Integration of modeling and simulation into hospital-based decision support systems guiding pediatric pharmacotherapy

Jeffrey S Barrett, John T Mondick, Mahesh Narayan, Kalpana Vijayakumar, Sundararajan Vijayakumar, Jeffrey S Barrett, John T Mondick, Mahesh Narayan, Kalpana Vijayakumar, Sundararajan Vijayakumar

Abstract

Background: Decision analysis in hospital-based settings is becoming more common place. The application of modeling and simulation approaches has likewise become more prevalent in order to support decision analytics. With respect to clinical decision making at the level of the patient, modeling and simulation approaches have been used to study and forecast treatment options, examine and rate caregiver performance and assign resources (staffing, beds, patient throughput). There us a great need to facilitate pharmacotherapeutic decision making in pediatrics given the often limited data available to guide dosing and manage patient response. We have employed nonlinear mixed effect models and Bayesian forecasting algorithms coupled with data summary and visualization tools to create drug-specific decision support systems that utilize individualized patient data from our electronic medical records systems.

Methods: Pharmacokinetic and pharmacodynamic nonlinear mixed-effect models of specific drugs are generated based on historical data in relevant pediatric populations or from adults when no pediatric data is available. These models are re-executed with individual patient data allowing for patient-specific guidance via a Bayesian forecasting approach. The models are called and executed in an interactive manner through our web-based dashboard environment which interfaces to the hospital's electronic medical records system.

Results: The methotrexate dashboard utilizes a two-compartment, population-based, PK mixed-effect model to project patient response to specific dosing events. Projected plasma concentrations are viewable against protocol-specific nomograms to provide dosing guidance for potential rescue therapy with leucovorin. These data are also viewable against common biomarkers used to assess patient safety (e.g., vital signs and plasma creatinine levels). As additional data become available via therapeutic drug monitoring, the model is re-executed and projections are revised.

Conclusion: The management of pediatric pharmacotherapy can be greatly enhanced via the immediate feedback provided by decision analytics which incorporate the current, best-available knowledge pertaining to dose-exposure and exposure-response relationships, especially for narrow therapeutic agents that are difficult to manage.

Figures

Figure 1
Figure 1
Typical progression of pharmacometric model development commonly used to support pharmacotherapeutic decision support systems.
Figure 2
Figure 2
Schematic of three-tier system architecture of hospital pharmacotherapy decision support system comprising a back end database tier, a business logic middle tier and data presentation/user interface front-end tier.
Figure 3
Figure 3
Diagnostic plots from preliminary methotrexate population pharmacokinetic model. (A) Observed versus population predicted concentrations. (B) Observed versus individual predicted concentrations. Open circles represent MTX plasma concentrations from patients predicted to have normal renal function. Open triangles represent patients predicted to have reduced MTX clearance.
Figure 4
Figure 4
Screen captures from the current MTX dashboard design showing (A) the most recent MTX dose event with the complementary monitored MTX plasma concentrations and safety markers, (B) the MTX exposure projected after the dosing guidance menu button is selected, (C) the view from Figure 4B overlaid against a nomogram used to assess the potential for MTX toxicity with consideration for drug rescue with leucovorin and (D) the update of the model fit when the additional blood collection time points were added to the patient data set.
Figure 5
Figure 5
Workflow of MTX dashboard operation.

References

    1. IOM . Building a Better Delivery system: A New Engineering/Health Care Partnership. National Academy of Engineering and Institute of Medicine; 2005.
    1. IOM Report: Patient safety – achieving a new standard for care. Acad Emerg Med. 2005;12:1011–2.
    1. Ferri F. User modeling techniques as support in the clinical decision-making process. Medinfo. 1995;8:926–30.
    1. Gardner SN. Modeling multi-drug chemotherapy: tailoring treatment to individuals. J Theor Biol. 2002;214:181–207. doi: 10.1006/jtbi.2001.2459.
    1. Starr JM, Campbell A. Mathematical modeling of Clostridium difficile infection. Clin Microbiol Infect. 2001;7:432–7. doi: 10.1046/j.1198-743x.2001.00291.x.
    1. Broderick A, et al. Nosocomial infections: validation of surveillance and computer modeling to identify patients at risk. Am J Epidemiol. 1990;131:734–42.
    1. Raboud J, et al. Modeling transmission of methicillin-resistant Staphylococcus aureus among patients admitted to a hospital. Infect Control Hosp Epidemiol. 2005;26:607–15. doi: 10.1086/502589.
    1. Shaw B, Marshall AH. Modeling the health care costs of geriatric inpatients. IEEE Trans Inf Technol Biomed. 2006;10:526–32. doi: 10.1109/TITB.2005.863821.
    1. Graves N, Nicholls TM, Morris AJ. Modeling the costs of hospital-acquired infections in New Zealand. Infect Control Hosp Epidemiol. 2003;24:214–23. doi: 10.1086/502192.
    1. Anderson JG, et al. Modeling the costs and outcomes of cardiovascular surgery. Health Care Manag Sci. 2002;5:103–11. doi: 10.1023/A:1014472731382.
    1. Hung GR, et al. Computer modeling of patient flow in a pediatric emergency department using discrete event simulation. Pediatr Emerg Care. 2007;23:5–10. doi: 10.1097/PEC.0b013e31802c611e.
    1. Costa AX, et al. Mathematical modelling and simulation for planning critical care capacity. Anaesthesia. 2003;58:320–7. doi: 10.1046/j.1365-2044.2003.03042.x.
    1. Koizumi N, Kuno E, Smith TE. Modeling patient flows using a queuing network with blocking. Health Care Manag Sci. 2005;8:49–60. doi: 10.1007/s10729-005-5216-3.
    1. Isken MW, Rajagopalan B. Data mining to support simulation modeling of patient flow in hospitals. J Med Syst. 2002;26:179–97. doi: 10.1023/A:1014814111524.
    1. Paul JA, et al. Transient modeling in simulation of hospital operations for emergency response. Prehospital Disaster Med. 2006;21:223–36.
    1. Van Houdenhoven M, et al. Optimizing intensive care capacity using individual length-of-stay prediction models. Crit Care. 2007;11:R42. doi: 10.1186/cc5730.
    1. Braithwaite RS, Roberts MS, Justice AC. Incorporating quality of evidence into decision analytic modeling. Ann Intern Med. 2007;146:133–41.
    1. Connelly LG, Bair AE. Discrete event simulation of emergency department activity: a platform for system-level operations research. Acad Emerg Med. 2004;11:1177–85.
    1. Slovensky DJ, Morin B. Learning through simulation: the next dimension in quality improvement. Qual Manag Health Care. 1997;5:72–9.
    1. Lucas CE, et al. Mathematical modeling to define optimum operating room staffing needs for trauma centers. J Am Coll Surg. 2001;192:559–65. doi: 10.1016/S1072-7515(01)00829-8.
    1. Verduijn M, et al. Modeling length of stay as an optimized two-class prediction problem. Methods Inf Med. 2007;46:352–9.
    1. Chien JY, et al. Pharmacokinetics/Pharmacodynamics and the stages of drug development: role of modeling and simulation. Aaps J. 2005;7:E544–59. doi: 10.1208/aapsj070355.
    1. Gobburu JV, Sekar VJ. Application of modeling and simulation to integrate clinical pharmacology knowledge across a new drug application. Int J Clin Pharmacol Ther. 2002;40:281–8.
    1. Meibohm B, et al. Population pharmacokinetic studies in pediatrics: issues in design and analysis. Aaps J. 2005;7:E475–87. doi: 10.1208/aapsj070248.
    1. Bondareva IB, et al. Nonparametric population modeling of valproate pharmacokinetics in epileptic patients using routine serum monitoring data: implications for dosage. J Clin Pharm Ther. 2004;29:105–20. doi: 10.1111/j.1365-2710.2003.00538.x.
    1. Jelliffe R. Goal-oriented, model-based drug regimens: setting individualized goals for each patient. Ther Drug Monit. 2000;22:325–9. doi: 10.1097/00007691-200006000-00016.
    1. Agresti A. A survey of models for repeated ordered categorical response data. Stat Med. 1989;8:1209–24. doi: 10.1002/sim.4780081005.
    1. Yano I, Beal SL, Sheiner LB. The need for mixed-effects modeling with population dichotomous data. J Pharmacokinet Pharmacodyn. 2001;28:389–412. doi: 10.1023/A:1011586814601.
    1. Borsi J, Revesz T, Schuler D. [Prognostic significance of systemic clearance of methotrexate in acute lymphoid leukemia in childhood] Orv Hetil. 1986;127:439–42.
    1. Evans WE, et al. Methotrexate systemic clearance influences probability of relapse in children with standard-risk acute lymphocytic leukaemia. Lancet. 1984;1:359–62. doi: 10.1016/S0140-6736(84)90411-2.
    1. Evans WE, et al. Clinical pharmacodynamics of high-dose methotrexate in acute lymphocytic leukemia. Identification of a relation between concentration and effect. N Engl J Med. 1986;314:471–7.
    1. Goldie JH, Price LA, Harrap KR. Methotrexate toxicity: correlation with duration of administration, plasma levels, dose and excretion pattern. Eur J Cancer. 1972;8:409–14.
    1. Tattersall MH, Brown B, Frei E., 3rd The reversal of methotrexate toxicity by thymidine with maintenance of antitumour effects. Nature. 1975;253:198–200. doi: 10.1038/253198a0.
    1. Isacoff WH, et al. Pharmacokinetics of high-dose methotrexate with citrovorum factor rescue. Cancer Treat Rep. 1977;61:1665–74.
    1. Stoller RG, et al. Use of plasma pharmacokinetics to predict and prevent methotrexate toxicity. N Engl J Med. 1977;297:630–4.
    1. Nirenberg A, et al. High-dose methotrexate with citrovorum factor rescue: predictive value of serum methotrexate concentrations and corrective measures to avert toxicity. Cancer Treat Rep. 1977;61:779–83.
    1. Perez C, et al. Significance of the 48-hour plasma level in high-dose methotrexate regimens. Cancer Clin Trials. 1978;1:107–11.
    1. Widemann BC, et al. High-dose methotrexate-induced nephrotoxicity in patients with osteosarcoma. Cancer. 2004;100:2222–32. doi: 10.1002/cncr.20255.
    1. Piard C, et al. A limited sampling strategy to estimate individual pharmacokinetic parameters of methotrexate in children with acute lymphoblastic leukemia. Cancer Chemother Pharmacol. 2007;60:609–20. doi: 10.1007/s00280-006-0394-3.
    1. Rousseau A, et al. Bayesian estimation of methotrexate pharmacokinetic parameters and area under the curve in children and young adults with localised osteosarcoma. Clin Pharmacokinet. 2002;41:1095–104. doi: 10.2165/00003088-200241130-00006.
    1. Odoul F, et al. Prediction of methotrexate elimination after high dose infusion in children with acute lymphoblastic leukaemia using a population pharmacokinetic approach. Fundam Clin Pharmacol. 1999;13:595–604.
    1. Pignon T, et al. Pharmacokinetics of high-dose methotrexate in adult osteogenic sarcoma. Cancer Chemother Pharmacol. 1994;33:420–4. doi: 10.1007/BF00686272.
    1. Bruno R, et al. Dosage predictions in high-dose methotrexate infusions. Part 2: Bayesian estimation of methotrexate clearance. Cancer Drug Deliv. 1985;2:277–83.
    1. Monjanel-Mouterde S, et al. Bayesian population model of methotrexate to guide dosage adjustments for folate rescue in patients with breast cancer. J Clin Pharm Ther. 2002;27:189–95. doi: 10.1046/j.1365-2710.2002.00402.x.
    1. Bauer RJ, Guzy S, Ng C. A survey of population analysis methods and software for complex pharmacokinetic and pharmacodynamic models with examples. Aaps J. 2007;9:E60–83. doi: 10.1208/aapsj0901007.
    1. Jelliffe RW, et al. Adaptive control of drug dosage regimens: basic foundations, relevant issues, and clinical examples. Int J Biomed Comput. 1994;36:1–23. doi: 10.1016/0020-7101(94)90091-4.
    1. Pestotnik SL. Expert clinical decision support systems to enhance antimicrobial stewardship programs: insights from the society of infectious diseases pharmacists. Pharmacotherapy. 2005;25:1116–25. doi: 10.1592/phco.2005.25.8.1116.
    1. Staes CJ, et al. A case for manual entry of structured, coded laboratory data from multiple sources into an ambulatory electronic health record. J Am Med Inform Assoc. 2006;13:12–5. doi: 10.1197/jamia.M1813.
    1. TheraDoc
    1. Cereplex
    1. Brossette SE, et al. A laboratory-based, hospital-wide, electronic marker for nosocomial infection: the future of infection control surveillance? Am J Clin Pathol. 2006;125:34–9. doi: 10.1309/502A-UPR8-VE67-MBDE.
    1. MedMined
    1. Evans RS, Pestotnik SL. Applications of medical informatics in antibiotic therapy. Adv Exp Med Biol. 1994;349:87–96.
    1. Evans RS, et al. Improving empiric antibiotic selection using computer decision support. Arch Intern Med. 1994;154:878–84. doi: 10.1001/archinte.154.8.878.
    1. Pestotnik SL, et al. Implementing antibiotic practice guidelines through computer-assisted decision support: clinical and financial outcomes. Ann Intern Med. 1996;124:884–90.

Source: PubMed

3
Abonner