Data Analysis Protocol for the Development and Evaluation of Population Pharmacokinetic Models for Incorporation Into the Web-Accessible Population Pharmacokinetic Service - Hemophilia (WAPPS-Hemo)

Alanna McEneny-King, Gary Foster, Alfonso Iorio, Andrea N Edginton, Alanna McEneny-King, Gary Foster, Alfonso Iorio, Andrea N Edginton

Abstract

Background: Hemophilia is an inherited bleeding disorder caused by a deficiency in a specific clotting factor. This results in spontaneous bleeding episodes and eventual arthropathy. The mainstay of hemophilia treatment is prophylactic replacement of the missing factor, but an optimal regimen remains to be determined. Rather, individualized prophylaxis has been suggested to improve both patient safety and resource utilization. However, uptake of this approach has been hampered by the demanding sampling schedules and complex calculations required to obtain individual estimates of pharmacokinetic (PK) parameters. The use of population pharmacokinetics (PopPK) can alleviate this burden by reducing the number of plasma samples required for accurate estimation, but few tools incorporating this approach are readily available to clinicians.

Objective: The Web-accessible Population Pharmacokinetic Service - Hemophilia (WAPPS-Hemo) project aims to bridge this gap by providing a Web-accessible service for the reliable estimation of individual PK parameters from only a few patient samples. This service is predicated on the development of validated brand-specific PopPK models.

Methods: We describe the data analysis plan for the development and evaluation of each PopPK model to be incorporated into the WAPPS-Hemo platform. The data sources and structure of the dataset are discussed first, followed by the procedures for handling both data below limit of quantification (BLQ) and absence of such BLQ data. Next, we outline the strategies for building the appropriate structural and covariate models, including the possible need for a process algorithm when PK behavior varies between subjects or significant covariates are not provided. Prior to use in a prospective manner, the models will undergo extensive evaluation using a variety of techniques such as diagnostic plots, bootstrap analysis and cross-validation. Finally, we describe the incorporation of a validated PopPK model into the Bayesian post hoc model to produce individualized estimates of PK parameters.

Results: Dense PK data has been collected for more than 20 brands of factor concentrate from both industry-sponsored and investigator-driven studies. The model development process is underway for the majority of molecules, with refinement and validation to be completed in 2017. Further, the WAPPS-Hemo co-investigator network has contributed more than 300 PK assessments for use in model development and evaluation. This constitutes the largest repository of this type of PK data globally.

Conclusions: The WAPPS-Hemo service aims to eliminate barriers to the uptake of individualized PK-tailored hemophilia treatment. By incorporating this tool into routine practice, clinicians can implement a personalized dosing strategy without performing rigorous sampling or complex calculations. This service is centred on validated models developed according to the robust approach to PopPK modeling described herein.

Clinicaltrial: ClinicalTrials.gov NCT02061072; https://ichgcp.net/clinical-trials-registry/NCT02061072 (Archived by WebCite at http://www.webcitation.org/6mRIXJh55).

Keywords: factor IX; factor VIII; hemophilia; population pharmacokinetics; tailored prophylaxis.

Conflict of interest statement

A Iorio is the principal investigator of the WAPPS project and has received research and consultancy funds from Bayer, Baxalta, Biogen, NovoNordisk, and Pfizer. All funds were granted to McMaster University and no funds were received for this manuscript. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

©Alanna McEneny-King, Gary Foster, Alfonso Iorio, Andrea N Edginton. Originally published in JMIR Research Protocols (http://www.researchprotocols.org), 07.12.2016.

Figures

Figure 1
Figure 1
The Web-Accessible Population Pharmacokinetic Service - Hemophilia (WAPPS-Hemo) platform uses brand-specific population pharmacokinetic models and submitted patient data to generate reports of individual pharmacokinetic profiles and estimates.
Figure 2
Figure 2
Illustration of the various components of the base model for a one-compartment model with exponential between subject variability and proportional residual unexplained variability.
Figure 3
Figure 3
Equations for defining different aspects of the population pharmacokinetic models including residual unexplained error (1-4), between subject variability (5-6), and covariates (7-10).
Figure 4
Figure 4
Factor concentration as a function of time (symbols: patient data, black line: predicted individual pharmacokinetic profile) where time to the lower 95% credibility interval bound for each of the 0.05 (green line), 0.02 (blue line) or 0.01 (red line) IU/mL thresholds is reported to the clinician. Time 0 represents time of dose initiation.

References

    1. Davie EW. Biochemical and molecular aspects of the coagulation cascade. Thromb Haemost. 1995 Jul;74(1):1–6.
    1. Mannucci PM, Tuddenham EG. The hemophilias--from royal genes to gene therapy. N Engl J Med. 2001 Jun 7;344(23):1773–9. doi: 10.1056/NEJM200106073442307.
    1. Castro HE, Briceño MF, Casas CP, Rueda JD. The history and evolution of the clinical effectiveness of haemophilia type a treatment: a systematic review. Indian J Hematol Blood Transfus. 2014 Mar;30(1):1–11. doi: 10.1007/s12288-012-0209-0.
    1. Lusher JM, Arkin S, Abildgaard CF, Schwartz RS. Recombinant factor VIII for the treatment of previously untreated patients with hemophilia A. Safety, efficacy, and development of inhibitors. Kogenate Previously Untreated Patient Study Group. N Engl J Med. 1993 Feb 18;328(7):453–9. doi: 10.1056/NEJM199302183280701.
    1. Schwartz RS, Abildgaard CF, Aledort LM, Arkin S, Bloom AL, Brackmann HH, Brettler DB, Fukui H, Hilgartner MW, Inwood MJ. Human recombinant DNA-derived antihemophilic factor (factor VIII) in the treatment of hemophilia A. recombinant Factor VIII Study Group. N Engl J Med. 1990 Dec 27;323(26):1800–5. doi: 10.1056/NEJM199012273232604.
    1. Nilsson IM, Hedner U, Ahlberg A. Haemophilia prophylaxis in Sweden. Acta Paediatr Scand. 1976 Mar;65(2):129–35.
    1. Nilsson IM, Blombäck M, Ahlberg A. Our experience in Sweden with prophylaxis on haemophilia. Bibl Haematol. 1970;34:111–24.
    1. Den Uijl IE, Mauser Bunschoten EP, Roosendaal G, Schutgens RE, Biesma DH, Grobbee DE, Fischer K. Clinical severity of haemophilia A: does the classification of the 1950s still stand? Haemophilia. 2011 Nov;17(6):849–53. doi: 10.1111/j.1365-2516.2011.02539.x.
    1. Oldenburg J. Prophylaxis in bleeding disorders. Thromb Res. 2011 Jan;127 Suppl 1:S14–7. doi: 10.1016/j.thromres.2010.10.005.
    1. Berntorp E, Spotts G, Patrone L, Ewenstein BM. Advancing personalized care in hemophilia A: ten years' experience with an advanced category antihemophilic factor prepared using a plasma/albumin-free method. Biologics. 2014;8:115–27. doi: 10.2147/BTT.S53456.
    1. Fischer K, Collins PW, Ozelo MC, Srivastava A, Young G, Blanchette VS. When and how to start prophylaxis in boys with severe hemophilia without inhibitors: communication from the SSC of the ISTH. J Thromb Haemost. 2016 May;14(5):1105–9. doi: 10.1111/jth.13298.
    1. Petrini P. Identifying and overcoming barriers to prophylaxis in the management of haemophilia. Haemophilia. 2007 Sep;13 Suppl 2:16–22. doi: 10.1111/j.1365-2516.2007.01501.x.
    1. Carcao M, Srivastava A. Factor VIII/factor IX prophylaxis for severe hemophilia. Semin Hematol. 2016 Jan;53(1):3–9. doi: 10.1053/j.seminhematol.2015.10.006.
    1. Björkman S, Berntorp E. Pharmacokinetics of coagulation factors: clinical relevance for patients with haemophilia. Clin Pharmacokinet. 2001;40(11):815–32. doi: 10.2165/00003088-200140110-00003.
    1. Björkman S, Folkesson A, Jönsson S. Pharmacokinetics and dose requirements of factor VIII over the age range 3-74 years: a population analysis based on 50 patients with long-term prophylactic treatment for haemophilia A. Eur J Clin Pharmacol. 2009 Oct;65(10):989–98. doi: 10.1007/s00228-009-0676-x.
    1. Collins PW, Björkman S, Fischer K, Blanchette V, Oh M, Schroth P, Fritsch S, Casey K, Spotts G, Ewenstein BM. Factor VIII requirement to maintain a target plasma level in the prophylactic treatment of severe hemophilia A: influences of variance in pharmacokinetics and treatment regimens. J Thromb Haemost. 2010 Feb;8(2):269–75. doi: 10.1111/j.1538-7836.2009.03703.x.
    1. Morfini M. Comparative pharmacokinetic studies in haemophilia. Haemophilia. 2002 Mar;8 Suppl 2:30–3.
    1. Wakefield J. The Bayesian analysis of population pharmacokinetic models. JASA. 1996 Mar;91(433):62. doi: 10.2307/2291383.
    1. Samara E, Granneman R. Role of population pharmacokinetics in drug development. A pharmaceutical industry perspective. Clin Pharmacokinet. 1997 Apr;32(4):294–312. doi: 10.2165/00003088-199732040-00003.
    1. Ette EI, Williams PJ. Population pharmacokinetics I: background, concepts, and models. Ann Pharmacother. 2004 Oct;38(10):1702–6. doi: 10.1345/aph.1D374.
    1. Holford NH, Buclin T. Safe and effective variability-a criterion for dose individualization. Ther Drug Monit. 2012 Oct;34(5):565–8. doi: 10.1097/FTD.0b013e31826aabc3.
    1. Sheiner LB, Beal S, Rosenberg B, Marathe VV. Forecasting individual pharmacokinetics. Clin Pharmacol Ther. 1979 Sep;26(3):294–305.
    1. Tod MM, Padoin C, Petitjean O. Individualising aminoglycoside dosage regimens after therapeutic drug monitoring: simple or complex pharmacokinetic methods? Clin Pharmacokinet. 2001;40(11):803–14. doi: 10.2165/00003088-200140110-00002.
    1. Ting LS, Villeneuve E, Ensom MH. Beyond cyclosporine: a systematic review of limited sampling strategies for other immunosuppressants. Ther Drug Monit. 2006 Jun;28(3):419–30. doi: 10.1097/01.ftd.0000211810.19935.44.
    1. Björkman S. Limited blood sampling for pharmacokinetic dose tailoring of FVIII in the prophylactic treatment of haemophilia A. Haemophilia. 2010 Jul 1;16(4):597–605. doi: 10.1111/j.1365-2516.2009.02191.x.
    1. Iorio A, Keepanasseril A, Foster G, Navarro-Ruan T, McEneny-King A, Edginton AN, Thabane L. Development of a Web Accessible Population Pharmacokinetic Service – Hemophilia (WAPPS-Hemo): Study Protocol (forthcoming) JMIR Res Protoc. 2016;5(4) doi: 10.2196/resprot.6558.
    1. Diao L, Li S, Ludden T, Gobburu J, Nestorov I, Jiang H. Population pharmacokinetic modelling of recombinant factor IX Fc fusion protein (rFIXFc) in patients with haemophilia B. Clin Pharmacokinet. 2014 May;53(5):467–77. doi: 10.1007/s40262-013-0129-7.
    1. Shapiro AD. Long-lasting recombinant factor VIII proteins for hemophilia A. Hematology Am Soc Hematol Educ Program. 2013;2013:37–43. doi: 10.1182/asheducation-2013.1.37.
    1. Barrowcliffe TW. Monitoring haemophilia severity and treatment: new or old laboratory tests? Haemophilia. 2004 Oct;10 Suppl 4:109–14. doi: 10.1111/j.1365-2516.2004.00985.x.
    1. Beal SL. Ways to fit a PK model with some data below the quantification limit. J Pharmacokinet Pharmacodyn. 2001 Oct;28(5):481–504.
    1. Ariano RE, Duke PC, Sitar DS. The influence of sparse data sampling on population pharmacokinetics: a post hoc analysis of a pharmacokinetic study of morphine in healthy volunteers. Clin Ther. 2012 Mar;34(3):668–76. doi: 10.1016/j.clinthera.2012.01.023.
    1. Meibohm B, Läer S, Panetta JC, Barrett JS. Population pharmacokinetic studies in pediatrics: issues in design and analysis. AAPS J. 2005 Oct 05;7(2):E475–87. doi: 10.1208/aapsj070248.
    1. Mesnil F, Mentré F, Dubruc C, Thénot JP, Mallet A. Population pharmacokinetic analysis of mizolastine and validation from sparse data on patients using the nonparametric maximum likelihood method. J Pharmacokinet Biopharm. 1998 Apr;26(2):133–61.
    1. US Department of Health and Human Services. Food and Drug Administration. Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research (CBER) Guidance for Industry: Population Pharmacokinetics. 1999. [2016-11-08]. .
    1. Rajagopalan P, Gastonguay MR. Population pharmacokinetics of ciprofloxacin in pediatric patients. J Clin Pharmacol. 2003 Jul;43(7):698–710.

Source: PubMed

3
Abonnieren