Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Oct 1.
Published in final edited form as: Thromb Haemost. 2010 Aug 5;104(4):750–754. doi: 10.1160/TH09-11-0763

Gamma-glutamyl carboxylase and its influence on warfarin dose

Cristi R King 1, Elena Deych 1, Paul Milligan 1, Charles Eby 1,2, Petra Lenzini 1, Gloria Grice 3, Rhonda M Porche-Sorbet 2, Paul M Ridker 4, Brian F Gage 1
PMCID: PMC2949522  NIHMSID: NIHMS225540  PMID: 20694283

Abstract

Background

Via generation of vitamin K-dependent proteins, gamma-glutamyl carboxylase (GGCX) plays a critical role in the vitamin K cycle. Single nucleotide polymorphisms (SNPs) in GGCX, therefore, may affect dosing of the vitamin K antagonist, warfarin.

Methods

In a multi-centered, cross-sectional study of 985 patients prescribed warfarin therapy, we genotyped for two GGCX SNPs (rs11676382 and rs12714145) and quantified their relationship to therapeutic dose.

Results

GGCX rs11676382 was a significant (p=0.03) predictor of residual dosing error and was associated with a 6.1% reduction in warfarin dose (95% CI: 0.6%-11.4%) per G allele. The prevalence was 14.1% in our predominantly (78%) Caucasian cohort, but the overall contribution to dosing accuracy was modest (partial R2 = 0.2%). GGCX rs12714145 was not a significant predictor of therapeutic dose (p = 0.26).

Conclusions

GGCX rs11676382 is a statistically significant predictor of warfarin dose, but the clinical relevance is modest. Given the potentially low marginal cost of adding this SNP to existing genotyping platforms, we have modified our non-profit website (www.WarfarinDosing.org) to accommodate knowledge of this variant.

Keywords: gamma-glutamyl carboxylase, warfarin, pharmacogenetics, algorithm

INTRODUCTION

Warfarin (Coumadin™ and others) is an anticoagulant with a narrow therapeutic window and wide inter-individual response. In 2007, and again in January 2010, the United States Food and Drug Administration approved label changes for warfarin to recommend lower initial doses for patients known to have single nucleotide polymorphisms (SNPs) in two genes: one affecting warfarin metabolism (CYP2C9), and the other affecting sensitivity (VKORC1) (1-3). Several pharmacogenetic algorithms incorporate SNPs in both genes (e.g. CYP2C9*2, *3 and VKORC1 -1639 G>A), but these SNPs alone explain only one-third of the variability in warfarin dose (4-8). The inclusion of non-genetic factors can explain an additional one-fifth of warfarin dosing variability (8, 9). Incorporating other SNPs may further improve the accuracy of warfarin dosing (10-13).

Because of the critical pharmacodynamic role gamma-glutamyl carboxylase (GGCX) plays in the generation of vitamin K-dependent proteins, SNPs in the GGCX gene may affect the dose of warfarin and other vitamin K antagonists. GGCX resides in the endoplasmic reticulum membrane, oxidizing reduced vitamin K to vitamin K-2,3-epoxide while adding a carboxyl residue to the gamma carbon on selected glutamic acids to produce functional clotting factors II, VII, IX, and X and other vitamin K dependent proteins (6, 14-16). Patients with defects in GGCX have elevated International Normalized Ratios (INRs), even in the absence of a vitamin K antagonist (17), and GGCX-knockout mice die at birth from massive hemorrhage (18).

Although previous investigators have examined GGCX polymorphisms in patients taking warfarin, initial reports of associations with warfarin dose have either been unsubstantiated or await validation. A SNP located in intron 2, rs12714145 G>A, was associated with a 3.3% increase in warfarin dose requirements per A allele in a Swedish study of 201 patients (6). This observation was not confirmed in two subsequent studies which genotyped only 186 European Americans and 318 Chinese patients, respectively (19, 20). However, if validated, the association would be important because this SNP explained 1.4-3.3% of the dose variability in the Swedish population (6).

More recently, GGCX rs11676382 C>G, located in intron 14, correlated with lower warfarin doses in a single-center study of 186 European Americans, explaining 2% of the total variance of the warfarin maintenance dose in a dominant genetic model (19). However, the putative association was not significant in an additive model or after adjusting for multiple comparisons. Furthermore, Lubitz et al. did not confirm the significance of this SNP in 145 multiethnic patients (21). The goal of the present study, therefore, was to further investigate the putative associations between warfarin therapeutic dose and GGCX rs12714145 and rs11676382 in a large, multiracial, multi-centered cohort.

MATERIALS AND METHODS

Study Design

This was a cross-sectional study of 985 patients who have been well characterized (8, 11, 22, 23). Patients provided written, informed consent for use of their DNA, and the participating institutions approved the protocol. We genotyped adult patients (Table 1) who had their warfarin therapy managed at one of three sites: the anticoagulation clinics affiliated with Barnes–Jewish Hospital at Washington University Medical Center in St. Louis, the Anticoagulation Clinics of North America (San Antonio), and subjects who participated in the PREVENT (PREvention of VENous Thromboembolism) study (23). A stable therapeutic warfarin dose was defined as a dose that was unchanged for at least six days and resulted in two or more consecutive therapeutic INR values at least 3 days apart.

Table 1.

Patient Demographics for GGCX analysis.

Variables
Demographic variables N = 985
        Age, mean (SD), y 60 (15)
        Gender:
            Women, N (%) 486 (49.3%)
            Men, N (%) 499 (50.7%)
        Race:
            Caucasian, N (%) 772 (78.4%)
            African-American, N (%) 180(18.3%)
            Asian or East Indian, N (%) 3(0.3%)
            Other or Unknown, N (%) 30 (3.0%)
Clinical variables
        Therapeutic warfarin dose, geometric mean (SD), mg/d 4.8 (1.6)
        Target INR, mean (SD), units 2.3 (0.4)
        Body surface area, mean (SD), m2 2.0 (0.3)
        Current smoker, N (%) 140 (14%)
        Takes amiodarone, N (%) 33 (3%)

SD = standard deviation; INR = international normalized ratio

Genotyping

DNA was extracted from patients’ whole blood or buffy coat samples using Puregene Purification Kits (Gentra, now Qiagen, Hilden, Germany). Genotyping was performed for GGCX rs11676382 and rs12714145 using PCR and Pyrosequencing® (24, 25). PCR primers were designed using the UCSC Golden Path Human Genome Browser March 2006 Build (http://genome.ucsc.edu/cgi-bin/hgGateway) and GGCX Refseq NM_000821 (Appendix 1).

Statistical analysis

To test an additive effect of GGCX SNPs on warfarin dose requirements, we coded each GGCX SNP as 0, 1, or 2 for the number of variant alleles present. We then regressed the number of GGCX SNPs onto the residual of the prediction according to a validated pharmacogenetic algorithm (8, 21): residual=In(observed therapeutic dose)-In(predicted dose)= βo+β1*(number of alleles), where ln is the natural logarithm. By exponentiating the residual error, we obtain the ratio of the observed dose/predicted dose, which can be used to correct the validated algorithm for GGCX, when genotype is known (Appendix 2).

To reduce the effect of extreme therapeutic dose values on regression coefficients, we Winsorized the 6 doses that fell outside of the usual (1-18 mg/day) range. We tested the model fit using standard regression diagnostic methods. All analyses were performed using SAS version 9.1 (SAS Institute, Cary, NC).

RESULTS

Participants had an average age of 60 years and half were female (Table 1). One hundred seventeen (12%) inherited one or two copies of the GGCX rs11676382 G allele (Table 2). The geometric means (SD) of the therapeutic dose (mg/day), stratified by rs11676382, were: C/C 4.9 (1.6); G/C 4.1 (1.6); and G/G 3.8 (2.2). GGCX rs11676382 was a significant predictor of residual dosing error (p=0.03), and was associated with a 6.1% (95% CI: 0.6%-11.4%) reduction in warfarin dose, per G allele (Table 3). Adding rs1167382 to the validated pharmacogenetic algorithm (8) increased the R2 by 0.2% in the total population and by 0.3% in Caucasians.

Table 2.

Genotype Frequencies.

GGCX Alleles Caucasians African-Americans Asian or East Indians Other or Unknown Race Association (95% CI) with dose residual, per allele
rs11676382
                C/C 662 (85.8%) 175 (97.2%) 3 28 (93.3%)
                G/C 103 (13.3%) 5 (2.8%) 0 2 (6.7%) 6.1% (0.6%-11.4%)
                G/G 7 (0.9%) 0 (0.0%) 0 0 (0%)
rs12714145
                G/G 394 (51.0%) 56 (31.1%) 3 14(46.7%)
                G/A 298 (38.6%) 91 (50.6%) 0 14 (46.7%) NS
                A/A 80 (10.4%) 33 (18.3%) 0 2 (6.7%)

NS = not significant; CI = confidence interval.

Table 3.

Adjustment Factors for Existing Pharmacogenetic Dosing Algorithms, Depending on GGCX Genotype.

GGCX rs11676382 genotype Formula for Correction Factor* Adjustment in African-Americans and Asians* Adjustment in Caucasians*
CC 1 / (PC2 + 2PCPG × 0.939 + PG2 × 0.9392) 1.00 1.01
GC 0.939 / (PC2 + 2PCPG × 0.939 + PG2 × 0.9392) 0.94 0.95
GG 0.9392 / (PC2 + 2PCPG × 0.939 + PG2 × 0.9392) 0.88 0.89
*

multiply predicted dose by this correction factor.

PC = allele frequency of the wildtype C allele.

PG= allele frequency of the G allele.

Half of the subjects (N = 492) carried either one or both copies of the GGCX rs12714145 A allele (Table 2), but it was not a significant predictor of dose residual (p=0.39). The geometric mean (SD) therapeutic dose was 4.7 (1.6) for subjects with either the GGCX rs12714145 A/A or A/G genotype, and was 5.1 (1.6) for those with G/G genotype.

DISCUSSION

Although GGCX rs12714145 was not confirmed as a predictor of therapeutic warfarin dose requirements, GGCX rs11676382 C>G was significantly associated with decreased warfarin requirements: each G allele was associated with a 6.1% reduction (e.g. 93.9% of prediction) in dose. The effect of GGCX rs11676382 C>G on R2 was modest, however, and could explain why Lubitz et. al were unable to confirm this association in their smaller population of 145 diverse patients (21). Given the allele frequency of rs11676382 C>G, we estimate about 14% of Caucasians would carry this SNP, and without genotyping for it, their initial pharmacogenetic-based therapeutic warfarin dose estimates would be 6%–12% too high. Because of warfarin's narrow therapeutic index, even this modest over-estimation of warfarin dose could contribute to supratherapeutic INR values or bleeding.

With CYP2C9 (*2 and *3) and VKORC1 −1639 G>A SNPs explaining about one-third of the variability in therapeutic warfarin dose (4-6, 8, 26), and emerging data from recent high throughput genotyping studies of Swedish and Chinese populations (27-29), few other genes are likely to have major effects on warfarin dose. However, by genotyping for GGCX rs11676382 and other SNPs that raise R2 modestly (10-13), one could increase the accuracy of existing pharmacogenetic algorithms significantly.

In their sample of 201 predominantly Swedish patients, Wadelius et al. 2005, found a 3.3% increase in dose per GGCX rs12714145 A allele (6, 30). While their univariate analysis was significant for this SNP (p=0.036), their multivariate analysis was not (6, 30). Reider et al., on the other hand, did not find any association with warfarin dose and rs10691423, a SNP that is in high linkage disequilibrium with rs12714145 (19). Similarly, our analysis of GGCX rs12714145 A SNP in a larger (n= 985), more diverse population found no association between this SNP and warfarin dose.

The scope of the present study was limited to validating two GGCX SNPs in a large and diverse cohort of therapeutic patients. We did not evaluate GGCX rs699664 (8016G>A; Q325R), which was associated with increased warfarin dosage in Japanese patients (31), even though prior studies in Caucasian populations did not confirm this putative association (19, 32, 33). A related limitation is that few Asian patients were included in the present study. However rs11676382 C>G has yet to be observed in Asian populations according to public databases such as HapMap Phase 3. Lastly, as with any warfarin related study, definitions of stable warfarin dose and selection of covariates vary among studies (34-38), and obfuscate attempts at repeatability.

In August, 2009 the United States Center for Medicare and Medicaid Services (CMS) announced it would reimburse for warfarin pharmacogenetic testing only in the setting of approved randomized clinical trials. We hope that CMS will remunerate laboratories involved in warfarin pharmacogenetic research not only for VKORC1 and CYP2C9 testing, but also for GGCX and other SNPs (10-13) that modestly improve accuracy. Although delaying warfarin therapy while waiting for a $400 genotype of 2 genes may not be cost effective now,(39) in the future, genotyping of multiple SNPs will become faster and less expensive, so the cost effectiveness of pharmacogenetic dosing will improve. Using all available pharmacogenetic information during warfarin initiation would allow for optimal dosing accuracy. With this vision in mind, we have updated our non-profit website, www.WarfarinDosing.org, to accommodate, but not require, GGCX rs11676382. We will use this website in our upcoming trial (ClinicalTrials.gov Identifier NCT01006733).

Supplementary Material

Supplementary Data

Table 4.

Warfarin Pharmacogenetics Snapshot: past, present and future.

1. What is known about this topic?
        • Warfarin is an anticoagulant with a narrow therapeutic window and wide intra-individual response.
        • In 2007 and again in 2010, the US FDA approved labeling changes for warfarin to recommend lower initial doses for patients known to have SNPs in genes affecting warfarin metabolism (CYP2C9) and sensitivity (VKORC1).
        • SNPs in CYP2C9 and VKORC1 explain one-third of the variability in warfarin dose.
2. What does this paper add?
        • GGCX rs11676382 was associated with a 6.1% reduction in warfarin dose (95% CI: 0.6%-11.4%) per G allele.
        • Knowledge of GGCX rs11676382 genotype has been incorporated in www.WarfarinDosing.org for pharmacogenetic dose initiation.
        • GGCX rs12714145 was not a significant predictor of warfarin dose

ACKNOWLEDGEMENTS

This study was supported by the NIH (R01 HL074724, R01 HL097036, R01 HL57951, R01 HL58036) and the American Heart Association.

REFERENCES

  • 1. [August 16, 2007];FDA Approves Updated Warfarin (Coumadin) Prescribing Information: New Genetic Information May Help Providers Improve Initial Dosing Estimates of the Anticoagulant for Individual Patients. FDA News 2007. Available from: http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/2007/ucm108967.htm.
  • 2. [2010 March 10];Safety Labeling Changes Approved By FDA Center for Drug Evaluation and Research (CDER) – January 2010. Safety 2010. Available from: http://www.fda.gov/Safety/MedWatch/SafetyInformation/ucm201100.htm.
  • 3.Wood S. [2007 Sept 4, 2007];New Warfarin Labeling Reminds Physicians About Genetic Tests to Help Guide Initial Warfarin Dosing. 2007. Available from: http://www.medscape.com/viewarticle/561608.
  • 4.Rieder MJ, Reiner AP, Gage BF, et al. Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. N Engl J Med. 2005;352(22):2285–93. doi: 10.1056/NEJMoa044503. [DOI] [PubMed] [Google Scholar]
  • 5.Sconce EA, Khan TI, Wynne HA, et al. The impact of CYP2C9 and VKORC1 genetic polymorphism and patient characteristics upon warfarin dose requirements: proposal for a new dosing regimen. Blood. 2005;106:2329–33. doi: 10.1182/blood-2005-03-1108. [DOI] [PubMed] [Google Scholar]
  • 6.Wadelius M, Chen LY, Downes K, et al. Common VKORC1 and GGCX polymorphisms associated with warfarin dose. The Pharmacogenomics Journal. 2005;5:262–70. doi: 10.1038/sj.tpj.6500313. [DOI] [PubMed] [Google Scholar]
  • 7.Herman D, Locatelli I, Grabnar I, et al. Influence of CYP2C9 polymorphisms, demographic factors and concomitant drug therapy on warfarin metabolism and maintenance dose. The Pharmacogenomics Journal. 2005;5(3):193–202. doi: 10.1038/sj.tpj.6500308. [DOI] [PubMed] [Google Scholar]
  • 8.Gage BF, Eby C, Johnson JA, et al. Use of pharmacogenetic and clinical factors to predict the therapeutic dose of warfarin. Clinical Pharmacology and Therapeutics. 2008;84(3):326–31. doi: 10.1038/clpt.2008.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Klein TE, Altman RB, Eriksson N, et al. Estimation of the warfarin dose with clinical and pharmacogenetic data. N Engl J Med. 2009;360(8):753–64. doi: 10.1056/NEJMoa0809329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Caldwell MD, Awad T, Johnson JA, et al. CYP4F2 genetic variant alters required warfarin dose. Blood. 2008;111(8):4106–12. doi: 10.1182/blood-2007-11-122010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Voora D, Koboldt DC, King CR, et al. A Polymorphism in the VKORC1 Regulator Calumenin Predicts Higher Warfarin Dose Requirements in African Americans. Clinical Pharmacology and Therapeutics. 2010 Mar 3; doi: 10.1038/clpt.2009.291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Cavallari LH, Langaee TY, Momary KM, et al. Genetic and Clinical Predictors of Warfarin Dose Requirements in African Americans. Clinical Pharmacology and Therapeutics. 2010 Jan 13; doi: 10.1038/clpt.2009.223. [DOI] [PubMed] [Google Scholar]
  • 13.Scott S, Edelmann L, Kornreich R, et al. Warfarin Pharmacogenetics: CYP2C9 and VKORC1 Genotypes Predict Different Sensitivity and Resistance Frequencies in the Ashkenazi and Sephardi Jewish Populations. AJHG. 2008;82(2):495–500. doi: 10.1016/j.ajhg.2007.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rost S, Fregin A, Koch D, et al. Compound heterozygous mutations in the gamma-glutamyl carboxylase gene cause combined deficiency of all vitamin K-dependent blood coagulation factors. Br J Haematol. 2004;126(4):546–9. doi: 10.1111/j.1365-2141.2004.05071.x. [DOI] [PubMed] [Google Scholar]
  • 15.Shikata E, Ieiri I, Ishiguro S, et al. Association of pharmacokinetic (CYP2C9) and pharmacodynamic (vitamin K-dependent protein-Factors II, VII, IX, and X, proteins S and C, and {gamma}-glutamyl carboxylase) gene variants with warfarin sensitivity. Blood. 2004;103:2630–5. doi: 10.1182/blood-2003-09-3043. [DOI] [PubMed] [Google Scholar]
  • 16.Gage BF, Eby CS. The genetics of vitamin K antagonists. The Pharmacogenomics Journal. 2004;4(4):224–5. doi: 10.1038/sj.tpj.6500258. [DOI] [PubMed] [Google Scholar]
  • 17.Vanakker OM, Martin L, Gheduzzi D, et al. Pseudoxanthoma elasticum-like phenotype with cutis laxa and multiple coagulation factor deficiency represents a separate genetic entity. J Invest Dermatol. 2007;127(3):581–7. doi: 10.1038/sj.jid.5700610. [DOI] [PubMed] [Google Scholar]
  • 18.Zhu A, Sun H, Raymond RM, Jr., et al. Fatal hemorrhage in mice lacking gamma-glutamyl carboxylase. Blood. 2007;109(12):5270–5. doi: 10.1182/blood-2006-12-064188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Rieder MJ, Reiner AP, Rettie AE. Gamma-glutamyl carboxylase (GGCX) tagSNPs have limited utility for predicting warfarin maintenance dose. J Thromb Haemost. 2007;5(11):2227–34. doi: 10.1111/j.1538-7836.2007.02744.x. [DOI] [PubMed] [Google Scholar]
  • 20.Wang TL, Li HL, Tjong WY, et al. Genetic factors contribute to patient-specific warfarin dose for Han Chinese. Clin Chim Acta. 2008;396(1-2):76–9. doi: 10.1016/j.cca.2008.07.005. [DOI] [PubMed] [Google Scholar]
  • 21.Lubitz SA, Scott SA, Rothlauf EB, et al. Comparative performance of gene-based warfarin dosing algorithms in a multiethnic population. J Thromb Haemost. 2010 Feb 2; doi: 10.1111/j.1538-7836.2010.03792.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lenzini PA, Grice GR, Milligan PE, et al. Laboratory and clinical outcomes of pharmacogenetic vs. clinical protocols for warfarin initiation in orthopedic patients. J Thromb Haemost. 2008;6(10):1655–62. doi: 10.1111/j.1538-7836.2008.03095.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ridker PM, Goldhaber SZ, Danielson E, et al. Long-term, low-intensity warfarin therapy for the prevention of recurrent venous thromboembolism. N Engl J Med. 2003;348(15):1425–34. doi: 10.1056/NEJMoa035029. [DOI] [PubMed] [Google Scholar]
  • 24.Marsh S, King CR, Garsa AA, et al. Pyrosequencing of clinically relevant polymorphisms. Methods in Molecular Biology (Clifton, NJ. 2005;311:97–114. doi: 10.1385/1-59259-957-5:097. [DOI] [PubMed] [Google Scholar]
  • 25.King CR, Porche-Sorbet RM, Gage BF, et al. Performance of commercial platforms for rapid genotyping of polymorphisms affecting warfarin dose. Am J Clin Pathol. 2008;129(6):876–83. doi: 10.1309/1E34UAPR06PJ6HML. [DOI] [PubMed] [Google Scholar]
  • 26.Herman D, Peternel P, Stegnar M, et al. The influence of sequence variations in factor VII, gamma-glutamyl carboxylase and vitamin K epoxide reductase complex genes on warfarin dose requirement. Thrombosis and Haemostasis. 2006;95(5):782–7. [PubMed] [Google Scholar]
  • 27.Takeuchi F, McGinnis R, Bourgeois S, et al. A genome-wide association study confirms VKORC1, CYP2C9, and CYP4F2 as principal genetic determinants of warfarin dose. PLoS Genet. 2009;5(3):e1000433. doi: 10.1371/journal.pgen.1000433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wadelius M, Chen LY, Lindh JD, et al. The largest prospective warfarin-treated cohort supports genetic forecasting. Blood. 2009;113(4):784–92. doi: 10.1182/blood-2008-04-149070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lee MT, Chen CH, Chou CH, et al. Genetic determinants of warfarin dosing in the Han-Chinese population. Pharmacogenomics. 2009;10(12):1905–13. doi: 10.2217/pgs.09.106. [DOI] [PubMed] [Google Scholar]
  • 30.Chen LY, Eriksson N, Gwilliam R, et al. Gamma-glutamyl carboxylase (GGCX) microsatellite and warfarin dosing. Blood. 2005;106(10):3673–4. doi: 10.1182/blood-2005-04-1711. [DOI] [PubMed] [Google Scholar]
  • 31.Kimura R, Miyashita K, Kokubo Y, et al. Genotypes of vitamin K epoxide reductase, gamma-glutamyl carboxylase, and cytochrome P450 2C9 as determinants of daily warfarin dose in Japanese patients. Thrombosis Research. 2007;120(2):181–6. doi: 10.1016/j.thromres.2006.09.007. [DOI] [PubMed] [Google Scholar]
  • 32.Loebstein R, Vecsler M, Kurnik D, et al. Common genetic variants of microsomal epoxide hydrolase affect warfarin dose requirements beyond the effect of cytochrome P450 2C9. Clinical Pharmacology and Therapeutics. 2005;77(5):365–72. doi: 10.1016/j.clpt.2005.01.010. [DOI] [PubMed] [Google Scholar]
  • 33.Vecsler M, Loebstein R, Almog S, et al. Combined genetic profiles of components and regulators of the vitamin K-dependent gamma-carboxylation system affect individual sensitivity to warfarin. Thrombosis and Haemostasis. 2006;95(2):205–11. doi: 10.1160/TH05-06-0446. [DOI] [PubMed] [Google Scholar]
  • 34.Voora D, Eby C, Linder MW, et al. Prospective dosing of warfarin based on cytochrome P-450 2C9 genotype. Thrombosis and haemostasis. 2005;93(4):700–5. doi: 10.1160/TH04-08-0542. [DOI] [PubMed] [Google Scholar]
  • 35.Crowther MA, Ginsberg JB, Kearon C, et al. A randomized trial comparing 5-mg and 10-mg warfarin loading doses. Arch Intern Med. 1999;159:46–8. doi: 10.1001/archinte.159.1.46. [DOI] [PubMed] [Google Scholar]
  • 36.Gage BF, Eby C, Milligan PE, et al. Use of pharmacogenetics and clinical factors to predict the maintenance dose of warfarin. Thrombosis and Haemostasis. 2004;91(1):87–94. doi: 10.1160/TH03-06-0379. [DOI] [PubMed] [Google Scholar]
  • 37.Siguret V, Gouin I, Debray M, et al. Initiation of warfarin therapy in elderly medical inpatients: a safe and accurate regimen. Am J Med. 2005;118(2):137–42. doi: 10.1016/j.amjmed.2004.07.053. [DOI] [PubMed] [Google Scholar]
  • 38.Fennerty A, Dolben J, Thomas P, et al. Flexible induction dose regimen for warfarin and prediction of maintenance dose. British Medical Journal (Clinical research ed. 1984;288(6426):1268–70. doi: 10.1136/bmj.288.6426.1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Eckman MH, Rosand J, Greenberg SM, et al. Cost-effectiveness of using pharmacogenetic information in warfarin dosing for patients with nonvalvular atrial fibrillation. Annals of Internal Medicine. 2009;150(2):73–83. doi: 10.7326/0003-4819-150-2-200901200-00005. [DOI] [PubMed] [Google Scholar]
  • 40.Knowlton CH, Thomas OV, Williamson A, et al. Establishing community pharmacy-based anticoagulation education and monitoring programs. J Am Pharm Assoc (Wash) 1999;39(3):368–74. doi: 10.1016/s1086-5802(16)30445-4. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Data

RESOURCES