Skip to main content
Log in

Assessing the relative potency of (S)- and (R)-warfarin with a new PK-PD model, in relation to VKORC1 genotypes

  • Pharmacodynamics
  • Published:
European Journal of Clinical Pharmacology Aims and scope Submit manuscript

Abstract

Purpose

The purpose of this study is to develop a new pharmacokinetic-pharmacodynamic (PK-PD) model to characterise the contribution of (S)- and (R)-warfarin to the anticoagulant effect on patients in treatment with rac-warfarin.

Methods

Fifty-seven patients starting warfarin (W) therapy were studied, from the first dose and during chronic treatment at INR stabilization. Plasma concentrations of (S)- and (R)-W and INRs were measured 12, 36 and 60 h after the first dose and at steady state 12–14 h after dosing. Patients were also genotyped for the G>A VKORC1 polymorphism. The PK-PD model assumed a linear relationship between W enantiomer concentration and INR and included a scaling factor k to account for a different potency of (R)-W. Two parallel compartment chains with different transit times (MTT1 and MTT2) were used to model the delay in the W effect. PD parameters were estimated with the maximum likelihood approach.

Results

The model satisfactorily described the mean time-course of INR, both after the initial dose and during long-term treatment. (R)-W contributed to the rac-W anticoagulant effect with a potency of about 27% that of (S)-W. This effect was independent of VKORC1 genotype. As expected, the slope of the PK/PD linear correlation increased stepwise from GG to GA and from GA to AA VKORC1 genotype (0.71, 0.90 and 1.49, respectively).

Conclusions

Our PK-PD linear model can quantify the partial pharmacodynamic activity of (R)-W in patients contemporaneously exposed to therapeutic (S)-W plasma levels. This concept may be useful in improving the performance of future algorithms aiming at identifying the most appropriate W maintenance dose.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Wu AH (2007) Use of genetic and nongenetic factors in warfarin dosing algorithms. Pharmacogenomics 8:851–861

    Article  CAS  PubMed  Google Scholar 

  2. Stergiopoulos K, Brown DL (2014) Genotype-guided vs clinical dosing of warfarin and its analogues: meta-analysis of randomized clinical trials. JAMA Intern Med 174:1330–1338

    Article  PubMed  Google Scholar 

  3. Wang ZQ, Zhang R, Zhang PP, Liu XH, Sun J, Wang J et al (2015) Pharmacogenetics-based warfarin dosing algorithm decreases time to stable anticoagulation and the risk of major hemorrhage: an updated meta-analysis of randomized controlled trials. J Cardiovasc Pharmacol 65:364–370

    Article  CAS  PubMed  Google Scholar 

  4. Li X, Yang J, Wang X, Xu Q, Zhang Y, Yin T (2015) Clinical benefits of pharmacogenetic algorithm-based warfarin dosing: meta-analysis of randomized controlled trials. Thromb Res 135:621–629

    Article  CAS  PubMed  Google Scholar 

  5. Tang HL, Shi WL, Li XG, Zhang T, Zhai SD, Xie HG (2015) Limited clinical utility of genotype-guided warfarin initiation dosing algorithms versus standard therapy: a meta-analysis and trial sequential analysis of 11 randomized controlled trials. Pharmacogenomics J 15:496–504

    Article  CAS  PubMed  Google Scholar 

  6. Dahal K, Sharma SP, Fung E, Lee J, Moore JH, Unterborn JN et al (2015) Meta-analysis of randomized controlled trials of genotype-guided vs standard dosing of warfarin. Chest 148:701–710

    Article  PubMed  PubMed Central  Google Scholar 

  7. Shi C, Yan W, Wang G, Wang F, Li Q, Lin N (2015) Pharmacogenetics-based versus conventional dosing of warfarin: a meta-analysis of randomized controlled trials. PLoS One 10:e0144511

    Article  PubMed  PubMed Central  Google Scholar 

  8. Hamberg AK, Dahl ML, Barban M, Scordo MG, Wadelius M, Pengo V et al (2007) A PK-PD model for predicting the impact of age, CYP2C9, and VKORC1 genotype on individualization of warfarin therapy. Clin Pharmacol Ther 81:529–538

    Article  CAS  PubMed  Google Scholar 

  9. Chan E, McLachlan A, O'Reilly R, Rowland M (1994) Stereochemical aspects of warfarin drug interactions: use of a combined pharmacokinetic-pharmacodynamic model. Clin Pharmacol Ther 56:286–294

    Article  CAS  PubMed  Google Scholar 

  10. Breckenridge A, Orme M, Wesseling H, Lewis RJ, Gibbons R (1974) Pharmacokinetics and pharmacodynamics of the enantiomers of warfarin in man. Clin Pharmacol Ther 15:424–430

    Article  CAS  PubMed  Google Scholar 

  11. O'Reilly RA (1974) Studies on the optical enantiomorphs of warfarin in man. Clin Pharmacol Ther 16:348–354

    Article  PubMed  Google Scholar 

  12. Xue L, Holford N, Ding XL, Shen ZY, Huang CR, Zhang H et al (2016) Theory-based pharmacokinetics and pharmacodynamics of S- and R-warfarin and effects on international normalized ratio: influence of body size, composition and genotype in cardiac surgery patients. Br J Clin Pharmacol. doi:10.1111/bcp.13157 [Epub ahead of print]

    Google Scholar 

  13. Maddison J, Somogyi AA, Jensen BP, James HM, Gentgall M, Rolan PE (2013) The pharmacokinetics and pharmacodynamics of single dose (R)- and (S)-warfarin administered separately and together: relationship to VKORC1 genotype. Br J Clin Pharmacol 75:208–216

    Article  CAS  PubMed  Google Scholar 

  14. Limdi NA, Wadelius M, Cavallari L, Eriksson N, Crawford DC, Lee MT et al (2010) Warfarin pharmacogenetics: a single VKORC1 polymorphism is predictive of dose across 3 racial groups. Blood 115:3827–3834

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Scordo MG, Pengo V, Spina E, Dahl ML, Gusella M, Padrini R (2007) Influence of CYP2C9 and CYP2C19 genetic polymorphisms on warfarin maintenance dose and metabolic clearance. Clin Pharmacol Ther 72:702–710

    Article  Google Scholar 

  16. Hamberg AK, Wadelius M, Lindh JD, Dahl ML, Padrini R, Deloukas P et al (2010) A pharmacometric model describing the relationship between warfarin dose and INR response with respect to variations in CYP2C9, VKORC1, and age. Clin Pharmacol Ther 87:727–734

    Article  CAS  PubMed  Google Scholar 

  17. Hirsh J, Dalen JE, Deykin D, Poller L, Bussey H (1995) Oral anticoagulants. Mechanism of action, clinical effectiveness, and optimal therapeutic range. Chest 108(4 Suppl):231S–246S

    Article  CAS  PubMed  Google Scholar 

  18. Galvanin F, Ballan CC, Barolo M, Bezzo F (2013) A general model-based design of experimental approach to achieve practical identifiability of pharmacokinetic and pharmacodynamic models. J Pharmacokinet Pharmacodyn 40:451–467

    Article  CAS  PubMed  Google Scholar 

  19. Galvanin F, Barolo M, Macchietto S, Bezzo F (2011) Optimal design of clinical trials for the identification of physiological models of type 1 diabetes in the presence of model mismatch. Med Biol Eng Comput 49:263–277

    Article  PubMed  Google Scholar 

  20. Walter E, Pronzato L (1997) Identification of parametric models from experimental data. Springer Publishing, NY

    Google Scholar 

  21. Holford NH (1986) Clinical pharmacokinetics and pharmacodynamics of warfarin. Understanding the dose-effect relationship. Clin Pharmacokinet 11:483–504

    Article  CAS  PubMed  Google Scholar 

  22. Wadelius M, Chen LY, Downes K, Ghori J, Hunt S, Eriksson N et al (2007) Common VKORC1 and GGCX polymorphisms associated with warfarin dose. Hum Genet 121:23–34

    Article  CAS  PubMed  Google Scholar 

  23. Lane S, Al-Zubiedi S, Hatch E, Matthews I, Jorgensen AL, Deloukas P et al (2012) The population pharmacokinetics of R- and S-warfarin: effect of genetic and clinical factors. Br J Clin Pharmacol 73:66–76

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Pitsiu M, Parker EM, Aarons L, Rowland M (2003) A Bayesian method based on clotting factor activity for the prediction of maintenance warfarin dosage regimens. Ther Drug Monit 25:36–40

    Article  CAS  PubMed  Google Scholar 

  25. Bard Y (1974) Nonlinear parameter estimation. Academic Press, NY

    Google Scholar 

Download references

Acknowledgements

The authors would like to thank Marja-Liisa Dahl and Maria Gabriella Scordo (Department of Laboratory Medicine, Division of Clinical Pharmacology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden) for genotyping VKORC1 polymorphisms.

Author information

Authors and Affiliations

Authors

Contributions

Myriam Ferrari: analysis of data and manuscript writing; Vittorio Pengo: acquisition of clinical data; Massimiliano Barolo: conception of the study and manuscript writing; Fabrizio Bezzo: conception of the study and manuscript writing; Roberto Padrini: conception of the study, warfarin assay and manuscript writing.

Corresponding author

Correspondence to Roberto Padrini.

Ethics declarations

The study was approved by the Ethics Committee of the Azienda Ospedaliera di Padova, and all patients gave their written informed consent.

Funding

The study was funded by the University of Padova, Italy (Fondi “ex-60%”, 2016).

Conflict of interest

The authors declare that they have no conflict of interest.

Appendix A

Appendix A

The PK model is represented by the following equations:

$$ {C}_i=\frac{k_a{D}_i}{V_i\left({k}_a-{K}_{e, i}\right)}\left({e}^{-{K_{e, i}}^t}-{e}^{-{K_a}^t}\right)\kern0.5em i= S, R $$
(1)

where C i is the (S)- or (R)- W concentration [mg/L]; k a is the absorption constant, set at 2 h−1 [24]; D i is the S- or R-W dose [50% that of rac-W], assuming 100% oral bioavailability; k e is the elimination constant, defined as k e,i  = CL i / V i ; CL i is clearance; and V i is compartment volume.

Parameters CL i and V i were estimated for each subject by a least squares approach, since the maximum likelihood approach used to estimate PD parameters was considered unsuitable for small sample sizes [25].

The PD model, shown in Fig. 1, may be represented by the following differential and algebraic equations:

$$ \frac{dF_1}{dt}={k}_{\mathrm{tr}1} m\left({C}_s+{ k C}_R\right)-{K}_{\mathrm{tr}1}{F}_1 $$
(2)
$$ \begin{array}{ll}\frac{dF_j}{dt}={k}_{\mathrm{tr}\mathrm{l}}{F}_{J-1}-{k}_{\mathrm{tr}1}{F}_J\hfill & J=2,3,4\hfill \end{array} $$
(3)
$$ \frac{dS_1}{dt}={k}_{\mathrm{tr}2} m\left({C}_s+{ k C}_R\right)-{K}_{\mathrm{tr}2}{S}_1 $$
(4)
$$ \begin{array}{ll}\frac{dS_j}{dt}={k}_{\mathrm{tr}2}{S}_{J-1}-{k}_{\mathrm{tr}2}{S}_J\hfill & J=2,3,4\hfill \end{array} $$
(5)
$$ \mathrm{INR}={\mathrm{INR}}_{\mathrm{base}}+\left({F}_4+{S}_4\right) $$
(6)

where F j and S j represent the response to W concentration in each compartment j of the fast and slow chains, respectively; k tr1 and k tr2 are rate constants [h−1] governing INR response delay [note that MTT1 = (k tr1)−1 and MTT2 = (k tr2)−1]; m is the slope of the linear correlation between W concentration and INR response; k is the correction coefficient to account for the different potency of the W R-form with respect to S-one; and INRbase is the basal INR value.

The model identification procedure was carried out iteratively according to a “two-step” maximum likelihood estimate (MLE) method [19, 25].

The procedure was as follows:

  1. Step 1.

    MTT1 and MTT2 were fixed at reasonable values (4.5 and 27.5 h, respectively); m and k were then estimated for each genotype group, with both single-dose and steady-state clinical data. The K values of the three genotype groups were averaged, weighting the estimates by the number of patients in each group.

  2. Step 2.

    K and m were fixed at the values estimated above, and MTT1 was estimated with single-dose data only, since steady-state data were minimally affected by MTT1.

The procedure was repeated starting from the estimated value of MTT1 (keeping MTT2 fixed at 27.5 h). The parameter estimates stabilized after two iterations. Parameter estimation and model simulations were carried out with the general purpose modelling tools gPROMS® v. 4.1.0 by Process Systems Enterprise (London, UK).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ferrari, M., Pengo, V., Barolo, M. et al. Assessing the relative potency of (S)- and (R)-warfarin with a new PK-PD model, in relation to VKORC1 genotypes. Eur J Clin Pharmacol 73, 699–707 (2017). https://doi.org/10.1007/s00228-017-2248-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00228-017-2248-9

Keywords

Navigation