Skip to main content
Log in

Challenges and Opportunities in Interdisciplinary Research and Real-World Data for Treatment Sequences in Health Technology Assessments

  • Review Article
  • Published:
PharmacoEconomics Aims and scope Submit manuscript

Abstract

With an ever-increasing number of treatment options, the assessment of treatment sequences has become crucial in health technology assessment (HTA). This review systematically explores the multifaceted challenges inherent in evaluating sequences, delving into their interplay and nuances that go beyond economic model structures. We synthesised a ‘roadmap’ of literature from key methodological studies, highlighting the evolution of recent advances and emerging research themes. These insights were compared against HTA guidelines to identify potential avenues for future research. Our findings reveal a spectrum of challenges in sequence evaluation, encompassing selecting appropriate decision-analytic modelling approaches and comparators, deriving appropriate clinical effectiveness evidence in the face of data scarcity, scrutinising effectiveness assumptions and statistical adjustments, considering treatment displacement, and optimising model computations. Integrating methodologies from diverse disciplines—statistics, epidemiology, causal inference, operational research and computer science—has demonstrated promise in addressing these challenges. An updated review of application studies is warranted to provide detailed insights into the extent and manner in which these methodologies have been implemented. Data scarcity on the effectiveness of treatment sequences emerged as a dominant concern, especially because treatment sequences are rarely compared in clinical trials. Real-world data (RWD) provide an alternative means for capturing evidence on effectiveness and future research should prioritise harnessing causal inference methods, particularly Target Trial Emulation, to evaluate treatment sequence effectiveness using RWD. This approach is also adaptable for analysing trials harbouring sequencing information and adjusting indirect comparisons when collating evidence from heterogeneous sources. Such investigative efforts could lend support to reviews of HTA recommendations and contribute to synthesising external control arms involving treatment sequences.

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

Similar content being viewed by others

References

  1. NICE. Position statement: consideration of products recommended for use in the Cancer Drugs Fund as comparators, or in a treatment sequence, in the appraisal of a new cancer product. Available at: https://www.nice.org.uk/Media/Default/About/what-we-do/NICE-guidance/NICE-technology-appraisal-guidance/cancer-drugs-fund/CDF-comparator-position-statement.pdf. Accessed 23 Mar 2020.

  2. Claxton K. Exploring uncertainty in cost-effectiveness analysis. Pharmacoeconomics. 2008;26:781–98.

    Article  PubMed  Google Scholar 

  3. Tosh J, Stevenson M, Akehurst R. Health economic modelling of treatment sequences for rheumatoid arthritis: a systematic review. Curr Rheumatol Rep. 2014;16:447.

    Article  PubMed  Google Scholar 

  4. Benedict A, Stevenson M, Sorensen S. ISPOR Europe 2017: Modeling separate lines of treatment versus treatment sequences in cancer (Workshop 8). Available at: https://www.ispor.org/docs/default-source/presentations/1221.pdf?sfvrsn=6f1f29a1_1. Accessed 20 June 2020.

  5. Al M, Jin H, Robins J, et al. Virtual ISPOR Europe 2020: novel methods for modeling treatment sequencing (spotlight session 3). Available at: https://www.ispor.org/conferences-education/conferences/past-conferences/ispor-europe-2020/program/spotlight-sessions. Accessed 19 Nov 2020.

  6. Kaufman HL, Atkins MB, Subedi P, et al. The promise of immuno-oncology: implications for defining the value of cancer treatment. J Immunother Cancer. 2019;7:1–11.

    Article  Google Scholar 

  7. Chehayeb RJ, Hood A, Wang X, et al. Treatment sequencing patterns and associated direct medical costs of metastatic breast cancer care in the United States, 2011 to 2021. JAMA Netw Open. 2022;5:e2244204–304.

    Article  Google Scholar 

  8. Roux J, Grimaud O, Leray E. Use of state sequence analysis for care pathway analysis: the example of multiple sclerosis. Stat Methods Med Res. 2019;28:1651–63.

    Article  PubMed  Google Scholar 

  9. Kim AH, Jang JE, Han J. Current status on the therapeutic strategies for heart failure and diabetic cardiomyopathy. Biomed Pharmacother. 2022;145: 112463.

    Article  CAS  PubMed  Google Scholar 

  10. Kern DM, Cepeda MS, Defalco F, et al. Treatment patterns and sequences of pharmacotherapy for patients diagnosed with depression in the United States: 2014 through 2019. BMC Psychiatry. 2020;20:1–10.

    Article  Google Scholar 

  11. Zheng Y, Pan F, Sorensen S. Modeling treatment sequences in pharmacoeconomic models. Pharmacoeconomics. 2017;35:15–24.

    Article  PubMed  Google Scholar 

  12. Viola MG, Diamantopoulos A. Determining the baseline strategy in a cost-effectiveness analysis with treatment sequences. Appl Health Econ Health Policy. 2020;18:17–29.

    Article  PubMed  Google Scholar 

  13. NICE. NICE Process and methods: guide to the methods of technology appraisal 2013 (PMG 9). Available at: https://www.nice.org.uk/process/pmg9/chapter/foreword. Accessed 2 June 2020.

  14. NICE. NICE health technology evaluations: the manual (PMG 36). Available at: https://www.nice.org.uk/process/pmg36/chapter/introduction-to-health-technology-evaluation. Accessed 17 Aug 2023.

  15. Canadian Agency for Drugs and Technologies in Health (CADTH). Guidelines for the economic evaluation of health technologies: Canada — 4th edition. Available at: https://www.cadth.ca/sites/default/files/pdf/guidelines_for_the_economic_evaluation_of_health_technologies_canada_4th_ed.pdf. Accessed 11 Mar 2023.

  16. Faria R. Problem structuring in economic evaluation. In: Oxford Research Encyclopedia of Economics and Finance. Oxford University Press, 2021.

  17. NICE. Briefing paper for methods review working party on choosing comparators. Available at: https://www.sheffield.ac.uk/media/34080/download?attachment. Accessed 26 Jan 2023.

  18. NICE. Briefing paper for methods review working party on treatment sequences and downstream costs. Available at: https://www.sheffield.ac.uk/media/34080/download?attachment. Accessed 26 Jan 2023.

  19. NICE. Taking a proportionate approach to technology appraisals. Available at: https://www.nice.org.uk/about/what-we-do/proportionate-approach-to-technology-appraisals. Accessed 26 Jan 2024.

  20. National Institute for Health and Care Excellence. NICE real-world evidence framework [ECD 9]. Available at: https://www.nice.org.uk/corporate/ecd9/chapter/overview. Accessed 30 June 2023.

  21. Greenhalgh T, Peacock R. Effectiveness and efficiency of search methods in systematic reviews of complex evidence: audit of primary sources. Br Med J. 2005;331:1064–5.

    Article  Google Scholar 

  22. Schlosser RW, Wendt O, Bhavnani S, et al. Use of information-seeking strategies for developing systematic reviews and engaging in evidence-based practice: the application of traditional and comprehensive Pearl Growing. A review. Int J Lang Commun Disord. 2006;41:567–82.

    Article  PubMed  Google Scholar 

  23. Hernandez L, O’Donnell M, Postma M. Modeling approaches in cost-effectiveness analysis of disease-modifying therapies for relapsing-remitting multiple sclerosis: an updated systematic review and recommendations for future economic evaluations. Pharmacoeconomics. 2018;36:1223–52.

    Article  PubMed  Google Scholar 

  24. Jansen JP, Incerti D, Mutebi A, et al. Cost-effectiveness of sequenced treatment of rheumatoid arthritis with targeted immune modulators. J Med Econ. 2017;20:703–14.

    Article  PubMed  Google Scholar 

  25. Badampudi D, Wohlin C, Petersen K. Experiences from using snowballing and database searches in systematic literature studies. In: Proceedings of the 19th International Conference on evaluation and assessment in software engineering (EASE). Nanjing, China: ACM Digital Library, 2015.

  26. Zwakman M, Verberne LM, Kars MC, et al. Introducing PALETTE: an iterative method for conducting a literature search for a review in palliative care. BMC Palliat Care. 2018;17:82.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Haywood P. Increasing complexity of cancer care: How displaced treatments impact efficiency, cost-effectiveness and equity [Doctoral Thesis]. University of Technology Sydney, 2018.

  28. Lewis R. Quantitative evidence synthesis methods for the assessment of the effectiveness of treatment sequences for clinical and economic decision-making [Doctoral Thesis]. Bangor University, 2019.

  29. Tosh J. Simulation optimisation to inform economic evaluations of sequential therapies for chronic conditions: a case study in rheumatoid arthritis [Doctoral Thesis]. University of Sheffield, 2015.

  30. Kim E. Sequential drug decision problems in long-term medical conditions: a case study of primary hypertension [Doctoral Thesis]. University of Sheffield, 2015.

  31. Hawkins N, Sculpher M, Epstein D. Cost-effectiveness analysis of treatments for chronic disease: using R to incorporate time dependency of treatment response. Med Decis Making. 2005;25:511–9.

    Article  PubMed  Google Scholar 

  32. Saramago P, Manca A, Sutton AJ. Deriving input parameters for cost-effectiveness modeling: taxonomy of data types and approaches to their statistical synthesis. Value Health. 2012;15:639–49.

    Article  PubMed  Google Scholar 

  33. Tordrup D, Chouaid C, Cuijpers P, et al. Priorities for health economic methodological research: results of an expert consultation. Int J Technol Assess Health Care. 2017;33:609–19.

    Article  PubMed  Google Scholar 

  34. Heeg BMS. Developing health economic models of chronic diseases for reimbursement purposes [Doctoral Thesis]. University of Groningen: Groningen, the Netherlands. 2015.

  35. Brixner DI, Watkins JB. Can CER be an effective tool for change in the development and assessment of new drugs and technologies? J Manag Care Pharm. 2012;18:S06-S11.

    PubMed  Google Scholar 

  36. Tappenden P, Chilcott J, Brennan A, et al. Whole disease modeling to inform resource allocation decisions in cancer: a methodological framework. Value Health. 2012;15:1127–36.

    Article  PubMed  Google Scholar 

  37. Lord J, Willis S, Eatock J, et al. Economic modelling of diagnostic and treatment pathways in National Institute for Health and Care Excellence clinical guidelines: the Modelling Algorithm Pathways in Guidelines (MAPGuide) project. Health Technol Assess. 2013;17(58):1–192.

  38. Brennan A, Chick SE, Davies R. A taxonomy of model structures for economic evaluation of health technologies. Health Econ. 2006;15:1295–310.

    Article  PubMed  Google Scholar 

  39. Rauner MS, Gutjahr WJ, Heidenberger K, et al. Dynamic policy modeling for chronic diseases: metaheuristic-based identification of pareto-optimal screening strategies. Oper Res. 2010;58:1269–86.

    Article  Google Scholar 

  40. Cressman S, Browman GP, Hoch JS, et al. A time-trend economic analysis of cancer drug trials. Oncologist. 2015;20:729–36.

    Article  PubMed  PubMed Central  Google Scholar 

  41. de Mello-Sampayo F. The timing and probability of treatment switch under cost uncertainty: an application to patients with gastrointestinal stromal tumor. Value Health. 2014;17:215–22.

    Article  PubMed  Google Scholar 

  42. Vernon JA, Hughen WK. A primer on dynamic optimization and optimal control in pharmacoeconomics. Value Health. 2006;9:106–13.

    Article  PubMed  Google Scholar 

  43. Chick SE, Inoue K. New two-stage and sequential procedures for selecting the best simulated system. Oper Res. 2001;49:732–43.

    Article  Google Scholar 

  44. Murphy SA, Oslin DW, Rush AJ, et al. Methodological challenges in constructing effective treatment sequences for chronic psychiatric disorders. Neuropsychopharmacology. 2007;32:257–62.

    Article  CAS  PubMed  Google Scholar 

  45. Cain LE, Saag MS, Petersen M, et al. Using observational data to emulate a randomized trial of dynamic treatment-switching strategies: an application to antiretroviral therapy. Int J Epidemiol. 2016;45:2038–49.

    Article  PubMed  Google Scholar 

  46. Messner DA. Evaluating the comparative effectiveness of treatment sequences in oncology: a new approach. J Comp Eff Res. 2015;4:537–40.

    Article  PubMed  Google Scholar 

  47. Dekker J, Van HL, Hendriksen M, et al. What is the best sequential treatment strategy in the treatment of depression? Adding pharmacotherapy to psychotherapy or vice versa? Psychother Psychosom. 2013;82:89–98.

    Article  PubMed  Google Scholar 

  48. Weintraub M, Waldman E, Koplewitz B, et al. A sequential treatment algorithm for infants with stage 4s neuroblastoma and massive hepatomegaly. Pediatr Blood Cancer. 2012;59:182–4.

    Article  PubMed  Google Scholar 

  49. Trappe R, Oertel S, Leblond V, et al. Sequential treatment with rituximab followed by CHOP chemotherapy in adult B-cell post-transplant lymphoproliferative disorder (PTLD): the prospective international multicentre phase 2 PTLD-1 trial. Lancet Oncol. 2012;13:196–206.

    Article  CAS  PubMed  Google Scholar 

  50. Grigor C, Capell H, Stirling A, et al. Effect of a treatment strategy of tight control for rheumatoid arthritis (the TICORA study): a single-blind randomised controlled trial. Lancet. 2004;364:263–9.

    Article  PubMed  Google Scholar 

  51. Wang L, Rotnitzky A, Lin X, et al. Evaluation of viable dynamic treatment regimes in a sequentially randomized trial of advanced prostate cancer. J Am Stat Assoc. 2012;107:493–508.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Briggs A, Sidhu M, Baker T. A framework for analysing treatment sequences: incorporating time dependent transitions through partitioned survival analysis. Value Health. 2014;17:A204–5.

    Google Scholar 

  53. Lewis RA, Hughes D, Sutton AJ, et al. Quantitative evidence synthesis methods for the assessment of the effectiveness of treatment sequences for clinical and economic decision making: a review and taxonomy of simplifying assumptions. Pharmacoeconomics. 2021;39:25–61.

  54. Welton N, Phillippo D, Owen R, et al. CHTE2020 sources and synthesis of evidence; update to evidence synthesis methods. Available at: https://rees-france.com/wp-content/uploads/2020/12/CHTE-2020_synthesis-of-evidence.pdf. Accessed 29 Aug 2022.

  55. Huang M, Ramsey S, Xue W, et al. Conceptual framework and methodological challenges for modeling effectiveness in oncology treatment sequence models. Pharmacoeconomics. 2022;40:257–268.

  56. Simpson A, Ramagopalan SV. R WE ready for reimbursement? A round up of developments in real-world evidence relating to HTA: part 5. J Comp Eff Res. 2022;11:213–5.

    Article  CAS  PubMed  Google Scholar 

  57. NICE. Enzalutamide for treating metastatic hormone-relapsed prostate cancer before chemotherapy is indicated (TA 377). Available at: https://www.nice.org.uk/guidance/ta377. Accessed 30 Apr 2021.

  58. NICE. Abiraterone for treating metastatic hormone-relapsed prostate cancer before chemotherapy is indicated (TA 387). Available at: https://www.nice.org.uk/guidance/ta387. Accessed 30 Apr 2021.

  59. Pan F, Reifsnider O, Zheng Y, et al. Modeling clinical outcomes in prostate cancer: application and validation of the discrete event simulation approach. Value Health. 2018;21:416–22.

    Article  PubMed  Google Scholar 

  60. Parikh RC, Du XL, Robert MO, et al. Cost-effectiveness of treatment sequences of chemotherapies and targeted biologics for elderly metastatic colorectal cancer patients. J Man Care Spec Pharm. 2017;23:64–73.

    Google Scholar 

  61. Rognoni C, Ciani O, Sommariva S, et al. Cost-effectiveness analysis of treatments involving radioembolization in intermediate-stage hepatocellular carcinoma. J Comp Eff Res. 2018;7:209–21.

    Article  PubMed  Google Scholar 

  62. Woods BS, Sideris E, Palmer S, et al. Partitioned survival and state transition models for healthcare decision making in oncology: where are we now? Value Health. 2020;23:1613–21.

    Article  PubMed  Google Scholar 

  63. Hernán MA, Robins JM. Using big data to emulate a target trial when a randomized trial is not available. Am J Epidemiol. 2016;183:758–64.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Spelman T, Magyari M, Piehl F, et al. Treatment escalation vs immediate initiation of highly effective treatment for patients with relapsing-remitting multiple sclerosis: data from 2 different national strategies. JAMA Neurol. 2021;78:1197–204.

    Article  PubMed  Google Scholar 

  65. Simonsen CS, Flemmen HØ, Broch L, et al. Early high efficacy treatment in multiple sclerosis is the best predictor of future disease activity over 1 and 2 years in a Norwegian population-based registry. Front Neurol. 2021;12: 693017.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Brown JWL, Coles A, Horakova D, et al. Association of initial disease-modifying therapy with later conversion to secondary progressive multiple sclerosis. JAMA. 2019;321:175–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. He A, Merkel B, Brown JWL, et al. Timing of high-efficacy therapy for multiple sclerosis: a retrospective observational cohort study. Lancet Neurol. 2020;19:307–16.

    Article  CAS  PubMed  Google Scholar 

  68. Vertosick EA, Assel M, Vickers AJ. A systematic review of instrumental variable analyses using geographic region as an instrument. Cancer Epidemiol. 2017;51:49–55.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Latimer NR, Abrams KR. NICE DSU Technical Support Document 16: Adjusting survival time estimates in the presence of treatment switching. Available at: http://nicedsu.org.uk/wp-content/uploads/2016/03/TSD16_Treatment_Switching.pdf. Accessed 2 June 2020

  70. National Institute for Clinical Excellence. Capecitabine and oxaliplatin in the adjuvant treatment of stage III (Dukes’ C) colon cancer (Technology Appraisal 100). Available at: https://www.nice.org.uk/guidance/ta100. Accessed 30 Jun 2020.

  71. Barton P, Jobanputra P, Wilson J, et al. The use of modelling to evaluate new drugs for patients with a chronic condition: the case of antibodies against tumour necrosis factor in rheumatoid arthritis. Health Technol Assess. 2004;8:iii (1-91).

    Article  CAS  PubMed  Google Scholar 

  72. Tran-Duy A, Boonen A, van de Laar MA, et al. A discrete event modelling framework for simulation of long-term outcomes of sequential treatment strategies for ankylosing spondylitis. Ann Rheum Dis. 2011;70:2111–8.

    Article  PubMed  Google Scholar 

  73. Drummond MF, Sculpher MJ, Claxton K, et al. Methods for the economic evaluation of health care programmes. 4th ed. Oxford: Oxford University Press; 2015.

    Google Scholar 

  74. Tappenden P. Conceptual modelling for health economic model development. Available at: https://eprints.whiterose.ac.uk/74464/1/HEDSDP1205.pdf. Accessed 30 Mar 2023.

  75. Kaltenthaler E, Tappenden P, Paisley S, et al. NICE DSU Technical Support Document 13: identifying and reviewing evidence to inform the conceptualisation and population of cost-effectiveness models. Available at: https://www.ncbi.nlm.nih.gov/books/NBK425832/. Accessed 30 Mar 2023.

  76. Dias S, Welton NJ, Sutton AJ, et al. NICE DSU Technical Support Document 1: introduction to evidence synthesis for decision making. Available at: https://www.ncbi.nlm.nih.gov/books/NBK395870/pdf/Bookshelf_NBK395870.pdf. Accessed 30 Mar 2023.

  77. Dias S, Sutton AJ, Welton NJ, et al. NICE DSU Technical Support Document 3: heterogeneity: subgroups, meta-regression, bias and bias-adjustment. Available at: https://www.ncbi.nlm.nih.gov/books/NBK395886/pdf/Bookshelf_NBK395886.pdf. Accessed 30 Mar 2023.

  78. Dias S, Welton NJ, Sutton AJ, et al. NICE DSU Technical Support Document 4: inconsistency in networks of evidence based on randomised controlled trials. Available at: https://www.ncbi.nlm.nih.gov/books/NBK310372/pdf/Bookshelf_NBK310372.pdf. Accessed 30 Mar 2023.

  79. Phillippo D, Ades T, Dias S, et al. NICE DSU Technical Support Document 18: Methods for population-adjusted indirect comparisons in submissions to NICE. Available at: https://www.sheffield.ac.uk/nice-dsu/tsds/population-adjusted. Accessed 15 Oct 2023.

  80. Woods B, Sideris E, Palmer S, et al. NICE DSU Technical Support Document 19: partitioned survival analysis for decision modelling in health care: a critical review. Available at: https://www.sheffield.ac.uk/media/34205/download?attachment. Accessed 4 July 2023.

  81. Faria R, Hernadez Alava M, Manca A, et al. NICE DSU Technical Support Document 17: the use of observational data to inform estimates of treatment effectiveness in technology appraisal: methods for comparative individual patient data. Available at: http://nicedsu.org.uk/wp-content/uploads/2016/03/TSD17-DSU-Observational-data-FINAL.pdf. Accessed 2 June 2020.

  82. Lawlor DA, Tilling K, Davey SG. Triangulation in aetiological epidemiology. Int J Epidemiol. 2017;45:1866–86.

    PubMed Central  Google Scholar 

  83. Hammerton G, Munafò MR. Causal inference with observational data: the need for triangulation of evidence. Psychol Med. 2021;51:563–78.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Vanni T, Karnon J, Madan J, et al. Calibrating models in economic evaluation. Pharmacoeconomics. 2011;29:35–49.

    Article  PubMed  Google Scholar 

  85. Mandrik O, Thomas C, Whyte S, et al. Calibrating natural history of cancer models in the presence of data incompatibility: problems and solutions. Pharmacoeconomics. 2022;40:359–66.

    Article  PubMed  Google Scholar 

  86. Ryan CJ, Smith MR, Fizazi K, et al. Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naive men with metastatic castration-resistant prostate cancer (COU-AA-302): final overall survival analysis of a randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 2015;16:152–60.

    Article  CAS  PubMed  Google Scholar 

  87. Latimer NR, White IR, Tilling K, et al. Improved two-stage estimation to adjust for treatment switching in randomised trials: g-estimation to address time-dependent confounding. Stat Methods Med Res. 2020;29:2900–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Jansen JP, Cope S. Meta-regression models to address heterogeneity and inconsistency in network meta-analysis of survival outcomes. BMC Med Res Methodol. 2012;12:1–16.

    Article  Google Scholar 

  89. Jansen JP, Incerti D, Trikalinos TA. Multi‐state network meta‐analysis of progression and survival data. Stat Med. 2023;42(19):3371–3391.

  90. NICE. Adalimumab, etanercept, infliximab, rituximab and abatacept for the treatment of rheumatoid arthritis after the failure of a TNF inhibitor (part review of NICE technology appraisal guidance 36, review of NICE technology appraisal guidance 126 and 141) (TA 195). Available at: https://www.nice.org.uk/guidance/ta195. Accessed 23 Mar 2020.

  91. National Institute for Health and Care Excellence. Rituximab in combination with glucocorticoids for treating anti-neutrophil cytoplasmic antibody-associated vasculitis (Technology Appraisal 308). at: https://www.nice.org.uk/guidance/ta308. Accessed 30 Jun 2020.

  92. National Institute for Health and Care Excellence. Apremilast for treating moderate to severe plaque psoriasis (Technology Appraisal 419). Available at: https://www.nice.org.uk/guidance/ta419. Accessed 30 Jun 2020.

  93. National Institute for Health and Care Excellence. Ixekizumab for treating moderate to severe plaque psoriasis (Technology Appraisal 442). Available at: https://www.nice.org.uk/guidance/ta442. Accessed 30 Jun 2020.

  94. Holland PW. Statistics and causal inference. J Am Stat Assoc. 1986;81:945–60.

    Article  Google Scholar 

  95. Hernán MA, Robins JM. Causal Inference: What If. Boca Raton: Chapman & Hall/CRC; 2020.

    Google Scholar 

  96. Robins J. A new approach to causal inference in mortality studies with a sustained exposure period—application to control of the healthy worker survivor effect. Mathematical Modelling. 1986;7:1393–512.

    Article  Google Scholar 

  97. Naimi AI, Cole SR, Kennedy EH. An introduction to g methods. Int J Epidemiol. 2017;46:756–62.

    PubMed  Google Scholar 

  98. Whyte S, Walsh C, Chilcott J. Bayesian calibration of a natural history model with application to a population model for colorectal cancer. Med Decis Making. 2011;31:625–41.

    Article  PubMed  Google Scholar 

  99. Bell Gorrod H, Kearns B, Stevens J, et al. A review of survival analysis methods used in NICE technology appraisals of cancer treatments: consistency, limitations, and areas for improvement. Med Decis Making. 2019;39:899–909.

    Article  PubMed  Google Scholar 

  100. Latimer NR, Adler AI. Extrapolation beyond the end of trials to estimate long term survival and cost effectiveness. BMJ Med. 2022;1:e000094

  101. Bullement A, Stevenson MD, Baio G, et al. A systematic review of methods to incorporate external evidence into trial-based survival extrapolations for health technology assessment. Med Decis Making. 2023;43(5):610–20.

  102. Jaksa A, Louder A, Maksymiuk C, et al. A comparison of seven oncology external control arm case studies: critiques from regulatory and health technology assessment agencies. Value Health. 2022;25:1967–76.

    Article  PubMed  Google Scholar 

  103. Burger HU, Gerlinger C, Harbron C, et al. The use of external controls: to what extent can it currently be recommended? Pharm Stat. 2021;20:1002–16.

    Article  PubMed  Google Scholar 

  104. Colnet B, Mayer I, Chen G, et al. Causal inference methods for combining randomized trials and observational studies: a review. arXiv preprint. Available at: https://doi.org/10.48550/arXiv.2011.08047. Accessed 30 June 2023.

  105. Dahabreh IJ, Hernán MA. Extending inferences from a randomized trial to a target population. Eur J Epidemiol. 2019;34:719–22.

    Article  CAS  PubMed  Google Scholar 

  106. NHS. Cancer Drugs Fund. Available at: https://www.england.nhs.uk/cancer/cdf/. Accessed 22 May 2020.

  107. Kang J, Cairns J. “Don’t think twice, It’s all right”: using additional data to reduce uncertainty regarding oncologic drugs provided through managed access agreements in England. PharmacoEconomics-Open. 2022;7:77–91.

  108. Chang J-YA, Chilcott JB, Latimer NR. Leveraging real-world data to assess treatment sequences in health economic evaluations: a study protocol for emulating target trials using the English Cancer Registry and US Electronic Health Records-Derived Database. HEDS Discussion Paper. 2024;24(01):1–61.

    Google Scholar 

  109. Franklin JM, Patorno E, Desai RJ, et al. Emulating randomized clinical trials with nonrandomized real-world evidence studies: first results from the RCT DUPLICATE initiative. Circulation. 2021;143:1002–13.

    Article  PubMed  Google Scholar 

  110. RCT Duplicate. RCT duplicate: randomized controlled trials duplicated using prospective longitudinal insurance claims: applying techniques of epidemiology. Available at: www.rctduplicate.org. Accessed 15 Jan 2021.

  111. Regan MM, Neven P, Giobbie-Hurder A, et al. Assessment of letrozole and tamoxifen alone and in sequence for postmenopausal women with steroid hormone receptor-positive breast cancer: the BIG 1–98 randomised clinical trial at 8·1 years median follow-up. Lancet Oncol. 2011;12:1101–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Eichelberg C, Vervenne WL, De Santis M, et al. SWITCH: A randomised, sequential, open-label study to evaluate the efficacy and safety of sorafenib-sunitinib versus sunitinib-sorafenib in the treatment of metastatic renal cell cancer. Eur Urol. 2015;68:837–47.

    Article  CAS  PubMed  Google Scholar 

  113. Retz M, Bedke J, Bogemann M, et al. SWITCH II: Phase III randomized, sequential, open-label study to evaluate the efficacy and safety of sorafenib-pazopanib versus pazopanib-sorafenib in the treatment of advanced or metastatic renal cell carcinoma (AUO AN 33/11). Eur J Cancer. 2019;107:37–45.

    Article  CAS  PubMed  Google Scholar 

  114. Lei H, Nahum-Shani I, Lynch K, et al. A “SMART” design for building individualized treatment sequences. Annu Rev Clin Psychol. 2012;8:21–48.

    Article  CAS  PubMed  Google Scholar 

  115. Bujkiewicz S, Singh J, Wheaton L, et al. Bridging disconnected networks of first and second lines of biologic therapies in rheumatoid arthritis with registry data: bayesian evidence synthesis with target trial emulation. J Clin Epidemiol. 2022;150:171–8.

    Article  PubMed  Google Scholar 

  116. Gomes M, Latimer N, Soares M, et al. Target trial emulation for transparent and robust estimation of treatment effects for health technology assessment using real-world data: opportunities and challenges. Pharmacoeconomics. 2022;40(6):577–86.

  117. Lunceford JK, Davidian M, Tsiatis AA. Estimation of survival distributions of treatment policies in two-stage randomization designs in clinical trials. Biometrics. 2002;58:48–57.

    Article  PubMed  Google Scholar 

  118. Robins J. Interview with Jamie Robins. Observational. Studies. 2022;8:37–76.

    Google Scholar 

  119. Zang Y, Lee JJ. Adaptive clinical trial designs in oncology. Chin Clin Oncoly. 2014;3(4):49.

  120. Pallmann P, Bedding AW, Choodari-Oskooei B, et al. Adaptive designs in clinical trials: why use them, and how to run and report them. BMC Med. 2018;16:29.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Mahajan R, Gupta K. Adaptive design clinical trials: Methodology, challenges and prospect. Indian J Pharmacol. 2010;42:201–7.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Thorlund K, Haggstrom J, Park JJ, et al. Key design considerations for adaptive clinical trials: a primer for clinicians. Br Med J. 2018;360:k698.

  123. Crown W, Buyukkaramikli N, Thokala P, et al. Constrained optimization methods in health services research—an introduction: report 1 of the ISPOR optimization methods emerging good practices task force. Value Health. 2017;20:310–9.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to express their gratitude to Professor Allan Wailoo and Professor Paul Tappenden from the University of Sheffield for their valuable feedback on the initial version of the work. The authors would also like to extend their appreciation to the Sheffield Centre for Health and Related Research (SCHARR) library service at the University of Sheffield for their guidance on review methodologies.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jen-Yu Amy Chang.

Ethics declarations

Funding

This study was supported by the Wellcome Trust [108903/B/15/Z]. The sponsor had no role in the study design, data collection, analysis, and decision to publish or preparation of the manuscript.

Conflicts of Interest

All authors have no conflicts of interest that are directly relevant to the content of this article.

Data Availability

Not applicable.

Ethics Approval

Not applicable.

Consent to Participate

Not applicable.

Consent for Publication

Not applicable.

Code Availability:

Not applicable.

Author Contributions

All authors contributed to the conceptualisation and design. J.Y.A.C. performed the literature search and selection, data extraction, evidence synthesis and drafted the original manuscript. J.C. and N.L. contributed insights for quality assessment and interpretation of results, revised the manuscript and provided supervision. All authors reviewed and approved the final manuscript.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chang, JY.A., Chilcott, J.B. & Latimer, N.R. Challenges and Opportunities in Interdisciplinary Research and Real-World Data for Treatment Sequences in Health Technology Assessments. PharmacoEconomics 42, 487–506 (2024). https://doi.org/10.1007/s40273-024-01363-1

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40273-024-01363-1

Navigation