Skip to main content

Advertisement

Log in

Optimal cholesterol treatment plans and genetic testing strategies for cardiovascular diseases

  • Published:
Health Care Management Science Aims and scope Submit manuscript

Abstract

Atherosclerotic cardiovascular disease (ASCVD) is among the leading causes of death in the US. Although research has shown that ASCVD has genetic elements, the understanding of how genetic testing influences its prevention and treatment has been limited. To this end, we model the health trajectory of patients stochastically and determine treatment and testing decisions simultaneously. Since the cholesterol level of patients is one controllable risk factor for ASCVD events, we model cholesterol treatment plans as Markov decision processes. We determine whether and when patients should receive a genetic test using value of information analysis. By simulating the health trajectory of over 64 million adult patients, we find that 6.73 million patients undergo genetic testing. The optimal treatment plans informed with clinical and genetic information save 5,487 more quality-adjusted life-years while costing $1.18 billion less than the optimal treatment plans informed with clinical information only. As precision medicine becomes increasingly important, understanding the impact of genetic information becomes essential.

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
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Kathiresan S, Srivastava D (2012) Genetics of human cardiovascular disease. Cell 148 (6):1242–1257. https://doi.org/10.1016/j.cell.2012.03.001

    Article  Google Scholar 

  2. The CARDIoGRAMplusC4D Consortium (2013) Large-scale association analysis identifies new risk loci for coronary artery disease. Nat Gen 45(1):25–33. https://doi.org/10.1038/ng.2480

    Article  Google Scholar 

  3. Abraham G, Kowalczyk A, Zobel J, Inouye M (2013) Performance and Robustness of Penalized and Unpenalized Methods for Genetic Prediction of Complex Human Disease. Genet Epidemiol 37 (2):184–195. https://doi.org/10.1002/gepi.21698

    Article  Google Scholar 

  4. Abraham G, Tye-Din J A, Bhalala O G, Kowalczyk A, Zobel J, Inouye M (2014) Accurate and Robust Genomic Prediction of Celiac Disease Using Statistical Learning. PLoS Genet 10(2):e1004137. https://doi.org/10.1371/journal.pgen.1004137, 1301.5948

    Article  Google Scholar 

  5. Sun Y, Goodison S, Li J, Liu L, Farmerie W (2007) Improved breast cancer prognosis through the combination of clinical and genetic markers. Bioinformatics 23(1):30–37. https://doi.org/10.1093/bioinformatics/btl543

    Article  Google Scholar 

  6. Vasan R S (2006) Biomarkers of cardiovascular disease: molecular basis and practical considerations. Circulation 113(19):2335–2362. https://doi.org/10.1161/CIRCULATIONAHA.104.482570

    Article  Google Scholar 

  7. Mega J L, Stitziel N O, Smith J G, Chasman D I, Caulfield M J, Devlin J J, Nordio F, Hyde C L, Cannon C P, Sacks F M, Poulter N R, Sever P S, Ridker P M, Braunwald E, Melander O, Kathiresan S, Sabatine M S (2015) Genetic risk, coronary heart disease events, and the clinical benefit of statin therapy: an analysis of primary and secondary prevention trials. Lancet 385(9984):2264–2271. https://doi.org/10.1016/S0140-6736(14)61730-X

    Article  Google Scholar 

  8. Bibbins-Domingo K, Grossman D C, Curry S J, Davidson K W, Epling J W, García F A R, Gillman M W, Kemper A R, Krist A H, Kurth A E, Landefeld C S, LeFevre M L, Mangione C M, Phillips W R, Owens D K, Phipps M G, Pignone M P (2016) Statin use for the primary prevention of cardiovascular disease in adults. J Am Med Assoc 316(19):1997–2007. https://doi.org/10.1001/jama.2016.15450

    Article  Google Scholar 

  9. Goff D C, Lloyd-Jones D M, Bennett G, Coady S, D’Agostino R B, Gibbons R, Greenland P, Lackland D T, Levy D, O’Donnell C J, Robinson J G, Schwartz J S, Shero S T, Smith S C, Sorlie P, Stone N J, Wilson P WF (2014) 2013 ACC/AHA guideline on the assessment of cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines. Circulation 129(25 SUPPL. 1). https://doi.org/10.1161/01.cir.0000437741.48606.98

  10. Heron M (2018) Deaths: leading causes for 2016. Natl Vital Stat Rep 67 (6):1–77. http://www.ncbi.nlm.nih.gov/pubmed/30248017

    Google Scholar 

  11. Benjamin E J, Virani S S, Callaway C W, Chamberlain A M, Chang A R, Cheng S, Chiuve S E, Cushman M, Delling F N, Deo R, de Ferranti S D, Ferguson J F, Fornage M, Gillespie C, Isasi C R, Jiménez M C, Jordan L C, Judd S E, Lackland D, Lichtman J H, Lisabeth L, Liu S, Longenecker C T, Lutsey P L, Mackey J S, Matchar D B, Matsushita K, Mussolino M E, Nasir K, O’Flaherty M, Palaniappan L P, Pandey A, Pandey D K, Reeves M J, Ritchey M D, Rodriguez C J, Roth G A, Rosamond W D, Sampson U KA, Satou G M, Shah S H, Spartano N L, Tirschwell D L, Tsao C W, Voeks J H, Willey J Z, Wilkins J T, Wu Jason HY, Alger H M, Wong S S, Muntner P (2018) Heart disease and stroke statistics - 2018 update: a report from the American Heart Association. Circulation 137(12):E67–E492. https://doi.org/10.1161/CIR.0000000000000558. NIHMS150003

    Article  Google Scholar 

  12. MacRae C A, Vasan R S (2016) The future of genetics and genomics. Circulation 133 (25):2634–2639. https://doi.org/10.1161/CIRCULATIONAHA.116.022547

    Article  Google Scholar 

  13. Jarmul J, Pletcher M J, Hassmiller Lich K, Wheeler S B, Weinberger M, Avery C L, Jonas D E, Earnshaw S, Pignone M (2018) Cardiovascular genetic risk testing for targeting statin therapy in the primary prevention of atherosclerotic cardiovascular disease. Circ Cardiovascul Qual Outcome 11(4):e004171. https://doi.org/10.1161/CIRCOUTCOMES.117.004171

    Article  Google Scholar 

  14. Khera A V, Emdin C A, Drake I, Natarajan P, Bick A G, Cook N R, Chasman D I, Baber U, Mehran R, Rader D J, Fuster V, Boerwinkle E, Melander O, Orho-Melander M, Ridker P M, Kathiresan S (2016) Genetic risk, adherence to a healthy lifestyle, and coronary disease. N Engl J Med 375(24):2349–2358. https://doi.org/10.1056/NEJMoa1605086

    Article  Google Scholar 

  15. Natarajan P, Young R, Stitziel N O, Padmanabhan S, Baber U, Mehran R, Sartori S, Fuster V, Reilly D F, Butterworth A, Rader D J, Ford I, Sattar N, Kathiresan S (2017) Polygenic risk score identifies subgroup with higher burden of atherosclerosis and greater relative benefit from statin therapy in the primary prevention setting. Circulation 135(22):2091–2101. https://doi.org/10.1161/CIRCULATIONAHA.116.024436

    Article  Google Scholar 

  16. Lewis C M, Vassos E (2020) Polygenic risk scores: From research tools to clinical instruments. Genome Med 12(1):1–11. https://doi.org/10.1186/s13073-020-00742-5

    Article  Google Scholar 

  17. Knowles J W, Ashley E A (2018) Cardiovascular disease: The rise of the genetic risk score. PLoS Med 15(3):1–7. https://doi.org/10.1371/journal.pmed.1002546

    Article  Google Scholar 

  18. Connor M J-, Bch M B, Natarajan P (2020) Current Clinical Implications of Coronary Artery Disease Polygenic Risk Scoring, pp 4–11

  19. Collins R, Reith C, Emberson J, Armitage J, Baigent C, Blackwell L, Blumenthal R, Danesh J, Smith G D, DeMets D, Evans S, Law M, MacMahon S, Martin S, Neal B, Poulter N, Preiss D, Ridker P, Roberts I, Rodgers A, Sandercock P, Schulz K, Sever P, Simes J, Smeeth L, Wald N, Yusuf S, Peto R (2016) Interpretation of the evidence for the efficacy and safety of statin therapy. Lancet 388(10059):2532–2561. https://doi.org/10.1016/S0140-6736(16)31357-5

    Article  Google Scholar 

  20. Stone N J, Robinson J G, Lichtenstein A H, Bairey Merz C N, Blum C B, Eckel R H, Goldberg A C, Gordon D, Levy D, Lloyd-Jones D M, McBride P, Schwartz J S, Shero S T, Smith S C, Watson K, Wilson P W F (2014) 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults. Circulation 129(25 suppl 2):S1–S45. https://doi.org/10.1161/01.cir.0000437738.63853.7a

    Google Scholar 

  21. Sussman J B, Wiitala W L, Zawistowski M, Hofer T P, Bentley D, Hayward R A (2017) The Veterans Affairs cardiac risk score: recalibrating the atherosclerotic cardiovascular disease score for applied use. Med Care 55(9):864–870. https://doi.org/10.1097/MLR.0000000000000781

    Article  Google Scholar 

  22. Puterman M L (2014) Markov decision processes: discrete stochastic dynamic programming. Wiley

  23. Raiffa H, Schlaifer R (1961) Applied statistical decision theory. Harvard University and MIT Press

  24. Glover MJ, Jones E, Masconi KL, Sweeting MJ, Thompson SG, Powell JT, Ulug P, Bown MJ (2018) Discrete event simulation for decision modeling in health care: lessons from abdominal aortic aneurysm screening. Med Dec Making 38(4):439–451. https://doi.org/10.1177/0272989X17753380

    Article  Google Scholar 

  25. Cipriano L E, Weber T A (2018) Population-level intervention and information collection in dynamic healthcare policy. Health Care Manag Sci 21(4):604–631. https://doi.org/10.1007/s10729-017-9415-5

    Article  Google Scholar 

  26. Yokota F, Thompson K M (2004) Value of information literature analysis: a review of applications in health risk management. Med Dec Making 24(3):287–298. https://doi.org/10.1177/0272989X04263157

    Article  Google Scholar 

  27. Ozcan Y A (2005) Quantitative methods in health care management: techniques and applications. Jossey-Bass.https://www.wiley.com/en-us/Quantitative+Methods+in+Health+Care+Management%3A+Techniques+and+Applications-p-9780787981341

  28. Steuten L, Van De Wetering G, Groothuis-Oudshoorn K, Retèl V (2013) A systematic and critical review of the evolving methods and applications of value of information in academia and practice. PharmacoEconomics 31(1):25–48. https://doi.org/10.1007/s40273-012-0008-3

  29. Heath A, Manolopoulou I, Baio G (2017) A review of methods for analysis of the expected value of information. Med Dec Making 37(7):747–758. https://doi.org/10.1177/0272989X17697692

    Article  Google Scholar 

  30. Thompson KM, Yokota F (2004) Value of information analysis in environmental health risk management decisions: Past, present, and future. Risk Anal 24(3):635–650

    Article  Google Scholar 

  31. Claxton K, Neumann P J, Araki S, Weinstein M C (2001) Bayesian value-of-information analysis: an application to a policy model of Alzheimer’s disease. Int J Technol Assess Health Care 17(1):38–55. https://doi.org/10.1017/S0266462301104058

    Article  Google Scholar 

  32. Felli J C, Hazen G B (1998) Sensitivity analysis and the expected value of perfect information. Med Dec Making 18(1):95–109. https://doi.org/10.1177/0272989X9801800117

    Article  Google Scholar 

  33. Felli J C, Hazen G B (1999) A Bayesian approach to sensitivity analysis. Health Econ 8 (3):263–268. https://doi.org/10.1002/(SICI)1099-1050(199905)8:3<263::AID-HEC426>3.0.CO;2-S

    Article  Google Scholar 

  34. Miller A C (1975) The value of sequential information. Manag Sci 22(1):1–11. https://doi.org/10.1287/mnsc.22.1.1

    Article  Google Scholar 

  35. Pozzi M, Der Kiureghian A (2011) Assessing the value of information for long-term structural health monitoring. In: Kundu T (ed) Proceedings of SPIE 7984, Health Monitoring of Structural and Biological Systems 2011. https://doi.org/10.1117/12.881918, p 79842W

  36. Eckermann S, Willan A R (2008) Time and expected value of sample information wait for no patient. Value Health 11(3):522–526. https://doi.org/10.1111/j.1524-4733.2007.00296.x,

    Article  Google Scholar 

  37. Memarzadeh M, Pozzi M (2016) Value of information in sequential decision making: component inspection, permanent monitoring and system-level scheduling. Reliab Eng Syst Saf 154:137–151. https://doi.org/10.1016/j.ress.2016.05.014

    Article  Google Scholar 

  38. Dong H, Coyle D, Buxton M (2007) Value of information analysis for a new technology: computer-assisted total knee replacement. Int J Technol Assess Health Care 3(3):337–342. https://doi.org/10.1017/S0266462307070419

    Article  Google Scholar 

  39. Martikainen J A, Kivioja A, Hallinen T, Vihinen P (2005) Economic evaluation of temozolomide in the treatment of recurrent glioblastoma multiforme. PharmacoEconomics 23(8):803–815. https://doi.org/10.2165/00019053-200523080-00006

    Article  Google Scholar 

  40. Ginnelly L, Claxton K, Sculpher M J, Golder S (2005) Using value of information analysis to inform publicly funded research priorities. Appl Health Econ Health Policy 4(1):37–46. https://doi.org/10.2165/00148365-200504010-00006

    Article  Google Scholar 

  41. Long E F, Vaidya N K, Brandeau M L (2008) Controlling co-epidemics: analysis of HIV and tuberculosis infection dynamics. Oper Res 56(6):1366–1381. https://doi.org/10.1287/opre.1080.0571

    Article  Google Scholar 

  42. Lee C P, Chertow G M, Zenios S A (2008) Optimal initiation and management of dialysis therapy. Oper Res 56(6):1428–1449. https://doi.org/10.1287/opre.1080.0613

    Article  Google Scholar 

  43. Chen Q, Ayer T, Chhatwal J (2018) Optimal M-switch surveillance policies for liver cancer in a hepatitis C-infected population. Oper Res 66(3):673–696. https://doi.org/10.1287/opre.2017.1706

    Article  Google Scholar 

  44. Chan T, Narasimhan C, Xie Y (2013) Treatment effectiveness and side effects: a model of physician learning. Manag Sci 59(6):1309–1325. https://doi.org/10.1287/mnsc.1120.1640

    Article  Google Scholar 

  45. Long E F, Nohdurft E, Spinler S (2018) Spatial resource allocation for emerging epidemics: a comparison of greedy, myopic, and dynamic policies. Manuf Serv Oper Manag 20(2):181–198. https://doi.org/10.1287/msom.2017.0681

    Article  Google Scholar 

  46. Liu S, Brandeau M L, Goldhaber-Fiebert J D (2017) Optimizing patient treatment decisions in an era of rapid technological advances: the case of hepatitis C treatment. Health Care Manag Sci 20(1):16–32. https://doi.org/10.1007/s10729-015-9330-6

    Article  Google Scholar 

  47. Negoescu D M, Bimpikis K, Brandeau M L, Iancu D A (2017) Dynamic learning of patient response types: an application to treating chronic diseases. Manag Sci. https://doi.org/10.1287/mnsc.2017.2793

  48. Denton B T, Alagoz O, Holder A, Lee E K (2011) Medical decision making: open research challenges

  49. Capan M, Khojandi A, Denton B T, Williams K D, Ayer T, Chhatwal J, Kurt M, Lobo J M, Roberts M S, Zaric G, Zhang S, Schwartz J S (2017) From data to improved decisions: operations research in healthcare delivery. Med Dec Making 37(8):849–859. https://doi.org/10.1177/0272989X17705636

    Article  Google Scholar 

  50. Saville C E, Smith H K, Bijak K (2018) Operational research techniques applied throughout cancer care services: a review. Health Syst 6965:1–22. https://doi.org/10.1080/20476965.2017.1414741

    Google Scholar 

  51. Cooper K, Brailsford S C, Davies R, Raftery J (2006) A review of health care models for coronary heart disease interventions. Health Care Manag Sci 9(4):311–324. https://doi.org/10.1007/s10729-006-9996-x

    Article  Google Scholar 

  52. Stanford R.E. (2004) A frontier analysis approach for benchmarking hospital performance in the treatment of acute myocardial infarction. Health Care Manag Sci 7:145–154. https://doi.org/10.1023/B:HCMS.0000020654.69499.50

    Article  Google Scholar 

  53. Zargoush M, Gümü M, Verter V, Daskalopoulou S S (2018) Designing risk-adjusted therapy for patients with hypertension. Prod Oper Manag 27(12):2291–2312. https://doi.org/10.1111/poms.12872

    Article  Google Scholar 

  54. Hauskrecht M, Fraser H (2000) Planning treatment of ischemic heart disease with partially observable Markov decision processes. Artif Intell Med 18:221–244. https://doi.org/10.1016/S0933-3657(99)00042-1

    Article  Google Scholar 

  55. Denton B T, Kurt M, Shah N D, Bryant S C, Smith S (2009) Optimizing the start time of statin therapy for patients with diabetes. Med Dec Making 29(3):351–367. https://doi.org/10.1177/0272989X08329462

    Article  Google Scholar 

  56. Kurt M, Denton B T, Schaefer A J, Shah N D, Smith S (2011) The structure of optimal statin initiation policies for patients with Type 2 diabetes. IIE Trans Healthcare Syst Eng 1:49–65. https://doi.org/10.1080/19488300.2010.550180

    Article  Google Scholar 

  57. Mason J E, Denton B T, Shah N D, Smith S A (2014) Optimizing the simultaneous management of blood pressure and cholesterol for Type 2 diabetes patients. Eur J Oper Res 233(3):727–738. https://doi.org/10.1016/j.ejor.2013.09.018

    Article  Google Scholar 

  58. Schell G J, Marrero W J, Lavieri M S, Sussman J B, Hayward R A (2016) Data-driven Markov decision process approximations for personalized hypertension treatment planning. MDM Policy Pract 1(1). https://doi.org/10.1177/2381468316674214

  59. Hicklin K, Ivy J S, Payton F C, Viswanathan M, Myerse E (2018) Exploring the value of waiting during labor. Serv Sci 10(3):334–353. https://doi.org/10.1017/S002205070001648X

    Article  Google Scholar 

  60. Chhatwal J, Alagoz O, Burnside E S (2010) Optimal breast biopsy decision-making based on mammographic features and demographic factors

  61. Suen S-, Brandeau M L, Goldhaber-Fiebert J D (2018) Optimal timing of drug sensitivity testing for patients on first-line tuberculosis treatment. Health Care Manag Sci 21(4):632–646. https://doi.org/10.1007/s10729-017-9416-4

    Article  Google Scholar 

  62. Agnihothri S, Cui L, Delasay M, Rajan B (2018) The value of mHealth for managing chronic conditions. Health Care Management Science. https://doi.org/10.1007/s10729-018-9458-2

  63. Onen Z, Sayin S, Gurvit (2018) Optimal population screening policies for Alzheimer’s disease. IISE Trans Healthcare Syst Eng 5579:1–36. https://doi.org/10.1080/24725579.2018.1543738

    Google Scholar 

  64. Ayer T, Alagoz O, Stout N K, Burnside E S (2016) Heterogeneity in women’s adherence and its role in optimal breast cancer screening policies. Manag Sci 62(5):1339–1362. https://doi.org/10.1287/mnsc.2015.2180

    Article  Google Scholar 

  65. Lee E, Lavieri M S, Volk M (2018) Optimal screening for hepatocellular carcinoma: a restless bandit model. Manuf Serv Oper Manag. https://doi.org/10.1287/msom.2017.0697

  66. Deo S, Rajaram K, Rath S, Karmarkar U S, Goetz M B (2015) Planning for HIV screening, testing, and care at the veterans health administration. Oper Res 63(2):287–304. https://doi.org/10.1287/opre.2015.1353

    Article  Google Scholar 

  67. Helm J E, Lavieri M S, Van Oyen M P, Stein J D, Musch D C (2015) Dynamic forecasting and control algorithms of glaucoma progression for clinician decision support. Oper Res 63(5):979–999. https://doi.org/10.1287/opre.2015.1405

    Article  Google Scholar 

  68. Maillart L M, Ivy J S, Ransom S, Diehl K (2008) Assessing dynamic breast cancer screening policies. Oper Res 56(6):1411–1427. https://doi.org/10.1287/opre.1080.0614

    Article  Google Scholar 

  69. Zhang J, Denton B T, Balasubramanian H, Shah N D, Inman B A (2012) Optimization of Prostate Biopsy Referral Decisions. Manuf Serv Oper Manag 14(4):529–547. https://doi.org/10.1287/msom.1120.0388

    Article  Google Scholar 

  70. Erenay F S, Alagoz O, Said A (2014) Optimizing colonoscopy screening for colorectal cancer prevention and surveillance. Manuf Servi Oper Manag 16(3):381–400. https://doi.org/10.1287/msom.2014.0484

    Article  Google Scholar 

  71. Skandari M R, Shechter S M, Zalunardo N (2015) Optimal vascular access choice for patients on hemodialysis. Manuf Serv Oper Manag 17(4):608–619. https://doi.org/10.1287/msom.2015.0552

    Article  Google Scholar 

  72. Sabouri A, Huh W T, Shechter S M (2017) Screening strategies for patients on the kidney transplant waiting list. Oper Res 65(5):1131–1146. https://doi.org/10.1287/opre.2017.1632

    Article  Google Scholar 

  73. Lin Y, Huang S, Simon G E, Liu S (2018) Data-based decision rules to personalize depression follow-up. Sci Rep 8(1):4–11. https://doi.org/10.1038/s41598-018-23326-1

    Google Scholar 

  74. Hutton D W, Tan D, So S K, Brandeau M L (2007) Cost-effectiveness of screening and vaccinating Asian and Pacific Islander adults for hepatitis B. Ann Intern Med 147(7):460. https://doi.org/10.7326/0003-4819-147-7-200710020-00004

    Article  Google Scholar 

  75. Hassmiller Lich K, Cornejo D A, Mayorga M E, Pignone M, Tangka F KL, Richardson L C, Kuo T-M, Meyer A-M, Hall I J, Smith J L, Durham T A, Chall S A, Crutchfield T M, Wheeler S B (2017) Cost-effectiveness analysis of four simulated colorectal cancer screening interventions, North Carolina. Prevent Chron Disease 14(1):160158. https://doi.org/10.5888/pcd14.160158

    Article  Google Scholar 

  76. Leshno M, Halpern Z, Arber N (2003) Cost-effectiveness of colorectal cancer screening in the average risk population. Health Care Manag Sci 6(3):165–174. https://doi.org/10.1023/A:1024488007043

    Article  Google Scholar 

  77. Chirikos T N (2003) Appraising the economic efficiency of cancer treatment: an exploratory analysis of lung cancer. Health Care Manag Sci 6(2):87–95. https://doi.org/10.1023/A:1023380918605

    Article  Google Scholar 

  78. Lin Y, Huang S, Simon G E, Liu S (2019) Cost-effectiveness analysis of prognostic-based depression monitoring. IISE Trans Healthcare Syst Eng 9(1):41–54. https://doi.org/10.1080/24725579.2019.1567627

    Article  Google Scholar 

  79. Robins J, Orellana L, Rotnitzky A (2008) Estimation and extrapolation of optimal treatment and testing strategies. Stat Med 27(23):4678–4721. https://doi.org/10.1002/sim.3301

    Article  Google Scholar 

  80. Kirkizlar E, Faissol D M, Griffin P M, Swann J L (2010) Timing of testing and treatment for asymptomatic diseases. Math Biosci 226(1):28–37. https://doi.org/10.1016/j.mbs.2010.03.007

    Article  Google Scholar 

  81. Kazemian P, Helm J E, Lavieri M S, Stein J D, Van Oyen M P (2018) Dynamic monitoring and control of irreversible chronic diseases with application to glaucoma. Prod Oper Manag 0(0):poms.12975. https://doi.org/10.1111/poms.12975

    Google Scholar 

  82. Yang Y, Goldhaber-Fiebert J D, Wein L M (2013) Analyzing screening policies for childhood obesity. Manag Sci 59(4):782–795. https://doi.org/10.1287/mnsc.1120.1587, NIHMS150003

    Article  Google Scholar 

  83. Ghamat S, Zaric G S, Pun H (2017) Contracts to promote optimal use of optional diagnostic tests in cancer treatment. Prod Oper Manag 27(12):2184–2200. https://doi.org/10.1111/poms.12780

    Article  Google Scholar 

  84. Harper P R, Jones S K (2005) Mathematical models for the early detection and treatment of colorectal cancer. Health Care Manag Sci 8(2):101–109. https://doi.org/10.1007/s10729-005-0393-7

    Article  Google Scholar 

  85. Brønnnum-Hansen H, Jørgensen T, Davidsen M, Madsen M, Osler M, Gerdes L U, Schroll M (2001) Survival and cause of death after myocardial infarction: the Danish MONICA study. J Clin Epidemiol 54(12):1244–1250. https://doi.org/10.1016/S0895-4356(01)00405-X

    Article  Google Scholar 

  86. Burn J, Dennis M, Bamford J, Sandercock P, Wade D, Warlow C (1994) Long-term risk of recurrent stroke after a first-ever stroke. The Oxfordshire Community Stroke Project. Stroke 25 (2):333–7. https://doi.org/10.1161/01.STR.25.2.333

    Article  Google Scholar 

  87. Khera A V, Chaffin M, Aragam K G, Haas M E, Roselli C, Choi S H, Natarajan P, Lander E S, Lubitz S A, Ellinor P T, Kathiresan S (2018) Genome-wide polygenic score to identify a monogenic risk-equivalent for coronary disease. Nat Genet 50(9):1219–1224. https://doi.org/10.1101/218388

    Article  Google Scholar 

  88. Gold M R, Stevenson D, Fryback D G (2002) HALYs and QALYs and DALYs, Oh My: Similarities and Differences in Summary Measures of Population Health. Annu Rev Public Health 23(1):115–134. https://doi.org/10.1146/annurev.publhealth.23.100901.140513

    Article  Google Scholar 

  89. Kleinbaum D G, Klein M (2005) Survival analysis: a self-learning text, 2nd edn. Springer, New York

    Book  Google Scholar 

  90. Hoel M, Iversen T, Nilssen T, Vislie J (2006) Genetic testing in competitive insurance markets with repulsion from chance: A welfare analysis. J Health Econ 25(5):847–860. https://doi.org/10.1016/j.jhealeco.2005.12.003

    Article  Google Scholar 

  91. Centers for Disease Control and Prevention (2017) National Health and Nutrition Examination Survey Data. https://wwwn.cdc.gov/nchs/nhanes/ContinuousNhanes/Default.aspx

  92. Stekhoven D J, Buhlmann P (2012) MissForest–non-parametric missing value imputation for mixed-type data. Bioinformatics 28(1):112–118. https://doi.org/10.1093/bioinformatics/btr597

    Article  Google Scholar 

  93. Nowok B, Raab G M, Dibben C (2016) synthpop: bespoke creation of synthetic data in R. J Stat Softw 74(11). https://doi.org/10.18637/jss.v074.i11, https://www.jstatsoft.org/article/view/v074i11

  94. National Center for Health Statistics (2017) Health, United States, 2016: with chartbook on long-term trends in health https://www.ncbi.nlm.nih.gov/books/NBK453378/

  95. Arias E, Xu J (2019) United States Life Tables, 2017. National Vital Statistics Reports 68(7)

  96. Fryback D G, Dasbach E J, Klein R, Klein B E, Dorn N, Peterson K, Martin P A (1993) The beaver dam health outcomes study: initial catalog of health-state quality factors. Med Dec Making 13 (2):89–102

    Article  Google Scholar 

  97. Pignone M, Earnshaw S, Tice J A, Pletcher M J (2006) Aspirin, statins, or both drugs for the primary prevention of coronary heart disease events in men: a cost-utility analysis. Ann Intern Med 144:326–336. https://doi.org/10.7326/0003-4819-144-5-200603070-00007

    Article  Google Scholar 

  98. Pignone M (2007) Aspirin for the primary prevention of cardiovascular disease in women. Arch Intern Med 167(3):290. https://doi.org/10.1001/archinte.167.3.290

    Article  Google Scholar 

  99. Pandya A, Sy S, Cho S, Weinstein M C, Gaziano T A (2015) Supplementary Online Content: Cost-effectiveness of 10-year risk thresholds for initiation of statin therapy for primary prevention of cardiovascular disease. J Am Med Assoc 314(2):142–150. https://doi.org/10.1001/jama.2015.6822

    Article  Google Scholar 

  100. O’Sullivan A K, Rubin J, Nyambose J, Kuznik A, Cohen D J, Thompson D (2011) Cost estimation of cardiovascular disease events in the US. PharmacoEconomics 29(8):693–704. https://doi.org/10.2165/11584620-000000000-00000

    Article  Google Scholar 

  101. Medical Expenditure Panel Survey (2015) MEPS HC-163: 2013 full year consolidated data file. https://meps.ahrq.gov/data_stats/download_data/pufs/h163/h163doc.shtml

  102. GoodRx (2017) Prescription prices, coupons & pharmacy information. https://www.goodrx.com/

  103. Color Genomics (2018) Genetic testing for inherited heart conditions. https://www.color.com/product/hereditary-heart-health-genetic-test

  104. Neumann PJ, Sanders GD, Russell LB, Siegel JE (2016) Cost-effectiveness in health and medicine. Oxford University Press

  105. Abraham G, Havulinna A S, Bhalala O G, Byars S G, De Livera A M, Yetukuri L, Tikkanen E, Perola M, Schunkert H, Sijbrands E J, Palotie A, Samani N J, Salomaa V, Ripatti S, Inouye M (2016) Genomic prediction of coronary heart disease. Eur Heart J 37(43):3267–3278. https://doi.org/10.1093/eurheartj/ehw450

    Article  Google Scholar 

  106. Kochanek K D, Murphy S L, Xu J, Arias E (2019) Deaths: final data for 2017. Natl Vital Stat Rep 68(9):1–18

    Google Scholar 

  107. Sussman J, Vijan S, Hayward R (2013) Using Benefit-Based Tailored Treatment to Improve the Use of Antihypertensive Medications. Circulation 128(21):2309–2317. https://doi.org/10.1161/CIRCULATIONAHA.113.002290

    Article  Google Scholar 

  108. Pletcher M J, Pignone M, Earnshaw S, McDade C, Phillips K A, Auer R, Zablotska L, Greenland P (2014) Using the coronary artery calcium score to guide statin therapy. Circul Cardiovascul Qual Outcomes 7 (2):276–284. https://doi.org/10.1161/CIRCOUTCOMES.113.000799 NIHMS150003

    Article  Google Scholar 

  109. Hayward R A, Krumholz H M, Zulman D M, Timbie J W, Vijan S (2010) Optimizing statin treatment for primary prevention of coronary artery disease. Ann Intern Med 152(2):69. https://doi.org/10.7326/0003-4819-152-2-201001190-00004

    Article  Google Scholar 

  110. Ibrahim J G, Chen M-H, Sinha D (2001) Bayesian survival analysis. Springer Series in Statistics. Springer, New York. https://doi.org/10.1007/978-1-4757-3447-8

    Google Scholar 

  111. Ben-Tal A, Ghaoui L E, Nemirovski A (2009) Robust optimization. Princeton University Press

  112. McNeil J J, Nelson M R, Woods R L, Lockery J E, Wolfe R, Reid C M, Kirpach B, Shah R C, Ives D G, Storey E, Ryan J, Tonkin A M, Newman A B, Williamson J D, Margolis K L, Ernst M E, Abhayaratna W P, Stocks N, Fitzgerald S M, Orchard S G, Trevaks R E, Beilin L J, Donnan G A, Gibbs P, Johnston C I, Radziszewska B, Grimm R, Murray A M (2018) Effect of aspirin on all-cause mortality in the healthy elderly. N Engl J Med 379(16):1519–1528. https://doi.org/10.1056/nejmoa1803955

    Article  Google Scholar 

  113. Williams B, Mancia G, Spiering W, Agabiti Rosei E, Azizi M, Burnier M, Clement D L, Coca A, de Simone G, Dominiczak A, Kahan T, Mahfoud F, Redon J, Ruilope L, Zanchetti A, Kerins M, Kjeldsen S E, Kreutz R, Laurent S, Lip G Y H, McManus R, Narkiewicz K, Ruschitzka F, Schmieder R E, Shlyakhto E, Tsioufis C, Aboyans V, Desormais I, De Backer G, Heagerty A M, Agewall S, Bochud M, Borghi C, Boutouyrie P, Brguljan J, Bueno H, Caiani E G, Carlberg B, Chapman N, Cífková R, Cleland J G F, Collet J-P, Coman I M, de Leeuw P W, Delgado V, Dendale P, Diener H-C, Dorobantu M, Fagard R, Farsang C, Ferrini M, Graham I M, Grassi G, Haller H, Hobbs F D R, Jelakovic B, Jennings C, Katus H A, Kroon A A, Leclercq C, Lovic D, Lurbe E, Manolis A J, McDonagh T A, Messerli F, Muiesan M L, Nixdorff U, Olsen M H, Parati G, Perk J, Piepoli M F, Polonia J, Ponikowski P, Richter D J, Rimoldi S F, Roffi M, Sattar N, Seferovic P M, Simpson I A, Sousa-Uva M, Stanton A V, van de Borne P, Vardas P, Volpe M, Wassmann S, Windecker S, Zamorano J L, Windecker S, Aboyans V, Agewall S, Barbato E, Bueno H, Coca A, Collet J-P, Coman I M, Dean V, Delgado V, Fitzsimons D, Gaemperli O, Hindricks G, Iung B, Jüni P, Katus H A, Knuuti J, Lancellotti P, Leclercq C, McDonagh T A, Piepoli M F, Ponikowski P, Richter D J, Roffi M, Shlyakhto E, Simpson I A, Sousa-Uva M, Zamorano J L, Tsioufis C, Lurbe E, Kreutz R, Bochud M, Rosei E A, Jelakovic B, Azizi M, Januszewics A, Kahan T, Polonia J, van de Borne P, Williams B, Borghi C, Mancia G, Parati G, Clement D L, Coca A, Manolis A, Lovic D, Benkhedda S, Zelveian P, Siostrzonek P, Najafov R, Pavlova O, De Pauw M, Dizdarevic-Hudic L, Raev D, Karpettas N, Linhart A, Olsen M H, Shaker A F, Viigimaa M, Metsärinne K, Vavlukis M, Halimi J-M, Pagava Z, Schunkert H, Thomopoulos C, Páll D, Andersen K, Shechter M, Mercuro G, Bajraktari G, Romanova T, Trušinskis K, Saade G A, Sakalyte G, Noppe S, DeMarco D C, Caraus A, Wittekoek J, Aksnes T A, Jankowski P, Polonia J, Vinereanu D, Baranova E I, Foscoli M, Dikic A D, Filipova S, Fras Z, Bertomeu-Martínez V, Carlberg B, Burkard T, Sdiri W, Aydogdu S, Sirenko Y, Brady A, Weber T, Lazareva I, Backer T D, Sokolovic S, Jelakovic B, Widimsky J, Viigimaa M, Pörsti I, Denolle T, Krämer B K, Stergiou G S, Parati G, Trušinskis K, Miglinas M, Gerdts E, Tykarski A, de Carvalho Rodrigues M, Dorobantu M, Chazova I, Lovic D, Filipova S, Brguljan J, Segura J, Gottsäter A, Pechère-Bertschi A, Erdine S, Sirenko Y, Brady A (2018) 2018 ESC/ESH guidelines for the management of arterial hypertension. Eur Heart J 39(33):3021–3104. https://doi.org/10.1093/eurheartj/ehy339

  114. James P A, Oparil S, Carter B L, Cushman W C, Dennison-Himmelfarb C, Handler J, Lackland D T, LeFevre M L, MacKenzie T D, Ogedegbe O, Smith S C, Svetkey L P, Taler S J, Townsend R R, Wright J T, Narva A S, Ortiz E (2014) Evidence-based guideline for the management of high blood pressure in adults. J Am Med Assoc 311(5):507–20. https://doi.org/10.1001/jama.2013.284427

    Article  Google Scholar 

  115. Kazi D S, Moran A E, Coxson P G, Penko J, Ollendorf D A, Pearson S D, Tice J A, Guzman D, Bibbins-Domingo K (2016) Cost-effectiveness of PCSK9 inhibitor therapy in patients with heterozygous familial hypercholesterolemia or atherosclerotic cardiovascular disease. J Am Med Assoc 316(7):743–. https://doi.org/10.1001/jama.2016.11004

  116. United States Department of Health and Human Services (2006) Coverage and Reimbursement of Genetic Tests and Services: Report of the Secretary’s Advisory Committee on Genetics, Health, and Society, pp 1–57

Download references

Funding

Wesley J. Marrero’s work on this study was supported in part by the National Science Foundation grant DGE 1256260. Mariel S. Lavieri’s work was supported in part by the National Science Foundation grant CMMI-1552545. Jeremy B. Sussman’s work was supported in part by the United States Department of Veterans Affairs grants VA HSR&D CDA13-021 and VA HSR&D IIR 15-432. The National Science Foundation and the Veterans Affairs had no influence on the study’s design, conduct, and/or reporting.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. Material preparation, data collection and analysis were performed by Wesley J. Marrero. The first draft of the manuscript was written by Wesley J. Marrero and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Wesley J. Marrero.

Ethics declarations

Conflict of interests

The authors declare that they have no conflict of interest.

Ethics Approval

In accordance with the policies of the University of Michigan, ethics approval was not needed.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Appendices

Appendix

1.1 A.1 Progression of Risk Factors Over Time

Table 4 Linear regression models coefficients

A.2 Convergence Analysis

Fig. 8
figure 8

Convergence of QALYs and cost saved in our simulation over the number of health trajectory replications under a single GRS realization per patient. Red line represents the QALYs and cost saved at 2,000 health trajectory replications

Fig. 9
figure 9

Convergence of QALYs and cost saved in our simulation over the number of GRS realizations with a fixed number of heath trajectory replications. Red line represents the QALYs and cost saved at 500 GRS realizations and 500 health trajectory replications

Fig. 10
figure 10

Convergence of QALYs and cost saved in our simulation over the number of health trajectory replications under 100 GRS realizations per patient. Red line represents the QALYs and cost saved at 750 health trajectory replications and 100 GRS realizations

A.3 Results of Sensitivity Analysis by Age Groups

Fig. 11
figure 11

Testing cost sensitivity analysis results by age group. Shaded boxplots represent the base case, asterisks represent outliers, and points in the center of the boxes represent the average testing year

Fig. 12
figure 12

Treatment cost sensitivity analysis results by age group. Shaded boxplots represent the base case, asterisks represent outliers, and points in the center of the boxes represent the average testing year

Fig. 13
figure 13

Treatment disutility sensitivity analysis results by age group. Shaded boxplots represent the base case, asterisks represent outliers, and points in the center of the boxes represent the average testing year

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Marrero, W.J., Lavieri, M.S. & Sussman, J.B. Optimal cholesterol treatment plans and genetic testing strategies for cardiovascular diseases. Health Care Manag Sci 24, 1–25 (2021). https://doi.org/10.1007/s10729-020-09537-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10729-020-09537-x

Keywords

Navigation