Perioperative Treatment Patient Taking Anticoagulation Medication

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Oral Maxillofacial Surg Clin N Am 18 (2006) 151 – 159

Perioperative Treatment of the Patient Taking


Anticoagulation Medication
Larry L. Cunningham, Jr, DDS, MDT, M. Todd Brandt, DDS, MD,
Eron Aldridge, MD
Department of Oral and Maxillofacial Surgery, University of Kentucky, 800 Rose Street, D-508, Lexington, KY 40536-0297, USA

Prolonged intraoperative and postoperative bleed- These events include cerebrovascular accident, tran-
ing is a concern for oral and maxillofacial surgeons sient ischemic attack, peripheral venous or arterial
who treat patients taking anticoagulation medication. thrombus or embolus, and clotted arteriovenous
In the United States alone, nearly 2.3 million patients grafts. Of these complications, oral and maxillofacial
have been found to have atrial fibrillation; of those, surgeons are probably most familiar with deep
40% receive anticoagulant medications [1]. Anti- venous thrombosis, which is a common occurrence
coagulation medications are often given prophylacti- among surgical patients. Its incidence in association
cally to patients who are at risk of thrombus formation with general and orthopedic surgery is 25% to 50%
and its sequelae as the result of acquired or hereditary in the absence of any form of prophylaxis [2].
medical disorders, most commonly coronary artery Various pharmacologic methods can be used to
disease, atrial fibrillation, and deep venous thrombo- treat or prevent complications, such as deep venous
sis. Patients who have undergone heart valve re- thrombosis, or other sequelae related to a hyper-
placement, particularly persons who have received coagulable state. Patients who take anticoagulation
mechanical valves, are treated with warfarin. Medi- medications may be seen by oral and maxillofacial
cal conditions such as cancer, ulcerative colitis, surgeons for evaluation and surgery in elective, urgent,
heparin-induced thrombocytopenia, antiphospholipid and emergent situations. The purpose of this article is
antibody syndrome, disseminated intravascular co- to familiarize readers with commonly encountered
agulation, and hyperhomocystinemia may predispose medications and recently published findings regarding
patients to a hypercoagulable state and the need for their use by patients at risk of thromboembolic events.
anticoagulation therapy. Oral and maxillofacial sur-
geons also may treat patients with hereditary con-
ditions. Patients who have deficiencies of protein C,
A review of hemostasis
protein S, or antithrombin III, patients who have
factor V Leiden thrombophilia, and patients who have
Hemostasis is the cessation of bleeding. To
abnormal plasminogen production may require anti-
achieve hemostasis, the body maintains a delicate
coagulation to prevent thrombotic sequelae.
balance between the risk of intravascular thrombus
Patients in a hypercoagulable state who do not
and the risk of hemorrhage. This balance depends on
receive prophylactic anticoagulation medications are
the normal functioning of the vascular endothelium,
at risk of venous or arterial thrombolic events that
the coagulation cascade, blood flow, platelets, anti-
may result in substantial morbidity or mortality rates.
clotting mechanisms, and the fibrinolytic system. It
is important to understand the overall concept of
T Corresponding author. hemostasis because multiple medications are used
E-mail address: llcunn2@email.uky.edu for anticoagulation, each with distinct characteristics
(L.L. Cunningham, Jr). and mechanisms by which they disrupt the coagu-

1042-3699/06/$ – see front matter D 2006 Elsevier Inc. All rights reserved.
doi:10.1016/j.coms.2005.12.009 oralmaxsurgery.theclinics.com
152 cunningham et al

lation cascade. A complete review of hemostasis proteins, factor XIIa, cleaves and activates prekalli-
is beyond the scope of this article, but the diagrams krein and factor XI into kallikrein and activated factor
that cover the intrinsic and extrinsic schemes and XIa. These proteins are anchored to the subendothe-
the physiologic pathway provide an overview. The lium by a high molecular weight kininogen. Kallikrein
process includes primary hemostasis (platelet plug then amplifies the activity of the intrinsic system by
formation), secondary hemostasis (coagulation), and activating neighboring molecules of factor XII. Once
the formation of a stable fibrin clot (Fig. 1). activated, factor XIa cleaves its anchoring cofactor,
In general, the coagulation cascade can be de- high molecular weight kininogen, and diffuses into
scribed as having three parts: the intrinsic system, the solution, where it activates factor IX in the presence of
extrinsic system, and the common pathway. Coagu- calcium. Activated factor IXa forms a complex with
lation proteins circulate in the bloodstream in inactive activated factor VIIIa and calcium. This complex then
forms. Unlike the subendothelium, the endothelium is binds to the surface of platelets that contain phospho-
devoid of thrombogenic tissue factor and collagen; lipids, in which the activation of factor X occurs. This
activation of platelets and the coagulation cascade event marks the end of the intrinsic pathway and the
are prevented. initiation of the common pathway of the coagulation
Maintaining normal, brisk flow through vessels cascade (Fig. 2) [3].
ensures that any activated coagulation proteins are Unlike the intrinsic system, the extrinsic system
swept away quickly for disposal in the liver. When a initiates the coagulation cascade with components
vessel wall is injured, however, the coagulation outside the blood. Disruption of the endothelial
apparatus is stimulated. The intrinsic system begins surface exposes the subendothelium, which expresses
with the exposure of subendothelial collagen through tissue factor on its cell surface. Once exposed to the
the defect at the site of injury, which activates platelets blood, circulating factor VII binds to the tissue factor
and other coagulation proteins. One of the activated and forms a tissue factor-factor VII complex that

Fig. 1. Normal hemostatic process.


perioperative treatment and anticoagulation medicine 153

Fig. 2. The classic coagulation cascade.

promotes the autoactivation of factor VII to activated alteration in the blood’s coagulability. These condi-
factor VIIa. In the presence of calcium and membrane tions produce a hypercoagulable state.
phospholipids, this complex activates factor X. Stasis, the first condition of Virchow’s triad, may
The activation of factor X is the point at which occur in large and small blood vessels but occurs
the intrinsic and extrinsic pathways converge to form more commonly in larger vessels, such as the deep
the common pathway of the coagulation cascade. veins of the calf [3]. When blood flow is rapid, small
Factor X is activated on the phospholipid-rich surface quantities of thrombin and other procoagulants are
of activated platelets. Activated factor X forms a mixed with large quantities of blood and are carried
complex with its activated cofactor, factor Va, in the by the blood to the liver, where they are removed
presence of calcium; it also converts prothrombin into mainly by Kupffer cells. When blood flow is too
thrombin [3]. slow, however, these procoagulants accumulate in
Thrombin is referred to as the primary regulator of local concentrations sufficient to initiate clotting [5].
the coagulation cascade. Thrombin cleaves fibrinogen The second condition of Virchow’s triad refers to
to form soluble fibrin monomers, which subsequently injury to a blood vessel wall. The injury causes dis-
polymerize to form an insoluble fibrin clot at the site of ruption of the vessel’s endothelial layer, which sets in
injury. Thrombin also activates circulating factor XIII, motion the intrinsic pathway of coagulation. This
which catalyzes the formation of cross-links between disruption exposes the underlying collagen to platelets
fibrin molecules. These cross-links provide the neces- and other coagulation proteins, which are activated by
sary structure for a stable fibrin clot. Thrombin also this exposure. Once activated, the platelets spread in
activates surrounding platelets that, in conjunction shape, and their procoagulant phospholipids become
with the fibrin clot, seal the defect in the vessel wall. externalized. This change allows the coagulation pro-
Finally, thrombin activates factor Vand factor VIII in a teins to assemble on the surfaces of the platelets and
positive feedback loop, which further amplifies the accelerates the coagulation reactions. Von Willebrand
activation of the coagulation cascade [4]. factor is synthesized and released by endothelial cells
Understanding thrombus formation—or the pre- and is exposed during this endothelial disruption.
vention thereof—requires a working knowledge of During this second condition of Virchow’s triad, von
the three conditions associated with this pathophysio- Willebrand’s factor assists platelets in attaching to
logic process that was first described by Virchow in collagen; the adherent platelets spread out, and their
1856 [2]. Known as Virchow’s triad, the three con- cytoplasmic granules release substances such as
ditions necessary for thrombus formation are venous adenosine 50-diphosphate (ADP), serotonin, and
or arterial stasis, endothelial wall damage (ie, damage thromboxane A2. These substances cause local vaso-
to the intima or inner layer of a vessel), and an constriction and platelet aggregation, which recruit
154 cunningham et al

more platelets. This initial hemostatic plug is referred factor VIII deficiency among patients with hemo-
to as primary hemostasis; its proper function requires philia A, for example, factor VIII concentrations
an adequate number of normally functioning platelets, actually may be increased rather than decreased
collagen, and von Willebrand’s factor, and products among patients with severe liver disease. This in-
of the coagulation cascade, such as thrombin and crease is probably an acute phase response [4].
fibrinogen [4]. Fibrinogen is the precursor of fibrin, from which
Defects of primary hemostasis are recognized in fibrin clots are built. Fibrinogen is produced by the
the operating room when normal avascular planes liver and is an acute phase reactant. An inflammatory
continue to ooze as small capillaries continue to bleed stimulus increases the hepatic output of fibrinogen to
despite the application of pressure and time. Coagu- as much as eight times its normal level [4].
lation continues through secondary hemostasis when Platelets are the disk-shaped fragments of mega-
the platelet plug is stabilized, and a mechanically karyocyte cytoplasm that are initially found in bone
strong clot composed of fibrin, platelets, and eryth- marrow. These fragments adhere to the site of injury,
rocytes is formed. become activated, and stimulate further aggregation.
Cross-linking of fibrin further strengthens the clot, Once platelets have been activated, they change their
which then contracts. Propagation of the clot is lim- shape, release the contents of their granules, and ex-
ited by three different mechanisms. First, tissue factor pose receptor sites that provide a surface for activation
pathway inhibitor limits the initiation of coagulation. and assembly of the coagulation complexes. As ma-
Second, the protein C pathway is activated by throm- ture cells, platelets circulate in the blood at a constant
bomodulin, which binds excess thrombin. Along with level, and approximately 33% of the platelet pool is
the cofactor protein S, this pathway inactivates fac- sequestered in the spleen. These sequestered platelets
tors Va and VIIa of the coagulation cascade, which are freely exchangeable with those in the blood and
limits amplification of the clot. Finally, thrombin is can be released in large numbers in response to epi-
inactivated by antithrombin III, which limits propa- nephrine or exercise. It is important to remember that
gation of the clot [4]. Chemotactic factors then stimu- among patients who have hypersplenism, an increase
late phagocytic leukocytes to clean up debris in the in the sequestration of platelets may result in throm-
region of injury. Platelet-derived growth factor, re- bocytopenia. Likewise, among patients who have
leased from degranulating platelets, stimulates vascu- undergone splenectomy, the entire platelet pool is con-
lar repair. When the continuity of the endothelium tained within the circulation, and thrombocytopenia
has been restored, the fibrinolytic system is activated may persist to some degree after this procedure [4].
and the occluding thrombus is lysed [4].
The final element of Virchow’s triad, the hyper-
coaguable state, reflects an imbalance between pro-
coagulant and anticoagulant tendencies. Patients who Preoperative evaluation
undergo oral or maxillofacial surgery may have
medical conditions that cause hypercoagulable state, Every preoperative evaluation begins with a
such as cancer, ulcerative colitis, heparin-induced comprehensive medical history, regardless of the
thrombocytopenia, antiphospholipid antibody syn- type of surgery planned. Questioning should assess
drome, disseminated intravascular coagulation, hyper- each patient’s individual bleeding risk by emphasiz-
homocystinemia, Factor V Leiden thrombophilia, ing past and current usage and dosage of medications,
abnormal plasminogen production, and deficiencies including vitamins, herbal remedies, over-the-counter
in antithrombin, protein C, and protein S. drugs, and prescription medications. Patients often
Because of the wide variety of diseases among forget to mention medications that they are currently
surgical patients, it is important to understand where taking. During the interview process, questions
the coagulation factors are synthesized. Most of them should focus on medical conditions that would
are produced in the liver. In fact, hepatic synthesis has warrant the use of anticoagulants or other medica-
been confirmed for fibrinogen, factor V, and the group tions that could cause prolonged bleeding after
of factors that require vitamin K for synthesis, namely, surgery, such as nonsteroidal anti-inflammatory
factor II (prothrombin), factor VII, factor IX, factor X, drugs. Once the history has been obtained, bleeding
protein C, and protein S [4]. Factor XIII is involved risk should be assessed by a consideration of medical
in fibrinogen cross-linkage and is synthesized parti- conditions, medications, previous surgical history or
ally in megakaryocytes and partially in the liver. complications, and the type of surgery planned. Al-
Factor VIII is believed to be produced at several though individually these factors may not arouse
sites. Although liver transplantation has corrected suspicion before surgery, their combination in certain
perioperative treatment and anticoagulation medicine 155

situations may pose a serious preoperative risk of determination of the PTT, PT, and thrombin time and
immediate or delayed hemorrhage. mixing studies and tests for inhibitors, disseminated
For example, dentists and physicians view a intravascular coagulation, and factor XIII deficiency.
simple extraction as minor surgery, after which pro- Testing for primary hemostasis begins with
longed bleeding would not be expected. When this determination of the platelet count. For this test, the
‘‘minor surgery’’ is combined with a history of poorly blood must be collected in citrated tubes without
monitored anticoagulation use that results in inad- exposure to ethylenediaminetetra-acetic acid, which
vertent overmedication of a patient, a lack of pre- may cause platelet clumping and an inaccurate count.
operative laboratory assessment, and a preoperative In the absence of overt thrombocytopenia, deter-
diagnosis of advanced periodontal disease, the out- mination of bleeding time is a means of assessing
come can change drastically. platelet dysfunction as a cause of symptoms. Tra-
In any clinical situation, numerous steps can be ditionally this is known as an Ivy bleeding time. A
taken preoperatively to reduce the risk of bleeding blood pressure cuff is placed on the upper arm and
intraoperatively and postoperatively. In addition to a inflated to 40 mm Hg. A small wound is made on the
proper history that details medication usage and forearm, filter paper is applied to the site, and the
underlying medical conditions that may increase the blood is pulled away by capillary action while bleed-
effects of anticoagulant medication or contribute to ing time is measured at 20-second intervals. A pro-
prolonged bleeding, a detailed, focused physical ex- longed bleeding time indicates a problem with platelet
amination should be performed. Certain anatomic function or perhaps with capillary integrity [6].
considerations and disease processes can predispose a Although some hospitals and laboratories still use
patient to bleeding complications. Surgery that re- the Ivy bleeding time to assess platelet function, a
quires a maxillary osteotomy or a neck dissection more recently developed test is the platelet function
poses a greater risk than surgery that does not traverse analysis. This laboratory test simulates the process of
such anatomic structures and does not increase the platelet adhesion and aggregation after a vascular
risk of severing a vessel that may or may not be injury in vitro. Using separate test cartridges known as
amenable to ligation or cautery. Inflammation at the the collagen/epinephrine test and the collagen/ADP
proposed surgical site may lead to fibrinolysis and test, the platelet function analysis is reported as a
prolonged bleeding. Excessive operative trauma also closure time. A closure time above the normal range
may predispose a patient to postoperative bleeding, may indicate the need for further testing to determine
especially from oral soft tissues. any possible causes of platelet dysfunction, including
Once a patient’s history has been obtained and the acquired, inherited, or induced by platelet-inhibiting
type of surgery and its potential operative complica- medications. The collagen/epinephrine test deter-
tions have been considered fully, clinicians should mines if the platelet dysfunction is induced by in-
assess each patient’s risk of intraoperative or post- trinsic platelet defects, von Willebrand’s disease, or
operative bleeding and determine whether pre- exposure to platelet-inhibiting agents. The collagen/
operative laboratory assessment is indicated. This ADP test is used to determine if an abnormal collagen/
determination does not depend entirely on the type epinephrine test was caused by either the effect of
or dose of anticoagulant medication but also is in- acetyl salicylic acid or medications that contain acetyl
fluenced by the type of surgery to be performed and salicylic acid.
a patient’s preoperative diagnosis. Laboratory assess- Screening for von Willebrand’s disease screen is
ment may include determining hemoglobin or he- appropriate for a patient with a history of bleeding, a
matocrit, platelet count, prothrombin time (PT), partial normal platelet count, and prolonged bleeding time.
thromboplastin time (PTT), an Ivy bleeding time, Finally, platelet function studies, such as secretion
platelet function analysis, International Normalized and aggregation in response to agonists, can implicate
Ratio (INR), or some combination of these factors. platelet dysfunction as a cause of bleeding. Such
Surgeons must assess a patient’s coagulation status studies are not indicated for the initial evaluation of
preoperatively. Surgeons commonly use a few basic anticoagulation status, however.
measures that deserve a brief explanation. For the Tests of secondary hemostasis include the deter-
purposes of this article we categorize these measures as mination of PT (often reported as INR). For this test,
tests of primary hemostasis or tests of secondary calcium and thromboplastin (a mixture of tissue
hemostasis. Tests of primary hemostasis include factor and phospholipid membrane fragments) are
determination of the platelet count, bleeding time, added to citrated blood, and the time required for a
and platelet function and screening for von Wille- clot to form is measured. This test measures the
brand’s disease. Tests of secondary hemostasis include extrinsic pathway of the coagulation cascade. The PT
156 cunningham et al

is prolonged by deficiencies in factors II (prothrom- Warfarin therapy


bin), V, VII, and X and in fibrinogen. The PT is used
to monitor the anticoagulation status of patients who Patients may require warfarin therapy for such
are taking warfarin [4]. diagnoses as atrial fibrillation, pulmonary embolism,
The determination of PTT measures the slower myocardial infarction, stroke, and deep venous
intrinsic pathway. In vitro, this pathway requires all thrombosis or because they have prosthetic heart
of the clotting factors except factor VII. Likewise, in valves. Warfarin causes anticoagulation by inhibiting
vitro, a reliable result can be obtained only when the the vitamin K – dependent coagulation factors II, VII,
concentrations of factor XII, prekallikrein, and high IX, and X. Its duration of action is 2 to 5 days.
molecular weight kininogen are normal. These Warfarin is 99% bound to plasma proteins. The
concentrations are not believed to be as important addition of other medications that are also protein
in vivo as they are in vitro, because patients who lack bound may result in decreased binding of warfarin,
these factors do not bleed abnormally [4]. The PTT is which increases the level of anticoagulation. PT or
used to monitor anticoagulation with heparin. When INR is checked regularly so that therapeutic levels of
PT and PTT are used as tools for measuring anti- the drug can be maintained in the blood.
coagulation, the results are generally not prolonged Several studies have evaluated dentoalveolar
until factor levels fall to less than 30% of normal [4]. surgery among anticoagulated patients. Souto and
Thrombin time measures the time to clot formation colleagues [7] prospectively studied bleeding com-
after thrombin is added to anticoagulated blood. This plications in 92 patients who were chronically treated
test is a good measure of quantitative and qualitative with acenocoumarol for valvular heart disease or
deficiencies in fibrinogen. cardiac valve prosthesis and who were scheduled for
Mixing studies should be one of the initial tests dental extractions (one or two teeth). At the time of
of coagulation status. A prolonged PT or PTT may surgery, the INR of all patients was between 2 and 3.
indicate either a factor deficiency or the presence of Patients were assigned to one of six groups. For three
an inhibitor. If the test plasma is mixed with an equal groups, the acenocoumarol dosage was decreased
amount of normal plasma, deficient factors are before surgery and one of three antifibrinolytic thera-
restored to at least 50% of normal levels. These pies was initiated: oral epsilon-amino-caproic acid
levels are sufficient to normalize the clotting results, (4 g orally) before surgery, tranexamic acid as a
but an excess of inhibitors remains, and test results are mouthwash, or oral epsilon-amino-caproic acid as a
not correct. In the event that mixing studies fail to mouthwash. The other three groups used the same
correct a prolonged coagulation time, confirmatory antifibrinolytic therapies but maintained the normal
testing for the specific inhibitors should be conducted. dosage of acenocoumarol. There was no statistically
Another test that may be clinically appropriate significant difference between groups on the basis of
is screening for disseminated intravascular coagu- gender, age, gingival hypertrophy, surgical trauma, or
lation. This test is especially helpful in differenti- number of extracted teeth. The authors concluded that
ating disseminated intravascular coagulation from the most desirable treatment was no change in the
liver failure. dose of anticoagulation medication and topical treat-
Finally, specific tests can be used to determine ment with tranexamic acid for 2 days after surgery.
which of several inherited thrombophilic conditions They also determined that heparin administration was
may be present, such as abnormalities in antithrom- an additional uncontrollable risk factor for hemor-
bin III activity, protein C level and activity, or pro- rhagic complications.
tein S level and activity. When patients have a history Blinder and colleagues [8] studied three types of
of venous thromboses and a positive family history of local hemostasis after extractions performed on
such events, depending on patient age it may be patients who were maintained on coumarin therapy.
appropriate to evaluate for inherited causes of the Reasons reported for anticoagulation included valvu-
hypercoagulable state [6]. lar disease, atrial fibrillation, ischemic heart disease,
The two most common types of chronic anti- and venous thromboembolism. Patients in group one
coagulation medications that may be used by patients were treated with a gelatin sponge in the extraction
seen by oral and maxillofacial surgeons are warfarin site and sutures; patients in group two were treated
and antiplatelet therapy. Understanding the mecha- with a gelatin sponge, sutures, and tranexamic acid
nism or pathophysiology of anticoagulation and the mouthwash for 4 days; patients in group three were
indications for which medicated patients are being treated with fibrin glue, gelatin sponges, and sutures.
treated greatly aids in decisions regarding the treat- Reasons for extraction were severe periodontitis and
ment of these patients. deep caries. The patients’ preoperative INRs ranged
perioperative treatment and anticoagulation medicine 157

from 1.5 to 4. There was no statistically significant associated with a comparatively low probability of
difference in postoperative bleeding between groups. embolic events in patients at high risk for embolism.
Postoperative bleeding was associated with advanced A review of 28 patients by Ananthasubramaniam
periodontal disease but was not associated with an and co-workers [13] led to similar conclusions. All
elevated INR. All postoperative bleeding episodes patients had mechanical heart valves and were
(13/150 patients) were controlled with local measures. receiving chronic anticoagulation therapy. This ther-
Blinder and colleagues [9] studied the association apy was discontinued because of severe bleeding
of INR and bleeding complications after dental complications. The mean duration of warfarin cessa-
extractions. In a study of 249 patients who underwent tion was 15 days. Four deaths occurred: two were
543 extractions, the authors separated the INR values thought to be related to the initial diagnosis, one was
into five ranges: 1.5 to 1.99, 2.0 to 2.49, 2.5 to 2.99, caused by intracerebral bleeding, and one was caused
3.0 to 3.49, and higher than 3.5. After the extractions, by massive hematemesis. Telephone follow-up at
patients were treated with gelatin sponges and sutures 6 months found that no clinically recognized throm-
at the extractions sites. Prolonged postoperative boembolic events had occurred in 19 of 21 patients.
bleeding occurred among 12% of patients, but there Another strategy that can be used to treat patients
was no statistically significant difference in bleeding undergoing long-term anticoagulation is decreasing a
time between the five groups. patient’s INR to 1.5 to 2.0 without discontinuing
The invasiveness of the procedure is important in treatment entirely. No study has shown that the risk
the determination of treatment for anticoagulated of bleeding is significantly increased when the INR
patients. Simple extractions can be accomplished is kept in this range. Larson and colleagues [14] re-
without cessation of anticoagulation therapy. The use ported the outcomes of 93 patients treated in this way.
of gelatin sponges and sutures after simple extrac- Most of the patients, all of whom were chronically
tions has been shown to control postoperative treated with warfarin, were at high risk of thrombo-
bleeding in patients with INRs as high as 3.5 [8,9]. embolic events during the perioperative period. Of the
For more invasive procedures, conventional therapy surgical procedures for which the INRs were adjusted,
has included hospitalization, heparinization, and daily 58% were considered to be substantially invasive
monitoring of PT, PTT, and INR. Such procedures (joint replacement, vascular surgery). For 35 patients,
are typically required for 3 to 4 days before surgery warfarin was supplemented with heparin (adminis-
so that a patient’s INR can normalize. Heparin ad- tered intravenously or subcutaneously) or with low
ministration is discontinued 6 hours before surgery molecular weight heparin (LMWH) (administered
and resumed postoperatively. Warfarin administration subcutaneously) because the INR fell below 1.5. The
is also resumed postoperatively, and heparin admin- mean INR was 2.1 on the day before and 1.8 on the day
istration can be discontinued when the INR reaches of surgery (range, 1.2 – 4.9). Complications were four
the therapeutic level [10,11]. minor bleeding episodes, two major bleeding episodes
The condition for which a patient is treated is (2% rate of major bleeding), two events of throm-
also important in the decision about the anticoagula- boembolism (one death), and the need for 34 trans-
tion process. For example, the risk of stroke in a fusions. Most of the transfusions occurred in patients
patient treated for atrial fibrillation is lower than the who received autogenous units of previously donated
risk of thrombus in a patient treated for a mechanical blood for joint replacement surgery. The theoretical
heart valve. These patients might be treated differ- advantage of continuing warfarin therapy and main-
ently with regard to discontinuation of anticoagu- taining the INR between 1.5 and 2 is the rebound
lation therapy. phenomenon and the hypercoagulable state that occurs
An interesting study by Pham and colleagues [12] when warfarin therapy is stopped and started [14].
examined the risks associated with discontinuing
warfarin therapy for high-risk patients who were being
treated for intracranial hemorrhage. Indications for
anticoagulation were prosthetic heart valves (group 1), Antiplatelet therapy
atrial fibrillation and cardioembolic stroke (group 2),
and recurrent transient ischemic attack or and ische- Multiple medications are used as antiplatelet ther-
mic stroke (group 3). The probability of an ischemic apy. Patients may be treated with antiplatelet drugs for
stroke 30 days (Kaplan-Meier curve) after the discon- various reasons. Antiplatelet therapy has been shown
tinuation of warfarin was 2.9% for group 1, 2.6% for to be effective in decreasing the risk of myocardial
group 2, and 4.8% for group 3. The authors concluded infarction and nonfatal stroke among patients who
that cessation of warfarin therapy for 1 to 2 weeks is have peripheral vascular disease [15,16].
158 cunningham et al

Clopidogrel, a commonly administered antiplate- complication is higher when antiplatelet therapy is not
let drug, inhibits ADP-induced platelet fibrinogen discontinued, however [20].
binding. Clopidogrel has been identified as an in- Other platelet-inhibiting medications are aspirin
dependent risk factor for re-exploration after coro- and ticlopidine. Aspirin inhibits the activity of cyclo-
nary artery bypass graft surgery [17]. In one study, oxygenase, and ticlopidine inhibits ADP-induced
the risk of surgical re-exploration because of post- platelet fibrinogen binding. Platelets are affected for
operative bleeding was 6.1% for patients treated the life of the cell, and complete reversal of anti-
with clopidogrel but only 1% for patients not treated platelet activity does not occur until after approxi-
with the drug (P = 0.058) [17]. The rate of trans- mately 2 weeks. It has been recommended that the
fusions was also higher for patients treated with administration of antiplatelet drugs should be dis-
clopidogrel. The findings of this study were con- continued 7 to 9 days before surgery, if indicated, so
firmed in 2005 by Kapetanakis and associates [15], that sufficient numbers of normal circulation platelets
who demonstrated that patients taking clopidogrel can be regenerated [11].
and undergoing coronary artery bypass graft surgery
had a higher risk of intraoperative hemorrhage,
the need for transfusions with various blood prod- Low molecular weight heparins
ucts (platelets, packed red blood cells, and fresh
frozen plasma), and the need for re-operation to con- An alternative to heparin for anticoagulation during
trol bleeding. warfarin cessation is the administration of LMWH.
In only a few instances would maxillofacial LMWHs are administered subcutaneously, have a
surgery be needed within 3 weeks of the insertion bioavailability of more than 90%, and offer a pre-
of a coronary artery stent. Should major maxillofacial dictable and reproducible anticoagulant response [21].
surgery be indicated during this 3-week period, There is no need to monitor the anticoagulation activ-
however, antiplatelet therapy should not be stopped. ity of the drugs [22]. LMWHs are not totally reversed
Sharma and colleagues [18] reviewed the records of by protamine, as is regular heparin [21]. The mecha-
patients who underwent major noncardiac surgery nism of action of this group of drugs is via binding to
after coronary stenting. They found that the risk of antithrombin III, which increases the ability of anti-
cardiac complications and death was greatest during thrombin III to inactivate factor Xa and factor II [22].
the 3 weeks after stent placement; six of seven deaths LMWHs have been used as bridging therapy for
occurred among patients whose antiplatelet therapy patients taking warfarin. The reported incidence of
had been discontinued. major bleeding episodes in patients treated with this
Many patients may receive a combination of anti- therapy ranges from 0% to 10% [14,23,24]. An ad-
platelet drugs (eg, aspirin and clopidogrel) [16,19]. vantage of this bridging therapy is that it avoids
This combination has been shown to cause a syn- the need for postoperative hospitalization while the
ergistic antiplatelet action; in other words, the risk of INR returns to normal [24]. Although LMWHs are
bleeding complications is much higher in association approved only for prophylaxis of venous thrombo-
with combined therapy than in association with embolism, they may be considered as an alternative
single-drug therapy. No large trials have been per- to intravenously administered heparin.
formed to evaluate the risk of bleeding complications
in association with combined antiplatelet therapy. A
recent survey of vascular surgeons showed that most Summary
did not stop the administration of antiplatelet drugs
preoperatively [20]. One might assume that the risk of Patients who are undergoing chronic anticoagula-
bleeding complications would be much higher in tion therapy and require maxillofacial surgery present
association with vascular procedures (carotid endar- a challenge to oral and maxillofacial surgeons.
terectomy or infrainguinal bypass) than in association Typical treatment previously required prolonged
with routine exodontia. For more invasive maxillo- hospital stays, complicated medication adjustments,
facial procedures, a surgeon may consider continuing multiple laboratory tests, and a high level of anxiety.
the administration of aspirin alone. The small studies A thorough surgeon continues to elicit a detailed
that have been reported indicate that the risk of myo- history, perform a thorough physical examination,
cardial infarction and stroke is lower for patients who and consult with a patient’s internist or cardiologist.
continue to receive antiplatelet therapy than for A growing body of literature indicates that routine
patients who stop such therapy. The results also dental extractions can be completed with only local
suggest that the risk additional surgery for a bleeding measures for hemostasis, without cessation of anti-
perioperative treatment and anticoagulation medicine 159

coagulation medications and without the risk of coagulated, oral surgery patient: a review and a pro-
thromboembolic events. posed nomogram. J Oral Maxillofac Surg 2000;58(2):
For patients who need more invasive surgery 198 – 202.
[11] Sear JW, Higham H. Issues in the perioperative
(eg, patients who have suffered trauma), options in-
management of the elderly patient with cardiovascular
clude discontinuation of warfarin with heparin bridg-
disease. Drugs Aging 2002;19(6):429 – 51.
ing, LMWH bridging, and maintaining the INR [12] Phan TG, Koh M, Wijdicks EF. Safety of discontinua-
between 1.5 and 2.0. Patients who receive antiplatelet tion of anticoagulation in patients with intracranial
therapy and require invasive maxillofacial surgery hemorrhage at high thromboembolic risk. Arch Neurol
can be treated with single-drug therapy with the use 2000;57(12):1710 – 3.
of local measures for hemostasis or can be considered [13] Ananthasubramaniam K, Beattie JN, Rosman HS, et al.
for aspirin therapy alone if on two-drug therapy. How safely and for how long can warfarin therapy be
withheld in prosthetic heart valve patients hospitalized
with a major hemorrhage? Chest 2001;119(2):478 – 84.
[14] Larson BJ, Zumberg MS, Kitchens CS. A feasibility
References study of continuing dose-reduced warfarin for invasive
procedures in patients with high thromboembolic risk.
[1] Dunn AS, Turpie AG. Perioperative management of Chest 2005;127(3):922 – 7.
patients receiving oral anticoagulants: a systematic [15] Kapetanakis EI, Medlam DA, Boyce SW, et al.
review. Arch Intern Med 2003;163(8):901 – 8. Clopidogrel administration prior to coronary artery by-
[2] Solomon JM, Schow SR. The potential risks, compli- pass grafting surgery: the cardiologist’s panacea or the
cations, and prevention of deep vein thrombosis in oral surgeon’s headache? Eur Heart J 2005;26(6):576 – 83.
and maxillofacial surgery patients. J Oral Maxillofac [16] Robless P, Mikhailidis DP, Stansby G. Systematic
Surg 1995;53(12):1441 – 7. review of antiplatelet therapy for the prevention of
[3] Wakefield TW. Hemostasis. In: Greenfield LJ, Oldham myocardial infarction, stroke or vascular death in pa-
KT, Mulholland MW, editors. Surgery: scientific tients with peripheral vascular disease. Br J Surg 2001;
principles and practice. Philadelphia7 Lippincott, Wil- 88(6):787 – 800.
liams & Wilkins; 2001. p. 86 – 107. [17] Yende S, Wunderink RG. Effect of clopidogrel on
[4] Scott-Conner CEH, Spence RK, Shander A, et al. bleeding after coronary artery bypass surgery. Crit Care
Hemostasis, thrombosis, hematopoiesis, and blood trans- Med 2001;29(12):2271 – 5.
fusions. In: O’Leary JP, editor. The physiologic basis of [18] Sharma AK, Ajani AE, Hamwi SM, et al. Major non-
surgery. Philadelphia7 Lippincott, Williams & Wilkins; cardiac surgery following coronary stenting: when is
2002. p. 531 – 47. it safe to operate? Catheter Cardiovasc Interv 2004;
[5] Guyton AC, Hall JE. Hemostasis and blood coagu- 63(2):141 – 5.
lation. In: Guyton AC, Hall JE, editors. Textbook of [19] Payne DA, Hayes PD, Jones CI, et al. Combined
medical physiology. WB Saunders7 Philadelphia; 2002. therapy with clopidogrel and aspirin significantly in-
p. 419 –29. creases the bleeding time through a synergistic anti-
[6] Andritsos L, Yusen RD, Eby C. Disorders of hemo- platelet action. J Vasc Surg 2002;35(6):1204 – 9.
stasis. In: Green GB, Harris IS, Lin GA, et al, editors. [20] Smout J, Stansby G. Current practice in the use of
The Washington manual of medical therapeutics. antiplatelet agents in the peri-operative period by UK
Philadelphia7 Lippincott, Williams & Wilkins; 2004. vascular surgeons. Ann R Coll Surg Engl 2003;
p. 398 – 422. 85(2):97 – 101.
[7] Souto JC, Oliver A, Zuazu-Jausoro I, et al. Oral sur- [21] Harder S, Klinkhardt U, Alvarez JM. Avoidance of
gery in anticoagulated patients without reducing the bleeding during surgery in patients receiving anti-
dose of oral anticoagulant: a prospective randomized coagulant and/or antiplatelet therapy: pharmacokinetic
study. J Oral Maxillofac Surg 1996;54(1):27 – 32 [dis- and pharmacodynamic considerations. Clin Pharmaco-
cussion 323]. kinet 2004;43(14):963 – 81.
[8] Blinder D, Manor Y, Martinowitz U, et al. Dental [22] Johnson Leong C, Rada RE. The use of low-
extractions in patients maintained on continued oral molecular-weight heparins in outpatient oral surgery
anticoagulant: comparison of local hemostatic mo- for patients receiving anticoagulation therapy. J Am
dalities. Oral Surg Oral Med Oral Pathol Oral Radiol Dent Assoc 2002;133(8):1083 – 7.
Endod 1999;88(2):137 – 40. [23] Spandorfer JM, Lynch S, Weitz HH, et al. Use of
[9] Blinder D, Manor Y, Martinowitz U, et al. Dental enoxaparin for the chronically anticoagulated patient
extractions in patients maintained on oral anticoagulant before and after procedures. Am J Cardiol 1999;84(4):
therapy: comparison of INR value with occurrence 478 – 80, A10.
of postoperative bleeding. Int J Oral Maxillofac Surg [24] Tinmouth AH, Morrow BH, Cruickshank MK, et al.
2001;30(6):518 – 21. Dalteparin as periprocedure anticoagulation for pa-
[10] Mehra P, Cottrell DA, Bestgen SC, et al. Management tients on warfarin and at high risk of thrombosis.
of heparin therapy in the high-risk, chronically anti- Ann Pharmacother 2001;35(6):669 – 74.

You might also like