Outpatient Oral Anticoagulant Therapy


The coumarin-type oral anticoagulants have been in use for more than 70 years and are well-established weapons in the armamentarium against thrombotic disease. Their discovery evolved from investigations into a hemorrhagic disease of cattle occurring early in the 20th century attributed to the consumption of spoiled sweet clover. Karl P. Link, a biochemist at the University of Wisconsin, eventually isolated the responsible agent in spoiled sweet clover, dicumarol (3-3′-methyl- bis -4-hydroxycoumarin), which quickly entered the clinical arena through work at the Mayo Clinic in 1941. Link subsequently synthesized a related compound (warfarin), which was initially popularized as a rodenticide in the late 1940s; it entered clinical practice in the 1950s and quickly became the major oral anticoagulant in clinical use. Little has changed in the formulation of the coumarin-type oral anticoagulants or vitamin K antagonists (VKAs). They have remained critically important drugs in the primary and secondary prevention of thromboembolism. In the last 20 years, the use of oral anticoagulants has grown considerably, commensurate with the increased understanding of the important role of thromboembolism in cardiovascular disorders. Now, for the first time, a new class of oral anticoagulants has been developed that can best be characterized as d irect, target- or factor-specific o ral a nti c oagulant s (DOACs, also known as NOACs or non-vitamin K oral anticoagulants). These agents are targeted to bind to and neutralize a specific coagulation factor. These drugs have favorable pharmacokinetic attributes, and phase 3 trials show that they are effective and safe compared with standard therapy. This chapter reviews both the VKAs and the new DOACs.

Vitamin K Antagonists

Mechanism of Action

Vitamin K is an essential cofactor in the posttranslational γ-carboxylation of several glutamic acid residues in the vitamin K-dependent coagulation factors II, VII, IX, and X ( Fig. 37.1 ), as well as protein C and protein S. In the absence of γ-carboxylation, these protein are unable to bind calcium and phospholipid, and depending on the level of carboxylation, they manifest a reduced coagulant (i.e., enzymatic) potential. Warfarin produces its anticoagulant effect by interfering with the cyclic interconversion and regeneration of reduced vitamin K from its 2,3-epoxide (vitamin K epoxide). Warfarin exerts this effect by inhibiting the enzymes, vitamin K epoxide reductase complex 1 (VKORC1) and vitamin K 1 reductase, responsible for this interconversion (see Fig. 37.1 ). Dietary vitamin K enters the body in a partially reduced state, bypassing the warfarin-sensitive reductase and replenishing fully reduced vitamin K stores in the presence of warfarin therapy.

FIG 37.1, Reduced vitamin K is oxidized in the posttranslational modification of prothrombin (and other vitamin K-dependent coagulation factors), introducing carboxyl groups to several glutamic acid moieties in the prothrombin polypeptide, and forming γ-carboxyglutamic acid. The oxidized form of vitamin K is recycled after a reduction mediated by VKORC (vitamin K epoxide reductase complex). VKORC is the target enzyme inhibited by the S isomer of warfarin. S-warfarin is the active portion of warfarin, whereas the R isomer has little effect. S-warfarin is metabolized by the cytochrome P-450 (CYP450) oxidase enzyme 2C9.

Pharmacokinetics and Pharmacodynamics

Because of its excellent bioavailability and favorable pharmacokinetics, warfarin is the most commonly used oral anticoagulant in North America. It is highly water soluble and rapidly absorbed from the gastrointestinal (GI) tract after oral ingestion. Peak absorption occurs in 60 to 90 minutes. Food may delay the rate of absorption but is said not to reduce the extent of absorption.

Warfarin is a racemic mixture of stereoisomers known as the R and S forms; the two have distinctive metabolic pathways, half-lives, and potencies. Racemic warfarin has an average plasma half-life of 36 to 42 hours, with a range of 15 to 60 hours. Variability in warfarin half-life due to natural differences in metabolism, disease- and/or drug-induced alterations in metabolic fate, or the sensitivity of the VKORC1 enzyme to warfarin, account for much of the variation in an individual's initial response to, and maintenance requirement for, warfarin. The S enantiomer of warfarin (five times more potent than the R enantiomer) is metabolized primarily by the CYP 2C9 enzyme of the cytochrome P-450 (CYP450) system. A number of genetic polymorphisms (single-nucleotide polymorphisms, or SNPs) in this enzyme lead to a reduced activity of the enzyme and may influence both the dosage required to achieve a therapeutic level and the bleeding risk with warfarin therapy. Specifically, the CYP2C9*2 and CYP2C9*3 alleles are associated with lower dosage requirements and higher bleeding complication rates compared with the wild-type CYP2C9*1. The prevalence of these polymorphisms varies in populations as indicated in Table 37.1 .

TABLE 37.1
Frequency of Genetic Polymorphisms of CYP2C9 and VKORC1 in Different Ethnic Groups
From Takahashi H, Echizen H. Pharmacogenetics of warfarin elimination and its clinical implications. Clin Pharmacokinet . 2001;40:587–603; and Rieder MJ, Reiner AP, Gage BF, et al. Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. N Engl J Med . 2005;352:2285–2293.
CYP2C9 Polymorphism a
CYP2C9*1 (%) CYP2C9*2 (%) CYP2C9*3 (%)
Whites 79–89 8–19 6–10
Native Canadians 91 3 6
African Americans 98 1.5–3.6 0.5–1.5
Asians 95–98 0 1.7–5
VKORC1 Haplotype b
H 1 H 2 (%) H 8 H 9 (%)
European Americans 37 58
African Americans 14 49
Asian Americans 89 10

a CYP2C9*1 is the wild type (i.e., common genotype) and CYP2C9*2 and CYP2C9*3 are polymorphisms associated with reduced functional capacity to metabolize the S enantiomer of warfarin.

b H 1 H 2 and H 8 H 9 are different haplotypes (combinations of polymorphisms) that are associated with either greater sensitivity (H 1 H 2 ) or less sensitivity (H 8 H 9 ) to warfarin inhibition.

A number of SNPs have also been discovered in the VKORC1 gene that lead to varying sensitivities of the enzyme to warfarin inhibition and have been shown to have a major impact on the pharmacodynamics of warfarin. A combination of SNPs leads to various haplotypes of the gene and gene product. Some of these haplotypes result in an enzyme that is sensitive to warfarin inhibition so that a lower dosage of warfarin is required, whereas others are more resistant, so that a higher dosage (and maintenance dosage) of warfarin is needed to achieve a therapeutic international normalized ratio (INR). The prevalence of these haplotypes varies in different populations as indicated in Table 37.1 . A combination of genetic alterations in either the CYP2C9 or VKORC1 genes has been shown to account for as much as 20% to 50% of the variability in warfarin maintenance dosing. The effect of warfarin also varies inversely with the amount of vitamin K absorbed (from the diet and from metabolic byproducts of GI bacteria) and varies directly with the amount of warfarin absorbed or available to exert its anticoagulant effect.

Warfarin and Drug Interactions

Drug interactions commonly occur by affecting the pharmacokinetic or pharmacodynamic behavior of warfarin. Drug interactions may interfere with GI absorption of warfarin resulting in a reduction in plasma levels, or interfere with the metabolism of warfarin leading to a reduction or increase in clearance and, consequently, higher or lower plasma warfarin levels. The latter effects may be stereospecific in that only one of the enantiomers may be affected or it may be nonspecific in that both enantiomers may be affected. Interference in the metabolism of the S enantiomer, usually through effects on the CYP450 system (CYP 2C9 enzyme), is more common and has a greater potential for enhancing the intensity of anticoagulation, because the S enantiomer is several times more potent than the R enantiomer. Drugs may also decrease plasma warfarin levels by enhancing the metabolic clearance of racemic warfarin. The pharmacodynamics of warfarin may also be altered when drugs interfere with other aspects of hemostasis or vitamin K homeostasis. Third-generation cephalosporins containing an N -methylthiotetrazole side chain are an example in that they interfere with the regeneration of reduced vitamin K from the 2,3-epoxide form. Some drugs or disease states (liver disease, hyperthyroidism) can alter the metabolism of coagulation factors, inhibit coagulation factor interactions by other mechanisms (heparin), or inhibit other aspects of hemostasis (aspirin effect on platelet function), and lead to a greater risk of bleeding. In general, such interactions are most problematic when interacting drugs are added to or deleted from a patient's regimen, or a dose change is made. Once a patient has achieved stability on warfarin and an interacting medication, there should be little problem in maintaining the stability of warfarin dosing.

Assessing the literature for warfarin-drug interactions is problematic because of the poor quality of the reports. Holbrook and colleagues performed a systematic review of warfarin and drug/food interactions, and found serious problems in the reporting of potential interactions. They reviewed 187 separate reports of interactions involving 120 drugs or foods and found that no report met their quality criteria as an excellent study. There were 33 small randomized controlled trials of fair or good quality. There were 148 reports that were rated as poor quality, and 130 of these were case reports (96% single case reports). Table 37.2 summarizes these interactions and categorizes them by mechanism of cause and effect on INR. The reader can also find useful summaries of warfarin-drug interactions at various sites on the internet such as: https://www.drugs.com/drug-interactions/warfarin , coumadin.html .

TABLE 37.2
Examples of Warfarin Drug Interactions by Mechanism and Effect on INR
With permission from Nutescu EA, Burnett A, Famikos J, Spinler S, Wittkowsky A: Pharmacology of anticoagulants used in the treatment of venous thromboembolism. J Thromb Thrombolyis 41:15-31, 2016.
Category Mechanism Effect Common Examples
Pharmacokinetic Interactions Induction of warfarin metabolism Decrease INR barbiturates
carbamazepine
nafcillin
rifampin
Reduced absorption of warfarin Decrease INR cholestyramine
colestipol
Inhibition of warfarin metabolism Increase INR amiodarone
azole antifungals
fluoroquinolone antibiotics
macrolide antibiotics
metronidazole
sulfa antibiotics
Pharmacodynamic Interactions Increased synthesis of clotting factors Decrease INR vitamin K
Decreased synthesis of clotting factors Increase INR cephalosporins
Reduced catabolism of clotting factors Decrease INR methimazole
propylthiouracil
Increased catabolism of clotting factors Increase INR thyroid hormones
Impaired vitamin K production by gut flora Increase INR aminoglycosides
tetracyclines
Additive anticoagulant response Increase bleeding risk without influencing INR anticoagulants
Concurrent antiplatelet therapy Increase bleeding risk without influencing INR antiplatelet agents

A problem of recent development is the widespread use of dietary supplements and herbal preparations. Unlike drug products, dietary supplements are not tested before marketing for safety, efficacy, dosing requirements, or interactions with other medications. They are not required to meet quality standards for labeling, nor are they required to meet US Pharmacopeia standards for tablet content uniformity. Consequently, patients may be exposed to different ingredients as well as different doses of those ingredients in similar products produced by different manufacturers and in different batches from the same manufacturer. There are a growing number of case reports describing interactions between warfarin and dietary supplements, but virtually none of these interactions has been systematically substantiated. Because of the uncertainty that exists, it is wise for patients taking warfarin to avoid the use of dietary supplements or, at least, to be carefully observed when beginning or stopping a supplement. For a listing of various drug interactions or reported herbal interactions, the reader is referred elsewhere.

Therapeutic Range and Monitoring

The anticoagulant and antithrombotic effects of warfarin may not be the same. Evidence suggests that it is a reduction in prothrombin and possibly factor X that are predominantly responsible for the antithrombotic effect, while the anticoagulant effect, as measured by the prothrombin time (PT), is heavily impacted by levels of factor VII. However, it is the easily measured and readily available PT that is universally used to establish therapeutic levels of antithrombotic effectiveness. The concept of a safe and effective therapeutic range evolved largely as a consequence of trial and error and clinical empiricism in the 1940s and 1950s. A PT ratio of 2.0 to 2.5 or 3.0, measured using a human brain thromboplastin reagent sensitive to a reduction in the vitamin K-dependent coagulation factors, was believed to represent this therapeutic range. However, less sensitive rabbit brain thromboplastin reagents came into use in the 1950s and 1960s, which resulted in the need for a higher average warfarin dose to achieve the same prolongation of the PT. Hull and coworkers demonstrated the consequences of this situation in a study of patients with deep vein thrombosis (DVT) treated with warfarin by documenting a higher incidence of bleeding in those monitored using the less sensitive rabbit brain thromboplastin, but no increase in recurrent thromboembolism in those monitored using the more sensitive reagent, when PTs for both groups were maintained in a similar therapeutic range.

To correct for differences in thromboplastin sensitivity, the World Health Organization recommended the use of an international standard PT. All thromboplastins are equilibrated against a sensitive international reference thromboplastin, and the equilibration factor (the International Sensitivity Index, or ISI) is used to convert PT ratios (patient PT divided by the mean of the normal range) to an international normalized ratio (the INR). The INR is essentially the PT ratio one would obtain if the international reference thromboplastin had been used to measure the PT. The calculation is made by taking the PT ratio and raising it to the power of the ISI, which is done automatically by laboratory instrumentation. By converting all PT ratios to INRs one can interpret a patient's PT result regardless of where the test is performed and then follow the guidelines of international consensus groups for therapeutic effectiveness as outlined in Table 37.3 . Use of the INR does not eliminate all discrepancies in PT reporting, but it significantly improves evaluation of PT results compared with using raw seconds or the PT ratio when monitoring patients taking warfarin.

TABLE 37.3
Indications for Vitamin K Antagonist Therapy and Recommended Intensity of Treatment
From Hirsh J, Dalen JE, Anderson DR, et al. Oral anticoagulants: mechanism of action, clinical effectiveness, and optimal therapeutic range. Chest . 2001;119(Suppl):8S–21S, 220S–227S, with permission.
Indication Target INR (Range)
Prophylaxis of venous thrombosis 2.5 (2.0–3.0)
Treatment of venous thrombosis 2.5 (2.0–3.0)
Treatment of pulmonary embolism 2.5 (2.0–3.0)
Prevention of systemic embolism
  • Atrial fibrillation

2.5 (2.0–3.0)
  • Recurrent systemic embolism

2.5 (2.0–3.0)
  • After myocardial infarction a

3.0 (2.5–3.5)
Bioprosthetic heart valves (M or Ao position) b 2.5 (2.0–3.0)
Mechanical prosthetic heart valves
  • Bileaflet valve in Ao position

2.5 (2.0–3.0)
  • Bileaflet or tilting valve in M position

3.0 (2.5–3.5)
  • Mechanical valve + atrial fibrillation (any position)

3.0 (2.5–3.5)
  • Mechanical valve + additional risk factors

3.0 (2.5–3.5) + aspirin (81 mg daily)
Ao, Aortic valve; INR, international normalized ratio; M, mitral valve; MI , myocardial infarction.

a For prevention of recurrent MI, an INR of 3.0 (2.5–3.5) is recommended.

b For St. Jude or CarboMedics bileaflet or Medtronic-Hall tilting-disk valve.

Practical Aspects of Warfarin Management

Initiation and Maintenance Dosing

The administration of a large loading dose of warfarin to initiate therapy (40 to 60 mg was used in the 1960s) is of historical interest only. It induces a rapid, but excessive, reduction in factor VII activity, predisposing patients to hemorrhage in the first few days of therapy, and it fails to achieve a more rapid decline of the other vitamin K-dependent coagulation factors (II, IX, and X). Therapy is properly initiated using an average maintenance dosage (about 5 mg daily) for the first 2 or 3 days. When an immediate effect is required, such as in the treatment of acute venous thrombosis, heparin should be given concurrently with warfarin for at least 5 days. Warfarin treatment should overlap with heparin therapy for a period of 4 to 5 days because it takes that long to lower the levels of prothrombin and factor X, those vitamin K-dependent coagulation factors with longer half-lives and that are mostly responsible for the antithrombotic effect of warfarin. Heparin is usually discontinued when the INR has been in the therapeutic range on two measurements taken at least 24 hours apart. If initiation of treatment is not urgent (e.g., in chronic stable atrial fibrillation [AF]), warfarin can be commenced out of hospital at an anticipated maintenance dosage of about 5 mg daily, which usually achieves a therapeutic anticoagulant effect in about 5 days, although a stable INR may take longer to achieve. Two studies showed that using an initial 10 mg dose of warfarin in outpatients achieved a therapeutic INR more rapidly than using a 5 mg dose without producing a higher rate of excessive anticoagulation. Using a 10 mg starting dose for outpatients is recommended by the 2012 American College of Chest Physicians (ACCP) Evidence-Based Clinical Practice Guidelines. A recent Cochrane Database Systematic Review of the optimal loading dose for warfarin examined 12 studies including studies that used pharmacogenetic dosing and failed to identify an optimal method or dosing schedule between an initial 5 and 10 mg dose. There was evidence in the elderly that using lower initiation doses led to fewer high INRs. The fear of creating a hypercoagulable state in patients with unrecognized protein C deficiency who are not simultaneously given heparin has not been substantiated in patients with AF; however, there is suggestive evidence that in patients with AF started on warfarin without overlapping heparin, there may be an increased risk of early stroke in the first 30 days. Certainly, in patients with a known protein C deficiency or other thrombophilic state, it would be prudent to begin heparin before, or at the same time as, warfarin. A starting dose lower than 5 mg might be appropriate in the elderly, in patients with impaired nutrition or liver disease, and in patients at high risk of bleeding. The physician should be aware of variables that influence the response to anticoagulation in the elderly. The dosage required to maintain a therapeutic range in patients older than 60 years of age has been shown to decrease with increasing age, and older patients are more likely to have other variables that might influence INR stability or the risk of bleeding, such as a greater number of other medical conditions or concurrent drug use. Consequently, it is advisable to monitor treatment more carefully in older patients to maximize their time in the therapeutic range.

Estimation of the maintenance dose is often based on observations of the INR response following administration of a fixed dose of warfarin over an interval of a few days. An individual who rapidly achieves a high therapeutic INR (above 1.5) after two doses of warfarin is likely to require a low maintenance dose. The opposite holds for a patient who shows little elevation of the INR (below 1.5) after two doses. It is now known that a major determinate influencing a patient's response to initial and maintenance dosing is the patient's genotype for the principal enzyme that metabolizes warfarin (CYP 2C9) and the target enzyme through which warfarin mediates its effect (VKORC1). Polymorphisms in the genes for these enzymes account for almost 50% of the variability in dosing requirements. They are also associated with a higher rate of bleeding events. Several recent, large, randomized trials using information about genetic polymorphisms to inform dosing have shown conflicting results. In a North American study, the COAG trial (Clarification of Optimal Anticoagulation through Genetics), investigators randomized 1015 patients in a double-blind fashion to receive initiation of warfarin therapy based on genetic and clinical variables versus initiation of therapy based only on clinical variables. They found no difference in time-in-range between groups in the first 15 days or at 4 weeks (genetic group 45.2% vs. standard group 45.4%; P = .91). Surprisingly, black patients managed by genotype-guided dosing had significantly less time-in-range than nonblack patients managed by standard care in a subgroup analysis. Lastly, they found no significant difference in the rates of the combined outcome of an INR of 4 or more, major bleeding, or thromboembolism based on dosing strategy. At the same time, a smaller European study of genotype-guided warfarin dosing found different outcomes in 455 patients randomized to genotype-guided versus standard dosing where the time-in-range was significantly better for the genotype group (67.4% vs. 60.3%; P < .001). They also found significantly fewer cases of INRs ≥4.0 and the median time to reach a therapeutic INR was 21 days in the genotype-guided group compared with 29 days in the standard dosing group. The divergent outcomes in these two landmark trials may have been due to different patient populations, different genetic frequencies, different dosing algorithms, and other factors. In a systematic review and meta-analysis, Franchini and colleagues primarily assessed the outcomes of major bleeding and thromboembolism in nine randomized trials comparing genotype-guided versus clinically-guided warfarin dosing. They found a significant reduction in major bleeding in the genotype-guided dosing compared with standard dosing (RR, 0.47; 95% CI, 0.23 to 0.96) but no differences in thromboembolism, deaths or the secondary endpoints of time-in-range or patients with an INR ≥4. To further complicate matters, two additional systematic reviews and meta-analyses comparing genotype-guided dosing with non-genotype-guided dosing found differing results. Belley-Cote et al. in an analysis of 12 trials, found no significant differences in the primary outcomes of mortality, thromboembolism, or major bleeding (RR, 0.85; 95% CI, 0.54 to 1.34; P = 0.35). However, they did note a significant difference between groups with time-in-range significantly higher in the genotype-guided group compared with fixed VKA dosing patients, but not when clinical dosing algorithms were used. In an analysis by Stergiopoulos and Brown of nine trials, investigators failed to find significant differences in percentage of time-in-range, patients with an INR greater than 4, or a reduction in major bleeding or thromboembolism. The results of cost-effectiveness studies using pharmacogenetic dosing are of borderline significance and are highly dependent on adverse event variables. There are a number of factors that diminish the value of pharmacogenetics-guided dosing. These include the limited availability of genetic assays, especially with rapid turnaround; cost of genetic assays; complex algorithms needed to utilize genetic information; and conflicting trial results about the usefulness of such information and the overall cost effectiveness. All of this must be compared with the relative inexpense and value of frequent monitoring of a simple INR test. At the present time, pharmacogenetics-based dosing is not recommended by the ACCP guidelines. Until appropriate information is available, frequent monitoring of the INR response is the best means of predicting and establishing the patient's maintenance dose requirements.

INR monitoring is usually performed daily for inpatients (somewhat less frequently for outpatients) until the therapeutic range has been achieved and maintained for at least 2 consecutive days; monitoring is then performed one to two times weekly for 1 to 2 weeks and less often thereafter, depending on the stability of INR results. It should be noted that immediately after hospital discharge is often the most unstable and dangerous time for patients owing to alterations in dietary habits and changes in other medications. Therefore monitoring should be performed frequently for the first 1 to 2 weeks after discharge.

It is well documented that the time-in-therapeutic range (TTR) is highly correlated with efficacy and safety outcomes of warfarin therapy. A number of investigators have identified patient characteristics or other factors that predict instability. These include characteristics such as young age, hospitalizations, pain medications, active cancer ; low weight, and secondary venous thromboembolism (VTE) ; and poor compliance with INR scheduling. Patients who exhibit these characteristics might benefit from more frequent monitoring.

If the PT response remains stable, the frequency of testing can be reduced to intervals as long as every 4 weeks. For patients who have demonstrated stable INR results (no change in dose in 3 months), recent studies suggest that monitoring may be extended to every 8 to 12 weeks. However, there is no guarantee that stable patients remain stable as demonstrated in a report from a large real-world registry of patients on warfarin for stroke prevention in AF. Of 968 patients with a stable INR over a 6-month period (80% or more INRs in range), only 34% remained stable over the subsequent 6 months. Even of those patients with 100% INRs in range over 6 months ( n = 376), only 37% remained stable in the next 6 months. If adjustments to the dose are required, then the cycle of more frequent monitoring is repeated until a stable dose response is again achieved.

Outpatient management of warfarin therapy should aim for simplicity and clarity to avoid patient confusion, poor compliance, and dosing errors that may result in complications. It is recommended that a limited number of warfarin tablet strengths be used in clinical practice, and that patients clearly understand the various dosing patterns that are used, such as alternate-day doses or dosing levels based on days of the week. One must also be aware that several different warfarin products are on the market, which can lead to confusion for the patient. There are generic preparations sold as warfarin, brand preparations sold as Coumadin, and branded generics sold under a different name (e.g., Jantoven). Patients may be given two different preparations and be taking both, not knowing that they are the same drug.

Management of Nontherapeutic International Normalized Ratios

Patients receiving long-term warfarin therapy often have unexpected fluctuations in dose response that require careful management. These may be due to inaccuracy in PT testing, changes in vitamin K intake (increased or decreased vitamin K in the diet), changes in vitamin K or warfarin absorption (GI factors or drug effects), changes in warfarin metabolism (liver disease or drug effects), changes in vitamin K-dependent coagulation factor synthesis or metabolism (liver disease, drug effects, worsening right-sided heart failure with increasing hepatic congestion, other medical conditions), or patient compliance issues (surreptitious self-medication, missed doses, miscommunication about dose adjustment, and so on).

A nontherapeutic (e.g., elevated) INR can be managed by briefly discontinuing warfarin, administering vitamin K, or infusing fresh frozen plasma (FFP) or a factor concentrate. The choice is based largely on the severity of the clinical situation (e.g., degree of elevation of the INR, presence of severe bleeding). Assuming an ongoing normal food intake and reasonable hepatic function, when warfarin administration is interrupted, it takes about 4 to 5 days for the INR to return to the normal range in patients whose INR is between 2.0 and 3.0. The INR will return to normal more quickly in patients requiring a larger daily maintenance dose than in those requiring a lower daily maintenance dose. After treatment with oral vitamin K, the INR declines substantially within 24 hours. Because the absolute daily risk of bleeding is low even when the INR is excessively prolonged, many physicians manage patients with INR values of 4.0 to 9.0 by simply holding warfarin and monitoring more frequently, unless the patient is at a higher risk of bleeding or bleeding has already developed. Vitamin K can be administered by the intravenous, subcutaneous, or oral route. Intravenous injection may be associated with anaphylactic reactions in rare cases, but it does lead to reversal of the INR more quickly than the oral or subcutaneous administration of vitamin K. The response to subcutaneous vitamin K may be unpredictable and sometimes delayed. Several studies confirm earlier reports that oral administration is predictably effective, more so than subcutaneous administration, and has the advantages of safety and convenience over parenteral routes. Ideally, vitamin K should be administered in a dose that will quickly lower the INR into a safe, but not subtherapeutic range, without causing resistance when warfarin is reinstated. High doses of vitamin K, although effective, may lower the INR more than is necessary and lead to warfarin resistance persisting for up to 1 week.

In the presence of significant bleeding, the INR must be reversed immediately. This can be done by replacing the vitamin K-dependent factors using FFP. At least 15 to 30 mL/kg of FFP should be given to have a significant impact on factor replenishment. The amount used should depend on the degree to which the INR is out of range and the severity of the bleeding. In the average 70 kg person, this equates to 4 to 8 units of FFP, which may take up to 6 to 24 hours to order, thaw, and infuse, and in some patients it may put an undue stress on the heart. In addition, the half-lives of the individual vitamin K-dependent factors govern the durability of any response. More FFP might be required. In a patient with life-threatening bleeding, especially intracranial hemorrhage, maximal factor replacement should be achieved in the shortest interval possible. This can be accomplished by using factor concentrates that have high factor concentrations in a small volume. Prothrombin complex concentrates (PCCs), containing the vitamin K-dependent factors, have traditionally been used. Studies have also shown that recombinant factor VIIa (rFVIIa) can reverse the coagulopathy and associated bleeding induced by warfarin, as well as other coagulopathies. Given the expense of these agents, one should use the lowest effective dose to control the bleeding. PCCs are often dosed in a range of 25 to 50 IU/kg, but the specific dose will depend on body weight, degree of INR prolongation, and desired level of correction. The same considerations are relevant for the dosing of rFVIIa, although the range of dosing reported empirically ranges from approximately 20 µg/kg to more than 100 µg/kg. Although these factor concentrates are shown to return the INR to normal more rapidly than with FFP, there is still debate as to how beneficial they are over FFP in terms of outcome. In a randomized, open-label trial comparing 4-factor PCC with FFP in patients taking a VKA and having major bleeding, Sarode et al. were unable to show any significant difference in serious adverse events, death or thromboembolic events between the two treatments. There was an increase in heart failure in the FFP group attributed to the high volume of plasma infusion. PCC infusion resulted in a significantly more rapid correction of the INR as others have shown. Both PCCs and rFVIIa have the potential to induce a prothrombotic state, and thromboembolism has occurred as a result of such therapy. Table 37.4 outlines the 2008 ACCP recommendations for managing patients receiving coumarin anticoagulants who need the INR lowered because of actual or potential bleeding. These recommendations have not significantly changed in the most recent guidelines.

TABLE 37.4
Recommendations for Managing Elevated International Normalized Ratios or Bleeding in Patients Receiving Vitamin K Antagonists
From Ansell J, Hirsh J, Poller L, et al. Pharmacology and management of the vitamin K antagonists: American College of Chest Physicians evidence-based clinical practice guidelines (8th edition). Chest. 2008;133(suppl):160S–198S.
Condition Description
INR above therapeutic range but <5.0; no significant bleeding Lower dose or omit dose, monitor more frequently, and resume treatment at lower dose when INR therapeutic; if only minimally above therapeutic range, no dose reduction may be required (Grade 2C)
INR ≥5.0 but <9.0; no significant bleeding Omit next one or two doses, monitor more frequently, and resume treatment at lower dose when INR in therapeutic range. Alternatively, omit dose and give vitamin K (≤5 mg orally), particularly if at increased risk of bleeding. If more rapid reversal is required because the patient requires urgent surgery, vitamin K (2 to 4 mg orally) can be given with the expectation that a reduction of the INR will occur in 24 h. If the INR is still high, additional vitamin K (1 to 2 mg orally) can be given (Grade 2C)
INR ≥9.0: no significant bleeding Hold warfarin therapy and give higher dose of vitamin K (5 to 10 mg orally) with the expectation that the INR will be reduced substantially in 24–48 h. Monitor more frequently and use additional vitamin K if necessary. Resume therapy at lower dose when INR is therapeutic (Grade 2C)
Serious bleeding at any elevation of INR Hold warfarin therapy and give vitamin K (10 mg by slow IV infusion), supplemented with fresh plasma or prothrombin complex concentrate, depending on the urgency of the situation; recombinant factor VIIa may be considered as alternative to prothrombin complex concentrate; vitamin K can be repeated every 12 h (Grade 1C)
Life-threatening bleeding Hold warfarin therapy and give prothrombin complex concentrate supplemented with vitamin K (10 mg by slow IV infusion); recombinant factor VIIa may be considered as an alternative to prothrombin complex concentrate; repeat if necessary, depending on INR (Grade 1C)
If continuation of warfarin therapy is indicated after high doses of vitamin K, then heparin or low-molecular-weight heparin can be given until the effects of vitamin K have been reversed and the patient becomes responsive to warfarin therapy. It should be noted that INR values of >4.5 are less reliable than values in or near the therapeutic range. Thus, these guidelines represent an approximate guide for high INRs. IV, Intravenous; INR , international normalized ratio.

The decision to provide coverage to a patient who comes to one's office with a subtherapeutic INR is similar to a bridging decision. If the thrombogenic risk is so high (such as within the first week or two of treating new DVTs and/or pulmonary embolisms [PEs]) that even a few days at a subtherapeutic level places the patient at high risk, then treating the patient with low-molecular-weight heparin (LMWH) for a few days until a therapeutic INR is achieved with higher doses of warfarin is appropriate. In most cases, this intervention is not needed, and simply raising the dose of warfarin is sufficient to reestablish a therapeutic range in a few days.

Management of Oral Anticoagulation During Invasive Procedures

Clinicians are often confronted with the challenge of managing anticoagulation in individuals requiring noncardiac surgery or other invasive procedures, especially individuals with prosthetic heart valves who are currently taking warfarin (see Chapter 35 ). There is a paucity of critical studies examining the alternative choices for anticoagulation (called bridging therapy ) in this setting, and until recently, there were no robust randomized trials to assess the benefit of bridging therapy versus no bridging therapy. At the present time, based on clinical evidence, there is a strong push against using bridging therapy except in those patients at very high risk of thromboembolism without adequate anticoagulation coverage. When bridging is contemplated, physicians must assess the risk of bleeding from a procedure if anticoagulation is continued versus the risk of thrombosis if anticoagulation is discontinued, as well as the risk of bleeding from an alternative rapidly acting anticoagulant, and the cost of alternative anticoagulation options. Reviews have addressed the management of these patients and the options available, and the ACCP has made formal recommendations. Full-dose intravenous unfractionated heparin (UFH) was originally the agent of choice to bridge a patient during an invasive procedure but has been replaced by LMWH, which offers a less complex alternative because it requires no monitoring and can be given at home. A particular problem with bridging therapy is the risk of post-procedure bleeding if a rapidly acting anticoagulant is started too soon. There is not only a risk of major bleeding but also a risk associated with the delayed reinstitution of long-term therapy because of the bleeding. Investigators have shown a 3-month cumulative risk of major post-procedure bleeding of approximately 3% with bridging versus 1% without such bridging, and selected patient characteristics may be associated with post-procedure bleeding. In a recent study, Douketis et al. randomized 1884 patients with AF at low to moderate risk of thromboembolism off anticoagulants to either LMWH bridging versus no bridging. The outcome of arterial thromboembolism was the same in each group (0.3% vs. 0.4%, respectively; P = .01 for noninferiority). The incidence of major bleeding was 3.2% in the bridging group versus 1.3% in the no bridging group (RR, 0.41; 95% CI, 0.20 to 0.78; P = .005 for superiority). The study population represented a wide spectrum of AF patients at risk except those in the highest risk group and also represented a wide spectrum of invasive procedures. In a recent retrospective analysis of 1178 Kaiser Permanente Colorado patients, mostly with VTE as opposed to AF, investigators found similar findings with a major bleeding incidence of 2.7% in bridged patients versus 0.2% in nonbridged patients (HR, 17.2; 95% CI, 3.9 to 75.1). There was no difference in the rate of VTE.

If bridging therapy is implemented in those at very high risk of thromboembolism, warfarin is usually discontinued 4 or 5 days before the procedure and the INR is allowed to decline. LMWH is started 2 or 3 days before the procedure, usually at a full treatment dose (100 to 150 anti-factor Xa U/kg subcutaneously) once or twice daily depending on the risk of thrombosis, with the last dose omitted the morning or night before the procedure (approximately 12 to 24 hours before the procedure). It is then restarted about 12 hours after the procedure along with warfarin. If the risk of postoperative bleeding from the procedure is high, LMWH can be restarted in 24 to 48 hours, rather than in 12 hours. When the INR becomes therapeutic, LMWH administration is stopped. Table 37.5 summarizes the author's approach integrated with various guideline recommendations for the management of oral anticoagulation during invasive procedures.

TABLE 37.5
Recommendations for Managing Anticoagulation Therapy in Patients Requiring an Invasive Procedure
Risk of Thromboembolism or Bleeding Example Recommendation or Suggestion a
Low risk of thromboembolism off anticoagulants with moderate to high risk of bleeding Bileaflet mechanical aortic valve without AF; AF with CHADS 2 score ≤2; VTE >12 months ago Stop warfarin approximately 5 days before procedure; allow the INR to return to normal; no need to bridge with rapidly acting anticoagulant; resume warfarin after procedure
Intermediate risk of thromboembolism off anticoagulants with moderate to high risk of bleeding Bileaflet mechanical aortic valve with other risk factors such as AF, prior stroke, heart failure, hypertension; VTE 3–12 months ago; recurrent VTE; active cancer Approach is based on individual patient and surgery-related factors. Based on results of the BRIDGE trial (77) bridging is generally not required. In some instances one might use a prophylactic dose of LMWH
High risk of thromboembolism off anticoagulants with moderate to high risk of bleeding Mechanical mitral valve; old-style aortic valve; AF with high CHADS 2 score (5 or 6); CVA/TIA <3 months; rheumatic valvular heart disease; recent (<3 months) VTE Stop warfarin approximately 5 days before procedure, allow the INR to return to normal; begin therapy with full-dose UFH or LMWH approximately 3 days before procedure; stop UFH approximately 6 h before procedure; omit last one or two doses of LMWH; restart UFH or LMWH ~12–24 h after procedure or when patient is in hemostatically stable condition
Low risk of bleeding on anticoagulants Dental work; screening colonoscopy; cataract surgery Continue warfarin without adjusting dose or INR if within therapeutic range; alternatively omit a dose or two of warfarin and allow INR to fall to a lower range (~1.5–2.0) and restart after procedure
Note: These guidelines represent the author's approach and are an amalgam of guideline recommendations and recent clinical trial results. The risk of thromboembolism with discontinuation of anticoagulants versus the risk of bleeding during the procedure if anticoagulants are continued must be assessed for each patient; restarting a rapidly acting anticoagulant too soon after a procedure can lead to bleeding, which will then prolong the resumption of warfarin anticoagulation and put the patient at risk of thromboembolism; deciding on a final course of action often requires a discussion with the physician performing the procedure.
AF, Atrial fibrillation; CHADS 2 , system for scoring thromboembolic risk in nonvalvular AF that assigns points for congestive heart failure, hypertension, age ≥75, diabetes mellitus, and prior stroke or transient ischemic attack; CVA , cerebrovascular accident; INR, international normalized ratio; LMWH, low-molecular-weight heparin; UFH, unfractionated heparin; TIA , transient ischemic attack; VTE, venous thromboembolism.

a Based on recommendations of the American College of Chest Physicians Consensus Conference on Antithrombotic Therapy. and Rechenmacher SJ, Fang JC. Bridging anticoagulation. J Am Coll Card . 2015;66:1392–1403.

Dental procedures represent a particularly common intervention in patients taking anticoagulants. A comprehensive review of the subject indicated that, in most cases, no change in the intensity of anticoagulation is needed (i.e., continue to keep and document an INR of 2.0 to 3.0). Should a need to control local bleeding arise, tranexamic acid or ε-aminocaproic acid mouthwash has been used successfully without interruption of anticoagulant therapy.

Diagnostic Evaluation of Bleeding

When bleeding occurs, especially from the GI or urinary tract, it is important to consider the possibility of a serious underlying occult lesion as the source of bleeding. A number of descriptive studies indicate that the chance of finding such a lesion is approximately 5% to 25%, which supports the need for further investigation in such cases. The significance of hematuria in a patient taking warfarin is less clear. There is a low prevalence of microscopic hematuria in patients not on anticoagulant therapy, and the likelihood of finding an underlying malignancy appears to be less.

Predicting and Managing the Risks of Oral Anticoagulant Therapy

Over the last 20 years, several developments have improved the safety and efficacy of oral anticoagulation. These include defining the optimal intensity of therapy by well-designed randomized trials and standardizing the reporting of PTs using the INR, which have led to more appropriate and standardized therapy. Identification of the risk factors associated with bleeding has also advanced. Time in the therapeutic range is an important measure of the quality of anticoagulation care, and a strong relationship exists between time in the therapeutic range and either bleeding or thrombosis rates as demonstrated by Cannegieter and colleagues, Hylek and coworkers, and others. In addition to time in the therapeutic range, a number of patient characteristics serve as risk factors for bleeding. Knowing these risk factors for an individual patient before initiating anticoagulant therapy or deciding to extend therapy is an essential aspect of care and the balance of risk and benefit. A number of risk indexes have been developed to help the clinician. These include the Bleeding Risk Index of Beyth and colleagues, the HEMORR2HAGES index of Gage and coworkers, the HAS-BLED score of Pisters and colleagues, and the ATRIA score of Fang and coworkers. All of these scoring systems incorporate many common risk factors, including age, history of bleeding, renal insufficiency, anemia, hypertension, and others. Unfortunately, the predictive ability of these scores when looked at by others has not always held up nor is one scoring system particularly superior to another. Moreover, none of them is so specific that other factors can be ignored, and in deciding the risk of bleeding in an individual patient, many factors need to be taken into consideration, including the living environment, other comorbid conditions, patient compliance, and others. Table 37.6 summarizes these bleeding risk indices.

TABLE 37.6
Commonly Used Bleeding Risk Indices to Assess Risk of Bleeding in Patients Receiving Vitamin K Antagonists
OBI HEMORR 2 HAGES HAS-BLED ATRIA ORBIT
Age ≥65 years 1 Abnormal renal/liver function 1 Hypertension 1 Anemia 3 Older age >74 1
Prior stroke 1 Alcohol abuse 1 Abnormal renal/liver function 1/2 Severe renal disease 3 Reduced Hgb/Hct/anemia 2
Prior gastrointestinal bleeding 1 Cancer 1 Stroke 1 Age ≥75 years 2 Bleeding history 2
Recent myocardial infarction, diabetes, hematocrit <30%, or creatinine >1.5 mg/dL 1–4 Age >75 1 Bleeding 1 Any prior bleed 1 Poor kidney function 1
Reduced platelet count or function 1 Labile INR 1 Hypertension 1 Antiplatelet therapy 1
Elderly (>65 years) 1
Rebleeding risk 1 Drugs or alcohol 1/2
Hypertension 1
Anemia 1
Genetic factors 1
Fall risk 1
Stroke 1
Low risk 0 ≤1 <3 <4 ≤2
High risk ≥3 ≥3 ≥3 ≥4 ≥4
INR, International normalized ratio; OBI, outpatient bleeding risk index.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here