoms or clinical and
radiographic ndings, such as:
Symptoms
Odontalgia not explained by an odontogenic cause.
Dull, aching bone pain in the jaw, which may radiate
to the temporomandibular joint region.
Sinus pain, which may be associated with inammation
and thickening of the maxillary sinus wall.
Altered neurosensory function.
Clinical Findings
Loosening of teeth not explained by chronic periodontal
disease.
Intraoral or extraoral swelling.
Radiographic Findings
Alveolar bone loss or resorption not attributable
to chronic periodontal disease.
Changes to trabecular pattern sclerotic bone and no new
bone in extraction sockets.
Regions of osteosclerosis involving the alveolar bone
and/or the surrounding basilar bone.
Thickening/obscuring of periodontal ligament
(thickening of the lamina dura, sclerosis and decreased
size of the periodontal ligament space).
40
These nonspecic ndings, which characterize this variant
of MRONJ without bone exposure, may occur in patients
with a prior history of Stage 1, 2 or 3 disease who have
been healed and have no clinical evidence of exposed
bone. Progression to Stage 1 disease has been reported
in up to 50 percent of patients with Stage 0 disease
41
and,
therefore, AAOMS deems it prudent to consider Stage 0
disease as a potential precursor to MRONJ.
Stage 1
Exposed and necrotic bone or stula that probes to the
bone in patients who are asymptomatic and have no
evidence of infection/inammation. These patients also
may present with radiographic ndings mentioned for
Stage 0 that are localized to the alveolar bone region.
Stage 2
Exposed and necrotic bone, or stula that probes to the
bone, with evidence of infection/inammation. These
patients are symptomatic. These patients also may present
with radiographic ndings mentioned for Stage 0 localized
to the alveolar bone region.
Stage 3
Exposed and necrotic bone or stulae that probes to the
bone, with evidence of infection, and one or more of the
following:
Exposed necrotic bone extending beyond the region
of alveolar bone (i.e., inferior border and ramus in the
mandible, maxillary sinus and zygoma in the maxilla).
Pathologic fracture.
Extraoral stula.
Oral antral/oral-nasal communication.
Osteolysis extending to the inferior border of the
mandible or sinus oor.
Causality
It is important to understand that patients at risk for or with
established MRONJ also can present with other common
clinical conditions not to be confused with MRONJ.
Commonly misdiagnosed conditions may include but
are not limited to alveolar osteitis, sinusitis, gingivitis/
periodontitis, caries, periapical pathology, odontalgia,
atypical neuralgias, bro-osseous lesions, sarcoma,
chronic sclerosing osteomyelitis, and temporomandibular
joint (TMJ) disorders. It also is important to remember
that delayed healing, exposed bone or sequestra (i.e.,
osteonecrosis [ONJ]), can occur in patients not exposed
to antiresorptive agents.
42
Proving causality of any medication-related complication
is challenging from an epidemiologic perspective. It is
well-known that MRONJ is a rare entity, multifactorial
in nature, and patients with the same clinical presentation
exist who have not been exposed to an antiresorptive
medication. Studies have reported jaw necrosis in
antiresorptive naïve patients in which necrosis was linked
to bacterial, viral or fungal infections, trauma, smoking,
steroids, immunocompromised host, autoimmune diseases,
diabetes and chemotherapy.
43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54,
55, 56, 57
Furthermore, patient populations – such as those
with osteogenesis imperfecta – have been treated with
antiresorptive agents without reports of MRONJ.
58
Many
patients receiving medications associated with MRONJ
have other comorbidities, which are likely exacerbating or
contributing factors. In combination, these confounding
variables make incidence and prevalence dicult to
estimate.
Clinical trials, while being the gold standard for ecacy
and safety data, are seldom powered to demonstrate
uncommon events. Prior to the discovery of MRONJ, large
randomized prospective trials of BPs with up to 10 years
of patient data did not reveal any jaw bone necrosis as a
complication.
17, 59
More recently, the HORIZON Pivotal
Fracture trial tested 3,889 randomized patients given
annual zoledronic acid versus placebo for three years; one
patient developed MRONJ in the intervention group and
one in the placebo group.
18
Extension of this trial for up to
six years resulted in one additional MRONJ patient in the
treatment group.
60
Extension to nice years resulted in no
additional conrmed cases of MRONJ.
61
Denitive causality, taken as a whole, remains a dicult
task to prove in general, let alone in individual patients
PAGE 4 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
PAGE 5 Medication-Related Osteonecrosis of the Jaw – 2022 Update
presenting with clinical symptoms. Clinicians should
be aware of these facts in decisions regarding treatment
recommendations.
Pathophysiology
Since the AAOMS position paper in 2014, signicant
knowledge has been gained regarding MRONJ
pathophysiology from both clinical and particularly
preclinical animal studies. It should be noted that animal
studies have a number of limitations, are most often using
supratherapeutic doses and likely do not truly mirror
the clinical environment. That said, they are critical in
understanding disease mechanisms and can serve as
one reference point to evidence-based clinical decision-
making.
Much debate persists among clinicians and researchers,
contributing to the various treatment protocols utilized
for patients today.
62, 63, 64, 65
Disease specicity unique
to the jaws has focused leading hypotheses to include
bone remodeling inhibition, inammation or infection,
angiogenesis inhibition, innate or acquired immune
dysfunction, as well as genetic predisposition.
3,65
Both
animal and human studies suggest that an antiresorptive
medication, coupled with inammation or infection, is
necessary and sucient to induce MRONJ. However, as
more knowledge is gained on the subject, it is becoming
increasingly apparent that MRONJ is multifactorial, and it
is likely that multiple hypotheses can explain the overall
pathophysiology of this disease.
3,65
Bone Remodeling Inhibition
The denition of MRONJ includes oral or parenteral
administration of antiresorptive medications, such that
bone remodeling suppression is a central hypothesis
in its pathophysiology. Antiresorptive medications,
including BPs and denosumab (DMB), have direct eects
on osteoclast formation, dierentiation or function. In
osteoporosis, BPs are a rst-line therapy to decrease bone
remodeling, increase bone mineral density, and decrease
vertebral and long bone fractures.
66,67
BPs, in higher doses,
also are utilized in primary bone malignancy and bone
metastases to decrease SREs, including hypercalcemia
of malignancy, reduce severe bone pain and improve
quality of life.
68, 69, 70, 71, 72
Although DMB has only been
approved for use since 2010, its use has increased
signicantly for both osteoporosis and malignancy
in the last decade. Prevalence of MRONJ with DMB
users is at least as high as BP users, likely due to its
increased potency to inhibit bone resorption.
30,73, 74, 75
This
is supported in the jaws as animal studies demonstrate
absent osteoclasts around the alveolar bone of DMB-
treated mice.
76
Human bone specimens also show an
increased number of nonfunctional osteoclasts surrounding
necrotic bone in BP-treated patients,
77
further reinforcing
bone remodeling inhibition as a leading hypothesis
in MRONJ pathophysiology. With the appearance of
MRONJ in DMB-treated patients, it becomes increasingly
apparent that the underlying pathophysiology involves
dysfunctional osteoclasts.
Animal studies evaluating withdrawal of BPs or DMB
further highlight the importance of bone remodeling
in MRONJ prevention and resolution. Rodents with
established ONJ failed to resolve when antiresorptives
were withdrawn. However, discontinuing DMB, but not
BPs, prior to tooth extraction successfully prevented
MRONJ development in rats.
78,79
Moreover, parathyroid
hormone, which acts directly on osteoblasts to induce
bone formation and indirectly increases osteoclastic bone
resorption and overall remodeling, has been shown to
prevent MRONJ and improve extraction socket healing
in rodents and preliminarily in patients.
80, 81, 82
This
observation provides further support for the central role
of osteoclast inhibition in MRONJ pathogenesis.
Inammation or Infection
Although most studies report tooth extraction as the
major inciting event for MRONJ development, it is clear
that most extracted teeth had pre-existing periodontal or
periapical disease.
3, 64, 83, 84
From this patient information,
animal models of inammation or infection were
developed to replicate clinical, radiographic and histologic
features of MRONJ.
85, 86, 87, 88
Presence of inammatory
cytokines, specically at the site of MRONJ, also
support the strong role of inammation.
89
As evidence of
increased systemic inammation and its contribution to
MRONJ development, mice with experimentally induced
rheumatoid arthritis demonstrated more severe MRONJ
with increased oral bone exposure, more pronounced
radiographic features, intense local inammatory inltrate
and larger areas of histologic necrosis.
90
Further support
for the inammatory etiology showed that removal of
the inammatory nidus in ligature-induced periodontitis
ameliorated MRONJ development in mice, demonstrating
Position Paper
PAGE 6 Medication-Related Osteonecrosis of the Jaw – 2022 Update
reduced inammation and prevention of disease
progression.
91
Moreover, transplantation of peripheral
blood mononuclear cells with anti-inammatory properties
reduced MRONJ prevalence by improving soft-tissue
healing, decreasing inammatory polymorphonuclear
cells and inammatory marker expression, as well as
enhancing vascularity.
92
These preclinical ndings conrm
the irrefutable role of inammation or infection in MRONJ
disease prevalence, severity and resolution.
The presence of bacteria on the exposed necrotic bone
also contributes to disease severity, where pain and signs
of infection dene Stage 2 MRONJ.
3, 93, 94
This is not
surprising since poor oral hygiene and biolm presence are
associated with MRONJ development,
95, 96
and oral health
maintenance and dental prophylaxis before initiating
antiresorptive therapy can decrease MRONJ prevalence.
97,
98
Importantly, clinical treatment protocols to reduce the
biolm and eradicate infection have emerged as important
alternatives to debridement and resection in patients who
may not be ideal surgical candidates.
63
Angiogenesis Inhibition
Osteonecrosis is traditionally dened as avascular necrosis
or aseptic necrosis, most commonly characterized as
osteocyte death after decreased blood ow to the femoral
head.
99
However, MRONJ is dened as necrotic bone
in the maxillofacial region after exposure to either
antiresorptive or antiangiogenic medications.
3
BPs such as
zoledronic acid directly inhibit angiogenesis in vitro and in
vivo
100, 101, 102, 103
and animal models demonstrate decreased
vascularity in sites of MRONJ and decreased microvessel
numbers during early stages of bone healing.
104
In
addition, angiogenesis normally seen during extraction
socket healing is inhibited by BPs, and both BPs and
DMB have been shown to decrease arterial area, venous
area and overall vascularity of periodontal tissues during
early and late MRONJ development.
105, 106
Importantly,
antiangiogenic medications, such as Vascular Endothelial
Growth Factor (VEGF) inhibitors, tyrosine kinase receptor
inhibitors and immunomodulatory drugs,
107, 108, 109
can be
associated with MRONJ. Moreover, patients with multiple
myeloma receiving both antiresorptive and antiangiogenic
medications, as shown in several studies,
110, 111, 112, 113, 114
have a higher MRONJ prevalence. Important aspects of
MRONJ treatment include determining disease margins,
which can be challenging as microvascular mucosal
abnormalities can be seen adjacent to frank MRONJ
lesions.
115
It is important to note that the incidence of
MRONJ in patients on antiangiogenics is much lower than
those taking antiresorptive medications.
Innate or Acquired Immune Dysfunction
Although animal studies conrm that an antiresorptive
medication—coupled with inammation or infection—
is necessary and sucient to produce MRONJ, not all
patients with dental infections develop the disease. It is
well-known that patients with medical comorbidities such
as diabetes or rheumatoid arthritis or immunocompromised
states are at signicantly higher risk for MRONJ with
or without exposure to antiresorptive agents.
3, 64, 114, 116
Patients with metastatic or primary bone malignancies
have a compromised immune system.
117
This also has
been conrmed with animal studies, where chemotherapy,
steroids and disease-modifying antirheumatic drugs
(DMARDs), combined with antiangiogenic medications
and an antiresorptive, increase MRONJ severity or
prevalence.
118, 119, 120
Moreover, higher rates of MRONJ
occur in patients with multiple myeloma who receive
multiple chemotherapeutic agents.
110,121
Replenishing the area of nonhealing MRONJ lesions with
mesenchymal stem cells (MSCs) to overcome immune
dysfunction is a potential area of therapeutic interest,
especially in patients who are immunocompromised.
A recent study showed altered numbers and patterns of
T-cells in human and rat MRONJ necrotic bone samples
as compared to healthy patients and non-MRONJ sites.
122
Preclinical studies also demonstrate healing or prevention
of MRONJ lesions after systemic infusion with adipose or
bone marrow-derived MSCs.
123, 124, 125
Genetic Factors
In the 2014 paper, the authors identied several reports
describing single-nucleotide polymorphisms (SNPs) that
were associated with the development of MRONJ. Most
of these SNPs were located within regions of the gene
associated with either bone turnover, collagen formation
or certain metabolic bone diseases. Indeed, increasing
evidence is available to support the role of SNPs with
MRONJ.
126, 127
Specic links to sirtuin-1 (SIRT1), a bone
remodeling regulator that promotes bone formation, may
be protective against MRONJ if upregulated.
128
SIRT1
also is involved in both reduction of inammation and
induction of angiogenesis, suggesting a role in several
Position Paper
PAGE 7 Medication-Related Osteonecrosis of the Jaw – 2022 Update
of the leading MRONJ hypotheses.
128
Other genes also
have been reported to increase MRONJ risk through
their role in angiogenesis, bone remodeling and immune
responses, including PPAR gamma, CYP2C8 and many
others.
129
Collectively, these studies suggest that MRONJ
is a multifactorial disease and that genetic factors may
play a role in its development.
130
Overall, however, current
studies document either a weak or no association between
genetic factors measured and risk for MRONJ.
131
To
determine predisposition, studies with larger sample sizes
should be performed, with genetic risks conrmed in both
BPs and DMB-treated patients who have breast or prostate
cancer metastases, multiple myeloma or osteoporosis.
Risk factors for MRONJ
Medication-related Risk Factors
To estimate the risk for medications associated with
MRONJ, the primary parameter to be considered is the
therapeutic indication for treatment (e.g., malignancy
or osteoporosis/osteopenia). The data suggest that
antiresorptive medications (i.e., BPs and DMB) are
associated with an increased risk for developing MRONJ.
The risk of MRONJ is considerably higher in the
malignancy group (<5%) than in the osteoporosis group
(<0.05%). Current data are insucient to identify other
medications as risk factors for developing MRONJ.
MRONJ risk among cancer patients
For estimating the risk for MRONJ among patients
exposed to a medication, the risk for MRONJ in patients
not exposed to antiresorptive medications must be
estimated (Table 1). The risk for MRONJ among cancer
patients enrolled in clinical trials and assigned to placebo
groups ranges from 0 percent to 0.7 percent.
132, 133, 134, 135, 136,
137, 138
a. Among cancer patients exposed to zoledronate,
the cumulative risk of MRONJ clusters in the low
single digits, <5 percent, and ranges from 0 percent
to 18 percent.
113,132,133,137, 138, 139, 140, 141, 142, 143, 144
The
wide variation in estimates may be explained by
the varying durations of follow-up, one to 10 years,
reported in the various studies. The risk of MRONJ
among cancer patients exposed to zoledronate
ranges between 2-10 times higher than cancer
patients treated with placebo.
b. Among cancer patients exposed to DMB, the
risk of MRONJ ranges from 0 percent to 6.9
percent, with most studies reporting rates <5
percent.
113,134,135,138,141,142,144,145
The risk for MRONJ
among cancer patients exposed to DMB is
comparable to the risk of MRONJ in cancer patients
exposed to zoledronate.
135,141,142,144,145
Since the 2014 update, investigators have implicated
numerous families of medications as risk factors for
MRONJ.
146, 147, 148, 149
These medications include tyrosine
kinase inhibitors (TKIs) such as sunitinib, monoclonal
antibodies (bevacizumab), fusion proteins (aibercept),
mTOR inhibitors (everolimus), radiopharmaceuticals
(radium 223), selective estrogen receptor modulators
(raloxifene) and immunosuppressants (methotrexate and
corticosteroids).
When compared to antiresorptive medications, the
level of evidence supporting other medication families
as risk factors for MRONJ is level 5 (e.g., isolated
case reports or mini-case series [<5 cases]).
146, 147, 148,
149
Given that the poly-pharmaceutical management of
cancer patients combined with the fact that cancer and
immunosuppression are risk factors for MRONJ without
exposure to antiresorptive agents, AAOMS believes that
identifying a single medication as being the etiologic agent
for MRONJ seems unlikely in case reports or mini-case
series. Further controlled prospective studies will be
required to measure the risk of MRONJ associated with
non-antiresorptive agents.
MRONJ Risk Among Osteoporosis Patients
Most dentists and oral and maxillofacial surgeons evaluate
patients in their practices exposed to antiresorptive therapy
for management of osteoporosis (Table 1).
a. Risk for MRONJ among osteoporotic patients
exposed to BPs.
The risk of MRONJ among study subjects assigned
to placebo groups enrolled in osteoporosis clinical
trials ranged from 0 percent to 0.02 percent.
26,150,151
Among study subjects treated with BPs, the risk
of MRONJ is 0.02 percent to 0.05 percent.
37,75,152
Among patients exposed to IV zoledronate,
the risk for MRONJ is estimated to be ≤0.02%
(≤ 2 per 10,000). For patients exposed to oral
bisphosphonates, MRONJ risk is estimated to be
≤0.05% (≤ 5 per 10,000).
Position Paper
PAGE 8 Medication-Related Osteonecrosis of the Jaw – 2022 Update
b. MRONJ risk among osteoporotic patients exposed
to RANK-L inhibitors.
After 10 years of follow-up, among patients
exposed to DMB, the risk for MRONJ was reported
to be 0.3 percent, almost an order of magnitude
higher than for BPs.
153
c. The risk for MRONJ when exposed to
romosozumab (0.03 percent to 0.05 percent) is
comparable to alendronate (0.05 percent).
37,151
In the
placebo group, there were no cases of MRONJ.
151
It will be important to continue to monitor
romosozumab to assess its role as a risk factor for
MRONJ.
The risk for MRONJ among osteoporosis patients
treated with BPs ranges from 0.02 percent to
0.05 percent and overlaps the risk for MRONJ
of patients enrolled in placebo groups (0 percent
to 0.02 percent). The risk for MRONJ among
patients treated with denosumab, however, has a
larger range—from 0.04 percent to 0.3 percent.
As such, additional research will be needed to
better estimate the risk of MRONJ among patients
receiving denosumab. The risk of MRONJ for
patients exposed to romosozumab (0.03 percent
to 0.05 percent) more closely aligns with the risk
associated with BPs.
37,151
However, given its recent
introduction as a therapeutic agent, additional
research will be needed to rene its association and
risk estimate for MRONJ.
Based on this current review of data, the risk of developing
MRONJ among osteoporotic patients exposed to BPs,
DMB and romosozumab is low. The occurrence of cases
seen is best explained by a rare event among a large
number of patients, 5.1 million over the age of 55, exposed
to these drugs.
154
MRONJ Risk Among Patients
with Nonmalignant Bone Disease
a. AAOMS identied two studies where DMB was
used to manage aggressive giant cell tumors of
bone.
155, 156
The risk of developing MRONJ in
the two studies was broad and ranged from 0.7
percent to 5 percent. This is comparable to the
risks of developing MRONJ in subjects treated
with DMB for malignancies (range = 0 percent
to 6.9 percent). Additional studies will be needed
to conrm the risk estimate for MRONJ among
patients with nonmalignant bone disease treated
with antiresorptives.
b. There are very limited data describing the
occurrence of MRONJ in the pediatric population
for osteogenesis imperfecta and other conditions. In
a systematic review estimating the risk of MRONJ
among children with osteogenesis imperfecta, there
were no cases of MRONJ identied in a sample
of 486 subjects treated for 4.5 to 6.8 years.
157
In a
dierent systematic review that estimated the risk
for MRONJ among those under the age of 24 for
several conditions treated using BPs, no cases of
MRONJ were reported.
158
The overall quality of
the studies included in both systematic reviews was
limited by small sample sizes or lack of MRONJ-
related risk factors.
Duration of Medication Therapy
as a Risk Factor for MRONJ
Regardless of indications for therapy, the duration of
antiresorptive therapy is a risk factor for developing
MRONJ. Among cancer patients exposed to zoledronate
or DMB (n = 5,723), the risk of developing MRONJ
was, respectively, 0.5 percent and 0.8 percent at 1 year,
1.0 percent and 1.8 percent at 2 years, and 1.3 percent
and 1.8 percent at 3 years.
141
In a study by Saad et al.,
the investigators combined three-blinded phase 3 trials
and found similar results, including a plateau after two
years for patients exposed to DMB.
5
In a more recent
systematic review by Ng et al., the risk of MRONJ among
cancer patients treated with zoledronate, was 1.6 percent
to 4 percent after two years of treatment and 3.8 percent
to 18 percent with more than two years of treatment.
145
Likewise, for DMB, the risks for developing MRONJ
were 1.9 percent and 6.9 percent with <24 months and >24
months of exposure, respectively.
145
For patients receiving bisphosphonate therapy to manage
osteoporosis, data regarding duration are mixed. Early on,
the prevalence of MRONJ was reported as increasing over
time from near 0 percent at baseline to 0.21 percent after
four or more years of BP exposure based on retrospective
analysis.
152,159
More recent data from a large prospective,
randomized placebo controlled trial demonstrate no
signicant increase in MRONJ in patients treated for up
to nine years.
18,60,61
In addition, there are no postmarketing
Position Paper
PAGE 9 Medication-Related Osteonecrosis of the Jaw – 2022 Update
data or general clinical experience to support an MRONJ
prevalence of 0.21 percent in any osteoporosis-treated
group. Therefore, while duration may be a risk factor, the
overall risk remains low.
Local Factors
Dentoalveolar Operations
Dentoalveolar operations are the most common identiable
predisposing factor for developing MRONJ. Several
studies report that among patients with MRONJ, tooth
extraction is cited as a predisposing event ranging from
62 percent to 82 percent.
5,75,160
While this information is
important, it is not what most patients or clinicians want to
know. Most providers and patients want an answer to the
following clinical question: “Among patients exposed to
antiresorptive medications, what is the risk for developing
MRONJ following tooth extraction (or other dentoalveolar
procedures such as implant placement or periodontal
procedures)?” Current estimates for the risk of MRONJ
among osteoporotic patients exposed to BPs following
tooth extraction range from 0 percent to 0.15 percent.
161,162
For osteoporotic patients exposed to DMB, the risk for
MRONJ following tooth extraction was 1 percent.
163
For cancer patients exposed to BPs, the risk of developing
MRONJ after tooth extraction ranges from 1.6 percent
to 14.8 percent.
164, 165, 166
In a small case series, n = 61
subjects having 102 extractions, the risk for MRONJ
after tooth extraction was 13.1 percent.
167
In a systematic
review by Gaudin et al., the risk for MRONJ after tooth
extraction (n = 564) was estimated to be 3.2 percent.
162
While the estimates for developing MRONJ in high-risk
patients undergoing tooth extraction vary, they cluster
between 1 percent and 5 percent, similar to estimates of
osteoradionecrosis following tooth extraction in irradiated
patients.
The risk of developing MRONJ among patients who
have been exposed to antiresorptive medications for
other dentoalveolar operations such as dental implant
placement and endodontic or periodontal procedures is
unknown.
168
The risk for MRONJ after implant placement
among patients treated with DMB has been reported to
be 0.5 percent.
163
Absent better data, AAOMS cautions
the use of these procedures in cancer patients exposed to
antiresorptive therapies and recommends osteoporosis
patients be informed of potential risks, albeit low,
including development of MRONJ, early and late implant
failure all of which have been described in case reports and
clinical trials.
Anatomic Factors
Limited new information regarding anatomic risk factors
for MRONJ is available. MRONJ is more likely to appear
in the mandible (75 percent) than the maxilla (25 percent)
but can appear in both jaws (4.5 percent).
5,75
Denture use
was associated with an increased risk for MRONJ among
cancer patients exposed to zoledronate (OR = 4.9; 95
percent CI = 1.2 to 20.1).
169
In a study by Vahtsevanos et
al., using a sample of 1,621 cancer patients treated with
intravenous zoledronate, ibandronate or pamidronate, there
was a twofold increased risk for MRONJ among denture
wearers.
170
Concomitant Oral Disease
Preexisting inammatory dental disease such as
periodontal disease or periapical pathology is cited as a
risk factor.
75,168
Among cancer patients with MRONJ, the
preexisting inammatory dental disease was a risk factor
among 50 percent of the cases.
5,165
Given that a common treatment of inammatory dental
disease is tooth extraction, pre-existing dental disease may
confound the relationship between tooth extraction and
risk for MRONJ. Tooth extraction may expose MRONJ
as opposed to being the precipitating event. It would be
valuable to see an estimate of the association between
tooth extraction and MRONJ adjusted for pre-existing
inammatory dental disease.
After tooth extraction and periodontal disease, the next
most common risk factor is reported as “spontaneous”
MRONJ with no identiable dental risk factor.
168
Demographic and Systemic Factors
and Other Medications
Age and sex are variably reported as risk factors
for MRONJ.
5, 165, 169, 170, 171
The higher prevalence of
MRONJ in the female population is likely a reection
of the underlying disease for which the agents are being
prescribed (e.g., osteoporosis, breast cancer).
As noted previously, those under the age of 24 treated
with antiresorptives for benign bone diseases have not
demonstrated any risk for MRONJ even after an extended
Position Paper
PAGE 10 Medication-Related Osteonecrosis of the Jaw – 2022 Update
duration of therapy. The overall quality of the studies
included even in systematic reviews is based on small
sample sizes and the lack of other MRONJ-related risk
factors. The risk of developing MRONJ in the pediatric
population requires continued surveillance.
Corticosteroids are associated with an increased risk for
MRONJ.
5,168,171
There are concerns that corticosteroids
increase the risk for MRONJ when given in conjunction
with antiresporptive agents.
Comorbid conditions are inconsistently reported to be
associated with an increased risk for MRONJ, including
anemia (hemoglobin < 10 g/dL) and diabetes.
5,171
Cancer
type also is variably reported as a risk factor.
170,172
Tobacco use is variably reported as a risk factor for
MRONJ. In a case-control study, tobacco use approached
statistical signicance as a risk factor for MRONJ in
cancer patients (OR = 3.0; 95 percent CI = 0.8 to 10.4).
169
In a more recent case-controlled study, tobacco use was
not associated with ONJ in a sample of cancer patients
exposed to zolendronate.
171
Vahtsevanos did not report an
association between tobacco use and MRONJ.
170
In brief, after chemotherapy and corticosteroid exposure,
the next most reported comorbidity is “no comorbidity.”
168
In summary, the current literature rearms that the risk
of MRONJ is signicantly greater in cancer patients
receiving antiresorptive therapy compared to patients
receiving antiresorptive therapy for osteoporosis.
Moreover, the risk of MRONJ in osteoporosis patients
receiving antiresorptive therapy continues to be very low
regardless of drug type (BPs, DMB, romoszumab) or
dosing schedule.
Management Strategies
Treatment Goals
The major goals of treatment for patients at risk of
developing or who have established MRONJ are:
Prevention of MRONJ (see section MRONJ risk
among cancer patients below).
Prioritization and support of continued oncologic
treatment in patients receiving antiresorptive therapy
alone or in combination with immune modulators or
antiangiogenic medications:
Oncology patients benet from the therapeutic
eect of antiresorptive therapy by controlling bone
pain and reducing the incidence of other SREs.
Prioritization and support of continued bone health and
the prevention of fragility fractures:
Patients with osteoporosis, osteopenia and other
metabolic bone diseases benet from antiresorptive
therapy by signicantly reducing the risk of fragility
fractures and other skeletal-related events.
Preservation of quality of life through:
Patient education and reassurance.
Control of pain.
Control of secondary infection.
Prevention of extension of lesion and development
of new areas of necrosis.
Prevention of MRONJ
Numerous studies demonstrate potentially modiable
factors for reducing the risk of MRONJ, including
performing high-risk surgical procedures prior to initiating
therapy,
95,173, 174, 175
using preoperative and postoperative
antibiotics and antimicrobial mouth rinses,
174,176, 177, 178, 179, 180
primarily closing extractions sites,
176, 177, 178
and maintaining
good oral hygiene.
95,166,176,177,181
Maximizing overall patient
health is always indicated, such as smoking cessation and
diabetes optimization. Although no individual strategy nor
collection of strategies eliminates all MRONJ risks, these
preventive procedures are recommended.
The prevention of MRONJ begins with the realization
that patients receiving antiresorptive therapies may have
altered osseous wound-healing capacity, which may also
be a risk for developing MRONJ. Similar to other common
preventive strategies in medicine and dentistry, healthcare
providers need to recognize the importance of coordinated
dental care and pretreatment management in minimizing
the risk of MRONJ. This requires a continuous eort
to educate patients, dentists and medical professionals
about the real risks associated with these therapies and
clinical prevention paradigms that can mitigate MRONJ
development.
AAOMS re-emphasizes the importance of a
multidisciplinary approach to the treatment of patients
who are receiving antiresorptive therapies. This may also
Position Paper
PAGE 11 Medication-Related Osteonecrosis of the Jaw – 2022 Update
apply to other immune modulators or targeted therapies
taken alone or in combination with antiresorptives. This
approach includes consultation with an appropriate dental
professional when it is determined a patient would benet
from these therapies.
Optimization of Oral Health
The 2014 AAOMS position paper identied valid
prophylactic treatment strategies that reduce the
incidence of MRONJ. The ecacies of these strategies
remain validated by subsequent studies that demonstrate
the importance of pretreatment dental screening and
regimented dental surveillance. There is a robust level of
support for early screening and initiation of appropriate
dental care prior to the initiation of antiresorptive
therapy.
38,182, 183, 184, 185, 186
These preventive management strategies not only decrease
the risk for MRONJ but accrue the benets that all patients
enjoy with optimum oral health.
186, 187, 188, 189, 190, 191, 192, 193
In a prospective study of prostate cancer patients with
bone metastasis, instituting a more regimented dental
health surveillance system resulted in a 2.5-fold reduction
in relative risk compared to symptomatically driven
dental treatment.
186
In a systematic review aimed at
identifying prevention strategies associated with tooth
extractions in patients at risk for MRONJ, no randomized
clinical trials were reported.
194
However, there are
many animal studies that demonstrate that periodontal
or periapical inammation plays a key role in creating
a local environment that supports the development of
bone necrosis in the context of systemic antiresorptive
therapy.
85,91,195,196
Treatment planning for patients at risk of developing
MRONJ should include a thorough examination of the
oral cavity and a radiographic assessment when indicated.
It is important to identify both acute infection and sites of
potential infection to prevent future sequelae that could
be exacerbated once drug therapies begin. Considerations
during the clinical and radiographic assessment include
patient motivation, patient education regarding dental care,
uoride application, chlorhexidine rinses, tooth mobility,
periodontal disease, presence of root fragments, caries,
periapical pathology, edentulism and denture stability.
197
An additional benet of early dental consultation, when the
use of antiresorptive therapy is being considered, is that
the patient is informed of the risk associated with these
drug therapies and the risk incurred by not undergoing
recommended dental preventive measures before
consenting to treatment.
Cessation of At-Risk Medication Therapy (Drug
Holiday) Prior to Tooth Extraction or Other
Procedures that Involve Osseous Injury (e.g.,
Dental Implant Placement, Periodontal or Apical
Endodontic Treatment)
The clinical practice of antiresorptive drug holidays to
mitigate MRONJ risk in patients undergoing dentoalveolar
surgery was controversial at the time of the previous
AAOMS position paper in 2014 and remained the case
in 2021. While the practice of a drug holiday has been
accepted and recommended by several international
professional societies,
3,38,182,183,198
the evidence to support or
refute such positions remains inconclusive. The diculty
in establishing or refuting the ecacy of drug holidays is
due to the rarity of MRONJ in these patient populations.
Therefore, since few events are reported, randomized-
controlled trials provide insucient data to create sound
treatment protocols. In a 2020 systematic review that
studied the ecacy of antiresorptive drug holiday in
preventing MRONJ, a variety of papers were identied
with diering conclusions suggesting that a high level of
evidence for supporting or refuting the use of a holiday is
missing.
199
The historical use of a drug holiday was intended to
decrease the prevalence of MRONJ subsequent to the
performance of high-risk surgical procedures. The
concern regarding this practice is the loss of ecacy of
antiresorptive therapy with the development of SREs and
fragility fractures. Among others, factors for consideration
may include disease-related risk (cancer vs. osteoporosis),
drug-dosing frequency, duration of therapy, comorbidities,
other medications (especially chemotherapy, steroids
or antiangiogenics), degree of underlying infection/
inammation and extent of surgery to be performed.
Of note, the working group was unable to reach a
consensus regarding a recommendation on drug
holidays and was evenly split between oering drug
holidays to patients on a case-by-case basis using prior
recommendations and those who never recommend drug
holidays, believing that the risks of potential deleterious
eects of suspending antiresorptive therapy may outweigh
a benet.
Position Paper
PAGE 12 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
A special concern should be considered for suspending
RANKL inhibitors in osteoporosis patients. Several studies
have demonstrated a rebound increase in bone resorption
following the discontinuation of DMB, resulting in an
increased risk of multilevel vertebral fractures.
200, 201, 202
If
DMB is to be suspended, the timing and duration of the
holiday should be optimized in order to minimize this
risk. The planned dentoalveolar surgery can be completed
3-4 months following the last dose of DMB when the
level of osteoclast inhibition is waning. It can then be
reinstituted 6-8 weeks postsurgery. This management
strategy minimizes the length of the drug holiday while
maintaining a favorable environment for bone healing.
Bone Turnover Markers
Since the 2014 AAOMS position paper, there has been a
shift away from bone turnover markers. No biomarkers
are validated for clinical decision-making, and continued
research and prospective studies are required before these
markers can be considered ecacious tools in estimating
MRONJ risk.
Other Biomarkers
Biomarkers related to angiogenesis, VEGF activity,
endocrine function and PTH have more recently been
described.
203, 204, 205
These markers remain at an exploratory
stage and are not yet validated for clinical decision-
making.
Prevention Strategies
Patients scheduled to initiate
antiresorptive treatment for cancer therapy
The treatment objective for this group of patients is
to minimize the risk of developing MRONJ (Table
2). Although a small percentage of patients receiving
antiresorptives develop osteonecrosis of the jaw
spontaneously, the majority of aected patients
experience this complication following dentoalveolar
surgery.
5,112,165,206,207
Therefore, if systemic conditions
permit, initiation of antiresorptive therapy should be
delayed until dental health is optimized.
173,208
This decision
must be made in conjunction with the treating physician
and dentist and other specialists involved in the care of
the patient. There is widespread consensus that optimizing
dental health prior to initiating therapy is ecacious and
of paramount importance.
38,185,186,209
Medical oncologists
should educate their patients about the importance of
dental health and the ecacy of prophylactic dental
treatment in the prevention of MRONJ. Similar to patients
who are to receive radiation therapy, optimizing the
dental health in patients receiving antiresorptives or other
therapies that can compromise bone healing is essential.
The pretreatment evaluation of dental health must extend
beyond a review of systems and include a physical and
radiographic exam. Therefore, a comprehensive dental
examination performed by a dental health professional
would be a prudent approach for all patients prior to
receiving antiresorptive therapy for malignant disease. This
level of dental health assessment is most appropriately
performed by a dental health professional.
The importance of minimizing the burden of dental
infection and inammation prior to dentoalveolar surgery
in this cohort of patients with an elevated MRONJ risk
cannot be over-emphasized. Nonrestorable teeth and those
with a poor prognosis should be extracted. Other necessary
elective dentoalveolar surgery also should be completed at
this time. It remains advisable that antiresorptive therapy
should be delayed, if systemic conditions permit, until the
surgical site(s) have mucosalized or until there is adequate
osseous healing. Dental prophylaxis, caries control,
conservative restorative dentistry and nonoperative
endodontic therapy are critical to maintaining functionally
sound teeth. This level of care must be continued on a
frequent and indenite basis.
185
The posterior lingual plate region is a common site for
trauma and mucosal irritation in denture wearers.
5,75,170
Therefore, patients with full or partial dentures should be
examined for areas of mucosal trauma, especially along
the lingual ange region. It also is critical that patients
be educated as to the importance of dental hygiene and
regular dental evaluations, and specically instructed to
report any pain, swelling or exposed bone.
Patients scheduled to initiate
antiresorptive treatment for osteoporosis
Patients who are scheduled to receive antiresorptive
therapy for the prevention of fragility fractures assume a
signicantly lower risk of MRONJ. Therefore, the urgency
and the timing of optimizing the dental health are not as
crucial. However, at the initiation of treatment, it would
be prudent to educate patients regarding the potential risks
PAGE 13 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
of MRONJ. The importance of optimizing dental health
throughout this treatment period and beyond cannot be
underestimated.
It is not uncommon for patients to seek the consultation of
an oral and maxillofacial surgeon in guiding their decision
about starting or continuing antiresorptive therapy. In this
scenario, the consulting oral and maxillofacial surgeon
should use this opportunity to place the risks and benets
into the proper perspective. More specically, patients
should be reminded of the benets associated with
antiresorptive therapies in preventing fragility fractures
and an acknowledgment of the rare occurrence of MRONJ.
The initial enthusiasm and attention associated with the
discovery of MRONJ have had unintended consequences.
When initially described, a “class eect” was observed,
suggesting that MRONJ rates for patients receiving
oncologic doses of BPs and those receiving osteoporotic
doses of BPs were similar. A plateau and a decline in
the use of BPs for osteoporosis was noted in 2006 and
is hypothesized to be associated with various safety
concerns, such as MRONJ. Patients are becoming
increasingly more reluctant to begin or comply with their
antiresorptive therapy. Current evidence also conrms an
increase in fragility fractures with signicant associated
morbidity. As one salient example, hip fracture rates in
the United States declined each year from 2002 to 2012
and then plateaued at levels higher than projected for
2013 to 2015, attributable to an “osteoporosis treatment
gap.”
210
Hip fracture carries signicant morbidity, with
only 40 percent to 60 percent of individuals recovering
their prefracture level of mobility and ability to perform
instrumental activities of daily living.
211
These data are
representative of a true health crisis. The documented
risk for developing MRONJ is low; however, the patient-
perceived risk is not. As such, patients are unwilling to
start or continue antiresorptive medical therapy. Patients
are irrationally denying themselves the tangible therapeutic
benet of antiresorptive therapy to minimize the risk of
fragility fractures in order to prevent a minuscule risk of
developing MRONJ.
It is clear the benet of fracture prevention outweighs
the risk of MRONJ development in osteoporotic
patients.
212
This benet is even more favorable in the
cancer population where bone-stabilizing medications
signicantly improve quality of life, and it is detrimental
when antiresorptives are withheld due to MRONJ safety
concerns.
Asymptomatic patients receiving
antiresorptive therapies for cancer
Maintaining good oral hygiene and dental care is of
paramount importance in preventing dental disease that
may require eventual extractions or other dentoalveolar
surgery. Procedures that involve direct osseous injury
should be avoided if possible. If a dentoalveolar surgical
procedure is unavoidable (e.g., fractured tooth, advanced
periodontal disease), patients should be informed of the
associated risks. The benet of a drug holiday remains
unsubstantiated in this setting. Nonrestorable teeth may be
treated by removal of the crowns and endodontic treatment
of the remaining roots.
213
Teeth may be extracted if
necessary. Placement of dental implants should be avoided
in the oncology patient receiving parenteral antiresorptive
therapy or antiangiogenic medications. Case series and
systematic reviews have reported necrosis associated with
antiresorptive therapy and implant placement.
194, 214, 215, 216
Asymptomatic patients receiving
antiresorptive therapy for osteoporosis
Since the 2014 position paper, epidemiologic data
regarding the risk of MRONJ in patients receiving
antiresorptive therapy for osteoporosis remain limited due
to the lack of sound prospective studies with sucient
power. Nevertheless, the risk for developing MRONJ is
between 0.02 percent and 0.04 percent for BPs and 0.3
percent for DMB. (see Table 1). Sound recommendations
based on strong clinical research design are still lacking
for patients taking oral BPs.
In general, elective dentoalveolar surgery does not appear
to be contraindicated in this group. Risk assessment for
the development of MRONJ in these patients includes the
above-stated data and the discussion above related to drug
holidays.
The placement of dental implants in the context of
antiresorptive therapy for osteoporosis continues to be an
area of research interest. Several systematic reviews have
acknowledged the lack of quality data and randomized
clinical trials. Some studies have recommended caution,
PAGE 14 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
especially with a longer duration of therapy or steroid
use.
194,216
For example, in their systematic review, Granate
et al.,
216
identied several studies that reported an elevated
MRONJ risk associated with implants placed in the
posterior jaw if the duration of bisphosphonate therapy
exceeded three years and if the patients were receiving
systemic corticosteroids. In contrast to these studies,
systematic reviews by Gelazius et al., and Stavropoulos et
al., reported no increase in risk.
214,217
A recent retrospective
propensity-matched cohort study of 44,900 patients
reported a decreased risk of ONJ in osteoporosis patients
receiving implants compared to matched controls who did
not have implants. Of note, 9,738 patients had a history of
BP use, and the results for implants was in contrast to risk
increase for patients who underwent tooth extraction.
218
Reports of implant-related (MRONJ) necrosis can be
divided into the early (implant surgery-triggered) or late
(implant presence-triggered) category.
215,219,220
In these
reviews, the majority of the implant-related necrosis were
not related to the initial implant surgery but occurred
late (>12 months) and often at sites where implants
were placed prior to the initiation of bisphosphonate
therapy. The common presentation was an en bloc failure,
where the osseointegration of the implants is maintained
within the sequestrum.
220,221
This has been recognized
as a separate pattern of failure that is distinct from the
common peri-implantitis failure and considered by some
to be pathognomonic of MRONJ. Although there are no
prospective studies or systematic reviews pertaining to
implant-related necrosis associated with RANKL inhibitors
or other targeted therapies, AAOMS considers this to have
a similar level of risk.
In summary, robust data do not exist, and available data
are conicting. Therefore, AAOMS suggests that if dental
implants are placed, informed consent should be provided
to include the low risk of MRONJ, as well as early and
late implant failure. These patients should be placed on a
regular long-term recall schedule.
Treatment Strategies
AAOMS has developed a series of treatment algorithms
to streamline the evaluation (Figure 1) and management
strategies (Figure 2, Figure 3, Figure 4) for patients with
MRONJ. These strategies are based on a current review of
nonoperative and operative therapies and their associated
outcomes. Emphasis is placed on both nonoperative and
operative management being acceptable for all stages of
disease based on surgical judgment and patient factors in a
shared decision-making model.
Nonoperative therapy
The ecacy of nonoperative therapies in the management
of MRONJ is documented in the literature and provides
a useful adjunct to the spectrum of management
strategies that also include operative treatment (Figure
2). Nonoperative strategies can be useful in all stages,
especially where signicant comorbidities preclude
operative treatment. They may also result in stabilization
of disease or cure in earlier stages. The goal of both
operative and nonoperative therapies remains the same:
curative therapy and quality-of-life improvement.
Nonoperative therapy heavily focuses on patient education,
reassurance, control of pain and control of secondary
infection to allow for sequestration of the exposed,
necrotic bone.
3,63
Decisions on operative versus nonoperative therapy should
be patient-specic and tailored to individual needs. The
risk versus benet ratio (including quality of life with their
current symptomology), ability to perform good wound
care to prevent infection and disease spread, morbidity
from a major surgical procedure, as well as oral function
or dental rehabilitation after marginal or segmental
resection should be considered. Radiographic imaging is
of utmost importance in the evaluation of MRONJ lesions.
Three-dimensional imaging can identify forming or fully
formed sequestra and potentially decrease the invasiveness
of a surgical procedure. Maintenance of maxillary or
mandibular integrity is desirable, as the reconstruction of
surgical defects in this population can be challenging.
63,222
Stage 1 patients can be managed with chlorhexidine
wound care and improved oral hygiene to remove the
biolm from the necrotic bone surface.
63
Surgery may not
be indicated in the absence of disease progression, with
patient adequate quality of life.
63, 223
Stage 2 patients may
struggle with local wound care and may require antibiotics
for symptom control. Those patients who remain refractory
to nonoperative treatment or those patients who cannot
maintain adequate hygiene may benet from operative
therapy. In the presence of developing or established bony
sequestra, nonoperative therapy may be indicated to allow
for ultimate sequestrectomy. Exfoliation of the exposed,
necrotic bone will often result in disease resolution.
63,224,225
PAGE 15 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
Therefore, for those patients with Stage 2 or 3 diseases
who are poor surgical candidates, nonoperative therapies
may be indicated (Figure 2).
There is little evidence to suggest that the use of adjunctive
therapies, such as hyperbaric oxygen or ozone therapy,
can lead to MRONJ resolution. Larger studies and
controlled trials have yet to demonstrate the ecacy of
the aforementioned treatments.
226, 227, 228, 229
Therefore, these
therapies should not be recommended as a mainstay of
treatment at this time.
The use of vitamin E and pentoxifylline as an adjunct to
standard MRONJ therapies have been reported only in case
studies. A randomized, prospective, placebo-controlled
trial of vitamin E and pentoxifylline is underway and
will provide additional information about this treatment
modality. Teriparatide, one of the few anabolic agents used
for the treatment of osteoporosis, also has shown promise
as an adjunct for the treatment of MRONJ in osteoporotic
patients.
230
Operative therapy
While nonoperative therapy continues to be a treatment
option for MRONJ, operative therapy is increasingly
reported as a viable option with high success rates for all
stages of the disease (Figures 3 and 4). Numerous reports
have identied high success rates associated with resection
of MRONJ lesions.
231, 232, 233, 234, 235, 236, 237
Importantly, one
must consider that MRONJ may progress over time, albeit
in an unpredictable manner.
238
Furthermore, adopting a
nonoperative approach to MRONJ does not uniformly
result in sequestration of the exposed necrotic bone with
disease resolution.
239
Thus, operative intervention should
be explored and presented as a treatment option in an
attempt to reduce the progression of disease with the
recognition that early surgical intervention can predict
benecial patient outcomes.
240
Segmental or marginal resection of the mandible and
partial maxillectomy are eective methods to control
MRONJ.
231, 232, 233, 234, 235, 236, 237, 238,241
This approach can be
applied to patients with all stages of MRONJ, including
Stage 1 disease.
169
These resections require margins
beyond the borders of the necrotic bone to an area of
vital, bleeding bone. Additional reports have identied
success when surgical resection of MRONJ was performed
by experienced surgeons.
242,243
Consistent with surgical
principles, control of comorbid conditions is paramount
in managing MRONJ.
241
Physiologically compromised
patients, such as those with an increasing burden of
distant metastatic disease, may not respond favorably to
resection of their osteonecrotic jaw and may occasionally
develop refractory disease.
241
Finally, surgical resection for
MRONJ in patients with metastatic cancer may identify
metastases in the jaw specimen, albeit in a minority of
patients.
62
Active clinical and radiographic surveillance is critical in
the nonoperative management of patients with Stage 1, 2
and 3 diseases to monitor for signs of disease progression.
In patients who demonstrate the failure of nonoperative
therapy, early operative intervention is recommended.
In patients with a progressive clinical or radiographic
disease or more advanced disease at presentation, surgical
resection of MRONJ should be performed without rst
instituting prolonged nonoperative measures. MRONJ
represents a complex wound whereby operative therapy
can be performed in a timely fashion.
241,244
Although
controversy between operative and nonoperative therapies
exist, operative treatment of patients has demonstrated
maintenance of mucosal coverage, improved quality of life
and expedient resumption of antiresorptive therapy for all
stages of MRONJ disease.
245
The benet of drug holidays
for the operative intervention of MRONJ has not been
substantiated.
Future Research
AAOMS realizes that MRONJ is a complex disease
process with a multifactorial etiology for which many
questions remain unanswered. Continued preclinical
and clinical data are required, especially in the form of
prospective studies. Continued research eorts and the
outcomes that result should be considered the foundation
upon which recommendations are developed that will
guide patients and providers. While the data supporting
the conclusion that antiresorptives represent genuine
risk factors are robust, this is not the case for other
classications of medications (e.g., antiangiogenics,
corticosteroids, immune modulators). Published studies
have reported a relationship of certain dosing practices
(e.g., transition from BPs to DMB) or a synergistic eect
between antiresorptive medications and antiangiogenic
medication with a risk of MRONJ. These associations,
however, are based on case reports and small case series.
It also has been hypothesized that the total exposure to an
PAGE 16 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
antiresorptive medication is a risk factor for developing
MRONJ. However, this has been dicult to demonstrate,
possibly as a result of not having a good measure of
exposure other than years of treatment. Similar to the
cancer risk associated with tobacco use (e.g., pack/years),
the antiresorptive exposure risk MRONJ may be better
dened as a cumulative dose load (e.g., mg equivalent
of BP/years of exposure) that would account for risk
associated with dierent medications and dosing schedules
over time. Dose-reduction protocols and individualized
strategies for antiresorptive therapy in long-term cancer
survivors with a metastatic bone disease are being
explored. It remains to be determined if these protocols
will reduce the risk of MRONJ in this patient cohort.
246
AAOMS acknowledges the challenge of elucidating
potential risks associated with non-antiresorptive therapies,
alone or in combination with antiresorptive medications,
and therefore considers it imperative that research eorts
continue in the form of prospective studies.
A review of the current literature also failed to provide
sound data in the form of randomized, controlled trials
that would establish the eectiveness of biomarkers
and drug holidays or validate a risk relationship with
genetic markers and MRONJ. Until these relationships
are established or refuted, AAOMS considers it prudent
to recognize that these factors may play a role in the
development and management of MRONJ.
PAGE 17 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
Table 1
MRONJ Disease Frequency Grouped by Disease Status vs Medication*
Indications for
treatment
Medication
Study
design
Placebo Zoledronate Oral BPs Denosumab Romosozumab
Malignancy
Coleman (2020)
138
0.2% (2,218)* 5% (2,241) RCT†
O’Carrigan, et al.
(2017)
137
0.7% (6,788) 0.4% (6,788)
Systematic
review
O’Carrigan et al.
(2017)
137
0% (3,060) 1% (3,078)
Systematic
review
Macherey, et al.
(2017)
136
0.7% (818) 1.5% (808)
Systematic
review
Gnant, et al.
(2015)
246
0% (903) 0% (900) RCT
Coleman, et al.
(2014)
133
0% (1,679) 1.7% (1,681) RCT
Valachis, et al.
(2013)
132
0% (3,039) 0.52% (4,774)
Systemic
review
Boquete-Castro, et
al. (2016)
135
0.1% 1.14% 1.7%
Systematic
review
Coleman (2020)
138
0.2% (2,218) 5.4% (2,214) RCT
Gnant, et al.
(2015)
134
0% (1,709) 0% (1,711) RCT
Raje, et al. (2018)
113
2.8% (82) 4.1% (850) RCT
Himelstein
(2017)
140
1.5% (1,822) RCT
Henry (2014)
141
1.1% (786) 0.8% (792) RCT
Yang, et al.
(2019)
247
2% (8,525)
Systematic
review
Peddi, et al.
(2013)
142
1.3% (2,846) 1.8% (2,885)
Systematic
review
Ng, et al. (2021)
145
1.6-4%‡
3.8-18%§
1.9%‡
6.9%§
Systematic
review
Wang, et al.
(2014)
144
1.4% (1,013) 2% (1,020)
Systematic
review
(continued on following page)
PAGE 18 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
(continued from previous page)
Indications for
treatment
Medications
Study
design
Placebo Zoledronate Oral BPs Denosumab Romosozumab
Osteoporosis
Papapoulos, et al.
(2012)
26
0% (3,383) 0.04% (4,549) RCT
Grbic, et al.
(2010)
150
0.02% (4,945) 0.02% (5,864)
Systematic
review
Cosman, et al.
(2016)
151
0% (3,322) RCT
Saag, et al. (2017)
37
0.05% (2,047) RCT
Bone, et al.
(2017)
153
0.3% (2,343)
10-yr f/u
RCT
Hallmer, et al.
(2018)
75
0.043%
Population
study
(50,000)
Non-malignant
bone disease
Chawla, et al.
(2019)
156
5% (532)
Prospective
case series
Rutkowski
155
0.7% (138)
Retrospective
case series
*Sample size in parentheses
†‡Randomized clinical trial
‡ <2 years of followup
§ >2 years of followup
PAGE 19 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
Table 2
MRONJ Prevention Strategies
Pretherapy (nonmalignant disease)
Educate patient about the potential risks associated
with long-term ART.*
Optimization of dental health can occur concurrent
with ART.
Pretherapy (malignant disease)
Educate patients about the higher risk of MRONJ
and the importance of regimented dental care.
Optimization of the dental health prior to the initiation
of ART if systemic conditions permit (extraction of
nonrestorable teeth or teeth with a poor prognosis).
During antiresorptive therapy
(nonmalignant disease)
No alteration of operative plan for most patients.
Considerations include drug schedule, duration of
therapy, comorbidities, other medications (especially
chemotherapy, steroids or antiangiogenics), degree of
underlying infection/inammation and extent of surgery
to be performed. Drug holidays are controversial.
BTM† are not a useful tool to assess MRONJ risk.
During antiresorptive therapy/targeted
therapies (malignant disease)
Educate patients about the higher MRONJ risk in the
setting of malignant disease.
Educate the patient about the importance of regimented
dental care and prevention.
Avoid dentoalveolar surgery if possible.
Consider root retention techniques to avoid extractions.
Dental implants are contraindicated.
Drug holidays are controversial.
*Antiresorptive therapies
† Bone turnover markers (CTX)
PAGE 20 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
Figure 1
PAGE 21 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
Figure 2
PAGE 22 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
Figure 3
PAGE 23 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
Figure 4
PAGE 24 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
References
1. American Association of Oral and Maxillofacial Surgeons position
paper on bisphosphonate-related osteonecrosis of the jaws. J Oral
Maxillofac Surg. 65:369, 2007.
2. Ruggiero SL, Dodson TB, Assael LA, et al. American
Association of Oral and Maxillofacial Surgeons position paper
on bisphosphonate-related osteonecrosis of the jaw - 2009 update.
J Oral Maxillofac Surg. 67, 2009, Supp 1.
3. Ruggiero SL, Dodson TB, Fantasia J, et al: American Association
of Oral and Maxillofacial Surgeons position paper on medication-
related osteonecrosis of the jaw–2014 update. J Oral Maxillofac
Surg. 72:1938, 2014.
4. Saad F, Gleason DM, Murray R, et al: Zoledronic acid prostate
cancer study G. A randomized, placebo-controlled trial of
zoledronic acid in patients with hormone-refractory metastatic
prostate carcinoma. J Natl Cancer Inst. 94:1458, 2002.
5. Saad F, Brown JE, Van Poznak C, et al. Incidence, risk factors,
and outcomes of osteonecrosis of the jaw: Integrated analysis from
three blinded active-controlled phase III trials in cancer patients
with bone metastases. Ann Oncol. 23:1341, 2012.
6. Rosen LS, Gordon D, Tchekmedyian S, et al. Zoledronic acid
versus placebo in the treatment of skeletal metastases in patients
with lung cancer and other solid tumors: A phase III, double-blind,
randomized trial–the zoledronic acid lung cancer and other solid
tumors study group. J Clin Oncol. 21:3150, 2003.
7. Hortobagyi GN, Theriault RL, Porter L, et al. Ecacy of
pamidronate in reducing skeletal complications in patients with
breast cancer and lytic bone metastases. Protocol 19 Aredia Breast
Cancer Study Group. N Engl J Med. 335:1785, 1996.
8. Hortobagyi GN, Theriault RL, Lipton A, et al. Long-term
prevention of skeletal complications of metastatic breast cancer
with pamidronate. Protocol 19 Aredia Breast Cancer Study Group.
J Clin Oncol. 16:2038, 1998.
9. Berenson JR, Lichtenstein A, Porter L, et al. Ecacy of
pamidronate in reducing skeletal events in patients with advanced
multiple myeloma. Myeloma Aredia Study Group. N Engl J Med.
334:488, 1996.
10. Berenson JR, Lichtenstein A, Porter L, et al: Long-term
pamidronate treatment of advanced multiple myeloma patients
reduces skeletal events. Myeloma Aredia Study Group. J Clin
Oncol. 16:593, 1998.
11. Berenson JR, Hillner BE, Kyle RA, et al. American Society of
Clinical Oncology Bisphosphonates Expert Panel. American
Society of Clinical Oncology clinical practice guidelines: The role
of bisphosphonates in multiple myeloma. J Clin Oncol. 20:3719,
2002.
12. Stopeck A, Brufsky A, Kennedy L, et al. Cost-eectiveness of
denosumab for the prevention of skeletal-related events in patients
with solid tumors and bone metastases in the United States. J Med
Econ. 23:37, 2020.
13. Jeon HL, Oh IS, Baek YH, et al. Zoledronic acid and skeletal-
related events in patients with bone metastatic cancer or multiple
myeloma. J Bone Miner Metab. 38:254, 2020.
14. Delmas PD. The use of bisphosphonates in the treatment
of osteoporosis. Curr Opin Rheumatol. 17:462, 2005.
15. Watts NB. Bisphosphonate treatment of osteoporosis. Clin Geriatr
Med. 19:395, 2003.
16. Gossiel F, Paggiosi MA, Naylor KE, et al. The eect of
bisphosphonates on bone turnover and bone balance in
postmenopausal women with osteoporosis: The T-score bone
marker approach in the TRIO study. Bone. 131:115158, 2020.
17. Black DM, Cummings SR, Karpf DB, et al. Randomized trial
of eect of alendronate on risk of fracture in women with existing
vertebral fractures. Fracture Intervention Trial Research Group.
Lancet. 348:1535, 1996.
18. Black DM, Delmas PD, Eastell R, et al. Once-yearly zoledronic
acid for treatment of postmenopausal osteoporosis. N Engl J Med.
356:1809, 2007.
19. Nakamura T, Fukunaga M, Nakano T, et al. Ecacy and safety
of once-yearly zoledronic acid in Japanese patients with primary
osteoporosis: Two-year results from a randomized placebo-
controlled double-blind study (ZOledroNate treatment in ecacy
to osteoporosis; ZONE study). Osteoporos Int. 28:389, 2017.
20. Cranney A, Wells G, Willan A, et al. Meta-analyses of therapies
for postmenopausal osteoporosis. II. Meta-analysis of alendronate
for the treatment of postmenopausal women. Endocr Rev. 23:508,
2002.
21. Delmas PD, Meunier PJ. The management of Paget’s disease of
bone. N Engl J Med. 336:558, 1997.
22. Letocha AD, Cintas HL, Troendle JF, et al. Controlled trial
of pamidronate in children with types III and IV osteogenesis
imperfecta conrms vertebral gains but not short-term functional
improvement. J Bone Miner Res. 20:977, 2005.
23. Florenzano P, Pan KS, Brown SM, et al. Age-related changes
and eects of bisphosphonates on bone turnover and disease
progression in brous dysplasia of bone. J Bone Miner Res.
34:653, 2019.
24. Boyce AM, Kelly MH, Brillante BA, et al. A randomized, double
blind, placebo-controlled trial of alendronate treatment for brous
dysplasia of bone. J Clin Endocrinol Metab. 99:4133, 2014.
PAGE 25 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
25. Cummings SR, San Martin J, McClung MR, et al. Denosumab
for prevention of fractures in postmenopausal women with
osteoporosis. N Engl J Med. 361:756, 2009.
26. Papapoulos S, Chapurlat R, Libanati C, et al. Five years of
denosumab exposure in women with postmenopausal osteoporosis:
Results from the rst two years of the FREEDOM extension.
J Bone Miner Res. 27:694, 2012.
27. Kanis JA, Harvey NC, Lorentzon M, et al. Combining fracture
outcomes in phase 3 trials of osteoporosis: An analysis of the
eects of denosumab in postmenopausal women. Osteoporos Int.
32:165, 2021.
28. Miller PD, Pannacciulli N, Malouf-Sierra J, et al. Ecacy and
safety of denosumab vs. bisphosphonates in postmenopausal
women previously treated with oral bisphosphonates. Osteoporos
Int. 31:181, 2020.
29. Terpos E, Raje N, Croucher P, et al. Denosumab compared with
zoledronic acid on PFS in multiple myeloma: Exploratory results
of an international phase 3 study. Blood Adv. 5:725, 2021.
30. Fizazi K, Carducci M, Smith M, et al. Denosumab versus
zoledronic acid for treatment of bone metastases in men with
castration-resistant prostate cancer: A randomized, double-blind
study. Lancet. 377:813, 2011.
31. Stopeck AT, Lipton A, Body JJ, et al. Denosumab compared with
zoledronic acid for the treatment of bone metastases in patients
with advanced breast cancer: A randomized, double-blind study.
J Clin Oncol. 28:5132, 2010.
32. Chawla S, Henshaw R, Seeger L, et al. Safety and ecacy of
denosumab for adults and skeletally mature adolescents with giant
cell tumour of bone: Interim analysis of an open-label, parallel-
group, phase 2 study. Lancet Oncol. 14:901, 2013.
33. Bredell M, Rordorf T, Kroiss S, et al. Denosumab as a treatment
alternative for central giant cell granuloma: A long-term
retrospective cohort study. J Oral Maxillofac Surg. 76:775, 2018.
34. Boyce AM. Denosumab: An emerging therapy in pediatric bone
Disorders. Curr Osteoporos Rep. 15:283, 2017.
35. de Castro LF, Burke AB, Wang HD, et al. Activation of RANK/
RANKL/OPG pathway is involved in the pathophysiology of
brous dysplasia and associated with disease burden. J Bone Miner
Res. 34:290, 2019.
36. Palmisano B, Spica E, Remoli C, et al. RANKL inhibition in
brous dysplasia of bone: A preclinical study in a mouse model of
the human disease. J Bone Miner Res. 34:2171, 2019.
37. Saag KG, Petersen J, Brandi ML, et al. Romosozumab or
alendronate for fracture prevention in women with osteoporosis.
N Engl J Med. 377:1417, 2017.
38. Campisi G, Mauceri R, Bertoldo F, et al. Medication-related
osteonecrosis of jaws (MRONJ) prevention and diagnosis: Italian
consensus update 2020. Int J Environ Res Public Health. 17, 2020.
39. Steinberg ME, Hayken GD, Steinberg DR. A quantitative system
for staging avascular necrosis. J Bone Joint Surg Br. 77:34, 1995.
40. Fleisher KE, Welch G, Kottal S, et al. Predicting risk for
bisphosphonate-related osteonecrosis of the jaws: CTX versus
radiographic markers. Oral Surg Oral Med Oral Pathol Oral Radiol
Endod. 110:509, 2010.
41. Fedele S, Porter SR, D’Aiuto F, et al. Nonexposed variant of
bisphosphonate-associated osteonecrosis of the jaw: A case series.
Am J Med. 123:1060, 2010.
42. Fleisher KE, Janal MN, Albstein N, et al. Comorbid conditions
are a risk for osteonecrosis of the jaw unrelated to antiresorptive
therapy. Oral Surg Oral Med Oral Pathol Oral Radiol. 127: 140,
2019.
43. Farah CS, Savage NW. Oral ulceration with bone sequestration.
Aust Dent J. 48:61, 2003.
44. Filippi A, Dreyer T, Bohle RM, et al. Sequestration of the alveolar
bone by invasive aspergillosis in acute myeloid leukemia. J Oral
Pathol Med. 26:437, 1997.
45. Friel P, Macintyre DR. Bone sequestration from lower 3rd molar
region. Br Dent J. 193:366, 2002.
46. Huang JS, Kok SH, Lee JJ, et al. Extensive maxillary sequestration
resulting from mucormycosis. Br J Oral Maxillofac Surg. 43:532,
2005.
47. Peters E, Daley T. American Academy of Oral and Maxillofacial
Pathology. Persistent painful ulcer of the posterior lingual
mandibular mucosa. J Contemp Dent Pract. 4:71, 2003.
48. Sonnier KE, Horning GM. Spontaneous bony exposure: A report
of 4 cases of idiopathic exposure and sequestration of alveolar
bone. J Periodontol. 68:758, 1997.
49. Peters E, Lovas GL, Wysocki GP. Lingual mandibular
sequestration and ulceration. Oral Surg Oral Med Oral Pathol.
75:739, 1993.
50. Nandakumar H, Shankaramba KB. Massive sequestration of the
upper jaw: A case report. Br J Oral Maxillofac Surg. 28:55, 1990.
51. Ramon Y, Oberman M, Horowitz I, Freedman. Extensive
maxillary sequestration resulting from rhinocerebral
mucormyocosis. 35:989, 1977.
52. Liao MT, Chien WC, Wang JC, et al. Increased risk of
bisphosphonate-related osteonecrosis of the jaw in patients with
Sjogren’s syndrome: Nationwide population-based cohort study.
BMJ Open. 9:e024655, 2019.
PAGE 26 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
53. Schwartz HC. Osteonecrosis of the jaws. A complication of cancer
chemotherapy. Head Neck Surg. 4:251, 1982.
54. Cooper JC. Tooth exfoliation and osteonecrosis of the jaw
following herpes zoster. Br Dent J. 143:297, 1977.
55. Schwartz O, Kvorning SA. Tooth exfoliation, osteonecrosis of the
jaw and neuralgia following herpes zoster of the trigeminal nerve.
Int J Oral Surg. 11:364, 1982.
56. Calhoun KH, Shapiro RD, Stiernberg CM, et al. Osteomyelitis of
the mandible. Arch Otolaryngol Head Neck Surg. 114:1157, 1988.
57. Koorbusch GF, Fotos P, Goll KT. Retrospective assessment
of osteomyelitis. Etiology, demographics, risk factors, and
management in 35 cases. Oral Surg Oral Med Oral Pathol. 74:149,
1992.
58. Maines E, Monti E, Doro F, et al. Children and adolescents treated
with neridronate for osteogenesis imperfecta show no evidence of
any osteonecrosis of the jaw. J Bone Miner Metab. 30:434, 2012.
59. Black DM, Schwartz AV, Ensrud KE, et al. Eects of continuing
or stopping alendronate after 5 years of treatment: The fracture
intervention trial long-term extension (FLEX): A randomized trial.
JAMA. 296:2927, 2006.
60. Black DM, Reid IR, Boonen S, et al. The eect of 3 versus 6
years of zoledronic acid treatment of osteoporosis: A randomized
extension to the HORIZON-Pivotal fracture trial (PFT). J Bone
Miner Res. 27:243, 2012.
61. Black DM, Reid IR, Cauley JA, et al. The eect of 6 versus 9 years
of zoledronic acid treatment in osteoporosis: A randomized second
extension to the HORIZON-Pivotal fracture trial (PFT). J Bone
Miner Res. 30:934, 2015.
62. Carlson ER, Fleisher KE, Ruggiero SL. Metastatic cancer
identied in osteonecrosis specimens of the jaws in patients
receiving intravenous bisphosphonate medications. J Oral
Maxillofac Surg. 71:2077, 2013.
63. Hadaya D, Soundia A, Freymiller E, et al. Nonsurgical
management of medication-related osteonecrosis of the jaws using
local wound care. J Oral Maxillofac Surg. 76:2332, 2018.
64. Marx RE, Sawatari Y, Fortin M, Broumand V. Bisphosphonate-
induced exposed bone (osteonecrosis/osteopetrosis) of the jaws:
Risk factors, recognition, prevention, and treatment. J Oral
Maxillofac Surg. 63:1567, 2005.
65. Aghaloo T, Hazboun R, Tetradis S. Pathophysiology of
osteonecrosis of the jaws. Oral Maxillofac Surg Clin North Am.
27:489, 2015.
66. Johnston CB, Dagar M. Osteoporosis in older adults. Med Clin
North Am 104:873, 2020.
67. Black DM, Rosen CJ. Clinical practice. Postmenopausal
Osteoporosis. N Engl J Med. 374:254, 2016.
68. Schwartz E, Reichert Z, Van Poznak C. Pharmacologic
management of metastatic bone disease. Bone. 115735, 2020.
69. Coleman R. Bisphosphonates and breast cancer from cautious
palliation to saving lives. Bone. 140:115570, 2020.
70. Lacey DL, Boyle WJ, Simonet WS, et al. Bench to bedside:
Elucidation of the OPG-RANK-RANKL pathway and the
development of denosumab. Nat Rev Drug Discov. 11:401, 2012.
71. Coleman RE, Major P, Lipton A, et a.: Predictive value of bone
resorption and formation markers in cancer patients with bone
metastases receiving the bisphosphonate zoledronic acid. J Clin
Oncol. 23:4925, 2005.
72. Stewart AF. Clinical practice. Hypercalcemia associated with
cancer. N Engl J Med. 352:373, 2005.
73. Benjamin B, Benjamin MA, Swe M, Sugathan S. Review
on the comparison of eectiveness between denosumab and
bisphosphonates in post-menopausal osteoporosis. Osteoporos
Sarcopenia. 2:77, 2016.
74. Limones A, Saez-Alcaide LM, Diaz-Parreno SA, et al. Medication-
related osteonecrosis of the jaws (MRONJ) in cancer patients
treated with denosumab vs.. zoledronic acid: A systematic review
and meta-analysis. Med Oral Patol Oral Cir Bucal. 25: e326, 2020.
75. Hallmer F, Andersson G, Gotrick B, et al. Prevalence, initiating
factor, and treatment outcome of medication-related osteonecrosis
of the jaw-a 4-year prospective study. Oral Surg Oral Med Oral
Pathol Oral Radiol. 126:477, 2018.
76. Soundia A, Hadaya D, Esfandi N, et al. Osteonecrosis of the jaws
(ONJ) in mice after extraction of teeth with periradicular disease.
Bone. 90:133, 2016.
77. Wehrhan F, Gross C, Creutzburg K, et al. Osteoclastic expression
of higher-level regulators NFATc1 and BCL6 in medication-related
osteonecrosis of the jaw secondary to bisphosphonate therapy: A
comparison with osteoradionecrosis and osteomyelitis. J Transl
Med. 17:69, 2019.
78. de Molon RS, Shimamoto H, Bezouglaia O, et al. OPG-Fc but not
zoledronic acid discontinuation reverses osteonecrosis of the jaws
(ONJ) in mice. J Bone Miner Res .30:1627, 2015.
79. Hadaya D, Soundia A, Gkouveris I, et al. Antiresorptive-type and
discontinuation-timing Aect ONJ burden. J Dent Res. 100:746,
2021.
PAGE 27 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
80. Kuroshima S, Entezami P, McCauley LK, Yamashita J. Early
eects of parathyroid hormone on bisphosphonate/steroid-
associated compromised osseous wound healing. Osteoporosis Int.
25:1141, 2014.
81. Dayisoylu EH, Senel FC, Ungor C, et al. The eects of adjunctive
parathyroid hormone injection on bisphosphonate-related
osteonecrosis of the jaws: An animal study. Int J Oral Maxillofac
Surg. 42:1475, 2013.
82. Chopra K, Malhan N. Teriparatide for the treatment of medication-
related osteonecrosis of the jaw. Am J Ther. 28: e469, 2021.
83. Ficarra G, Beninati F, Rubino I, et al. Osteonecrosis of the jaws
in periodontal patients with a history of bisphosphonates treatment.
J Clin Periodontol. 32:1123, 2005.
84. Thumbigere-Math V, Michalowicz BS, Hodges JS, et al.
Periodontal disease as a risk factor for bisphosphonate-related
osteonecrosis of the jaw. J Periodontol. 85:226, 2014.
85. Aghaloo TL, Kang B, Sung EC, et al. Periodontal disease and
bisphosphonates induce osteonecrosis of the jaws in the rat. J Bone
Miner Res. 26:1871, 2011.
86. Kang B, Cheong S, Chaichanasakul T, et al. Periapical disease and
bisphosphonates induce osteonecrosis of the jaws in mice. J Bone
Miner Res. 28:1631, 2013.
87. Hadaya D, Soundia A, Gkouveris I, et al. Development of
medication-related osteonecrosis of the jaw after extraction of
teeth with experimental periapical disease. J Oral Maxillofac Surg.
77:71, 2019.
88. Aguirre JI, Akhter MP, Kimmel DB, et al. Oncologic doses of
zoledronic acid induce osteonecrosis of the jaw-like lesions in rice
rats (Oryzomys palustris) with periodontitis. J Bone Miner Res.
27:2130, 2012.
89. Soma T, Iwasaki R, Sato Y, et al. Tooth extraction in mice
administered zoledronate increases inammatory cytokine levels
and promotes osteonecrosis of the jaw. J Bone Miner Metab.
39:372, 2021
90. de Molon RS, Hsu C, Bezouglaia O, et al. Rheumatoid arthritis
Exacerbates the severity of osteonecrosis of the jaws (ONJ) in
mice. A randomized, prospective, controlled animal study. J Bone
Miner Res. 31:1596, 2016.
91. Kim T, Kim S, Song M, et al. Removal of pre-existing periodontal
inammatory condition before tooth extraction Ameliorates
medication-related osteonecrosis of the jaw-like lesion in mice.
Am J Pathol. 188:2318, 2018.
92. Kuroshima S, Nakajima K, Sasaki M, et al. Systemic
administration of quality and quantity-controlled PBMNCs reduces
bisphosphonate-related osteonecrosis of jaw-like lesions in mice.
Stem Cell Res Ther. 10:209, 2019.
93. Hansen T, Kunkel M, Weber A, James Kirkpatrick C.
Osteonecrosis of the jaws in patients treated with bisphosphonates
histomorphologic analysis in comparison with infected
osteoradionecrosis. J Oral Pathol Med. 35:155, 2006.
94. Mawardi H, Giro G, Kajiya M, et al. A role of oral bacteria in
bisphosphonate-induced osteonecrosis of the jaw. J Dent Res.
90:1339, 2011.
95. Vandone AM, Donadio M, Mozzati M, et al. Impact of dental care
in the prevention of bisphosphonate-associated osteonecrosis of the
jaw: A single-center clinical experience. Ann Oncol. 23:193, 2012.
96. Sedghizadeh PP, Kumar SK, Gorur A, et al. Microbial biolms in
osteomyelitis of the jaw and osteonecrosis of the jaw secondary to
bisphosphonate therapy. J Am Dent Assoc. 140:1259, 2009.
97. Francini F, Pascucci A, Francini E, et al. Osteonecrosis of the
jaw in patients with cancer who received zoledronic acid and
bevacizumab. J Am Dent Assoc. 142:506, 2011.
98. Migliorati CA, Schubert MM, Peterson DE, Seneda LM.
Bisphosphonate-associated osteonecrosis of mandibular and
maxillary bone: An emerging oral complication of supportive
cancer therapy. Cancer. 104:83, 2005.
99. Petek D, Hannouche D, Suva D. Osteonecrosis of the femoral
head: Pathophysiology and current concepts of treatment. EFORT
Open Rev. 4:85, 2019.
100. Wood J, Bonjean K, Ruetz S, et al. Novel antiangiogenic eects of
the bisphosphonate compound zoledronic acid. J Pharmacol Exp
Ther. 302:1055, 2002.
101. Santini D, Vincenzi B, Dicuonzo G, et al. Zoledronic acid induces
signicant and long-lasting modications of circulating angiogenic
factors in cancer patients. Clin Cancer Res. 9: 2893, 2003.
102. Ishtiaq S, Edwards S, Sankaralingam A, et al. The eect of
nitrogen containing bisphosphonates, zoledronate and alendronate,
on the production of pro-angiogenic factors by osteoblastic cells.
Cytokine. 71:154, 2015.
103. Kun-Darbois JD, Libouban H, Mabilleau G, et a.: Bone
mineralization and vascularization in bisphosphonate-related
osteonecrosis of the jaw: An experimental study in the rat. Clin
Oral Investig. 22:2997, 2018.
104. Gao SY, Lin RB, Huang SH, et al. PDGF-BB exhibited therapeutic
eects on rat model of bisphosphonate-related osteonecrosis
of the jaw by enhancing angiogenesis and osteogenesis. Bone.
144:115117, 2021.
105. Bi Y, Gao Y, Ehirchiou D, et al. Bisphosphonates cause
osteonecrosis of the jaw-like disease in mice. Am J Pathol.
177:280, 2010.
106. Gkouveris I, Hadaya D, Soundia A, et a.: Vasculature submucosal
changes at early stages of osteonecrosis of the jaw (ONJ). Bone.
123:234, 2019.
PAGE 28 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
107. Vallina C, Ramirez L, Torres J, et al. Osteonecrosis of the jaws
produced by sunitinib: A systematic review. Med Oral Patol Oral
Cir Bucal. 24:e326, 2019.
108. Gacche RN, Meshram RJ. Angiogenic factors as potential drug
target: Ecacy and limitations of anti-angiogenic therapy. Biochim
Biophys Acta. 1846(1):161, 2014.
109. Akita Y, Kuroshima S, Nakajima K, et al. Eect of anti-
angiogenesis induced by chemotherapeutic monotherapy,
chemotherapeutic/bisphosphonate combination therapy and
anti-VEGFA mAb therapy on tooth extraction socket healing
in mice. J Bone Miner Metab. 36:547, 2018.
110. Rugani P, Walter C, Kirnbauer B, et al. Prevalence of medication-
related osteonecrosis of the jaw in patients with breast cancer,
prostate cancer, and multiple myeloma. Dent J (Basel). 4:32, 2016.
111. Dimopoulos MA, Kastritis E, Anagnostopoulos A, et al.
Osteonecrosis of the jaw in patients with multiple myeloma treated
with bisphosphonates: Evidence of increased risk after treatment
with zoledronic acid. Haematologica. 91:968, 2006.
112. Badros A, Weikel D, Salama A, et al. Osteonecrosis of the jaw
in multiple myeloma patients: Clinical features and risk factors.
J Clin Oncol. 24:945, 2006.
113. Raje N, Terpos E, Willenbacher W, et al. Denosumab versus
zoledronic acid in bone disease treatment of newly diagnosed
multiple myeloma: An international, double-blind, double-dummy,
randomised, controlled, phase 3 study. Lancet Oncol. 19:370,
2018.
114. Filleul O, Crompot E, Saussez S. Bisphosphonate-induced
osteonecrosis of the jaw: A review of 2,400 patient cases. J Cancer
Res Clin Oncol. 136:1117, 2010.
115. Bastos P, Patel V, Festy F, et al. In-vivo imaging of the
microvasculature of the soft tissue margins of osteonecrotic jaw
lesions. Br Dent J. 223:699, 2017.
116. Zhang Q, Yu W, Lee S, et al. Bisphosphonate induces osteonecrosis
of the jaw in diabetic mice via NLRP3/Caspase-1Dependent IL-1
beta mechanism. J Bone Miner Res. 30:2300, 2015.
117. Kabilova TO, Kovtonyuk LV, Zonov EV, et al. Immunotherapy of
hepatocellular carcinoma with small double-stranded RNA. BMC
Cancer. 14:338, 2014.
118. Hayano H, Kuroshima S, Sasaki M, et al. Distinct
immunopathology in the early stages between dierent
antiresorptives-related osteonecrosis of the jaw-like lesions in
mice. Bone. 135:115308, 2020.
119. Rao NJ, Yu RQ, Wang JY, et al. Eect of periapical diseases in
development of MRONJ in immunocompromised mouse model.
Biomed Res Int. 2019:1271492, 2019.
120. Aghaloo TL, Tetradis S. Osteonecrosis of the jaw in the absence
of antiresorptive or antiangiogenic exposure: A series of 6 cases.
J Oral Maxillofac Surg. 75:129, 2017.
121. Bamias A, Kastritis E, Bamia C, et a.: Osteonecrosis of the jaw in
cancer after treatment with bisphosphonates: Incidence and risk
factors. J Clin Oncol. 23:8580, 2005.
122. Qu X, Wang Z, Zhou T, Shan L. Determination of the molecular
mechanism by which macrophages and gammadelta-T cells
contribute to ZOL-induced ONJ. Aging (Albany NY). 12: 20743,
2020.
123. Rodriguez-Lozano FJ, Onate-Sanchez R, Gonzalvez-Garcia M, et
al. Allogeneic bone marrow mesenchymal stem cell transplantation
in tooth extractions sites Ameliorates the incidence of osteonecrotic
jaw-like lesions in zoledronic acid-treated rats. J Clin Med. 9:1649,
2020.
124. Alonso-Rodriguez E, Gonzalez-Martin-Moro J, Cebrian-Carretero
JL, et al. Bisphosphonate-related osteonecrosis. Application of
adipose-derived stem cells in an experimental murine model. Med
Oral Patol Oral Cir Bucal. 24: e529, 2019.
125. Barba-Recreo P, Del Castillo Pardo de Vera JL, Georgiev-Hristov
T, et al. Adipose-derived stem cells and platelet-rich plasma for
preventive treatment of bisphosphonate-related osteonecrosis of
the jaw in a murine model. J Craniomaxillofac Surg. 43:1161,
2015.
126. Sarasquete ME, Garcia-Sanz R, Marin L, et al. Bisphosphonate-
related osteonecrosis of the jaw is associated with polymorphisms
of the cytochrome P450 CYP2C8 in multiple myeloma: A genome-
wide single nucleotide polymorphism analysis. Blood. 112:2709,
2008.
127. Yang G, Hamadeh IS, Katz J, et al. SIRT1/HERC4 Locus
associated with bisphosphonate-induced osteonecrosis of the jaw:
An Exome-wide association analysis. J Bone Miner Res. 33:91,
2018.
128. Yang G, Collins JM, Raee R, et al. SIRT1 gene SNP rs932658 is
associated with medication-related osteonecrosis of the jaw. J Bone
Miner Res. 36:347, 2021.
129. Lee KH, Kim SH, Kim CH, et a.: Identifying genetic variants
underlying medication-induced osteonecrosis of the jaw in cancer
and osteoporosis: A case control study. J Transl Med. 17: 381,
2019.
130. Kastritis E, Melea P, Bagratuni T, et a.: Genetic factors related with
early onset of osteonecrosis of the jaw in patients with multiple
myeloma under zoledronic acid therapy. Leuk Lymphoma.
58:2304, 2017.
131. Guo Z, Cui W, Que L, et al. Pharmacogenetics of medication-
related osteonecrosis of the jaw: A systematic review and meta-
analysis. Int J Oral Maxillofac Surg. 49:298, 2020.
PAGE 29 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
132. Valachis A, Polyzos NP, Coleman RE, et al. Adjuvant therapy with
zoledronic acid in patients with breast cancer: A systematic review
and meta-analysis. Oncologist. 18:353, 2013.
133. Coleman R, Cameron D, Dodwell D, et al. Adjuvant zoledronic
acid in patients with early breast cancer: Final ecacy analysis
of the AZURE (BIG 01/04) randomised open-label phase 3 trial.
Lancet Oncol. 15:997, 2014.
134. Gnant M, Pfeiler G, Dubsky PC et al. Adjuvant denosumab in
breast cancer (ABCSG-18): A multicentre, randomised, double
blind, placebo-controlled trial. Lancet. 386:433, 2015.
135. Boquete-Castro A, Gomez-Moreno G, Calvo-Guirado JL, et al:
Denosumab and osteonecrosis of the jaw. A systematic analysis of
events reported in clinical trials. Clin Oral Implants Res. 27: 367,
2016.
136. Macherey S, Monsef I, Jahn F, et al. Bisphosphonates for advanced
prostate cancer. Cochrane Database Syst Rev. 12: CD006250,
2017.
137. O’Carrigan B, Wong MH, Willson ML, et a.: Bisphosphonates and
other bone agents for breast cancer. Cochrane Database Syst Rev.
10:CD003474, 2017.
138. Coleman R, Finkelstein DM, Barrios C, et al. Adjuvant denosumab
in early breast cancer (D-CARE): An international, multicentre,
randomised, controlled, phase 3 trial. Lancet Oncol. 21:60, 2020.
139. Barrett-Lee P, Casbard A, Abraham J, et al. Oral ibandronic acid
versus intravenous zoledronic acid in treatment of bone metastases
from breast cancer: A randomised, open label, noninferiority phase
3 trial. Lancet Oncol. 15:114, 2014.
140. Himelstein AL, Foster JC, Khatcheressian JL, et al. Eect of
longer-Interval vs standard dosing of zoledronic acid on skeletal
events in patients with bone metastases: A randomized clinical
trial. JAMA. 317:48, 2017.
141. Henry D, Vadhan-Raj S, Hirsh V, et al. Delaying skeletal-related
events in a randomized phase 3 study of denosumab versus
zoledronic acid in patients with advanced cancer: An analysis of
data from patients with solid tumors. Support Care Cancer. 22:
679, 2014.
142. Peddi P, Lopez-Olivo MA, Pratt GF, Suarez-Almazor ME.
Denosumab in patients with cancer and skeletal metastases: A
systematic review and meta-analysis. Cancer Treat Rev. 39: 97,
2013.
143. Jackson GH, Morgan GJ, Davies FE, et al. Osteonecrosis of the
jaw and renal safety in patients with newly diagnosed multiple
myeloma: Medical Research Council Myeloma IX Study results.
Br J Haematol. 166:109, 2014.
144. Wang X, Yang KH, Wanyan P, Tian JH. Comparison of the
ecacy and safety of denosumab versus bisphosphonates in
breast cancer and bone metastases treatment: A meta-analysis of
randomized controlled trials. Oncol Lett. 7: 1997, 2014.
145. Ng TL, Tu MM, Ibrahim MFK, et al. Long-term impact
of bone-modifying agents for the treatment of bone metastases:
A systematic review. Support Care Cancer. 29:925, 2021.
146. Fusco V, Santini D, Armento G, et al. Osteonecrosis of jaw beyond
antiresorptive (bone-targeted) agents: New horizons in oncology.
Expert Opin Drug Saf. 15:925, 2016.
147. Nicolatou-Galitis O, Kouri M, Papadopoulou E, et al.
Osteonecrosis of the jaw related to non-antiresorptive medications:
A systematic review. Support Care Cancer. 27:383, 2019.
148. King R, Tanna N, Patel V. Medication-related osteonecrosis of the
jaw unrelated to bisphosphonates and denosumab-a review. Oral
Surg Oral Med Oral Pathol Oral Radiol. 127:289, 2019.
149. Sacco R, Shah S, Leeson R, et al. Osteonecrosis and osteomyelitis
of the jaw associated with tumour necrosis factor-alpha (TNF-
alpha) inhibitors: A systematic review. Br J Oral Maxillofac Surg.
58:25, 2020
150. Grbic JT, Black DM, Lyles KW, et al. The incidence of
osteonecrosis of the jaw in patients receiving 5 milligrams of
zoledronic acid: Data from the health outcomes and reduced
incidence with zoledronic acid once yearly clinical trials program.
J Am Dent Assoc. 141:1365, 2010..
151. Cosman F, Crittenden DB, Adachi JD, et al. Romosozumab
treatment in postmenopausal women with osteoporosis. N Engl J
Med. 375:1532, 2016.
152. Administration USFaD. Background Document for Meeting of
Advisory Committee for Reproductive Health Drugs and Drug
Safety and Risk Management Advisory Committee. Adelphi, MD,
FDA, HHS, 2011.
153. Bone HG, Wagman RB, Brandi ML, et al. 10 years of denosumab
treatment in postmenopausal women with osteoporosis: Results
from the phase 3 randomised FREEDOM trial and open-label
extension. Lancet Diabetes Endocrinol. 5:513, 2017.
154. FDA. Brieng Information for the September 9, 2011 Joint
Meeting of the Reproductive Health Drugs Advisory Committee
and the Drug Safety and Risk Management Advisory Committee.
Adelphi, MD, FDA, HHS, 2011.
155. Rutkowski P, Gaston L, Borkowska A, et al. Denosumab
treatment of inoperable or locally advanced giant cell tumor of
bone multicenter analysis outside clinical trial. Eur J Surg Oncol.
44:1384, 2018.
156. Chawla S, Blay JY, Rutkowski P, et al. Denosumab in patients with
giant-cell tumour of bone: A multicentre, open-label, phase 2 study.
Lancet Oncol. 20:1719, 2019.
PAGE 30 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
157. Hennedige AA, Jayasinghe J, Khajeh J, Macfarlane TV. Systematic
review on the incidence of bisphosphonate related osteonecrosis of
the jaw in children diagnosed with osteogenesis imperfecta. J Oral
Maxillofac Res. 4:e1, 2013.
158. Duarte NT, Rech BO, Martins IG, et al. Can children be aected
by bisphosphonate-related osteonecrosis of the jaw? A systematic
review. Int J Oral Maxillofac Surg. 49:183, 2020.
159. Lo JC, O'Ryan FS, Gordon NP et al. Prevalence of osteonecrosis
of the jaw in patients with oral bisphosphonate exposure. J Oral
Maxillofac Surg. 68: 243, 2010.
160. Aljohani S, Fliefel R, Ihbe J, et al. What is the eect of anti-
resorptive drugs (ARDs) on the development of medication-related
osteonecrosis of the jaw (MRONJ) in osteoporosis patients: A
systematic review. J Craniomaxillofac Surg. 45:1493, 2017.
161. Shudo A, Kishimoto H, Takaoka K, Noguchi K. Long-term oral
bisphosphonates delay healing after tooth extraction: A single
institutional prospective study. Osteoporos Int. 29:2315, 2018.
162. Gaudin E, Seidel L, Bacevic M, et al. Occurrence and risk
indicators of medication-related osteonecrosis of the jaw after
dental extraction: A systematic review and meta-analysis. J Clin
Periodontol. 42:922, 2015.
163. Watts NB, Grbic JT, Binkley N, et al. Invasive oral procedures and
events in postmenopausal women with osteoporosis treated with
denosumab for up to 10 Years. J Clin Endocrinol Metab. 104:2443,
2019.
164. Mozzati M, Arata V, Gallesio G. Tooth extraction in patients on
zoledronic acid therapy. Oral Oncol. 48:817, 2012.
165. Yamazaki T, Yamori M, Ishizaki T, et al. Increased incidence of
osteonecrosis of the jaw after tooth extraction in patients treated
with bisphosphonates: A cohort study. Int J Oral Maxillofac Surg.
41:1397, 2012.
166. Scoletta M, Arata V, Arduino PG, et al. Tooth extractions in
intravenous bisphosphonate-treated patients: A rened protocol.
J Oral Maxillofac Surg. 71:994, 2013.
167. Bodem JP, Kargus S, Eckstein S, et al. Incidence of
bisphosphonate-related osteonecrosis of the jaw in high-risk
patients undergoing surgical tooth extraction. J Craniomaxillofac
Surg. 43:510, 2015.
168. McGowan K, McGowan T, Ivanovski S. Risk factors for
medication-related osteonecrosis of the jaws: A systematic review.
Oral Dis. 24:527, 2018.
169. Kyrgidis A, Vahtsevanos K, Koloutsos G, et al. Bisphosphonate-
related osteonecrosis of the jaws: A case-control study of risk
factors in breast cancer patients. J Clin Oncol. 26:4634, 2008.
170. Vahtsevanos K, Kyrgidis A, Verrou E, et al. Longitudinal cohort
study of risk factors in cancer patients of bisphosphonate-related
osteonecrosis of the jaw. J Clin Oncol. 27:5356, 2009.
171. Tsao C, Darby I, Ebeling PR, et al. Oral health risk factors for
bisphosphonate-associated jaw osteonecrosis. J Oral Maxillofac
Surg. 71:1360, 2013.
172. Qi WX, Tang LN, He AN, et al. Risk of osteonecrosis of the jaw
in cancer patients receiving denosumab: A meta-analysis of seven
randomized controlled trials. Int J Clin Oncol. 19:403, 2014.
173. Ripamonti CI, Maniezzo M, Campa T, et al. Decreased occurrence
of osteonecrosis of the jaw after implementation of dental
preventive measures in solid tumour patients with bone metastases
treated with bisphosphonates. The experience of the National
Cancer Institute of Milan. Ann Oncol. 20:137, 2009.
174. Montefusco V, Gay F, Spina F, et al. Antibiotic prophylaxis before
dental procedures may reduce the incidence of osteonecrosis
of the jaw in patients with multiple myeloma treated with
bisphosphonates. Leuk Lymphoma. 49:2156, 2008.
175. Bantis A, Zissimopoulos A, Sountoulides P, et al. Bisphosphonate-
induced osteonecrosis of the jaw in patients with bone metastatic,
hormone-sensitive prostate cancer. Risk factors and prevention
strategies. Tumori. 97:479, 2011.
176. Kunchur R, Goss AN. The oral health status of patients on oral
bisphosphonates for osteoporosis. Aust Dent J. 53:354, 2008.
177. Lodi G, Sardella A, Salis A, et al. Tooth extraction in patients
taking intravenous bisphosphonates: A preventive protocol and
case series. J Oral Maxillofac Surg. 68:107, 2010.
178. Mozzati M, Gallesio G, Arata V, et al. Platelet-rich therapies in the
treatment of intravenous bisphosphonate-related osteonecrosis of
the jaw: A report of 32 cases. Oral Oncol. 48:469, 2012.
179. Ferlito S, Puzzo S, Liardo C. Preventive protocol for tooth
extractions in patients treated with zoledronate: A case series.
J Oral Maxillofac Surg. 69:e1, 2011.
180. Schubert M, Klatte I, Linek W, et al. The saxon bisphosphonate
register - therapy and prevention of bisphosphonate-related
osteonecrosis of the jaws. Oral Oncol. 48:349, 2012.
181. Bonacina R, Mariani U, Villa F, Villa A. Preventive strategies and
clinical implications for bisphosphonate-related osteonecrosis of
the jaw: A review of 282 patients. J Can Dent Assoc. 77:b147,
2011.
182. Khan AA, Morrison A, Hanley DA, et al. International Task
Force on Osteonecrosis of the Jaw. Diagnosis and management
of osteonecrosis of the jaw: a systematic review and international
consensus. J Bone Miner Res. 30:3, 2015.
PAGE 31 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
183. Japanese Allied Committee on Osteonecrosis of the Jaw, Yoneda
T, Hagino H, Sugimoto T, et al. Antiresorptive agent-related
osteonecrosis of the jaw: Position paper 2017 of the Japanese
Allied Committee on osteonecrosis of the jaw. J Bone Miner
Metab. 35: 6, 2017.
184. Aparecida Cariolatto F, Carelli J, de Campos Moreira T, et al.
Recommendations for the prevention of bisphosphonate-related
osteonecrosis of the jaw: A systematic review. J Evid Based Dent
Pract. 18:142, 2018.
185. Beth-Tasdogan NH, Mayer B, Hussein H, Zolk O. Interventions
for managing medication-related osteonecrosis of the jaw.
Cochrane Database Syst Rev. 10:CD012432, 2017.
186. Mucke T, Deppe H, Hein J, et al. Prevention of bisphosphonate-
related osteonecrosis of the jaws in patients with prostate cancer
treated with zoledronic acid - a prospective study over 6 years.
J Craniomaxillofac Surg. 44:1689, 2016.
187. Smidt-Hansen T, Folkmar TB, Fode K, et al. Combination of
zoledronic Acid and targeted therapy is active but may induce
osteonecrosis of the jaw in patients with metastatic renal cell
carcinoma. J Oral Maxillofac Surg. 71:1532, 2013.
188. Sivolella S, Lumachi F, Stellini E, Favero L. Denosumab and anti-
angiogenetic drug-related osteonecrosis of the jaw: An uncommon
but potentially severe disease. Anticancer Res. 33:1793, 2013.
189. Patel V, McLeod NM, Rogers SN, Brennan PA. Bisphosphonate
osteonecrosis of the jaw–a literature review of UK policies
versus international policies on bisphosphonates, risk factors and
prevention. Br J Oral Maxillofac Surg. 49:251, 2011.
190. Hellstein JW, Adler RA, Edwards B, et al. Ristic H, American
Dental Association Council on scientic Aairs Expert Panel on
antiresorptive Agents: Managing the care of patients receiving
antiresorptive therapy for prevention and treatment of osteoporosis:
Executive summary of recommendations from the American
Dental Association Council on scientic Aairs. J Am Dent Assoc.
142:1243, 2011.
191. Atalay B, Yalcin S, Emes Y, et al. Bisphosphonate-related
osteonecrosis: Laser-assisted surgical treatment or conventional
surgery? Lasers Med Sci. 26:815, 2011.
192. Guarneri V, Miles D, Robert N, et al. Bevacizumab and
osteonecrosis of the jaw: Incidence and association with
bisphosphonate therapy in three large prospective trials in
advanced breast cancer. Breast Cancer Res Treat. 122:181, 2010.
193. Edwards BJ, Hellstein JW, Jacobsen PL, et al. American Dental
Association Council on scientic Aairs Expert Panel on
bisphosphonate-associated osteonecrosis of the Jaw: Updated
recommendations for managing the care of patients receiving oral
bisphosphonate therapy: An advisory statement from the American
Dental Association Council on scientic Aairs. J Am Dent Assoc.
139:1674, 2008.
194. de-Freitas NR, Lima LB, de-Moura MB, et al. Bisphosphonate
treatment and dental implants: A systematic review. Med Oral
Patol Oral Cir Bucal. 21:e644, 2016.
195. de Molon RS, Cheong S, Bezouglaia O, et al. Spontaneous
osteonecrosis of the jaws in the maxilla of mice on antiresorptive
treatment: A novel ONJ mouse model. Bone. 68:11, 2014.
196. Messer JG, Jiron JM, Mendieta Calle JL, et al. Zoledronate
treatment duration is linked to bisphosphonate-related
osteonecrosis of the jaw prevalence in rice rats with generalized
periodontitis. Oral Dis. 25:1116, 2019.
197. Hinchy NV, Jayaprakash V, Rossitto. RA, et al. Osteonecrosis
of the jaw - prevention and treatment strategies for oral health
professionals. Oral Oncol. 49:878, 2013.
198. Khan AA, Morrison A, Kendler DL, et al. International task
Force on osteonecrosis of the Jaw: Case-based review of
osteonecrosis of the jaw (ONJ) and application of the international
recommendations for management from the international task
Force on ONJ. J Clin Densitom. 20:8, 2017.
199. Ottesen C, Schiodt M, Gotfredsen K. Ecacy of a high-dose
antiresorptive drug holiday to reduce the risk of medication-related
osteonecrosis of the jaw (MRONJ): A systematic review. Heliyon.
6:e03795, 2020.
200. Anastasilakis AD, Polyzos SA, Makras P, et al. Clinical features
of 24 patients with rebound-associated vertebral fractures after
denosumab discontinuation: Systematic review and additional
cases. J Bone Miner Res. 32:1291, 2017.
201. Cummings SR, Ferrari S, Eastell R, et al. Vertebral fractures
after discontinuation of denosumab: A post Hoc analysis of the
randomized placebo-controlled FREEDOM trial and its extension.
J Bone Miner Res. 33:190, 2018.
202. Tsourdi E, Langdahl B, Cohen-Solal M, et al. Discontinuation
of denosumab therapy for osteoporosis: A systematic review and
position statement by ECTS. Bone. 105:11, 2017.
203. Lorenzo-Pouso AI, Perez-Sayans M, Gonzalez-Palanca S, et
al. Biomarkers to predict the onset of biphosphonate-related
osteonecrosis of the jaw: A systematic review. Med Oral Patol Oral
Cir Bucal. 24:e26, 2019.
204. Musolino C, Oteri G, Allegra A, et al. Altered microRNA
expression prole in the peripheral lymphoid compartment
of multiple myeloma patients with bisphosphonate-induced
osteonecrosis of the jaw. Ann Hematol. 97:1259, 2018.
205. Yang R, Tao Y, Wang C, et al. Circulating microRNA Panel as a
novel biomarker to Diagnose bisphosphonate-related osteonecrosis
of the jaw. Int J Med Sci. 15:1694, 2018.
PAGE 32 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
206. Durie BG, Katz M, Crowley J. Osteonecrosis of the jaw and
bisphosphonates. N Engl J Med. 353:99, 2005.
207. Ho AO, Toth BB, Altundag K, et al. Frequency and risk factors
associated with osteonecrosis of the jaw in cancer patients treated
with intravenous bisphosphonates. J Bone Miner Res. 23:826,
2008.
208. Dimopoulos MA, Kastritis E, Bamia C, et al. Reduction of
osteonecrosis of the jaw (ONJ) after implementation of preventive
measures in patients with multiple myeloma treated with
zoledronic acid. Ann Oncol. 20:117, 2009.
209. Yarom N, Shapiro CL, Peterson DE, et al. Medication-related
osteonecrosis of the jaw: MASCC/ISOO/ASCO clinical practice
guideline. J Clin Oncol. 37:2270, 2019.
210. Lewiecki EM, Wright NC, Curtis JR, et al. Hip fracture trends in
the United States, 2002 to 2015. Osteoporos Int. 29:717, 2018.
211. Guzon-Illescas O, Perez Fernandez E, Crespi Villarias N, et al.
Mortality after osteoporotic hip fracture: Incidence, trends, and
associated factors. J Orthop Surg Res. 14:203, 2019.
212. McCauley LK. Clinical recommendations for prevention of
secondary fractures in patients with osteoporosis: Implications for
dental care. J Am Dent Assoc. 151:311, 2020.
213. Moinzadeh AT, Shemesh H, Neirynck NA, et al. Bisphosphonates
and their clinical implications in endodontic therapy. Int Endod J.
46:391, 2013.
214. Gelazius R, Poskevicius L, Sakavicius D, et al. Dental implant
placement in patients on bisphosphonate therapy: A systematic
review. J Oral Maxillofac Res. 9:e2, 2018.
215. Holzinger D, Seemann R, Matoni N, et al. Eect of dental implants
on bisphosphonate-related osteonecrosis of the jaws. J Oral
Maxillofac Surg. 72:1937.e1, 2014.
216. Granate-Marques A, Polis-Yanes C, Seminario-Amez M, et al.
Medication-related osteonecrosis of the jaw associated with
implant and regenerative treatments: Systematic review. Med Oral
Patol Oral Cir Bucal. 24:e195, 2019.
217. Stavropoulos A, Bertl K, Pietschmann P, Pandis N, Schiødt M,
Klinge. The eect of antiresorptive drugs on implant therapy:
Systematic review and meta-analysis. B Clin Oral Implants Res.
2018 Oct;29 Suppl 18:54-92.
218. Ryu JI, Kim HY, Kwon YD. Is implant surgery a risk factor for
osteonecrosis of the jaw in older adult patients with osteoporosis?
A national cohort propensity score-matched study. Clin Oral
Implants Res. 32:437, 2021.
219. Giovannacci I, Meleti M, Manfredi M, et al. Medication-related
osteonecrosis of the jaw around dental implants: Implant surgery-
triggered or implant presence-triggered osteonecrosis? J Craniofac
Surg. 27:697, 2016.
220. Kwon TG, Lee CO, Park JW, et al. Osteonecrosis associated with
dental implants in patients undergoing bisphosphonate treatment.
Clin Oral Implants Res. 25:632, 2014.
221. Pogrel MA, Ruggiero SL. Previously successful dental implants
can fail when patients commence anti-resorptive therapy-a case
series. Int J Oral Maxillofac Surg. 47:220, 2018.
222. Walton K, Grogan TR, Eshaghzadeh E, et al. Medication related
osteonecrosis of the jaw in osteoporotic vs oncologic patients -
quantifying radiographic appearance and relationship to clinical
ndings. Dentomaxillofac Radiol. 48:20180128, 2019.
223. Coropciuc RG, Grisar K, Aerden T, et al. Medication-related
osteonecrosis of the jaw in oncological patients with skeletal
metastases: Conservative treatment is eective up to stage 2.
Br J Oral Maxillofac Surg. 55:787, 2017.
224. Akrivoula Soundia, Danny Hadaya, Sanjay M Mallya, Tara L
Aghaloo, Sotirios Tetradis. Radiographic predictors of bone
exposure in patients with stage 0 medication-related osteonecrosis
of the jaws. Surg Oral Med Oral Pathol Oral Radiol. 126(6), 2018.
225. Varoni EM, Lombardi N, Villa G, et al. Conservative management
of medication-related osteonecrosis of the jaws (MRONJ): A
retrospective cohort study. Antibiotics (Basel). 10, 2021.
226. Ripamonti CI, Cislaghi E, Mariani L, Maniezzo M. Ecacy
and safety of medical ozone (O(3)) delivered in oil suspension
applications for the treatment of osteonecrosis of the jaw in
patients with bone metastases treated with bisphosphonates:
Preliminary results of a phase I-II study. Oral Oncol. 47:185, 2011.
227. Freiberger JJ, Padilla-Burgos R, McGraw T, et al. What is the
role of hyperbaric oxygen in the management of bisphosphonate-
related osteonecrosis of the jaw: A randomized controlled trial of
hyperbaric oxygen as an adjunct to surgery and antibiotics. J Oral
Maxillofac Surg. 70:1573, 2012.
228. Epstein MS, Wicknick FW, Epstein JB, et al. Management of
bisphosphonate-associated osteonecrosis: Pentoxifylline and
tocopherol in addition to antimicrobial therapy. An initial case
series. Oral Surg Oral Med Oral Pathol Oral Radiol Endod.
110:593, 2010.
229. Morishita K, Yamada SI, Kawakita A, et al. Treatment outcomes of
adjunctive teriparatide therapy for medication-related osteonecrosis
of the jaw (MRONJ): A multicenter retrospective analysis in Japan.
J Orthop Sci. 25:1079, 2020.
230. Sim IW, Borromeo GL, Tsao C, et al. Teriparatide promotes bone
healing in medication-related osteonecrosis of the jaw: A placebo-
controlled, randomized trial. J Clin Oncol. 38:2971, 2020.
231. Carlson ER, Basile JD. The role of surgical resection in the
management of bisphosphonate-related osteonecrosis of the jaws.
J Oral Maxillofac Surg. 67:85, 2009.
PAGE 33 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
232. Abu-Id MH, Warnke PH, Gottschalk J, et al. "Bisphossy jaws" -
high and low risk factors for bisphosphonate-induced osteonecrosis
of the jaw. J Craniomaxillofac Surg. 36:95, 2008.
233. Wutzl A, Biedermann E, Wanschitz F, et al. Treatment results of
bisphosphonate-related osteonecrosis of the jaws. Head Neck.
30:1224, 2008.
234. Stanton DC, Balasanian E. Outcome of surgical management of
bisphosphonate-related osteonecrosis of the jaws: Review of 33
surgical cases. J Oral Maxillofac Surg. 67:943, 2009.
235. Wilde F, Heufelder M, Winter K, et al. The role of surgical therapy
in the management of intravenous bisphosphonates-related
osteonecrosis of the jaw. Oral Surg Oral Med Oral Pathol Oral
Radiol Endod. 111:153, 2011.
236. Adornato MC, Morcos I, Rozanski J. The treatment of
bisphosphonate-associated osteonecrosis of the jaws with bone
resection and autologous platelet-derived growth factors. J Am
Dent Assoc. 138:971, 2007.
237. Williamson R. Surgical management of bisphosphonate induced
osteonecrosis of the jaws. Int J Oral Maxillofac Surg. 39.:251,
2010.
238. Carlson ER. Management of antiresorptive osteonecrosis of the
jaws with primary surgical resection. J Oral Maxillofac Surg. 72:
655, 2014.
239. Watters AL, Hansen HJ, Williams T, et al. Intravenous
bisphosphonate-related osteonecrosis of the jaw: Long-term
follow-up of 109 patients. Oral Surg Oral Med Oral Pathol Oral
Radiol. 115:192, 2013.
240. Ristow O, Ruckschloss T, Muller M, et al. Is the conservative non-
surgical management of medication-related osteonecrosis of the
jaw an appropriate treatment option for early stages? A long-term
single-center cohort study. J Craniomaxillofac Surg. 47:491, 2019.
241. Carlson ER, Schlott BJ: Anti-resorptive osteonecrosis of the
jaws. Facts forgotten, questions answered, lessons learned. Oral
Maxillofac Surg Clin North Am. 26:171, 2014.
242. Klingelhoer C, Zeman F, Meier J, et al. Evaluation of surgical
outcome and inuencing risk factors in patients with medication-
related osteonecrosis of the jaws. J Craniomaxillofac Surg.
44:1694, 2016.
243. Nisi M, La Ferla F, Karapetsa D, et al. Conservative surgical
management of patients with bisphosphonate-related osteonecrosis
of the jaws: A series of 120 patients. Br J Oral Maxillofac Surg.
54:930, 2016
244. Park H, Copeland C, Henry S, Barbul A: Complex wounds and
their management. Surg Clin North Am. 90:1181, 2010.
245. Giudice A, Barone S, Diodati F, et al: Can surgical management
improve resolution of medication-related osteonecrosis of the jaw
at early stages? A prospective cohort study. J Oral Maxillofac Surg.
78:1986, 2020.
246. Rachner TD, Coleman R, Hadji P, Hofbauer LC: Individualized
bone-protective management in long-term cancer survivors with
bone metastases. J Bone Miner Res. 36:1906, 2021.
247. Gnant M, Mlineritsch B, Stoeger H, et al. Colorectal cancer study
group VA: Zoledronic acid combined with adjuvant endocrine
therapy of tamoxifen versus anastrozol plus ovarian function
suppression in premenopausal early breast cancer: Final analysis of
the Austrian breast and Colorectal cancer study group trial 12. Ann
Oncol 26:313, 2015.
248. Yang YL, Xiang ZJ, Yang JH, et al. The incidence and relative risk
of adverse events in patients treated with bisphosphonate therapy
for breast cancer: A systematic review and meta-analysis. Ther Adv
Med Oncol. June 9;11, 2019.
PAGE 34 Medication-Related Osteonecrosis of the Jaw – 2022 Update
Position Paper
DISCLAIMER
The American Association of Oral and Maxillofacial Surgeons
(AAOMS) is providing this position paper on Medication Related
Osteonecrosis of the Jaw (MRONJ) to inform practitioners,
patients and other interested parties. The position paper is based
on a review of the existing literature and the clinical observations
of a Special Committee composed of oral and maxillofacial
surgeons, oral pathologists, and oncologists experienced in the
diagnosis, surgical and adjunctive treatment of diseases, injuries
and defects involving both the functional and esthetic aspects
of the hard and soft tissues of the oral and maxillofacial regions,
epidemiologists, and basic researchers.
The position paper is informational in nature and is not intended
to set any standards of care. AAOMS cautions all readers that
the strategies described in the position paper are NOT practice
parameters or guidelines and may NOT be suitable for every, or
any, purpose or application. This position paper cannot substitute
for the individual judgment brought to each clinical situation by
the patient's oral and maxillofacial surgeon. As with all clinical
materials, the position paper reects the science related to
MRONJ at the time of the paper’s development, and it should
be used with the clear understanding that continued research
and practice may result in new knowledge or recommendations.
AAOMS makes no express or implied warranty regarding the
accuracy, content, completeness, reliability, operability, or legality
of information contained within the position paper, including,
without limitation, the warranties of merchantability, tness for a
particular purpose, and non-infringement of proprietary rights. In
no event shall the AAOMS be liable to the user of the position
paper or anyone else for any decision made or action taken by
him or her in reliance on such information.