Skip to main content
Log in

Developing a Framework for the Health Technology Assessment of Histology-independent Precision Oncology Therapies

  • Current Opinion
  • Published:
Applied Health Economics and Health Policy Aims and scope Submit manuscript

Abstract

The arrival of precision oncology is challenging the evidence standards under which technologies are evaluated for regulatory approval as well as for health technology assessment (HTA) purposes. Several key concepts are discussed to highlight the source of the challenges in evaluating these products, particularly those impacting the HTA of histology-independent therapies. These include the basket trial design, high uncertainty in (potentially substantial) benefits for histology-independent therapies, and the inability to identify and quantify benefits of standard of care in daily practice when the biomarker is not currently used in practice. There is little precedent for a technology with the unique mixture of challenges for HTA of histology-independent therapies and they will be evaluated using standard HTA, as there currently is no evidence suggesting the standard HTA framework is not appropriate. A number of questions proposed to help guide HTA bodies when assessing the appropriateness of local processes to optimally evaluate histology-independent therapies. Pragmatic solutions are further proposed to decrease uncertainty in the benefits of histology independent therapies as well as fill gaps in comparative evidence. The proposed solutions ensure a consistent and streamlined approach to evaluation across histology-independent products, although with varying strengths and limitations. Alongside these solutions, sponsors should engage early with HTA bodies/payers and regulatory agencies through parallel/joint scientific advice to facilitate the integration of both regulatory and HTA perspectives into one clinical development programme, potentially reconciling evidence requirements.

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

Similar content being viewed by others

References

  1. Guo Y, Cao R, Zhang X, Huang L, Sun L, Zhao J, et al. Recent progress in rare oncogenic drivers and targeted therapy for non-small cell lung cancer. Onco Targets Ther. 2019;12:10343.

    Article  CAS  Google Scholar 

  2. Torres-Ayuso P, Brognard J. Combing the cancer genome for novel kinase drivers and new therapeutic targets. Cancers. 2019;11(12):1972.

    Article  CAS  Google Scholar 

  3. Hyman DM, Taylor BS, Baselga J. Implementing genome-driven oncology. Cell. 2017;168(4):584–99.

    Article  CAS  Google Scholar 

  4. Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9(1):34.

    Article  Google Scholar 

  5. Suh JH, Johnson A, Albacker L, Wang K, Chmielecki J, Frampton G, et al. Comprehensive genomic profiling facilitates implementation of the National Comprehensive Cancer Network guidelines for lung cancer biomarker testing and identifies patients who may benefit from enrollment in mechanism-driven clinical trials. Oncologist. 2016;21(6):684.

    Article  CAS  Google Scholar 

  6. Kunz M, Jeromin J, Fuchs M, Christoph J, Veronesi G, Flentje M, et al. In silico signaling modeling to understand cancer pathways and treatment responses. Brief Bioinform. 2020;21(3):1115–7.

    Article  CAS  Google Scholar 

  7. Love-Koh J, Peel A, Rejon-Parrilla JC, Ennis K, Lovett R, Manca A, et al. The future of precision medicine: potential impacts for health technology assessment. Pharmacoeconomics. 2018;36(12):1439–51.

    Article  Google Scholar 

  8. Hatswell AJ, Baio G, Berlin JA, Irs A, Freemantle N. Regulatory approval of pharmaceuticals without a randomised controlled study: analysis of EMA and FDA approvals 1999–2014. BMJ Open. 2016;6(6):e011666.

    Article  Google Scholar 

  9. Yan L, Zhang W. Precision medicine becomes reality—tumor type-agnostic therapy. Cancer Commun. 2018;38(1):6.

    Article  Google Scholar 

  10. European Medicines Agency. First ‘histology-independent’ treatment for solid tumors with a specific gene mutation. 2019. https://www.ema.europa.eu/en/documents/press-release/first-histology-independent-treatment-solid-tumours-specific-gene-mutation_en.pdf. Accessed June 2020.

  11. Lemery S, Keegan P, Pazdur R. First FDA approval agnostic of cancer site—when a biomarker defines the indication. N Engl J Med. 2017;377(15):1409–12.

    Article  Google Scholar 

  12. University of Colorado Cancer Center. With the right care, patients now living a median 6.8 years after stage IV ALK+ lung cancer diagnosis. 2018. https://coloradocancerblogs.org/with-the-right-care-patients-now-living-a-median-6-8-years-after-stage-iv-alk-lung-cancer-diagnosis/. Accessed June 2020.

  13. Khan M, Lin J, Liao G, Tian Y, Liang Y, Li R, et al. ALK inhibitors in the treatment of ALK positive NSCLC. Front Oncol. 2019;8:557.

    Article  Google Scholar 

  14. Pacheco JM, Gao D, Smith D, Purcell T, Hancock M, Bunn P, et al. Natural history and factors associated with overall survival in stage IV ALK-rearranged non–small cell lung cancer. J Thorac Oncol. 2019;14(4):691–700.

    Article  CAS  Google Scholar 

  15. Kaufman HL, Atkins MB, Subedi P, Wu J, Chambers J, Mattingly TJ, et al. The promise of Immuno-oncology: implications for defining the value of cancer treatment. J Immunother Cancer. 2019;7(1):129.

    Article  Google Scholar 

  16. Luo SY, Lam DC. Oncogenic driver mutations in lung cancer. Transla Respir Med. 2013;1(1):6.

    Article  Google Scholar 

  17. Schwaederle M, Zhao M, Lee JJ, Lazar V, Leyland-Jones B, Schilsky RL, et al. Association of biomarker-based treatment strategies with response rates and progression-free survival in refractory malignant neoplasms: a meta-analysis. JAMA Oncol. 2016;2(11):1452–9.

    Article  Google Scholar 

  18. Hazim A, Prasad V. A pooled analysis of published, basket trials in cancer medicine. Eur J Cancer. 2018;101:244–50.

    Article  Google Scholar 

  19. Janiaud P, Serghiou S, Ioannidis JP. New clinical trial designs in the era of precision medicine: an overview of definitions, strengths, weaknesses, and current use in oncology. Cancer Treat Rev. 2019;73:20–30.

    Article  Google Scholar 

  20. Qin B-D, Jiao X-D, Liu K, Wu Y, He X, Liu J, et al. Basket trials for intractable cancer. Front Oncol. 2019;9:229.

    Article  Google Scholar 

  21. Cunanan KM, Gonen M, Shen R, Hyman DM, Riely GJ, Begg CB, et al. Basket trials in oncology: a trade-off between complexity and efficiency. J Clin Oncol. 2017;35(3):271.

    Article  Google Scholar 

  22. Canadian Agency for Drugs and Technologies in Health (CADTH). pCODR Expert Review Committee—Final Decision for Larotrectinib. 2019. https://cadth.ca/sites/default/files/pcodr/Reviews2019/10159LarotrectinibNTRK%2BSolidTumours_fnRec_REDACT_31Oct201_ChairApproved_final.pdf. Accessed June 2020.

  23. National Institute for Health and Care Excellence (NICE). Larotrectinib for treating advanced solid tumours with TRK fusions. 2020. https://www.nice.org.uk/guidance/indevelopment/gid-ta10229. Accessed June 2020.

  24. Zia MI, Siu LL, Pond GR, Chen EX. Comparison of outcomes of phase II studies and subsequent randomized control studies using identical chemotherapeutic regimens. J Clin Oncol. 2005;23(28):6982–91.

    Article  CAS  Google Scholar 

  25. Gibson E, Koblbauer I, Begum N, Dranitsaris G, Liew D, McEwan P, et al. Modelling the survival outcomes of immuno-oncology drugs in economic evaluations: a systematic approach to data analysis and extrapolation. Pharmacoeconomics. 2017;35(12):1257–70.

    Article  Google Scholar 

  26. 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 Effectiveness Resour Allocation. 2017;15(1):8.

    Article  Google Scholar 

  27. Jackson CH, Sharples LD, Thompson SG. Survival models in health economic evaluations: balancing fit and parsimony to improve prediction. Int J Biostat. 2010;6(1):34. https://doi.org/10.2202/1557-4679.1269.

    Article  Google Scholar 

  28. Royston P, Parmar MK. Flexible parametric proportional-hazards and proportional-odds models for censored survival data, with application to prognostic modelling and estimation of treatment effects. Stat Med. 2002;21(15):2175–97.

    Article  Google Scholar 

  29. Davies J, Martinec M, Delmar P, Coudert M, Bordogna W, Golding S, et al. Comparative effectiveness from a single-arm trial and real-world data: alectinib versus ceritinib. J Comp Effectiveness Res. 2018;7(09):855–65.

    Article  Google Scholar 

  30. 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(6):1563–72.

    Article  Google Scholar 

  31. Von Hoff DD. There are no bad anticancer agents, only bad clinical trial designs–twenty-first Richard and Hinda Rosenthal Foundation Award Lecture. Clin Cancer Res. 1998;4(5):1079–86.

    Google Scholar 

  32. Von Hoff DD, Stephenson JJ Jr, Rosen P, Loesch DM, Borad MJ, Anthony S, et al. Pilot study using molecular profiling of patients’ tumors to find potential targets and select treatments for their refractory cancers. J Clin Oncol. 2010;28(33):4877–83.

    Article  Google Scholar 

  33. Penel N, Demetri G, Blay JY, Cousin S, Maki RG, Chawla S, et al. Growth modulation index as metric of clinical benefit assessment among advanced soft tissue sarcoma patients receiving trabectedin as a salvage therapy. Ann Oncol. 2013;24(2):537–42.

    Article  CAS  Google Scholar 

  34. Tafuri G, Lucas I, Estevão S, Moseley J, d’Andon A, Bruehl H, et al. The impact of parallel regulatory–health technology assessment scientific advice on clinical development. Assessing the uptake of regulatory and health technology assessment recommendations. Br J Clin Pharmacol. 2018;84(5):1013–9.

    Article  Google Scholar 

  35. Goncalves E. Advanced therapy medicinal products: value judgement and ethical evaluation in health technology assessment. Eur J Health Econ. 2020;21(3):311–20.

    Article  Google Scholar 

  36. Bullement A., Hatswell AJ, Parkinson B, Bharmal M. Health technology assessment of curative interventions—an old problem with new issues. ISPOR Value & Outcomes Spotlight. 2019. https://www.ispor.org/docs/default-source/publications/value-outcomes-spotlight/may-june-2019/featurehta-of-curative-interventions.pdf?sfvrsn=4ad9f533_2. Accessed Mar 2021.

Download references

Acknowledgements

The authors would like to thank Adam Lloyd, Beth Wehler, Qianyi Li and Peter Rouse for their contribution to this manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jennifer G. Gaultney.

Ethics declarations

Funding and conflict of interest

JG and SK received funding for medical writing assistance from Bayer. JC has no funding sources or conflicts of interest to declare. RC has received research grants from Blue Shield of California Foundation, California Healthcare Foundation, Laura and John Arnold Foundation, and NE States Consortium Systems Organization, outside the submitted work; other support received from Aetna, AHIP, Allergan, Alnylam, Anthem, AstraZeneca, Biogen, Blue Shield of California, Boehringer-Ingelheim, Cambia Health Services, CVS, Editas, Express Scripts, Genentech/Roche, GlaxoSmithKline, Harvard Pilgrim Health Care, Health Care Service Corporation, Health Partners, Johnson & Johnson (Janssen), Kaiser Permanente, LEO, Mallinckrodt, Merck, National Pharmaceutical Council, Novartis, Premera, Prime Therapeutics, Regeneron, Sanofi, Spark Therapeutics, and United Healthcare, as dues for annual Policy Summit meeting. AU is an employee of Bayer. CB has received speaking honoraria from Merck KGaA, Sanofi, Roche, Bayer, BMS, AstraZeneca and MSD. CB has received consulting fees from, Lilly, Merck Serono, Sanofi, Bayer, MSD, GSO and AOK Health Insurance. CB has further received research funding from Abbvie, ADC Therapeutics, Agile Therapeutics, Alexion Pharmaceuticals, Amgen, Apellis Pharmaceuticals, AstraZeneca, Bayer, BarGen Bio, Blueprint Medicines, BMS, Boehringer Ingelheim, Celgene, Daiichi Sankyo, Eisai, Gilead, Glycotope, GSK, Incyte, IO Biotech, Isofol Medical, Janssen, Karyopharm Therapeutics, Lilly, Millennium, MSD, Nektar, Novartis, Rafael Pharmaceuticals, Roche, Springworks Therapeutics, and Taiho Pharmaceutical. CB has received travel grants from Merck Serono, Sanofi, Pfizer, and BMS. JW has received advisory board and lecture fees from Amgen, AstraZeneca, Bayer, Blueprint, BMS, Boehringer-Ingelheim, Chugai, Daiichi Sankyo, Ignyta, Janssen, Lilly, Loxo, MSD, Novartis, Pfizer, Roche, Seattle Genetics, and Takeda. JW has received research grants from BMS, Janssen, Novartis, and Pfizer. OSM and AB have received consulting fees, travel grants and speaking honoraria from Bayer.

Author contributions

All authors were involved in the manuscript conception. JG and SK drafted the article. JB, RC, AU, CB, OSM, JW and AB provided critical revision of the article. All authors provided final approval of the version to be published.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gaultney, J.G., Bouvy, J.C., Chapman, R.H. et al. Developing a Framework for the Health Technology Assessment of Histology-independent Precision Oncology Therapies. Appl Health Econ Health Policy 19, 625–634 (2021). https://doi.org/10.1007/s40258-021-00654-4

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40258-021-00654-4

Navigation