Display options
Share it on

Health Technol Assess. 2021 Dec;25(76):1-228. doi: 10.3310/hta25760.

Modelling approaches for histology-independent cancer drugs to inform NICE appraisals: a systematic review and decision-framework.

Health technology assessment (Winchester, England)

Peter Murphy, David Glynn, Sofia Dias, Robert Hodgson, Lindsay Claxton, Lucy Beresford, Katy Cooper, Paul Tappenden, Kate Ennis, Alessandro Grosso, Kath Wright, Anna Cantrell, Matt Stevenson, Stephen Palmer

Affiliations

  1. Centre for Reviews and Dissemination, University of York, York, UK.
  2. Centre for Health Economics, University of York, York, UK.
  3. School of Health and Related Research (ScHARR) Technology Assessment Group, University of Sheffield, Sheffield, UK.

PMID: 34990339 DOI: 10.3310/hta25760

Abstract

BACKGROUND: The first histology-independent marketing authorisation in Europe was granted in 2019. This was the first time that a cancer treatment was approved based on a common biomarker rather than the location in the body at which the tumour originated. This research aims to explore the implications for National Institute for Health and Care Excellence appraisals.

METHODS: Targeted reviews were undertaken to determine the type of evidence that is likely to be available at the point of marketing authorisation and the analyses required to support National Institute for Health and Care Excellence appraisals. Several challenges were identified concerning the design and conduct of trials for histology-independent products, the greater levels of heterogeneity within the licensed population and the use of surrogate end points. We identified approaches to address these challenges by reviewing key statistical literature that focuses on the design and analysis of histology-independent trials and by undertaking a systematic review to evaluate the use of response end points as surrogate outcomes for survival end points. We developed a decision framework to help to inform approval and research policies for histology-independent products. The framework explored the uncertainties and risks associated with different approval policies, including the role of further data collection, pricing schemes and stratified decision-making.

RESULTS: We found that the potential for heterogeneity in treatment effects, across tumour types or other characteristics, is likely to be a central issue for National Institute for Health and Care Excellence appraisals. Bayesian hierarchical methods may serve as a useful vehicle to assess the level of heterogeneity across tumours and to estimate the pooled treatment effects for each tumour, which can inform whether or not the assumption of homogeneity is reasonable. Our review suggests that response end points may not be reliable surrogates for survival end points. However, a surrogate-based modelling approach, which captures all relevant uncertainty, may be preferable to the use of immature survival data. Several additional sources of heterogeneity were identified as presenting potential challenges to National Institute for Health and Care Excellence appraisal, including the cost of testing, baseline risk, quality of life and routine management costs. We concluded that a range of alternative approaches will be required to address different sources of heterogeneity to support National Institute for Health and Care Excellence appraisals. An exemplar case study was developed to illustrate the nature of the assessments that may be required.

CONCLUSIONS: Adequately designed and analysed basket studies that assess the homogeneity of outcomes and allow borrowing of information across baskets, where appropriate, are recommended. Where there is evidence of heterogeneity in treatment effects and estimates of cost-effectiveness, consideration should be given to optimised recommendations. Routine presentation of the scale of the consequences of heterogeneity and decision uncertainty may provide an important additional approach to the assessments specified in the current National Institute for Health and Care Excellence methods guide.

FURTHER RESEARCH: Further exploration of Bayesian hierarchical methods could help to inform decision-makers on whether or not there is sufficient evidence of homogeneity to support pooled analyses. Further research is also required to determine the appropriate basis for apportioning genomic testing costs where there are multiple targets and to address the challenges of uncontrolled Phase II studies, including the role and use of surrogate end points.

FUNDING: This project was funded by the National Institute for Health Research (NIHR) Evidence Synthesis programme and will be published in full in

Keywords: BAYES THEOREM; BIOMARKERS; COST–BENEFIT ANALYSIS; DATA COLLECTION; GENETIC TESTING; NEOPLASMS; POLICY; UNCERTAINTY

References

  1. US Food and Drug Administration (FDA), Center for Drug Evaluation and Research. Approval Package for Application Number 125514Orig1s014. Trade Name: Keytruda. Generic or Proper Name: Pembrolizumab. Rockville, MD: FDA; 2017. - PubMed
  2. European Medicines Agency (EMA). Assessment Report: VITRAKVI. International Non-Proprietary Name: Larotrectinib. Amsterdam: EMA; 2019. - PubMed
  3. Drilon A, Laetsch TW, Kummar S, DuBois SG, Lassen UN, Demetri GD, et al. Efficacy of Larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med 2018;378:731–9. https://doi.org/10.1056/NEJMoa1714448 - PubMed
  4. National Institute for Health and Care Excellence (NICE). Guide to the Methods of Technology Appraisal. London: NICE; 2013. - PubMed
  5. U.S. Food and Drug Administration (FDA), Center for Drug Evaluation and Research, Center for Biologics Evaluation and Research. Developing Targeted Therapies in Low-Frequency Molecular Subsets of a Disease. Guidance for Industry. Silver Spring, MD: FDA; 2018. - PubMed
  6. U.S. Food and Drug Administration (FDA), Center for Drug Evaluation and Research, Center for Biologics Evaluation and Research, Oncology Center of Excellence. Master Protocols: Efficient Clinical Trial Design Strategies to Expedite Development of Oncology Drugs and Biologics. Guidance for Industry. Silver Spring, MD: FDA; 2018. - PubMed
  7. Simon R. Optimal two-stage designs for phase II clinical trials. Control Clin Trials 1989;10:1–10. https://doi.org/10.1016/0197-2456(89)90015-9 - PubMed
  8. U.S. Food and Drug Administration (FDA), Center for Drug Evaluation and Research, Center for Biologics Evaluation and Research. Guidance for Industry Expedited Programs for Serious Conditions – Drugs and Biologics. Silver Spring, MD: FDA; 2014. - PubMed
  9. U.S. Food and Drug Administration (FDA). Drug Approval Package: Vitrakvi (Larotrectinib). Rockville, MD: FDA; 2018. URL: www.accessdata.fda.gov/drugsatfda_docs/nda/2018/210861Orig1s000_21171Orig1s000TOC.cfm (accessed 17 January 2020). - PubMed
  10. U.S. Food and Drug Administration (FDA), Center for Drug Evaluation and Research. Approval Package for Application Number 212725Orig1s000, 212726Orig1s000. Trade Name: ROZLYTREK® Capsules, 100 mg and 200 mg (Entrectinib). Rockville, MD: FDA; 2019. - PubMed
  11. Kim C, Prasad V. Strength of validation for surrogate end points used in the US Food and Drug Administration’s approval of oncology drugs. Mayo Clin Proc 2016;91:713–25. https://doi.org/10.1016/j.mayocp.2016.02.012 - PubMed
  12. Doebele RC, Drilon A, Paz-Ares L, Siena S, Shaw AT, Farago AF, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol 2020;21:271–82. [Erratum published in Lancet Oncol 2020;21:e70.] https://doi.org/10.1016/S1470-2045(19)30691-6 - PubMed
  13. European Medicines Agency (EMA). Workshop on Site and Histology - Independent Indications in Oncology. Amsterdam: EMA; 2017. URL: www.ema.europa.eu/en/events/workshop-site-histology-independent-indications-oncology (accessed 17 January 2020). - PubMed
  14. European Medicines Agency (EMA). Workshop on Single-arm Trials in Oncology. Amsterdam: EMA; 2016. URL: www.ema.europa.eu/en/events/workshop-single-arm-trials-oncology (accessed 17 January 2020). - PubMed
  15. European Medicines Agency (EMA). Guideline on the Evaluation of Anticancer Medicinal Products in Man. Amsterdam: EMA; 2017. - PubMed
  16. European Medicines Agency (EMA). Concept Paper on the Revision of the Guideline on the Evaluation of Anticancer Medicinal Products in Man. Amsterdam: EMA; 2018. - PubMed
  17. Laetsch TW, DuBois SG, Mascarenhas L, Turpin B, Federman N, Albert CM, et al. Larotrectinib for paediatric solid tumours harbouring NTRK fusions: phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol 2018;19:705–14. [Erratum published in Lancet Oncol 2018;19:e229.] https://doi.org/10.1016/S1470-2045(18)30119-0 - PubMed
  18. Gehan EA. The determination of the number of patients required in a preliminary and a follow-up trial of a new chemotherapeutic agent. J Chronic Dis 1961;13:346–53. https://doi.org/10.1016/0021-9681(61)90060-1 - PubMed
  19. Fleming TR. One-sample multiple testing procedure for phase II clinical trial. Biometrics 1982;38:143–51. https://doi.org/10.2307/2530297 - PubMed
  20. Lin Y, Shih WJ. Adaptive two-stage designs for single-arm phase IIA cancer clinical trials. Biometrics 2004;60:482–90. https://doi.org/10.1111/j.0006-341X.2004.00193.x - PubMed
  21. Herson J. Predictive probability early termination plans for phase II clinical trials. Biometrics 1979;35:775–83. https://doi.org/10.2307/2530109 - PubMed
  22. Thall PF, Simon R. Practical Bayesian guidelines for phase IIB clinical trials. Biometrics 1994;50:337–49. https://doi.org/10.2307/2533377 - PubMed
  23. Heitjan DF. Bayesian interim analysis of phase II cancer clinical trials. Stat Med 1997;16:1791–802. https://doi.org/10.1002/(SICI)1097-0258(19970830)16:16<1791::AID-SIM609>3.0.CO;2-E - PubMed
  24. Tan SB, Machin D. Bayesian two-stage designs for phase II clinical trials. Stat Med 2002;21:1991–2012. https://doi.org/10.1002/sim.1176 - PubMed
  25. Blagden SP, Billingham L, Brown LC, Buckland SW, Cooper AM, Ellis S, et al. Effective delivery of Complex Innovative Design (CID) cancer trials – a consensus statement. Br J Cancer 2020;122:473–82. https://doi.org/10.1038/s41416-019-0653-9 - PubMed
  26. Redig AJ, Jänne PA. Basket trials and the evolution of clinical trial design in an era of genomic medicine. J Clin Oncol 2015;33:975–7. https://doi.org/10.1200/JCO.2014.59.8433 - PubMed
  27. Renfro LA, Sargent DJ. Statistical controversies in clinical research: basket trials, umbrella trials, and other master protocols: a review and examples. Ann Oncol 2017;28:34–43. https://doi.org/10.1093/annonc/mdw413 - PubMed
  28. Hyman DM, Taylor BS, Baselga J. Implementing genome-driven oncology. Cell 2017;168:584–99. https://doi.org/10.1016/j.cell.2016.12.015 - PubMed
  29. Renfro LA, Mandrekar SJ. Definitions and statistical properties of master protocols for personalized medicine in oncology. J Biopharm Stat 2018;28:217–28. https://doi.org/10.1080/10543406.2017.1372778 - PubMed
  30. Simon R. New designs for basket clinical trials in oncology. J Biopharm Stat 2018;28:245–55. https://doi.org/10.1080/10543406.2017.1372779 - PubMed
  31. Lopez-Chavez A, Thomas A, Rajan A, Raffeld M, Morrow B, Kelly R, et al. Molecular profiling and targeted therapy for advanced thoracic malignancies: a biomarker-derived, multiarm, multihistology phase II basket trial. J Clin Oncol 2015;33:1000–7. https://doi.org/10.1200/JCO.2014.58.2007 - PubMed
  32. Hyman DM, Puzanov I, Subbiah V, Faris JE, Chau I, Blay JY, et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N Engl J Med 2015;373:726–36. https://doi.org/10.1056/NEJMoa1502309 - PubMed
  33. Le Tourneau C, Delord JP, Gonçalves A, Gavoille C, Dubot C, Isambert N, et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol 2015;16:1324–34. https://doi.org/10.1016/S1470-2045(15)00188-6 - PubMed
  34. Beckman RA, Antonijevic Z, Kalamegham R, Chen C. Adaptive design for a confirmatory basket trial in multiple tumor types based on a putative predictive biomarker. Clin Pharmacol Ther 2016;100:617–25. https://doi.org/10.1002/cpt.446 - PubMed
  35. Antonijevic Z, Beckman R. Platform Trial Designs in Drug Development: Umbrella Trials and Basket Trials. Boca Raton, FL: CRD Press; 2019. https://doi.org/10.1201/9781315167756 - PubMed
  36. Freidlin B, Korn EL. Borrowing information across subgroups in phase II trials: is it useful? Clin Cancer Res 2013;19:1326–34. https://doi.org/10.1158/1078-0432.CCR-12-1223 - PubMed
  37. Simon R. Critical review of umbrella, basket, and platform designs for oncology clinical trials. Clin Pharmacol Ther 2017;102:934–41. https://doi.org/10.1002/cpt.814 - PubMed
  38. Simon RM. Primary Site Independent Clinical Trials in Oncology. In Antonijevic Z, Beckman R, editors. Platform Trials in Drug Development: Umbrella Trials and Basket Trials. Boca Raton, FL: CRC Press; 2019. pp. 167–80. https://doi.org/10.1201/9781315167756-12 - PubMed
  39. London WB, Chang MN. One- and two-stage designs for stratified phase II clinical trials. Stat Med 2005;24:2597–611. https://doi.org/10.1002/sim.2139 - PubMed
  40. Leblanc M, Rankin C, Crowley J. Multiple histology phase II trials. Clin Cancer Res 2009;15:4256–62. https://doi.org/10.1158/1078-0432.CCR-08-2069 - PubMed
  41. Jung SH, Chang MN, Kang SJ. Phase II cancer clinical trials with heterogeneous patient populations. J Biopharm Stat 2012;22:312–28. https://doi.org/10.1080/10543406.2010.536873 - PubMed
  42. Cunanan KM, Iasonos A, Shen R, Begg CB, Gönen M. An efficient basket trial design. Stat Med 2017;36:1568–79. https://doi.org/10.1002/sim.7227 - PubMed
  43. Simon R, Geyer S, Subramanian J, Roychowdhury S. The Bayesian basket design for genomic variant-driven phase II trials. Semin Oncol 2016;43:13–18. https://doi.org/10.1053/j.seminoncol.2016.01.002 - PubMed
  44. Palmer AC, Plana D, Sorger PK. Comparing the efficacy of cancer therapies between subgroups in basket trials. Cell Syst 2020;11:449–60.E2. https://doi.org/10.1101/401620 - PubMed
  45. Spiegelhalter DJ, Abrams KR, Myles J. Bayesian Approaches to Clinical Trials and Health-Care Evaluation. New York, NY: Wiley; 2004. https://doi.org/10.1002/0470092602 - PubMed
  46. Thall PF, Wathen JK, Bekele BN, Champlin RE, Baker LH, Benjamin RS. Hierarchical Bayesian approaches to phase II trials in diseases with multiple subtypes. Stat Med 2003;22:763–80. https://doi.org/10.1002/sim.1399 - PubMed
  47. Thall PF, Sung HG. Some extensions and applications of a Bayesian strategy for monitoring multiple outcomes in clinical trials. Stat Med 1998;17:1563–80. https://doi.org/10.1002/(SICI)1097-0258(19980730)17:14%3C1563::AID-SIM873%3E3.0.CO;2-L - PubMed
  48. Chugh R, Wathen JK, Maki RG, Benjamin RS, Patel SR, Meyers PA, et al. Phase II multicenter trial of imatinib in 10 histologic subtypes of sarcoma using a Bayesian hierarchical statistical model. J Clin Oncol 2009;27:3148–53. https://doi.org/10.1200/JCO.2008.20.5054 - PubMed
  49. Berry SM, Broglio KR, Groshen S, Berry DA. Bayesian hierarchical modeling of patient subpopulations: efficient designs of Phase II oncology clinical trials. Clin Trials 2013;10:720–34. https://doi.org/10.1177/1740774513497539 - PubMed
  50. Liu R, Liu Z, Ghadessi M, Vonk R. Increasing the efficiency of oncology basket trials using a Bayesian approach. Contemp Clin Trials 2017;63:67–72. https://doi.org/10.1016/j.cct.2017.06.009 - PubMed
  51. Neuenschwander B, Wandel S, Roychoudhury S, Bailey S. Robust exchangeability designs for early phase clinical trials with multiple strata. Pharm Stat 2016;15:123–34. https://doi.org/10.1002/pst.1730 - PubMed
  52. Cunanan KM, Iasonos A, Shen R, Gönen M. Variance prior specification for a basket trial design using Bayesian hierarchical modeling. Clin Trials 2019;16:142–53. https://doi.org/10.1177/1740774518812779 - PubMed
  53. Leon-Novelo LG, Bekele BN, Müller P, Quintana F, Wathen K. Borrowing strength with nonexchangeable priors over subpopulations. Biometrics 2012;68:550–8. https://doi.org/10.1111/j.1541-0420.2011.01693.x - PubMed
  54. Chu Y, Yuan Y. BLAST: Bayesian latent subgroup design for basket trials accounting for patient heterogeneity. J R Stat Soc Ser C Appl Stat 2018;67:723–40. https://doi.org/10.1111/rssc.12255 - PubMed
  55. Chu Y, Yuan Y. A Bayesian basket trial design using a calibrated Bayesian hierarchical model. Clin Trials 2018;15:149–58. https://doi.org/10.1177/1740774518755122 - PubMed
  56. Fujikawa K, Teramukai S, Yokota I, Daimon T. A Bayesian basket trial design that borrows information across strata based on the similarity between the posterior distributions of the response probability. Biom J 2020;62:330–8. https://doi.org/10.1002/bimj.201800404 - PubMed
  57. Hobbs BP, Landin R. Bayesian basket trial design with exchangeability monitoring. Stat Med 2018;37:3557–72. https://doi.org/10.1002/sim.7893 - PubMed
  58. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med 2010;363:809–19. https://doi.org/10.1056/NEJMoa1002011 - PubMed
  59. Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 2012;483:100–3. https://doi.org/10.1038/nature10868 - PubMed
  60. Heinrich MC, Joensuu H, Demetri GD, Corless CL, Apperley J, Fletcher JA, et al. Phase II, open-label study evaluating the activity of imatinib in treating life-threatening malignancies known to be associated with imatinib-sensitive tyrosine kinases. Clin Cancer Res 2008;14:2717–25. https://doi.org/10.1158/1078-0432.CCR-07-4575 - PubMed
  61. Hudis CA. Trastuzumab – mechanism of action and use in clinical practice. N Engl J Med 2007;357:39–51. https://doi.org/10.1056/NEJMra043186 - PubMed
  62. Fleming GF, Sill MW, Darcy KM, McMeekin DS, Thigpen JT, Adler LM, et al. Phase II trial of trastuzumab in women with advanced or recurrent, HER2-positive endometrial carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol 2010;116:15–20. https://doi.org/10.1016/j.ygyno.2009.09.025 - PubMed
  63. Gatzemeier U, Groth G, Butts C, van Zandwijk N, Shepherd F, Ardizzoni A, et al. Randomized phase II trial of gemcitabine-cisplatin with or without trastuzumab in HER2-positive non-small-cell lung cancer. Ann Oncol 2004;15:19–27. https://doi.org/10.1093/annonc/mdh031 - PubMed
  64. Cooper K, Tappenden P, Cantrell A, Ennis K. A systematic review of meta-analyses assessing the validity of tumour response endpoints as surrogates for progression-free or overall survival in cancer. Br J Cancer 2020;123:1686–96. https://doi.org/10.1038/s41416-020-01050-w - PubMed
  65. Fleming TR, Powers JH. Biomarkers and surrogate endpoints in clinical trials. Stat Med 2012;31:2973–84. https://doi.org/10.1002/sim.5403 - PubMed
  66. Taylor RS, Elston J. The use of surrogate outcomes in model-based cost-effectiveness analyses: a survey of UK Health Technology Assessment reports. Health Technol Assess 2009;13(8). https://doi.org/10.3310/hta13080 - PubMed
  67. Fleming TR, DeMets DL. Surrogate end points in clinical trials: are we being misled? Ann Intern Med 1996;125:605–13. https://doi.org/10.7326/0003-4819-125-7-199610010-00011 - PubMed
  68. Prentice RL. Surrogate endpoints in clinical trials: definition and operational criteria. Stat Med 1989;8:431–40. https://doi.org/10.1002/sim.4780080407 - PubMed
  69. Buyse M, Molenberghs G, Burzykowski T, Renard D, Geys H. The validation of surrogate endpoints in meta-analyses of randomized experiments. Biostatistics 2000;1:49–67. https://doi.org/10.1093/biostatistics/1.1.49 - PubMed
  70. Heller G. Statistical controversies in clinical research: an initial evaluation of a surrogate end point using a single randomized clinical trial and the Prentice criteria. Ann Oncol 2015;26:2012–16. https://doi.org/10.1093/annonc/mdv333 - PubMed
  71. Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 2001;69:89–95. https://doi.org/10.1067/mcp.2001.113989 - PubMed
  72. Bucher HC, Guyatt GH, Cook DJ, Holbrook A, McAlister FA. Users’ guides to the medical literature: XIX. Applying clinical trial results. A. How to use an article measuring the effect of an intervention on surrogate end points. JAMA 1999;282:771–8. https://doi.org/10.1001/jama.282.8.771 - PubMed
  73. Elston J, Taylor RS. Use of surrogate outcomes in cost-effectiveness models: a review of United Kingdom health technology assessment reports. Int J Technol Assess Health Care 2009;25:6–13. https://doi.org/10.1017/S0266462309090023 - PubMed
  74. German Institute of Quality and Efficiency in Health Care (IQWiG). Validity of Surrogate Endpoints in Oncology. Cologne: IQWiG; 2011. - PubMed
  75. Lassere MN, Johnson KR, Schiff M, Rees D. Is blood pressure reduction a valid surrogate endpoint for stroke prevention? An analysis incorporating a systematic review of randomised controlled trials, a by-trial weighted errors-in-variables regression, the surrogate threshold effect (STE) and the Biomarker-Surrogacy (BioSurrogate) Evaluation Schema (BSES). BMC Med Res Methodol 2012;12:27. https://doi.org/10.1186/1471-2288-12-27 - PubMed
  76. Buyse M, Molenberghs G, Paoletti X, Oba K, Alonso A, van der Elst W, Burzykowski T. Statistical evaluation of surrogate endpoints with examples from cancer clinical trials. Biom J 2016;58:104–32. https://doi.org/10.1002/bimj.201400049 - PubMed
  77. Fischer A, Hernandez-Villafuerte K, Latimer N, Henshall C. Extrapolation from Progression-Free Survival to Overall Survival in Oncology. London: Office of Health Economics; 2016. - PubMed
  78. Davis S, Tappenden P, Cantrell A. A Review of Studies Examining the Relationship Between Progression-free Survival and Overall Survival in Advanced or Metastatic Cancer. Sheffield: University of Sheffield Report for NICE Decision Support Unit; 2012. https://doi.org/10.1016/S0084-3873(12)00197-6 - PubMed
  79. Savina M, Gourgou S, Italiano A, Dinart D, Rondeau V, Penel N, et al. Meta-analyses evaluating surrogate endpoints for overall survival in cancer randomized trials: a critical review. Crit Rev Oncol Hematol 2018;123:21–41. https://doi.org/10.1016/j.critrevonc.2017.11.014 - PubMed
  80. Haslam A, Hey SP, Gill J, Prasad V. A systematic review of trial-level meta-analyses measuring the strength of association between surrogate end-points and overall survival in oncology. Eur J Cancer 2019;106:196–211. https://doi.org/10.1016/j.ejca.2018.11.012 - PubMed
  81. Abdel-Rahman O. Surrogate end points for overall survival in trials of PD-(L)1 inhibitors for urinary cancers: a systematic review. Immunotherapy 2018;10:139–48. https://doi.org/10.2217/imt-2017-0115 - PubMed
  82. Agarwal N, Bellmunt J, Maughan BL, Boucher KM, Choueiri TK, Qu AQ, et al. Six-month progression-free survival as the primary endpoint to evaluate the activity of new agents as second-line therapy for advanced urothelial carcinoma. Clin Genitourin Cancer 2014;12:130–7. https://doi.org/10.1016/j.clgc.2013.09.002 - PubMed
  83. Agarwal SK, Mangal N, Menon RM, Freise KJ, Salem AH. Response rates as predictors of overall survival: a meta-analysis of acute myeloid leukemia trials. J Cancer 2017;8:1562–7. https://doi.org/10.7150/jca.18686 - PubMed
  84. Blumenthal GM, Karuri SW, Zhang H, Zhang L, Khozin S, Kazandjian D, et al. Overall response rate, progression-free survival, and overall survival with targeted and standard therapies in advanced non-small-cell lung cancer: US Food and Drug Administration trial-level and patient-level analyses. J Clin Oncol 2015;33:1008–14. https://doi.org/10.1200/JCO.2014.59.0489 - PubMed
  85. Blumenthal GM, Zhang L, Zhang H, Kazandjian D, Khozin S, Tang S, et al. Milestone analyses of immune checkpoint inhibitors, targeted therapy, and conventional therapy in metastatic non-small cell lung cancer trials: a meta-analysis. JAMA Oncol 2017;3:e171029. https://doi.org/10.1001/jamaoncol.2017.1029 - PubMed
  86. Bruzzi P, Del Mastro L, Sormani MP, Bastholt L, Danova M, Focan C, et al. Objective response to chemotherapy as a potential surrogate end point of survival in metastatic breast cancer patients. J Clin Oncol 2005;23:5117–25. https://doi.org/10.1200/JCO.2005.02.106 - PubMed
  87. Burzykowski T, Buyse M, Piccart-Gebhart MJ, Sledge G, Carmichael J, Lück HJ, et al. Evaluation of tumor response, disease control, progression-free survival, and time to progression as potential surrogate end points in metastatic breast cancer. J Clin Oncol 2008;26:1987–92. https://doi.org/10.1200/JCO.2007.10.8407 - PubMed
  88. Buyse M, Thirion P, Carlson RW, Burzykowski T, Molenberghs G, Piedbois P. Relation between tumour response to first-line chemotherapy and survival in advanced colorectal cancer: a meta-analysis. Meta-Analysis Group in Cancer. Lancet 2000;356:373–8. https://doi.org/10.1016/S0140-6736(00)02528-9 - PubMed
  89. Ciani O, Buyse M, Garside R, Peters J, Saad ED, Stein K, Taylor RS. Meta-analyses of randomized controlled trials show suboptimal validity of surrogate outcomes for overall survival in advanced colorectal cancer. J Clin Epidemiol 2015;68:833–42. https://doi.org/10.1016/j.jclinepi.2015.02.016 - PubMed
  90. Colloca G, Venturino A. Trial-level analysis of progression-free survival and response rate as end points of trials of first-line chemotherapy in advanced ovarian cancer. Med Oncol 2017;34:87. https://doi.org/10.1007/s12032-017-0939-9 - PubMed
  91. Colloca G, Venturino A, Guarneri D. Analysis of response-related and time-to-event endpoints in randomized trials of gemcitabine-based treatment versus gemcitabine alone as first-line treatment of patients with advanced pancreatic cancer. Clin Colorectal Cancer 2016;15:264–76. https://doi.org/10.1016/j.clcc.2015.11.006 - PubMed
  92. Colloca G, Venturino A, Guarneri D. Analysis of clinical end points of randomised trials including bevacizumab and chemotherapy versus chemotherapy as first-line treatment of metastatic colorectal cancer. Clin Oncol 2016;28:e155–64. https://doi.org/10.1016/j.clon.2016.05.001 - PubMed
  93. Colloca G, Vitucci P, Venturino A. Trial level analysis of prostate-specific antigen-related versus unrelated endpoints in phase III trials of first-line and second-line medical treatments of patients with metastatic castration-resistant prostate cancer. Clin Genitourin Cancer 2016;14:389–97. https://doi.org/10.1016/j.clgc.2016.03.022 - PubMed
  94. Cremolini C, Antoniotti C, Pietrantonio F, Berenato R, Tampellini M, Baratelli C, et al. Surrogate endpoints in second-line trials of targeted agents in metastatic colorectal cancer: a literature-based systematic review and meta-analysis. Cancer Res Treat 2017;49:834–45. https://doi.org/10.4143/crt.2016.249 - PubMed
  95. Delea TE, Khuu A, Heng DY, Haas T, Soulières D. Association between treatment effects on disease progression end points and overall survival in clinical studies of patients with metastatic renal cell carcinoma. Br J Cancer 2012;107:1059–68. https://doi.org/10.1038/bjc.2012.367 - PubMed
  96. Elia EG, Städler N, Ciani O, Taylor RS, Bujkiewicz S. Combining tumour response and progression free survival as surrogate endpoints for overall survival in advanced colorectal cancer. Cancer Epidemiol 2020;64:101665. https://doi.org/10.1016/j.canep.2019.101665 - PubMed
  97. Foster NR, Qi Y, Shi Q, Krook JE, Kugler JW, Jett JR, et al. Tumor response and progression-free survival as potential surrogate endpoints for overall survival in extensive stage small-cell lung cancer: findings on the basis of North Central Cancer Treatment Group trials. Cancer 2011;117:1262–71. https://doi.org/10.1002/cncr.25526 - PubMed
  98. Giessen C, Laubender RP, Ankerst DP, Stintzing S, Modest DP, Schulz C, et al. Surrogate endpoints in second-line treatment for mCRC: a systematic literature-based analysis from 23 randomised trials. Acta Oncol 2015;54:187–93. https://doi.org/10.3109/0284186X.2014.938830 - PubMed
  99. Hackshaw A, Knight A, Barrett-Lee P, Leonard R. Surrogate markers and survival in women receiving first-line combination anthracycline chemotherapy for advanced breast cancer. Br J Cancer 2005;93:1215–21. https://doi.org/10.1038/sj.bjc.6602858 - PubMed
  100. Hamada T, Nakai Y, Isayama H, Yasunaga H, Matsui H, Takahara N, et al. Progression-free survival as a surrogate for overall survival in first-line chemotherapy for advanced pancreatic cancer. Eur J Cancer 2016;65:11–20. https://doi.org/10.1016/j.ejca.2016.05.016 - PubMed
  101. Han K, Ren M, Wick W, Abrey L, Das A, Jin J, Reardon DA. Progression-free survival as a surrogate endpoint for overall survival in glioblastoma: a literature-based meta-analysis from 91 trials. Neuro Oncol 2014;16:696–706. https://doi.org/10.1093/neuonc/not236 - PubMed
  102. Hashim M, Pfeiffer BM, Bartsch R, Postma M, Heeg B. Do surrogate endpoints better correlate with overall survival in studies that did not allow for crossover or reported balanced postprogression treatments? An application in advanced non-small cell lung cancer. Value Health 2018;21:9–17. https://doi.org/10.1016/j.jval.2017.07.011 - PubMed
  103. Hotta K, Kato Y, Leighl N, Takigawa N, Gaafar RM, Kayatani H, et al. Magnitude of the benefit of progression-free survival as a potential surrogate marker in phase 3 trials assessing targeted agents in molecularly selected patients with advanced non-small cell lung cancer: systematic review. PLOS ONE 2015;10:e0121211. https://doi.org/10.1371/journal.pone.0121211 - PubMed
  104. Hotta K, Kiura K, Fujiwara Y, Takigawa N, Oze I, Ochi N, et al. Association between incremental gains in the objective response rate and survival improvement in phase III trials of first-line chemotherapy for extensive disease small-cell lung cancer. Ann Oncol 2009;20:829–34. https://doi.org/10.1093/annonc/mdp020 - PubMed
  105. Ichikawa W, Sasaki Y. Correlation between tumor response to first-line chemotherapy and prognosis in advanced gastric cancer patients. Ann Oncol 2006;17:1665–72. https://doi.org/10.1093/annonc/mdl174 - PubMed
  106. Imaoka H, Sasaki M, Takahashi H, Hashimoto Y, Ohno I, Mitsunaga S, et al. Progression-free survival as a surrogate endpoint in advanced neuroendocrine neoplasms. Endocr Relat Cancer 2017;24:475–83. https://doi.org/10.1530/ERC-17-0197 - PubMed
  107. Imaoka H, Sasaki M, Takahashi H, Hashimoto Y, Ohno I, Mitsunaga S, et al. Alternate endpoints for phase II trials in advanced neuroendocrine tumors. Oncologist 2019;24:47–53. https://doi.org/10.1634/theoncologist.2017-0651 - PubMed
  108. Iezzi R, Pompili M, Rinninella E, Annicchiarico E, Garcovich M, Cerrito L, et al. TACE with degradable starch microspheres (DSM-TACE) as second-line treatment in HCC patients dismissing or ineligible for sorafenib. Eur Radiol 2019;29:1285–92. https://doi.org/10.1007/s00330-018-5692-8 - PubMed
  109. Johnson KR, Ringland C, Stokes BJ, Anthony DM, Freemantle N, Irs A, et al. Response rate or time to progression as predictors of survival in trials of metastatic colorectal cancer or non-small-cell lung cancer: a meta-analysis. Lancet Oncol 2006;7:741–6. https://doi.org/10.1016/S1470-2045(06)70800-2 - PubMed
  110. Kaufman HL, Schwartz LH, William WN, Sznol M, Fahrbach K, Xu Y, et al. Evaluation of classical clinical endpoints as surrogates for overall survival in patients treated with immune checkpoint blockers: a systematic review and meta-analysis. J Cancer Res Clin Oncol 2018;144:2245–61. https://doi.org/10.1007/s00432-018-2738-x - PubMed
  111. Lee L, Wang L, Crump M. Identification of potential surrogate end points in randomized clinical trials of aggressive and indolent non-Hodgkin’s lymphoma: correlation of complete response, time-to-event and overall survival end points. Ann Oncol 2011;22:1392–403. https://doi.org/10.1093/annonc/mdq615 - PubMed
  112. Li J, He Q, Yu X, Khan K, Weng X, Guan M. Complete response associated with immune checkpoint inhibitors in advanced non-small-cell lung cancer: a meta-analysis of nine randomized controlled trials. Cancer Manag Res 2019;11:1623–9. https://doi.org/10.2147/CMAR.S188551 - PubMed
  113. Li X, Liu S, Gu H, Wang D. Surrogate end points for survival in the target treatment of advanced non-small-cell lung cancer with gefitinib or erlotinib. J Cancer Res Clin Oncol 2012;138:1963–9. https://doi.org/10.1007/s00432-012-1278-z - PubMed
  114. Liu L, Chen F, Zhao J, Yu H. Correlation between overall survival and other endpoints in metastatic breast cancer with second- or third-line chemotherapy: literature-based analysis of 24 randomized trials. Bull Cancer 2016;103:336–44. https://doi.org/10.1016/j.bulcan.2016.01.002 - PubMed
  115. Louvet C, de Gramont A, Tournigand C, Artru P, Maindrault-Goebel F, Krulik M. Correlation between progression free survival and response rate in patients with metastatic colorectal carcinoma. Cancer 2001;91:2033–8. https://doi.org/10.1002/1097-0142(20010601)91:11<2033::AID-CNCR1229>3.0.CO;2-J - PubMed
  116. Makris EA, MacBarb R, Harvey DJ, Poultsides GA. Surrogate end points for overall survival in metastatic, locally advanced, or unresectable pancreatic cancer: a systematic review and meta-analysis of 24 randomized controlled trials. Ann Surg Oncol 2017;24:2371–8. https://doi.org/10.1245/s10434-017-5826-2 - PubMed
  117. Mangal N, Salem AH, Li M, Menon R, Freise KJ. Relationship between response rates and median progression-free survival in non-Hodgkin’s lymphoma: a meta-analysis of published clinical trials. Hematol Oncol 2018;36:37–43. https://doi.org/10.1002/hon.2463 - PubMed
  118. Mangal N, Salem AH, Menon RM, Freise KJ. Use of depth of response to predict progression-free survival in relapsed or refractory multiple myeloma: evaluation of results from 102 clinical trials. Hematol Oncol 2018;36:547–53. https://doi.org/10.1002/hon.2514 - PubMed
  119. Moriwaki T, Yamamoto Y, Gosho M, Kobayashi M, Sugaya A, Yamada T, et al. Correlations of survival with progression-free survival, response rate, and disease control rate in advanced biliary tract cancer: a meta-analysis of randomised trials of first-line chemotherapy. Br J Cancer 2016;114:881–8. https://doi.org/10.1038/bjc.2016.83 - PubMed
  120. Mushti SL, Mulkey F, Sridhara R. Evaluation of overall response rate and progression-free survival as potential surrogate endpoints for overall survival in immunotherapy trials. Clin Cancer Res 2018;24:2268–75. https://doi.org/10.1158/1078-0432.CCR-17-1902 - PubMed
  121. Nakashima K, Horita N, Nagai K, Manabe S, Murakami S, Ota E, Kaneko T. Progression-free survival, response rate, and disease control rate as predictors of overall survival in phase III randomized controlled trials evaluating the first-line chemotherapy for advanced, locally advanced, and recurrent non-small cell lung carcinoma. J Thorac Oncol 2016;11:1574–85. https://doi.org/10.1016/j.jtho.2016.04.025 - PubMed
  122. Nickolich M, Babakoohi S, Fu P, Dowlati A. Clinical trial design in small cell lung cancer: surrogate end points and statistical evolution. Clin Lung Cancer 2014;15:207–12. https://doi.org/10.1016/j.cllc.2013.12.001 - PubMed
  123. Nie RC, Chen FP, Yuan SQ, Luo YS, Chen S, Chen YM, et al. Evaluation of objective response, disease control and progression-free survival as surrogate end-points for overall survival in anti-programmed death-1 and anti-programmed death ligand 1 trials. Eur J Cancer 2019;106:1–11. https://doi.org/10.1016/j.ejca.2018.10.011 - PubMed
  124. Pang Y, Shen Z, Sun J, Wang W. Does the use of targeted agents in advanced gastroesophageal cancer increase complete response? A meta-analysis of 18 randomized controlled trials. Cancer Manag Res 2018;10:5505–14. https://doi.org/10.2147/CMAR.S174063 - PubMed
  125. Penel N, Ryckewaert T, Kramar A. What is an active regimen in carcinoma of unknown primary sites? Analysis of correlation between activity endpoints reported in phase II trials. Correlation of activity endpoints in phase II trials. Bull Cancer 2014;101:E19–24. https://doi.org/10.1684/bdc.2014.1934 - PubMed
  126. Petrelli F, Barni S. Surrogate end points and postprogression survival in renal cell carcinoma: an analysis of first-line trials with targeted therapies. Clin Genitourin Cancer 2013;11:385–9. https://doi.org/10.1016/j.clgc.2013.07.012 - PubMed
  127. Petrelli F, Barni S. Surrogate endpoints in metastatic breast cancer treated with targeted therapies: an analysis of the first-line phase III trials. Med Oncol 2014;31:776. https://doi.org/10.1007/s12032-013-0776-4 - PubMed
  128. Ritchie G, Gasper H, Man J, Lord S, Marschner I, Friedlander M, Lee CK. Defining the most appropriate primary end point in phase 2 trials of immune checkpoint inhibitors for advanced solid cancers: a systematic review and meta-analysis. JAMA Oncol 2018;4:522–8. https://doi.org/10.1001/jamaoncol.2017.5236 - PubMed
  129. Rose PG, Tian C, Bookman MA. Assessment of tumor response as a surrogate endpoint of survival in recurrent/platinum-resistant ovarian carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol 2010;117:324–9. https://doi.org/10.1016/j.ygyno.2010.01.040 - PubMed
  130. Roviello G, Andre F, Venturini S, Pistilli B, Curigliano G, Cristofanilli M, et al. Response rate as a potential surrogate for survival and efficacy in patients treated with novel immune checkpoint inhibitors: a meta-regression of randomised prospective studies. Eur J Cancer 2017;86:257–65. https://doi.org/10.1016/j.ejca.2017.09.018 - PubMed
  131. Sekine I, Tamura T, Kunitoh H, Kubota K, Shinkai T, Kamiya Y, Saijo N. Progressive disease rate as a surrogate endpoint of phase II trials for non-small-cell lung cancer. Ann Oncol 1999;10:731–3. https://doi.org/10.1023/a:1008303921033 - PubMed
  132. Shi Q, Flowers CR, Hiddemann W, Marcus R, Herold M, Hagenbeek A, et al. Thirty-month complete response as a surrogate end point in first-line follicular lymphoma therapy: an individual patient-level analysis of multiple randomized trials. J Clin Oncol 2017;35:552–60. https://doi.org/10.1200/JCO.2016.70.8651 - PubMed
  133. Shitara K, Matsuo K, Muro K, Doi T, Ohtsu A. Correlation between overall survival and other endpoints in clinical trials of second-line chemotherapy for patients with advanced gastric cancer. Gastric Cancer 2014;17:362–70. https://doi.org/10.1007/s10120-013-0274-6 - PubMed
  134. Shukuya T, Mori K, Amann JM, Bertino EM, Otterson GA, Shields PG, et al. Relationship between overall survival and response or progression-free survival in advanced non-small cell lung cancer patients treated with anti-PD-1/PD-L1 antibodies. J Thorac Oncol 2016;11:1927–39. https://doi.org/10.1016/j.jtho.2016.07.017 - PubMed
  135. Siddiqui MK, Tyczynski J, Pahwa A, Fernandes AW. Objective response rate is a possible surrogate endpoint for survival in patients with advanced, recurrent ovarian cancer. Gynecol Oncol 2017;146:44–51. https://doi.org/10.1016/j.ygyno.2017.03.515 - PubMed
  136. Sidhu R, Rong A, Dahlberg S. Evaluation of progression-free survival as a surrogate endpoint for survival in chemotherapy and targeted agent metastatic colorectal cancer trials. Clin Cancer Res 2013;19:969–76. https://doi.org/10.1158/1078-0432.CCR-12-2502 - PubMed
  137. Tanaka K, Kawano M, Iwasaki T, Itonaga I, Tsumura H. Surrogacy of intermediate endpoints for overall survival in randomized controlled trials of first-line treatment for advanced soft tissue sarcoma in the pre- and post-pazopanib era: a meta-analytic evaluation. BMC Cancer 2019;19:56. https://doi.org/10.1186/s12885-019-5268-2 - PubMed
  138. Tang PA, Bentzen SM, Chen EX, Siu LL. Surrogate end points for median overall survival in metastatic colorectal cancer: literature-based analysis from 39 randomized controlled trials of first-line chemotherapy. J Clin Oncol 2007;25:4562–8. https://doi.org/10.1200/JCO.2006.08.1935 - PubMed
  139. Tsujino K, Kawaguchi T, Kubo A, Aono N, Nakao K, Koh Y, et al. Response rate is associated with prolonged survival in patients with advanced non-small cell lung cancer treated with gefitinib or erlotinib. J Thorac Oncol 2009;4:994–1001. https://doi.org/10.1097/JTO.0b013e3181a94a2f - PubMed
  140. Tsujino K, Shiraishi J, Tsuji T, Kurata T, Kawaguchi T, Kubo A, et al. Is response rate increment obtained by molecular targeted agents related to survival benefit in the phase III trials of advanced cancer? Ann Oncol 2010;21:1668–74. - PubMed
  141. Vidaurre T, Wilkerson J, Simon R, Bates SE, Fojo T. Stable disease is not preferentially observed with targeted therapies and as currently defined has limited value in drug development. Cancer J 2009;15:366–73. https://doi.org/10.1097/PPO.0b013e3181b9d37b - PubMed
  142. Wilkerson J, Fojo T. Progression-free survival is simply a measure of a drug’s effect while administered and is not a surrogate for overall survival. Cancer J 2009;15:379–85. https://doi.org/10.1097/PPO.0b013e3181bef8cd - PubMed
  143. Zer A, Prince RM, Amir E, Abdul Razak A. Evolution of randomized trials in advanced/metastatic soft tissue sarcoma: end point selection, surrogacy, and quality of reporting. J Clin Oncol 2016;34:1469–75. https://doi.org/10.1200/JCO.2015.64.3437 - PubMed
  144. Zhu R, Lu D, Chu YW, Chai A, Green M, Zhang N, Jin JY. Assessment of correlation between early and late efficacy endpoints to identify potential surrogacy relationships in non-Hodgkin lymphoma: a literature-based meta-analysis of 108 phase II and phase III studies. AAPS J 2017;19:669–81. https://doi.org/10.1208/s12248-017-0056-x - PubMed
  145. Ito K, Miura S, Sakaguchi T, Murotani K, Horita N, Akamatsu H, et al. The impact of high PD-L1 expression on the surrogate endpoints and clinical outcomes of anti-PD-1/PD-L1 antibodies in non-small cell lung cancer. Lung Cancer 2019;128:113–19. https://doi.org/10.1016/j.lungcan.2018.12.023 - PubMed
  146. Bujkiewicz S, Achana F, Papanikos T, Riley RD, Abrams KR. NICE DSU Technical Support Document 20: Multivariate Meta-analysis of Summary Data for Combining Treatment Effects on Correlated Outcomes and Evaluating Surrogate Endpoints. Report by The Decision Support Unit. Sheffield: Decision Support Unit, ScHARR, University of Sheffield; 2019. - PubMed
  147. Bujkiewicz S, Jackson D, Thompson JR, Turner RM, Städler N, Abrams KR, White IR. Bivariate network meta-analysis for surrogate endpoint evaluation. Stat Med 2019;38:3322–41. https://doi.org/10.1002/sim.8187 - PubMed
  148. Murphy P, Claxton L, Hodgson R, Glynn D, Beresford L, Walton M, et al. Exploring heterogeneity in histology-independent technologies and the implications for cost-effectiveness. Med Decis Making 2021;41:165–78. https://doi.org/10.1177/0272989X20980327 - PubMed
  149. Phelps CE, Mushlin AI. Focusing technology assessment using medical decision theory. Med Decis Making 1988;8:279–89. https://doi.org/10.1177/0272989X8800800409 - PubMed
  150. Coyle D, Buxton MJ, O’Brien BJ. Stratified cost-effectiveness analysis: a framework for establishing efficient limited use criteria. Health Econ 2003;12:421–7. https://doi.org/10.1002/hec.788 - PubMed
  151. Espinoza MA, Manca A, Claxton K, Sculpher MJ. The value of heterogeneity for cost-effectiveness subgroup analysis: conceptual framework and application. Med Decis Making 2014;34:951–64. https://doi.org/10.1177/0272989X14538705 - PubMed
  152. Basu A, Meltzer D. Value of information on preference heterogeneity and individualized care. Med Decis Making 2007;27:112–27. https://doi.org/10.1177/0272989X06297393 - PubMed
  153. Cancer Research UK. Cancer Research UK Policy Statement: Patient Access to Molecular Diagnostics and Targeted Medicines in England. London: Cancer Research UK; 2018. - PubMed
  154. Lunn D, Jackson C, Best N, Thomas A, Spiegelhalter D. The BUGS Book. Boca Raton, FL: CRC Press; 2013. https://doi.org/10.1201/b13613 - PubMed
  155. Sturtz S, Ligges U, Gelman A. R2WinBUGS: a package for running WinBUGS from R. J Stat Softw 2005;12:1–16. URL: www.jstatsoft.org/v2/i03 - PubMed
  156. Brooks SP, Gelman A. General methods for monitoring convergence of iterative simulations. J Comput Graph Stat 1998;7:434–55. https://doi.org/10.1080/10618600.1998.10474787 - PubMed
  157. Gelman A, Rubin DB. Inferences from iterative simulation using multiple sequences. Stat Sci 1992;7:457–72. https://doi.org/10.1214/ss/1177011136 - PubMed
  158. National Institute for Health and Care Excellence (NICE). Larotrectinib for Treating NTRK Fusion-positive Advanced Solid Tumours. Technology Appraisal Guidance [TA630]. London: NICE; 2019. - PubMed
  159. Bongarzone I, Vigneri P, Mariani L, Collini P, Pilotti S, Pierotti MA. RET/NTRK1 rearrangements in thyroid gland tumors of the papillary carcinoma family: correlation with clinicopathological features. Clin Cancer Res 1998;4:223–8. - PubMed
  160. Prasad ML, Vyas M, Horne MJ, Virk RK, Morotti R, Liu Z, et al. NTRK fusion oncogenes in pediatric papillary thyroid carcinoma in northeast United States. Cancer 2016;122:1097–107. https://doi.org/10.1002/cncr.29887 - PubMed
  161. Vokuhl C, Nourkami-Tutdibi N, Furtwängler R, Gessler M, Graf N, Leuschner I. ETV6–NTRK3 in congenital mesoblastic nephroma: a report of the SIOP/GPOH nephroblastoma study. Pediatr Blood Cancer 2018;65:e26925. https://doi.org/10.1002/pbc.26925 - PubMed
  162. Phillippo DM, Ades AE, Dias S, Palmer S, Abrams KR, Welton NJ. NICE DSU Technical Support Document 18: Methods for Population-adjusted Indirect Comparisons in Submissions to NICE. Sheffield: ScHARR, University of Sheffield; 2016. - PubMed
  163. Hatswell AJ, Thompson GJ, Maroudas PA, Sofrygin O, Delea TE. Estimating outcomes and cost effectiveness using a single-arm clinical trial: ofatumumab for double-refractory chronic lymphocytic leukemia. Cost Eff Resour Alloc 2017;15:8. https://doi.org/10.1186/s12962-017-0071-x - PubMed
  164. Hatswell AJ, Sullivan WG. Creating historical controls using data from a previous line of treatment – two non-standard approaches. Stat Methods Med Res 2020;29:1563–72. https://doi.org/10.1177/0962280219826609 - PubMed
  165. Hsiao SJ, Zehir A, Sireci AN, Aisner DL. Detection of tumor NTRK gene fusions to identify patients who may benefit from tyrosine kinase (TRK) inhibitor therapy. J Mol Diagn 2019;21:553–71. https://doi.org/10.1016/j.jmoldx.2019.03.008 - PubMed
  166. NHS England. National Genomic Test Directory. 2019. URL: www.england.nhs.uk/publication/national-genomic-test-directories/ (accessed 15 May 2019). - PubMed
  167. Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med 2004;10:789–99. https://doi.org/10.1038/nm1087 - PubMed
  168. Morganti S, Tarantino P, Ferraro E, D’Amico P, Viale G, Trapani D, et al. Complexity of genome sequencing and reporting: next generation sequencing (NGS) technologies and implementation of precision medicine in real life. Crit Rev Oncol Hematol 2019;133:171–82. https://doi.org/10.1016/j.critrevonc.2018.11.008 - PubMed
  169. Eifert C, Powers RS. From cancer genomes to oncogenic drivers, tumour dependencies and therapeutic targets. Nat Rev Cancer 2012;12:572–8. https://doi.org/10.1038/nrc3299 - PubMed
  170. Sheikine Y, Kuo FC, Lindeman NI. Clinical and technical aspects of genomic diagnostics for precision oncology. J Clin Oncol 2017;35:929–33. https://doi.org/10.1200/JCO.2016.70.7539 - PubMed
  171. Solomon JP, Hechtman JF. Detection of NTRK fusions: merits and limitations of current diagnostic platforms. Cancer Res 2019;79:3163–8. https://doi.org/10.1158/0008-5472.CAN-19-0372 - PubMed
  172. Penault-Llorca F, Rudzinski ER, Sepulveda AR. Testing algorithm for identification of patients with TRK fusion cancer. J Clin Pathol 2019;72:460–7. https://doi.org/10.1136/jclinpath-2018-205679 - PubMed
  173. Wright C. Long Noncoding RNAs and Cancer. In Gray S, editor. Epigenetic Cancer Therapy. London: Academic Press; 2015. pp. 91–114. https://doi.org/10.1016/B978-0-12-800206-3.00005-7 - PubMed
  174. Marchiò C, Scaltriti M, Ladanyi M, Iafrate AJ, Bibeau F, Dietel M, et al. ESMO recommendations on the standard methods to detect NTRK fusions in daily practice and clinical research. Ann Oncol 2019;30:1417–27. https://doi.org/10.1093/annonc/mdz204 - PubMed
  175. Sigal DS, Bhangoo MS, Hermel JA, Pavlick DC, Frampton G, Miller VA, et al. Comprehensive genomic profiling identifies novel NTRK fusions in neuroendocrine tumors. Oncotarget 2018;9:35809–12. https://doi.org/10.18632/oncotarget.26260 - PubMed
  176. Okamura R, Boichard A, Kato S, Sicklick JK, Bazhenova L, Kurzrock R. Analysis of NTRK alterations in pan-cancer adult and pediatric malignancies: implications for NTRK-targeted therapeutics. JCO Precis Oncol 2018;2018:1PO.18.00183. https://doi.org/10.1200/PO.18.00183 - PubMed
  177. U.S. Food and Drug Administration (FDA), Center for Drug Evaluation and Research. NDA Multidisciplinary Review and Evaluation NDA 210861 and NDA 211710 VITRAKVI (Larotrectinib). Silver Spring, MD: FDA; 2016. - PubMed
  178. Amatu A, Sartore-Bianchi A, Siena S. NTRK gene fusions as novel targets of cancer therapy across multiple tumour types. ESMO Open 2016;1:e000023. https://doi.org/10.1136/esmoopen-2015-000023 - PubMed
  179. Williams HL, Walsh K, Diamond A, Oniscu A, Deans ZC. Validation of the Oncomine™ focus panel for next-generation sequencing of clinical tumour samples. Virchows Arch 2018;473:489–503. https://doi.org/10.1007/s00428-018-2411-4 - PubMed
  180. Solomon JP, Linkov I, Rosado A, Mullaney K, Rosen EY, Frosina D, et al. NTRK fusion detection across multiple assays and 33,997 cases: diagnostic implications and pitfalls. Mod Pathol 2020;33:38–46. https://doi.org/10.1038/s41379-019-0324-7 - PubMed
  181. Makretsov N, He M, Hayes M, Chia S, Horsman DE, Sorensen PH, Huntsman DG. A fluorescence in situ hybridization study of ETV6–NTRK3 fusion gene in secretory breast carcinoma. Genes Chromosomes Cancer 2004;40:152–7. https://doi.org/10.1002/gcc.20028 - PubMed
  182. Scottish Science Advisory Council (SSAC). Informing the Future of Genomic Medicine in Scotland. Edinburgh: SSAC; 2019. - PubMed
  183. Canadian Agency for Drugs and Techonologies in Health (CADTH). Larotrectinib (Vitrakvi) for Neurotrophic Tyrosine Receptor Kinase (NTRK) Positive Solid Tumours: Pan-Canadian Oncology Drug Review. Final Economic Guidance Report. Toronto, ON: CADTH; 2019. - PubMed
  184. Woods B, Sideris E, Palmer S, Latimer N, Soares M. Partitioned Survival Analysis for Decision Modelling in Health Care: A Critical Review. Sheffield: Decision Support Unit, ScHARR, University of Sheffield; 2017. - PubMed
  185. Latimer NJ. NICE DSU Technical Support Document 14: Survival Analysis for Economic Evaluations alongside Clinical Trials - Extrapolation with Patient-level Data. Sheffield: Decision Support Unit, ScHARR, University of Sheffield; 2011. - PubMed
  186. Ouwens MJNM, Mukhopadhyay P, Zhang Y, Huang M, Latimer N, Briggs A. Estimating lifetime benefits associated with immuno-oncology therapies: challenges and approaches for overall survival extrapolations. PharmacoEconomics 2019;37:1129–38. https://doi.org/10.1007/s40273-019-00806-4 - PubMed
  187. Price MJ, Welton NJ, Briggs AH, Ades AE. Model averaging in the presence of structural uncertainty about treatment effects: influence on treatment decision and expected value of information. Value Health 2011;14:205–18. https://doi.org/10.1016/j.jval.2010.08.001 - PubMed
  188. Jackson CH, Bojke L, Thompson SG, Claxton K, Sharples LD. A framework for addressing structural uncertainty in decision models. Med Decis Making 2011;31:662–74. https://doi.org/10.1177/0272989X11406986 - PubMed
  189. Australian Government Department of Health. Co-dependent and Hybrid Technologies. 2017. URL: www1.health.gov.au/internet/hta/publishing.nsf/Content/co-1 (accessed 17 January 2020). - PubMed
  190. Soares MO, Walker S, Palmer SJ, Sculpher MJ. Establishing the value of diagnostic and prognostic tests in health technology assessment. Med Decis Making 2018;38:495–508. https://doi.org/10.1177/0272989X17749829 - PubMed
  191. Albert CM, Davis JL, Federman N, Casanova M, Laetsch TW. TRK fusion cancers in children: a clinical review and recommendations for screening. J Clin Oncol 2019;37:513–24. https://doi.org/10.1200/JCO.18.00573 - PubMed
  192. Stinnett AA, Mullahy J. Net health benefits: a new framework for the analysis of uncertainty in cost-effectiveness analysis. Med Decis Making 1998;18(Suppl. 2):68–80. https://doi.org/10.1177/0272989X98018002S09 - PubMed
  193. Briggs A, Claxton K, Sculpher MJ. Decision Modelling for Health Economic Evaluation. Oxford: Oxford University Press; 2006. - PubMed
  194. Hettle R, Corbett M, Hinde S, Hodgson R, Jones-Diette J, Woolacott N, Palmer S. The assessment and appraisal of regenerative medicines and cell therapy products: an exploration of methods for review, economic evaluation and appraisal. Health Technol Assess 2017;21(7). https://doi.org/10.3310/hta21070 - PubMed
  195. Grimm S, Strong M, Brennan A, Wailoo A. Framework for Analysing Risk in Health Technology Assessments and Its Application to Managed Entry Agreements. Report by the Decision Support Unit. Sheffield: Decision Support Unit, ScHARR, University of Sheffield; 2016. - PubMed
  196. Claxton K. The irrelevance of inference: a decision-making approach to the stochastic evaluation of health care technologies. J Health Econ 1999;18:341–64. https://doi.org/10.1016/S0167-6296(98)00039-3 - PubMed
  197. Claxton K, Palmer S, Longworth L, Bojke L, Griffin S, Soares M, et al. A comprehensive algorithm for approval of health technologies with, without, or only in research: the key principles for informing coverage decisions. Value Health 2016;19:885–91. https://doi.org/10.1016/j.jval.2016.03.2003 - PubMed
  198. Ades AE, Lu G, Claxton K. Expected value of sample information calculations in medical decision modeling. Med Decis Making 2004;24:207–27. https://doi.org/10.1177/0272989X04263162 - PubMed
  199. NHS England Cancer Drugs Fund Team. Appraisal and Funding of Cancer Drugs from July 2016 (Including the New Cancer Drugs Fund): A New Deal for Patients, Taxpayers and Industry. London: NHS England; 2016. - PubMed
  200. Claxton K. Pharmaceutical Pricing: Early Access, The Cancer Drugs Fund and the Role of NICE. York: Centre for Health Economics, University of York; 2016. - PubMed
  201. Grieve R, Abrams K, Claxton K, Goldacre B, James N, Nicholl J, et al. Cancer Drugs Fund requires further reform. BMJ 2016;354:i5090. https://doi.org/10.1136/bmj.i5090 - PubMed
  202. Morrell L, Wordsworth S, Schuh A, Middleton MR, Rees S, Barker RW. Will the reformed Cancer Drugs Fund address the most common types of uncertainty? An analysis of NICE cancer drug appraisals. BMC Health Serv Res 2018;18:393. https://doi.org/10.1186/s12913-018-3162-2 - PubMed
  203. Strong M, Oakley JE, Brennan A. Estimating multiparameter partial expected value of perfect information from a probabilistic sensitivity analysis sample: a nonparametric regression approach. Med Decis Making 2014;34:311–26. https://doi.org/10.1177/0272989X13505910 - PubMed
  204. Strong M, Oakley JE, Brennan A, Breeze P. Estimating the expected value of sample information using the probabilistic sensitivity analysis sample: a fast, nonparametric regression-based method. Med Decis Making 2015;35:570–83. https://doi.org/10.1177/0272989X15575286 - PubMed
  205. Willan AR, Pinto EM. The value of information and optimal clinical trial design. Stat Med 2005;24:1791–806. https://doi.org/10.1002/sim.2069 - PubMed
  206. National Institute for Health and Care Excellence (NICE). Guide to the Processes of Technology Appraisal. London: NICE; 2018. - PubMed
  207. Kim DD, Basu A. New metrics for economic evaluation in the presence of heterogeneity: focusing on evaluating policy alternatives rather than treatment alternatives. Med Decis Making 2017;37:930–41. https://doi.org/10.1177/0272989X17702379 - PubMed
  208. Drummond MF, Sculpher MJ, Claxton K, Stoddart GL, Torrance GW. Methods for the Economic Evaluation of Health Care Programmes. Oxford: Oxford University Press; 2015. - PubMed
  209. Claxton K. OFT, VBP: QED? Health Econ 2007;16:545–58. - PubMed
  210. Rothery C, Claxton K, Palmer S, Epstein D, Tarricone R, Sculpher M. Characterising uncertainty in the assessment of medical devices and determining future research needs. Health Econ 2017;26(Suppl. 1):109–23. https://doi.org/10.1002/hec.3467 - PubMed
  211. Kennedy-Martin T, Curtis S, Faries D, Robinson S, Johnston J. A literature review on the representativeness of randomized controlled trial samples and implications for the external validity of trial results. Trials 2015;16:495. https://doi.org/10.1186/s13063-015-1023-4 - PubMed
  212. National Institute for Health and Care Excellence (NICE). Pembrolizumab with Carboplatin and Paclitaxel for Untreated Metastatic Squamous Non-small-cell Lung Cancer. Technology Appraisal Guidance [TA600]. London: NICE; 2019. - PubMed
  213. National Institute for Health and Care Excellence (NICE). Pembrolizumab for Untreated PD-L1-positive Locally Advanced or Metastatic Urothelial Cancer when Cisplatin is Unsuitable. Technology Appraisal [TA674]. London: NICE; 2018. - PubMed
  214. National Institute for Health and Care Excellence (NICE). Pembrolizumab for Untreated Metastatic or Unresectable Recurrent Squamous Cell Head and Neck Cancer. Technology Appraisal Guidance [TA661]. London: NICE; 2020. URL: www.nice.org.uk/guidance/TA661 (accessed 20 February 2020). - PubMed
  215. European Medicines Agency. Workshop on Site and Histology – Independent Indications of Oncology. 2018. URL: www.ema.europa.eu/en/events/workshop-site-histology-independent-indications-oncology (accessed October 2021). - PubMed
  216. European Medicines Agency. Workshop on Single-arm Studies in Oncology. 2016. www.ema.europa.eu/en/events/workshop-single-arm-trials-oncology (accessed October 2021). - PubMed
  217. US Food and Drug Administration. Developing Targeted Therapies in Low-Frequency Molecular Subsets of a Disease. 2018. www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM588884.pdf (accessed October 2021). - PubMed
  218. US Food and Drug Administration. Master Protocols: Efficient Clinical Trial Design Strategies to Expedite Development of Oncology Drugs and Biologics. 2018. www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM621817.pdf (accessed October 2021). - PubMed
  219. US Department of Health and Human Services, U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Center for Biologics Evaluation and Research. Guidance for Industry: Expedited Programs for Serious Conditions – Drugs and Biologics. 2014. www.fda.gov/downloads/Drugs/Guidances/UCM358301.pdf (accessed October 2021). - PubMed
  220. Food and Drug Administration. Table of Surrogate Endpoints That Were the Basis of Drug Approval or Licensure. URL: www.fda.gov/drugs/development-resources/table-surrogate-endpoints-were-basis-drug-approval-or-licensure (accessed 17 January 2020). - PubMed
  221. Food and Drug Administration (FDA). Clinical Trial Endpoints for the Approval of Cancer Drugs and Biologics Guidance for Industry. December 2018. URL: www.fda.gov/regulatory-information/search-fda-guidance-documents/clinical-trial-endpoints-approval-cancer-drugs-and-biologics (accessed 17 January 2020). - PubMed
  222. US Food and Drug Administration. Tissue Agnostic Therapies in Oncology: Regulatory Considerations for Orphan Drug Designation. www.fda.gov/downloads/NewsEvents/MeetingsConferencesWorkshops/UCM598186.pdf (accessed October 2021). - PubMed
  223. European Medicines Agency. Essential Considerations for Successful Qualification of Novel Methodologies. 2017. www.ema.europa.eu/documents/other/essential-considerations-successful-qualification-novel-methodologies_en.pdf (accessed October 2021). - PubMed
  224. European Medicines Agency. Biostatistics. 2021. www.ema.europa.eu/en/human-regulatory/research-development/scientific-guidelines/clinical-efficacy-safety/biostatistics (accessed October 2021). - PubMed
  225. European Medicines Agency. Predictive Biomarker-based Assay Development in the Context of Drug Development and Lifecycle. 2017. www.ema.europa.eu/en/predictive-biomarker-based-assay-development-context-drug-development-lifecycle (accessed October 2021). - PubMed
  226. European Medicines Agency. Guideline on the Evaluation of Anticancer Medicinal Products in Man Rev. 5. 2017. www.ema.europa.eu/documents/scientific-guideline/guideline-evaluation-anticancer-medicinal-products-man-revision-5_en.pdf (accessed October 2021). - PubMed
  227. European Medicines Agency. Appendix 4 to the Guideline on the Evaluation of Anticancer Medicinal Products in Man. 2015. www.ema.europa.eu/documents/scientific-guideline/evaluation-anticancer-medicinal-products-man-appendix-4-condition-specific-guidance-rev2_en.pdf (accessed October 2021). - PubMed
  228. National Institute for Health and Care Excellence (NICE). Ceritinib for Previously Treated Anaplastic Lymphoma Kinase Positive Non-Small-Cell Lung Cancer. Technology Appraisal Guidance [TA395]. London: NICE; 2016. - PubMed
  229. Kim DW, Mehra R, Tan DS, Felip E, Chow LQ, Camidge DR, Vansteenkiste J, et al. Activity and safety in ceritinib in patients with ALK-rearranged non-small-cell lung cancer (ASCEND-1): updated results from the multicentre, open-label, phase 1 trial. Lancet Oncol 2016;17:452–63. https://doi.org/10.1016/S1470-2045(15)00614-2 - PubMed
  230. Crinò L, Ahn MJ, De Marinis F, Groen HJ, Wakelee H, Hida T, et al. Multicenter phase II study of whole-body and intracranial activity with ceritinib in patients with ALK-rearranged non-small-cell lung cancer previously treated with chemotherapy and crizotinib: results from ASCEND-2. J Clin Oncol 2016;34:2866–78. https://doi.org/10.1200/JCO.2015.65.5936 - PubMed
  231. National Institute for Health and Care Excellence (NICE). Osimertinib for Treating Locally Advanced or Metastatic EGFR T790M Mutation-positive Non-small-cell Lung Cancer. Technology Appraisal Guidance [TA416]. London: NICE; 2016. - PubMed
  232. Yang JC, Ahn MJ, Kim DW, Ramalingam SS, Sequist LV, Su WC, et al. Osimertinib in pretreated T790M-positive advanced non-small-cell lung cancer: AURA study phase II extension component. J Clin Oncol 2017;35:1288–96. https://doi.org/10.1200/JCO.2016.70.3223 - PubMed
  233. Goss G, Tsai CM, Shepherd FA, Bazhenova L, Lee JS, Chang GC, et al. Osimertinib for pretreated EGFR Thr790Met-positive advanced non-small-cell lung cancer (AURA2): a multicentre, open-label, single-arm, phase 2 study. Lancet Oncol 2016;17:1643–52. https://doi.org/10.1016/S1470-2045(16)30508-3 - PubMed
  234. Ahn MJ, Tsai CM, Shepherd FA, Bazhenova L, Sequist LV, Hida T, et al. Osimertinib in patients with T790M mutation-positive, advanced non–small cell lung cancer: long-term follow-up from a pooled analysis of 2 phase 2 studies. Cancer 2019;125:892–901. https://doi.org/10.1002/cncr.31891 - PubMed
  235. National Institute for Health and Care Excellence (NICE). Nivolumab for Treating Relapsed or Refractory Classical Hodgkin Lymphoma. Technology Appraisal Guidance [TA462]. London: NICE; 2017. - PubMed
  236. Younes A, Santoro A, Shipp M, Zinzani PL, Timmerman JM, Ansell S, et al. Nivolumab for classical Hodgkin's lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial. Lancet Oncol 2016;17:1283–94. https://doi.org/10.1016/S1470-2045(16)30167-X - PubMed
  237. Lesokhin AM, Ansell SM, Arman P, Scott EC, Halwani A, Gutierrez M, et al. Nivolumab in patients with relapsed or refractory hematologic malignancy: preliminary results of a phase 1b study. J Clin Oncol 2016;34:2698–704. https://doi.org/10.1200/JCO.2015.65.9789 - PubMed
  238. National Institute for Health and Care Excellence (NICE). Venetoclax for Treating Chronic Lymphocytic Leukaemia. Technology Appraisal Guidance [TA487]. London: NICE; 2017. - PubMed
  239. Davids MS, Roberts AW, Seymour JK, Pagel JM, Kahl BS, Wierda WG, et al. Phase I first-in-human study of venetoclax in patients with relapsed or refractory non-Hodgkin lymphoma. J Clin Oncol 2017;35:826–33. https://doi.org/10.1200/JCO.2016.70.4320 - PubMed
  240. Stilgenbauer S, Eichhorst B, Schetelig J, Coutre S, Seymour JF, Munir T, et al. Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: a multicentre, open-label, phase 2 study. Lancet Oncol 2016;17:768–78. https://doi.org/10.1016/S1470-2045(16)30019-5 - PubMed
  241. Stilgenbauer S, Eichhorst B, Schetelig J, Hillmen P, Seymour JF, Coutre S, et al. Venetoclax for patients with chronic lymphocytic leukemia with 17p deletion: results from the full population of a phase II pivotal trial. J Clin Oncol 2018;36:1973–80. https://doi.org/10.1200/JCO.2017.76.6840 - PubMed
  242. Coutre S, Choi M, Furman RR, Eradat H, Heffner L, Jones JA, et al. Venetoclax for patients with chronic lymphocytic leukemia who progressed during or after idelalisib therapy. Blood 2018;131:1704–11. https://doi.org/10.1182/blood-2017-06-788133 - PubMed
  243. Jones JA, Mato AR, Wierda WG, Davids MS, Choi M, Cheson BD, et al. Venetoclax for chronic lymphocytic leukaemia progressing after ibrutinib: an interim analysis of a multicentre, open-label, phase 2 trial. Lancet Oncol 2018;19:65–75. https://doi.org/10.1016/S1470-2045(17)30909-9 - PubMed
  244. National Institute for Health and Care Excellence (NICE). Atezolizumab for Untreated PD-L1-Positive Locally Advanced or Metastatic Urothelial Cancer When Cisplatin is Unsuitable. Technology Appraisal Guidance [TA492]. London: NICE; 2018. - PubMed
  245. Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt J, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet Oncol 2017;389:67–76. https://doi.org/10.1016/S0140-6736(16)32455-2 - PubMed
  246. Necchi A, Joseph RW, Loriot Y, Hoffman-Censits J, Perez-Gracia JL, Petrylak DP, et al. Atezolizumab in platinum-treated locally advanced or metastatic urothelial carcinoma: post-progression outcomes from the phase II IMvigor210 study. Ann Oncol 2017;28:3044–50. https://doi.org/10.1093/annonc/mdx518 - PubMed
  247. Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 2016;387:1909–20. https://doi.org/10.1016/S0140-6736(16)00561-4 - PubMed
  248. National Institute for Health and Care Excellence (NICE). Daratumumab Monotherapy for Treating Relapsed and Refractory Multiple Myeloma. Technology Appraisal Guidance [TA510]. London: NICE; 2018. - PubMed
  249. Lonial S, Weiss BM, Usmani SZ, Singhai S, Chari A, Bahlis NJ, et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet 2016;387:1551–60. https://doi.org/10.1016/S0140-6736(15)01120-4 - PubMed
  250. Lokhorst HM, Plesner T, Laubach JP, Nahi H, Gimsing P, Hansson M, et al. Targeting CD38 with daratumumab monotherapy in multiple myeloma. New Engl J Med 2015;373:1207–19. https://doi.org/10.1056/NEJMoa1506348 - PubMed
  251. Usmani SZ, Weiss BM, Plesner T, Bahlis NJ, Belch A, Lonial S, et al. Clinical efficacy of daratumumab monotherapy in patients with heavily pretreated relapsed or refractory multiple myeloma. Blood 2016;128:37–44. https://doi.org/10.1182/blood-2016-03-705210 - PubMed
  252. National Institute for Health and Care Excellence (NICE). Avelumab for Treating Metastatic Merkel Cell Carcinoma. Technology Appraisal Guidance [TA517]. London: NICE; 2017. - PubMed
  253. Kaufman HL, Russell J, Hamid O, Bhatia S, Terheyden P, D’Angelo SP, et al. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. Lancet Oncol 2016;17:1374–85. https://doi.org/10.1016/S1470-2045(16)30364-3 - PubMed
  254. D’Angelo SP, Russell J, Lebbé C, Chmielowski B, Gambichler T, Grob JJ, et al. Efficacy and safety of first-line avelumab treatment in patients with stage IV metastatic Merkel cell carcinoma: a preplanned interim analysis of a clinical trial. JAMA Oncol 2018;4:e180077. https://doi.org/10.1001/jamaoncol.2018.0077 - PubMed
  255. National Institute for Health and Care Excellence (NICE). Crizotinib for Treating ROS1-positive Advanced Non-small-cell Lung Cancer. Technology Appraisal Guidance [TA529]. London: NICE; 2018. - PubMed
  256. Shaw AT, Ou SHI, Bang YJ, Camidge DR, Solomon BJ, Salgia R, et al. Crizotinib in ROS1-rearranged non–small-cell lung cancer. New Engl J Med 2014;371:1963–71. https://doi.org/10.1056/NEJMoa1406766 - PubMed
  257. Shaw AT, Riely GJ, Bang YJ, Kim DW, Camidge DR, Solomon BJ, et al. Crizotinib in ROS1-rearranged advanced non-small-cell lung cancer (NSCLC): updated results, including overall survival, from PROFILE 1001. Ann Oncol 2019;30:1121–6. https://doi.org/10.1093/annonc/mdz131 - PubMed
  258. National Institute for Health and Care Excellence (NICE). Pembrolizumab for Treating Relapsed or Refractory Classical Hodgkin Lymphoma. Technology Appraisal Guidance [TA540]. London: NICE; 2018. - PubMed
  259. Chen R, Zinzani PL, Fanale MA, Armand P, Johnson NA, Brice P, et al. Phase II study of the efficacy and safety of pembrolizumab for relapsed/refractory classic Hodgkin lymphoma. J Clin Oncol 2017;35:2125–32. https://doi.org/10.1200/JCO.2016.72.1316 - PubMed
  260. National Institute for Health and Care Excellence (NICE). Brigatinib for Treating ALK-positive Non-small-cell Lung Cancer after Crizotinib. Technology Appraisal Guidance [TA571]. London: NICE; 2019. - PubMed
  261. Kim DW, Tiseo M, Ahn MJ, Reckamp KL, Hansen KH, Kim SW, et al. Brigatinib in patients with crizotinib-refractory anaplastic lymphoma kinase-positive non-small-cell lung cancer: a randomized, multicenter phase II trial. J Clin Oncol 2017;35:2490–8. https://doi.org/10.1200/JCO.2016.71.5904 - PubMed
  262. Gettinger SN, Bazhenova LA, Langer CJ, Salgia R, Gold KA, Rosell R, et al. Activity and safety of brigatinib in ALK-rearranged non-small-cell lung cancer and other malignancies: a single-arm, open-label, phase 1/2 trial. Lancet Oncol 2016;17:1683–96. https://doi.org/10.1016/S1470-2045(16)30392-8 - PubMed
  263. Hatswell AJ, Freemantle N, Baio G. Economic evaluations of pharmaceuticals granted a marketing authorisation without the results of randomised trials: a systematic review and taxonomy. PharmacoEconomics 2017;35:163–76. https://doi.org/10.1007/s40273-016-0460-6 - PubMed
  264. Faria R, Hernandez Alava M, Manca A, Wailoo AJ. The Use of Observational Data to Inform Estimates of Treatment Effectiveness for Technology Appraisal: Methods for Comparative Individual Patient Data. Report by the Decision Support Unit. Sheffield: Decision Support Unit, ScHARR, University of Sheffield; 2015. - PubMed
  265. Bell H, Wailoo A, Hernandez M, Grieve R, Faria R, Gibson L, et al. The Use of Real World Data for the Estimation of Treatment Effects in NICE Decision Making. Report by the Decision Support Unit. Sheffield: Decision Support Unit, ScHARR, University of Sheffield; 2016. - PubMed
  266. Wright SJ, Newman WG, Payne K. Accounting for capacity constraints in economic evaluations of precision medicine: a systematic review. PharmacoEconomics 2019;37:1011–27. https://doi.org/10.1007/s40273-019-00801-9 - PubMed
  267. Marmor S, Portschy PR, Tuttle TM, Virnig BA. The rise in appendiceal cancer incidence: 2000–2009. J Gastrointest Surg 2015;19:743–50. https://doi.org/10.1007/s11605-014-2726-7 - PubMed
  268. World Population Review. Western European Countries 2020. 2019. URL: http://worldpopulationreview.com/countries/western-european-countries/ (accessed 12 December 2019). - PubMed
  269. Office for National Statistics. Cancer Registration Statistics, England: 2017. 2019. URL: www.ons.gov.uk/releases/cancerregistrationstatisticsengland2017 (accessed June 2019). - PubMed
  270. Cancer Research UK. Breast Cancer Incidence by Stage at Diagnosis. London: Cancer Research UK; 2014. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/breast-cancer/incidence (accessed June 2019). - PubMed
  271. Coombes N, Elliss-Brookes L, Johnson S, Lyons J, Mackay C, Morement H, et al. National Cancer Intelligence Network. Rare and Less Common Cancers: Incidence and Mortality in England, 2010 to 2013. London: Public Health England; 2015. - PubMed
  272. Tsuchiya N, Sawada Y, Endo I, Saito K, Uemura Y, Nakatsura T. Biomarkers for the early diagnosis of hepatocellular carcinoma. World J Gastroenterol 2015;21:10573–83. https://doi.org/10.3748/wjg.v21.i37.10573 - PubMed
  273. Thrumurthy SG, Thrumurthy SS, Gilbert CE, Ross P, Haji A. Colorectal adenocarcinoma: risks, prevention and diagnosis. BMJ 2016;354:i3590. https://doi.org/10.1136/bmj.i3590 - PubMed
  274. Cancer Research UK. Bowel Cancer Incidence by Stage at Diagnosis. London: Cancer Research UK; 2016. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/bowel-cancer (accessed June 2019). - PubMed
  275. Starczewska Amelio JM, Cid Ruzafa J, Desai K, Tzivelekis S, Muston D, Khalid JM, et al. Prevalence of gastrointestinal stromal tumour (GIST) in the United Kingdom at different therapeutic lines: an epidemiologic model. BMC Cancer 2014;14:364. https://doi.org/10.1186/1471-2407-14-364 - PubMed
  276. PDQ Adult Treatment Editorial Board. Gastrointestinal Stromal Tumors Treatment (Adult) (PDQ®): Patient Version. 2020. URL: https://www.ncbi.nlm.nih.gov/books/NBK65929/ (accessed November 2021). - PubMed
  277. Orbach D, Rey A, Cecchetto G, Oberlin O, Casanova M, Thebaud E, et al. Infantile fibrosarcoma: management based on the European experience. J Clin Oncol 2010;28:318–23. https://doi.org/10.1200/JCO.2009.21.9972 - PubMed
  278. Skálová A, Vanecek T, Sima R, Laco J, Weinreb I, Perez-Ordonez B, et al. Mammary analogue secretory carcinoma of salivary glands, containing the ETV6–NTRK3 fusion gene: a hitherto undescribed salivary gland tumor entity. Am J Surg Pathol 2010;34:599–608. https://doi.org/10.1097/PAS.0b013e3181d9efcc - PubMed
  279. Luk PP, Selinger CI, Eviston TJ, Lum T, Yu B, O’Toole SA, et al. Mammary analogue secretory carcinoma: an evaluation of its clinicopathological and genetic characteristics. Pathology 2015;47:659–66. https://doi.org/10.1097/PAT.0000000000000322 - PubMed
  280. Sethi R, Kozin E, Remenschneider A, Meier J, VanderLaan P, Faquin W, et al. Mammary analogue secretory carcinoma: update on a new diagnosis of salivary gland malignancy. Laryngoscope 2014;124:188–95. https://doi.org/10.1002/lary.24254 - PubMed
  281. Cancer Research UK. Melanoma Skin Cancer Incidence by Stage at Diagnosis. London: Cancer Research UK; 2016. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/melanoma-skin-cancer/incidence (accessed June 2019). - PubMed
  282. Care Quality Improvement Department, Royal College of Physicians. National Lung Cancer Audit Annual Report 2018 (for the Audit Period 2017). London: Healthcare Quality Improvement Partnership (HQIP); 2019. - PubMed
  283. Pancreatic Cancer UK. Types of Pancreatic Cancer. London: Pancreatic Cancer UK; 2018. URL: www.pancreaticcancer.org.uk/information-and-support/facts-about-pancreatic-cancer/types-of-pancreatic-cancer/ (accessed 16 December 2019). - PubMed
  284. Cancer Research UK. Pancreatic Cancer Incidence by Stage at Diagnosis. London: Cancer Research UK; 2016. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/pancreatic-cancer/incidence#heading-Three (accessed June 2019). - PubMed
  285. Cancer Research UK. Soft Tissue Sarcoma Incidence Statistics. 2010. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/soft-tissue-sarcoma/incidence#heading-Four (accessed 27 February 2019). - PubMed
  286. American Cancer Society. Cancer Facts and Figures. 2017. URL: www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2017.html (accessed 24 January 2020). - PubMed
  287. Deen MH, Burke KM, Janitz A, Campbell J. Cancers of the thyroid: overview and statistics in the United States and Oklahoma. J Okla State Med Assoc 2016;109:333–8. - PubMed
  288. Cancer Research UK. Cervical Cancer Incidence by Stage at Diagnosis. London: Cancer Research UK; 2016. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/cervical-cancer/incidence#heading-Three (accessed June 2019). - PubMed
  289. Gooskens SL, Houwing ME, Vujanic GM, Dome JS, Diertens T, Coulomb-l’Herminé A, et al. Congenital mesoblastic nephroma 50 years after its recognition: a narrative review. Pediatr Blood Cancer 2017;64:e26437. https://doi.org/10.1002/pbc.26437 - PubMed
  290. National Institute for Health and Care Excellence (NICE). Pembrolizumab for Previously Treated Oesophageal or Gastrooesophageal Junction Cancer Draft Scope. London: NICE; 2018. - PubMed
  291. Cancer Research UK. Oesophageal Cancer Incidence by Stage at Diagnosis. London: Cancer Research UK; 2016. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/oesophageal-cancer/incidence (accessed June 2019). - PubMed
  292. Cancer Research UK. Head and Neck Cancers Statistics. 2016. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/head-and-neck-cancers (accessed June 2019). - PubMed
  293. Public Health England. Glioblastoma in England 2007-2011. London: Public Health England; 2016. - PubMed
  294. Wang Y, Jiang T. Understanding high grade glioma: molecular mechanism, therapy and comprehensive management. Cancer Lett 2013;331:139–46. https://doi.org/10.1016/j.canlet.2012.12.024 - PubMed
  295. UK and Ireland Neuroendcrine Tumour Society (UKI NETS). Incidence and Prevalence of Neuroendocrine Tumours in England. London: Endocrine Abstracts; 2017. - PubMed
  296. Cancer Research UK. Ovarian Cancer Incidence by Stage at Diagnosis. London: Cancer Research UK; 2016. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/ovarian-cancer/incidence (accessed June 2019). - PubMed
  297. Farrimond J. Analysis of the Cancer Registry Combined Database for Use with the Brain and CNS Registry. London: National Cancer Intelligence Network, Eastern Cancer Registration and Information Centre; 2010. - PubMed
  298. Cancer Research UK. Children’s Cancer Incidence Statistics. London: Cancer Research UK; 2014. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/childrens-cancers (accessed June 2019). - PubMed
  299. Austin MT, Xing Y, Hayes-Jordan AA, Lally KP, Cormier JN. Melanoma incidence rises for children and adolescents: an epidemiologic review of pediatric melanoma in the United States. J Pediatr Surg 2013;48:2207–13. https://doi.org/10.1016/j.jpedsurg.2013.06.002 - PubMed
  300. Brzeziańska E, Karbownik M, Migdalska-Sek M, Pastuszak-Lewandoska D, Włoch J, Lewiński A. Molecular analysis of the RET and NTRK1 gene rearrangements in papillary thyroid carcinoma in the Polish population. Mutat Res 2006;599:26–35. https://doi.org/10.1016/j.mrfmmm.2005.12.013 - PubMed
  301. Cancer Research UK. Prostate Cancer Incidence by Stage at Diagnosis. London: Cancer Research UK; 2016. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/prostate-cancer/incidence#heading-Three (accessed June 2019). - PubMed
  302. Cancer Research UK. Kidney Cancer: Stages, Types and Grades. London: Cancer Research UK; 2016. URL: www.cancerresearchuk.org/about-cancer/kidney-cancer/stages-types-grades/types-grades (accessed December 2019). - PubMed
  303. Horowitz DP, Sharma CS, Connolly E, Gidea-Addeo D, Deutsch I. Secretory carcinoma of the breast: results from the survival, epidemiology and end results database. Breast 2012;21:350–3. https://doi.org/10.1016/j.breast.2012.02.013 - PubMed
  304. Jacob JD, Hodge C, Franko J, Pezzi CM, Goldman CD, Klimberg VS. Rare breast cancer: 246 invasive secretory carcinomas from the National Cancer Data Base. J Surg Oncol 2016;113:721–5. https://doi.org/10.1002/jso.24241 - PubMed
  305. Rushton L, Bagga S, Bevan R, Brown T, Cherrie J, Holmes P, et al. The Burden of Occupational Cancer in Great Britain. London: Health & Safety Executive; 2010. - PubMed
  306. Cancer Research UK. Uterine Cancer Incidence By Stage At Diagnosis. 2016. URL: www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/uterine-cancer/incidence#heading-Three (accessed June 2019). - PubMed
  307. Lee SJ, Kim NKD, Lee S-H, Kim ST, Park SH, Park JO, et al. NTRK gene amplification in patients with metastatic cancer. Precision Future Med 2017;1:129–37. https://doi.org/10.23838/pfm.2017.00142 - PubMed
  308. Lange AM, Lo HW. Inhibiting TRK proteins in clinical cancer therapy. Cancers 2018;10:E105. - PubMed
  309. National Cancer Institute. Gastrointestinal Stromal Tumors Treatment (Adult) (PDQ®)–Health Professional Version. Rockville, MD: National Cancer Institute; 2018. URL: www.cancer.gov/types/soft-tissue-sarcoma/hp/gist-treatment-pdq (accessed June 2019). - PubMed
  310. Demetri GD, Paz-Ares L, Farago AF, Liu SV, Chawla SP, Tosi D, et al. Efficacy and safety of entrectinib in patients with NTRK fusion-positive tumours: pooled analysis of STARTRK-2, STARTRK-1, and ALKA-372-001. Ann Oncol 2018;29:ix175. https://doi.org/10.1093/annonc/mdy483.003 - PubMed
  311. Hyman D, van Tilburg C, Albert C, Tan D, Geoerger B, Farago A, et al. 445PD Durability of response with larotrectinib in adult and pediatric patients with TRK fusion cancer. Ann Oncol 2019;30:v162–3. https://doi.org/10.1093/annonc/mdz244.007 - PubMed
  312. Georghiou T, Bardsley M. Exploring the Cost of End of Life Care. London: Nuffield Trust; 2014. - PubMed

Publication Types