15.05.2014 Views

Laboratory Monitoring of Warfarin Therapy - Pathology

Laboratory Monitoring of Warfarin Therapy - Pathology

Laboratory Monitoring of Warfarin Therapy - Pathology

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>Laboratory</strong> <strong>Monitoring</strong><br />

<strong>of</strong> <strong>Warfarin</strong> <strong>Therapy</strong><br />

David Williams, M.D., Ph.D.<br />

Roger S. Riley, M.D., Ph.D.<br />

Ann Tidwell, M.T. (ASCP) SH


<strong>Warfarin</strong> <strong>Monitoring</strong><br />

Crystalline warfarin sodium (Coumadin,<br />

Panwarfin, S<strong>of</strong>arin, Coufarin, Athrombin-K) is<br />

the most widely used oral anticoagulant in<br />

the world. <strong>Warfarin</strong> interferes with the hepatic<br />

synthesis <strong>of</strong> the vitamin-K dependent<br />

coagulation factors by interfering with the<br />

vitamin K cycle. <strong>Laboratory</strong> monitoring <strong>of</strong><br />

warfarin therapy is mandatory, since the<br />

agent has a relatively narrow therapeutic<br />

range. The therapeutic efficacy is <strong>of</strong> warfarin<br />

is reduced with an inadequate dose, while there<br />

is a serious risk <strong>of</strong> bleeding with an excess<br />

amount <strong>of</strong> the drug. In addition, the pharmacokinetics<br />

<strong>of</strong> warfarin are significantly influenced<br />

by the diet, other medications, and<br />

many other factors. During the past two decades,<br />

several developments in laboratory science<br />

have helped the clinician to ensure a<br />

proper dosage intensity that decreases the<br />

risk <strong>of</strong> bleeding while maintaining therapeutic<br />

efficacy. These developments include improved<br />

laboratory reagents and instrumentation,<br />

as well as a standardized method for reporting<br />

the prothrombin time (i.e., International<br />

Normalized Ratio, INR). However, a comprehensive<br />

system <strong>of</strong> quality assurance is still<br />

necessary for optimal patient care.<br />

It is imparative that physicians monitoring<br />

warfarinized patients must understand the biological<br />

effect and pharmacokinetics <strong>of</strong> warfarin,<br />

its interaction with other medications, and the<br />

factors affecting the prothrombin time/INR assay,<br />

including the recently discovered impact <strong>of</strong><br />

the CYP2C9 and VKORC1 genetic polymorphisms.<br />

The physician must also work with<br />

other members <strong>of</strong> the patient care team to ensure<br />

that patient identification procedures are<br />

followed and the proper type and amount <strong>of</strong><br />

specimen is submitted for laboratory testing.<br />

<strong>Warfarin</strong> is a synthetic derivative <strong>of</strong> the<br />

naturally occurring anti-coagulant dicumarol.<br />

The anti-coagulant properties <strong>of</strong> dicumarol<br />

were first observed in cattle that suffered a<br />

hemorrhagic disorder after being fed spoiled<br />

sweet clover hay during the 1920s. Karl Link<br />

identified the causative agent by 1939 and<br />

later developed warfarin as a rat poison before<br />

it was used in humans in the 1950s. However<br />

the anticoagulant mechanism <strong>of</strong> warfarin<br />

was not elucidated until twenty years<br />

later.<br />

<strong>Warfarin</strong> is a specific inhibitor <strong>of</strong> the vitamin<br />

K epoxide reductase necessary for the<br />

regeneration <strong>of</strong> vitamin K from vitamin K<br />

2,3-epoxide in vivo (Fig. 1). Vitamin K acts as<br />

a c<strong>of</strong>actor for !-glutamyl carboxylase, which<br />

carboxylates specific glutamic acid residues<br />

in vitamin K dependent proteins. The vitamin<br />

K dependent proteins include coagulation<br />

factors V, VII, IX, and X as well as protein C<br />

and protein S. This post-translational modification<br />

forms !-carboxyglutamic acid residues<br />

that chelate metal ions and allow the proteins<br />

to bind specific c<strong>of</strong>actors on phospholipid<br />

surfaces necessary for normal coagulation.<br />

The carboxylation reaction generates vitamin<br />

K 2,3-epoxide, which must be converted back<br />

to vitamin K by an epoxide reductase to<br />

maintain stores. Inhibition <strong>of</strong> vitamin K epoxide<br />

reductase reduces the availability <strong>of</strong> vitamin<br />

K, which leads to a decrease in the active<br />

forms <strong>of</strong> vitamin K dependent coagulation<br />

factors and ultimately anti-coagulation.<br />

<strong>Warfarin</strong> is currently the only FDA approved<br />

orally available anti-coagulant medication.<br />

It is rapidly absorbed from the gastrointestinal<br />

tract, reaching maximum concentration<br />

90 minutes after administration, with<br />

a half-life <strong>of</strong> 36 to 42 hours. However, the<br />

pharmacokinetics <strong>of</strong> warfarin can by highly<br />

variable. It circulates bound to albumin and is<br />

metabolized in the liver. Genetic variation in<br />

a cytochrome P450 enzyme alters the dose-<br />

<strong>Warfarin</strong> Structure, Metabolism, Pharmacokinetics 2


<strong>Warfarin</strong> <strong>Monitoring</strong><br />

O<br />

OH<br />

O<br />

O<br />

CH 3<br />

Fig. 1. The biochemistry <strong>of</strong> warfarin.<br />

(A) The molecular structure <strong>of</strong> warfarin.<br />

(B) A simplied schematic <strong>of</strong> the<br />

vitamin K cycle, showing the effect <strong>of</strong><br />

warfarin on the synthesis <strong>of</strong> vitamin-K<br />

dependent coagulation factors. In the<br />

cycle, vitamin K1 or K2 are reduced to<br />

their hydroquinone forms (KH2). A further<br />

stepwise oxidation <strong>of</strong> KH2 to vitamin<br />

K epoxide (KO) occurs, during<br />

which descarboxyprothrombin<br />

(descarboxy-II) is converted to<br />

prothrombin by carboyxlation <strong>of</strong> glutamate<br />

residues (Glu) to g-<br />

carboxyglutamate (Gla). Enzymatic<br />

reduction <strong>of</strong> the epoxide requires reduced<br />

nicotinamide adenine dinucleotide<br />

(NADH) as a c<strong>of</strong>actor and is sensitive<br />

to warfarin (Stop sign). The R<br />

sidechain on the vitamin K molecule is<br />

a 20-carbon phytyl in vitamin K1 and a<br />

5- to 65-carbon prenyl side chain in<br />

vitamin K2.<br />

response relationship as well as a number <strong>of</strong><br />

environmental, dietary, and disease related<br />

factors. A long list <strong>of</strong> commonly prescribed<br />

medications can either enhance or interfere<br />

with warfarin anti-coagulation by various<br />

mechanisms. For example, amiodarone inhibits<br />

clearance via hepatic metabolism and<br />

enhances anti-coagulation while carabamezepine,<br />

barbiturates, and rifampicin<br />

increase metabolic clearance and inhibit<br />

anti-coagulation. Interestingly, there are specific<br />

inherited mutations <strong>of</strong> factor IX that<br />

render it particularly sensitive to decreased<br />

carboxylation such that the anti-coagulation<br />

effect <strong>of</strong> warfarin is enhanced without prolonging<br />

the prothrombin time. Furthermore,<br />

dietary changes can have a pr<strong>of</strong>ound effect<br />

on anti-coagulation. Vitamin K is largely<br />

found in green vegetables such that increased<br />

intake <strong>of</strong> leafy green vegetables can<br />

provide vitamin K bypassing the need to regenerate<br />

stores with epoxide reductase.<br />

The variable pharmacokinetics <strong>of</strong> warfarin<br />

requires careful monitoring and dose<br />

adjustments to achieve a therapeutic range.<br />

Even after stabilizing within a therapeutic<br />

range, monitoring is still necessary since so<br />

<strong>Warfarin</strong> <strong>Monitoring</strong> 3


<strong>Warfarin</strong> <strong>Monitoring</strong><br />

many environmental and dietary factors can<br />

change the dose-response. Measuring the<br />

prothrombin time (PT) remains the primary<br />

mechanism by which warfarin anticoagulation<br />

is monitored. Prolongation <strong>of</strong><br />

the PT, however, depends on the responsiveness<br />

<strong>of</strong> the thromboplastin used to initiate<br />

coagulation. Hence, the WHO has currently<br />

established and international standard by<br />

which manufacturers or individual labs can<br />

determine an international sensitivity index<br />

(ISI) for each lot <strong>of</strong> thromboplastin. A therapeutic<br />

range <strong>of</strong> PT is then defined by the international<br />

normalized ratio (INR), which is<br />

the ratio between the patients PT to the mean<br />

normal PT for the lab raised to the power <strong>of</strong><br />

the ISI:<br />

Fig. 2. A nomogram for obtaining<br />

an INR from a measured<br />

prothrombin ratio (i.e., ratio <strong>of</strong><br />

patient PT/mean normal PT). The<br />

ISI is provided by the manufacturer<br />

<strong>of</strong> each lot <strong>of</strong> prothrombin<br />

reagent (thromboplastin). In the<br />

example shown, an INR <strong>of</strong> 3.0 is<br />

obtained from a plasma sample<br />

with a PTR <strong>of</strong> 2.0 if the ISI is 1.6.<br />

INR = (Patient PT/Mean Normal PT) ISI<br />

where the “mean normal PT” is the geometric<br />

mean prothrombin time for the laboratory.<br />

The ISI is a correction factor, specific for each<br />

lot <strong>of</strong> laboratory prothrombin reagent, that<br />

correlates the activity <strong>of</strong> the reagent to a<br />

standard maintained by the World Health Organization.<br />

A smaller ISI reflects a more sensitive<br />

thromboplastin reagent such that a<br />

more prolonged PT will be observed for the<br />

same therapeutic effect.<br />

In most medical institutions, the INR is<br />

automatatically calculated by the laboratory<br />

information system. The INR can also be calculated<br />

from the above formula or derived<br />

from an INR nomogram (Fig. 2).<br />

The dosing <strong>of</strong> warfarin is driven by<br />

monitoring the prothrombin time to achieve<br />

a PT in the appropriate therapeutic range.<br />

Typically 5 to 10 mg <strong>of</strong> warfarin is administered<br />

daily with daily monitoring <strong>of</strong> the INR.<br />

The dose is changed gradually until therapeutic<br />

and then the INR is followed on a regular<br />

but less frequent basis. Adjustments in dosage<br />

should be based on the weekly dose since<br />

<strong>Warfarin</strong> <strong>Monitoring</strong> 4


<strong>Warfarin</strong> <strong>Monitoring</strong><br />

warfarin has a long half-life. For most<br />

indications the INR should be maintained<br />

between 2.0 to 3.0. Exceptions include<br />

anticoagulation in patients with<br />

mechanical prosthetic heart valves and<br />

patients with the antiphospholipid antibody<br />

syndrome, in which the INR is usually<br />

kept between 2.5 and 3.5. However, it<br />

must be remembered that the INR was<br />

developed for deep venous thrombosis<br />

prophylaxis and may not be the most appropriate<br />

monitoring mechanism for<br />

other situations. Furthermore, inhibitors<br />

such as lupus anticoagulants can prolong<br />

the PT obscuring the appropriate therapeutic<br />

range such that it may be necessary<br />

to use alternative assays such as the<br />

direct quantitation <strong>of</strong> the plasma warfarin<br />

concentration.<br />

Fig. 3. Recommended therapeutic<br />

ranges for oral anticoagulant therapy.<br />

Most recent recommendations<br />

<strong>of</strong> the American College <strong>of</strong> chest<br />

Physicians and the National Heart,<br />

Lung, and Blood institute. From<br />

Ansell et al. 2004.<br />

As with all anticoagulant medications,<br />

the primary risk for warfarin therapy is<br />

hemorrhage. The risk is greater the<br />

higher the INR, highlighting the importance<br />

<strong>of</strong> regular monitoring. The risk <strong>of</strong><br />

a major bleed is 1 to 2 percent per year<br />

with only a 0.1 to 0.5 percent per year<br />

risk <strong>of</strong> intracranial hemorrhage. If a pa-<br />

<strong>Warfarin</strong> Risks & Side Effects 5


<strong>Warfarin</strong> <strong>Monitoring</strong><br />

Table I<br />

Clinical Management <strong>of</strong> the Supratherapeutic INR*<br />

*Data from Ansell, 2004 and other sources. The purpose <strong>of</strong> this table is to convey general guidelines for the treatment <strong>of</strong> a<br />

supratherapeutic INR. Clinical judgment is required for the individual patient.<br />

<strong>Warfarin</strong> Risks and Side Effects 6


<strong>Warfarin</strong> <strong>Monitoring</strong><br />

tient is actively bleeding and the INR needs to<br />

be corrected acutely, fresh frozen plasma can<br />

be administered with vitamin K and in extreme<br />

12cases prothrombin complex concentrates<br />

can be given. When a patient is found<br />

to have a markedly elevated PT without active<br />

bleeding, a more conservative approach is<br />

warranted such as simply skipping dosages<br />

until the INR returns to the appropriate range<br />

(Table I).<br />

<strong>Warfarin</strong>-induced skin necrosis is a rare<br />

but significant side effect. It follows administration<br />

by one to ten days, is heralded by<br />

parasthesia and an erythematous flush, which<br />

is followed by petechiae and hemorrhage<br />

which leads to skin necrosis. Skin necrosis is<br />

most commonly associated with proctein C<br />

deficiency and occasionally protein S deficiency.<br />

Protein C has a very short half-life<br />

and a rapid decline in activity from over 50%<br />

to less than 5% apparently initiates the development<br />

<strong>of</strong> skin necrosis in these patients.<br />

Therefore, screening for protein C and S deficiencies<br />

in patients with deep venous thrombosis<br />

may help to identify those with an increased<br />

risk <strong>of</strong> skin necrosis.<br />

Ansell, J. et al. The Pharmacology and Management<br />

<strong>of</strong> the Vitamin K Antagonists: The<br />

Seventh ACCP Conference on Antithrombotic<br />

and Thrombolytic <strong>Therapy</strong>. Chest 126: 204-<br />

233, 2004.<br />

DeLoughery, T.G. “Oral anticoagulants” in<br />

Disorders <strong>of</strong> Hemostasis and Thrombosis, a<br />

Clinical Guide, Second Edition. Eds Goodnight,<br />

S.H. Jr. & Hathaway, W.E.. The McGraw-<br />

Hill Companies, New York, pp. 553-566, 2001.<br />

Fairweather, R.B., Ansell, J., van den Besselaar,<br />

A.M.H.P., Brandt, J.T., Bussey, H.I., Poller, L.,<br />

Triplett, D.A. & White, R.H. College <strong>of</strong> American<br />

Pathologists Conference XXXI on laboratory<br />

monitoring <strong>of</strong> anticoagulant therapy.<br />

<strong>Laboratory</strong> monitoring <strong>of</strong> oral anticoagulant<br />

therapy. Arch. Pathol. Lab. Med. 122:768-781,<br />

1998.<br />

Gage, B.F. et al. Management and dosing <strong>of</strong><br />

warfarin therapy. Am. J. Med. 109:481– 488,<br />

2000.<br />

Hirsh, J. Oral anticoagulants: Mechanism <strong>of</strong><br />

action, clinical effectiveness, and optimal<br />

therapeutic range. Chest 119: 8S-21S, 2001.<br />

Jaffer, A.K. et al. Low–Molecular-Weight-<br />

Heparins as Periprocedural Anticoagulation<br />

for Patients on Long-Term <strong>Warfarin</strong> <strong>Therapy</strong>:<br />

A Standardized Bridging <strong>Therapy</strong> Protocol. J.<br />

Thromb. Thrombolysis. 20:11-16, 2005.<br />

Mueller, R.L. First-generation agents: aspirin,<br />

heparin and coumarins. Best Pract. Res. Clin.<br />

Haematol. 17:23-53, 2004.<br />

Riley, R.S., Fisher, L.M., and Rowe, D. Clinical<br />

utilization <strong>of</strong> the International Normalized<br />

Ratio (INR). J. Clin. Lab. Anal. 14:101-114, 2000.<br />

Sconce, E.A. The impact <strong>of</strong> CYP2C9 and<br />

VKORC1 genetic polymorphism and patient<br />

characteristics upon warfarin dose requirements:<br />

proposal for a new dosing regimen.<br />

Blood. 106:2329-2333, 2005.<br />

Tie, J.K., Nicchitta, C., von Heijne, G. & Stafford,<br />

D.W. Membrane topology mapping <strong>of</strong><br />

vitamin K epoxide reductase by in vitro<br />

translation/cotranslocation. J. Biol. Chem. 280:<br />

16410-16416, 2005.<br />

Wallin, R. and Hutson, S.M. <strong>Warfarin</strong> and the<br />

Vitamin K-Dependent !-Carboxylation System.<br />

Trends Mol. Med. 10: 299-302, 2004.<br />

References 7

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!