Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Perspective
  • Published:

The value of anticancer drugs — a regulatory view

Abstract

The high prices of new anticancer drugs and the marginal added benefit perceived by some stakeholders have fuelled a debate on the value of anticancer drugs in the European Union, even though an agreed definition of what constitutes a drug’s value does not exist. In this Perspective, we discuss the value of drugs from different viewpoints and objectives of decision makers: for regulators, assessment of the benefit–risk balance of a drug is a cornerstone for approval; payers rely on cost-effectiveness analyses carried out by health technology assessment agencies for reimbursement decisions; for patients, treatment choices are based on personal preferences and attitudes to risk; and clinicians can use several scales (such as the ESMO Magnitude of Clinical Benefit Scale (ESMO–MCBS)) that have been developed as an attempt to measure value objectively. Although a unique definition that fully captures the concept of value is unlikely to emerge, herein we discuss the importance of understanding different perspectives, and how regulators can help to inform different decision makers.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Clinical trials versus real-world evidence.

Similar content being viewed by others

References

  1. Garattini, S. Evaluation of benefit-risk. Pharmacoeconomics 28, 981–986 (2010).

    Article  PubMed  Google Scholar 

  2. Prasad, V., McCabe, C. & Mailankody, S. Low-value approvals and high prices might incentivize ineffective drug development. Nat. Rev. Clin. Oncol. 15, 399–400 (2018).

    Article  PubMed  Google Scholar 

  3. Vokinger, K. et al. Prices and clinical benefit of cancer drugs in the USA and Europe: a cost-benefit analysis. Lancet Oncol. 21, 664–670 (2020).

    Article  CAS  PubMed  Google Scholar 

  4. Jonsson, B., Martinalbo, J. & Pignatti, F. European Medicines Agency perspective on oncology study design for marketing authorization and beyond. Clin. Pharmacol. Ther. 101, 577–579 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. McCabe, C. et al. Market and patient access to new oncology products in Europe: a current, multidisciplinary perspective. Ann. Oncol. 20, 403–412 (2009).

    Article  CAS  PubMed  Google Scholar 

  6. Vogler, S., Kilpatrick, K. & Babar, Z. Analysis of medicine prices in New Zealand and 16 European countries. Value Health 18, 484–492 (2015).

    Article  PubMed  Google Scholar 

  7. Pignatti, F. et al. Structured frameworks to increase the transparency of the assessment of benefits and risks of medicines: current status and possible future directions. Clin. Pharmacol. Ther. 98, 522–533 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Davis, C. et al. Availability of evidence of benefits on overall survival and quality of life of cancer drugs approved by European Medicines Agency: retrospective cohort study of drug approvals 2009-13. BMJ 359, j4530 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Eichler, H. G., Hurts, H., Broich, K. & Rasi, G. Drug regulation and pricing–can regulators influence affordability? N. Engl. J. Med. 374, 1807–1809 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Pinilla-Dominguez, P., Naci, H., Osipenko, L. & Mossialos, E. NICE’s evaluations of medicines authorized by EMA with conditional marketing authorization or under exceptional circumstances. Int. J. Technol. Assess. Health Care 36, 426–433 (2020).

    Article  Google Scholar 

  11. Mason, A. & Drummond, M. Public funding of new cancer drugs: is NICE getting nastier? Eur. J. Cancer 45, 1188–1192 (2009).

    Article  PubMed  Google Scholar 

  12. Squires, H., Stevenson, M., Simpson, E., Harvey, R. & Stevens, J. Trastuzumab emtansine for treating HER2-positive, unresectable, locally advanced or metastatic breast cancer after treatment with trastuzumab and a taxane: an Evidence Review Group perspective of a NICE Single Technology appraisal. Pharmacoeconomics 34, 673–680 (2016).

    Article  PubMed  Google Scholar 

  13. Aggarwal, A., Fojo, T., Chamberlain, C., Davis, C. & Sullivan, R. Do patient access schemes for high-cost cancer drugs deliver value to society? – lessons from the NHS Cancer Drugs Fund. Ann. Oncol. 28, 1738–1750 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ades, F. et al. An exploratory analysis of the factors leading to delays in cancer drug reimbursement in the European Union: the trastuzumab case. Eur. J. Cancer 50, 3089–3097 (2014).

    Article  PubMed  Google Scholar 

  15. Wilking, N. et al. Achieving equal and timely access to innovative anticancer drugs in the European Union (EU): summary of a multidisciplinary CECOG-driven roundtable discussion with a focus on Eastern and South-Eastern EU countries. ESMO Open 4, e000550 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Prasad, V. & Vandross, A. Characteristics of exceptional or super responders to cancer drugs. Mayo Clin. Proc. 90, 1639–1649 (2015).

    Article  PubMed  Google Scholar 

  17. Simon, T., Khouri, M., Kou, T. & Gomez-Caminero, A. Realizing the potential of the patient perspective. Patient Prefer. Adherence 14, 2001–2007 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Delyon, J., Maio, M. & Lebbé, C. The ipilimumab lesson in melanoma: achieving long-term survival. Semin. Oncol. 42, 387–401 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Cherny, N. et al. A standardised, generic, validated approach to stratify the magnitude of clinical benefit that can be anticipated from anti-cancer therapies: the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS). Ann. Oncol. 26, 1547–1573 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Cherny, N. et al. ESMO–Magnitude of Clinical Benefit Scale version 1.1. Ann. Oncol. 28, 2340–2366 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Schnipper, L. et al. American Society of Clinical Oncology statement: a conceptual framework to assess the value of cancer treatment options. J. Clin. Oncol. 33, 2563–2577 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Schnipper, L. et al. Updating the American Society of Clinical Oncology value framework: revisions and reflections in response to comments received. J. Clin. Oncol. 34, 2925–2933 (2016).

    Article  PubMed  Google Scholar 

  23. Slomiany, M., Madhavan, P., Kuehn, M. & Richardson, S. Value frameworks in oncology: comparative analysis and implications to the pharmaceutical industry. Am. Heal. Drug Benefits 10, 253–260 (2017).

    Google Scholar 

  24. Shah-Manek, B., Wong, W., Ravelo, A. & DiBonaventura, M. Oncologists’ perceptions of drug affordability using NCCN evidence blocks: results from a national survey. J. Manag. Care Spec. Pharm. 24, 565–571 (2018).

    PubMed  Google Scholar 

  25. Eichler, H. G., Thomson, A., Eichler, I. & Schneeweiss, S. Assessing the relative efficacy of new drugs: an emerging opportunity. Nat. Rev. Drug Discov. 14, 443–444 (2015).

    Article  CAS  PubMed  Google Scholar 

  26. Eichler, H. G. et al. Relative efficacy of drugs: an emerging issue between regulatory agencies and third-party payers. Nat. Rev. Drug Discov. 9, 277–291 (2010).

    Article  CAS  PubMed  Google Scholar 

  27. Hilal, T., Sonbol, M. & Prasad, V. Analysis of control arm quality in randomized clinical trials leading to anticancer drug approval by the US Food and Drug Administration. JAMA Oncol. 5, 887–892 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Tao, D. & Prasad, V. Choice of control group in randomised trials of cancer medicine: are we testing trivialities? Lancet Oncol. 19, 1150–1152 (2018).

    Article  PubMed  Google Scholar 

  29. Bartoli, L., Ferracane, E., Trippoli, S. & Messori, A. First-line treatments for chronic lymphocytic leukemia: analysis of 7 trials based on the restricted mean survival time. Int. J. Clin. Pharmacol. Ther. 59, 322–327 (2021).

    Article  CAS  PubMed  Google Scholar 

  30. Kuhr, K., Wirth, D., Srivastava, K., Lehmacher, W. & Hellmich, M. First-line therapy for non-transplant eligible patients with multiple myeloma: direct and adjusted indirect comparison of treatment regimens on the existing market in Germany. Eur. J. Clin. Pharmacol. 72, 257–265 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Slevin, M. et al. Attitudes to chemotherapy: comparing views of patients with cancer with those of doctors, nurses, and general public. BMJ 300, 1458–1460 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Pignatti, F. et al. The European Medicines Agency: an overview of its mission, responsibilities, and recent initiatives in cancer drug regulation. Clin. Cancer Res. 17, 5220–5225 (2011).

    Article  PubMed  Google Scholar 

  33. Schulman, K. A., Greicius, M. D. & Richman, B. Will CMS find aducanumab reasonable and necessary for Alzheimer disease after FDA approval? JAMA 326, 383–384 (2021).

    Article  PubMed  Google Scholar 

  34. Crosson, F. J., Covinsky, K. & Redberg, R. F. Medicare and the shocking US Food and Drug Administration approval of aducanumab: crisis or opportunity? JAMA Intern. Med. 181, 1278–1280 (2021).

    Article  PubMed  Google Scholar 

  35. Kleijnen, S. et al. European collaboration on relative effectiveness assessments: what is needed to be successful? Health Policy 119, 569–576 (2015).

    Article  PubMed  Google Scholar 

  36. Kristensen, F. et al. European network for Health Technology Assessment, EUnetHTA: planning, development, and implementation of a sustainable European network for Health Technology Assessment. Int. J. Technol. Assess. Health Care 25, 107–116 (2009).

    Article  PubMed  Google Scholar 

  37. Berntgen, M. et al. Improving the contribution of regulatory assessment reports to health technology assesments – a collaboration betwen the European Medicines Agency and the European network for Health Technology Assessment. Value Heal. 17, 634–641 (2014).

    Article  Google Scholar 

  38. Schulthess, D. et al. Medicines adaptive pathways to patients (MAPPs): a story of international collaboration leading to implementation. Ther. Innov. Regul. Sci. 50, 347–354 (2016).

    Article  PubMed  Google Scholar 

  39. Tenhunen, O., Lasch, F., Schiel, A. & Turpeinen, M. Single-arm clinical trials as pivotal evidence for cancer drug approval: a retrospective cohort study of centralized European marketing authorizations between 2010 and 2019. Clin. Pharmacol. Ther. 108, 653–660 (2020).

    Article  PubMed  Google Scholar 

  40. Postmus, D. et al. Incorporating patient preferences into drug development and regulatory decision making: results from a quantitative pilot study with cancer patients, carers, and regulators. Clin. Pharmacol. Ther. 99, 548–554 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Topp, M. S. et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 16, 57–66 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Kim, D.-W. et al. Activity and safety of 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. 17, 452–463 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kantarjian, H. et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N. Engl. J. Med. 376, 836–847 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Shaw, A. T. et al. Ceritinib versus chemotherapy in patients with ALK-rearranged non-small-cell lung cancer previously given chemotherapy and crizotinib (ASCEND-5): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 18, 874–886 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Shaw, A. et al. Ceritinib in ALK-rearranged non-small-cell lung cancer. N. Engl. J. Med. 370, 1189–1197 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wallerstedt, S. & Henriksson, M. Balancing early access with uncertainties in evidence for drugs authorized by prospective case series – systematic review of reimbursement decisions. Br. J. Clin. Pharmacol. 84, 1146–1155 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Johnson, K., Liauw, W. & Lassere, M. Evaluating surrogacy metrics and investigating approval decisions of progression-free survival (PFS) in metastatic renal cell cancer: a systematic review. Ann. Oncol. 26, 485–496 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Early Breast Cancer Trialists’ Collaborative Group. Systemic treatment of early breast cancer by hormonal, cytotoxic, or immune therapy. 133 randomised trials involving 31,000 recurrences and 24,000 deaths among 75,000 women. Lancet 339, 71–85 (1992).

    Article  Google Scholar 

  49. Bryson, H. M. & Plosker, G. L. Tamoxifen: a review of pharmacoeconomic and quality-of-life considerations for its use as adjuvant therapy in women with breast cancer. Pharmacoeconomics 4, 40–66 (1993).

    Article  CAS  PubMed  Google Scholar 

  50. Neven, P. & Vergote, I. Tamoxifen, screening and new oestrogen receptor modulators. Best. Pract. Res. Clin. Obstet. Gynaecol. 15, 365–380 (2001).

    Article  CAS  PubMed  Google Scholar 

  51. Hind, D. et al. Hormonal therapies for early breast cancer: systematic review and economic evaluation. Health Technol. Assess. 11, 1–134 (2007).

    Article  PubMed  Google Scholar 

  52. Garsen, M., Steenhof, M. & Zwiers, A. A decade of marketing authorization applications of anticancer drugs in the European Union: an analysis of procedural timelines. Ther. Innov. Regul. Sci. 55, 633–642 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Piatkiewicz, T., Traulsen, J. & Holm-Larsen, T. Risk-sharing agreements in the EU: a systematic review of major trends. Pharmacoecon. Open 2, 109–123 (2018).

    Article  PubMed  Google Scholar 

  54. Gaultney, J. G. et al. Experience with outcomes research into the real-world effectiveness of novel therapies in Dutch daily practice from the context of conditional reimbursement. Health Policy 119, 186–194 (2015).

    Article  PubMed  Google Scholar 

  55. Ermisch, M. et al. Payers’ views of the changes arising through the possible adoption of adaptive pathways. Front. Pharmacol. 7, 305 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Vella Bonanno, P. et al. Adaptive pathways: possible next steps for payers in preparation for their potential implementation. Front. Pharmacol. 8, 497 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Keyter, A., Salek, S., Banoo, S. & Walker, S. Can standardisation of the public assessment report improve benefit-risk communication? Front. Pharmacol. 11, 855 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Kleijnen, S. et al. Standardized reporting for rapid relative effectiveness assessments of pharmaceuticals. Int. J. Technol. Assess. Health Care 30, 488–496 (2014).

    Article  PubMed  Google Scholar 

  59. Walker, S., McAuslane, N., Liberti, L., Leong, J. & Salek, S. A universal framework for the benefit-risk assessment of medicines: is this the way forward? Ther. Innov. Regul. Sci. 49, 17–25 (2015).

    Article  PubMed  Google Scholar 

  60. Dittrich, C., Negrouk, A. & Casali, P. An ESMO-EORTC position paper on the EU clinical trials regulation and EMA’s transparency policy: making European research more competitive again. Ann. Oncol. 26, 829–832 (2015).

    Article  CAS  PubMed  Google Scholar 

  61. Bonini, S., Eichler, H., Wathion, N. & Rasi, G. Transparency and the European Medicines Agency–sharing of clinical trial data. N. Engl. J. Med. 371, 2452–2455 (2014).

    Article  PubMed  Google Scholar 

  62. Orsini, L. et al. Improving transparency to build trust in real-world secondary data studies for hypothesis testing–why, what, and how: recommendations and a road map from the real-world evidence transparency initiative. Value Health 23, 1128–1136 (2020).

    Article  PubMed  Google Scholar 

  63. Jaksa, A. et al. Organized structure of real-world evidence best practices: moving from fragmented recommendations to comprehensive guidance. J. Comp. Eff. Res. 10, 711–731 (2021).

    Article  PubMed  Google Scholar 

  64. Eichler, H. et al. From adaptive licensing to adaptive pathways: delivering a flexible life-span approach to bring new drugs to patients. Clin. Pharmacol. Ther. 97, 234–246 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Saesen, R., Lacombe, D. & Huys, I. Design, organisation and impact of treatment optimisation studies in breast, lung and colorectal cancer: the experience of the European Organisation for Research and Treatment of Cancer. Eur. J. Cancer 151, 221–232 (2021).

    Article  PubMed  Google Scholar 

  66. European Medicines Agency. Guideline on the Clinical Evaluation of Anticancer Medicinal Products– revision 6. https://www.ema.europa.eu/en/documents/scientific-guideline/draft-guideline-evaluation-anticancer-medicinal-products-man-revision-6_en.pdf (2020).

  67. Sequist, L. et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J. Clin. Oncol. 31, 3327–3334 (2013).

    Article  CAS  PubMed  Google Scholar 

  68. Shaw, A. et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N. Engl. J. Med. 368, 2385–2394 (2013).

    Article  CAS  PubMed  Google Scholar 

  69. Hodi, F. et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363, 711–723 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Chapman, P. et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N. Engl. J. Med. 364, 2507–2516 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. McArthur, G. et al. Safety and efficacy of vemurafenib in BRAF(V600E) and BRAF(V600K) mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study. Lancet Oncol. 15, 323–332 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Hauschild, A. et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 380, 358–365 (2012).

    Article  CAS  PubMed  Google Scholar 

  73. Cortes, J. et al. Eribulin monotherapy versus treatment of physician’s choice in patients with metastatic breast cancer (EMBRACE): a phase 3 open-label randomised study. Lancet 377, 914–923 (2011).

    Article  CAS  PubMed  Google Scholar 

  74. Baselga, J. et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N. Engl. J. Med. 366, 109–119 (2012).

    Article  CAS  PubMed  Google Scholar 

  75. Swain, S. et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N. Engl. J. Med. 372, 724–734 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Verma, S. et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N. Engl. J. Med. 367, 1783–1791 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. de Bono, J. et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet 376, 1147–1154 (2010).

    Article  PubMed  Google Scholar 

  78. de Bono, J. et al. Abiraterone and increased survival in metastatic prostate cancer. N. Engl. J. Med. 364, 1995–2005 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Scher, H. et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N. Engl. J. Med. 367, 1187–1197 (2012).

    Article  CAS  PubMed  Google Scholar 

  80. Parker, C. et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N. Engl. J. Med. 369, 213–223 (2013).

    Article  CAS  PubMed  Google Scholar 

  81. Van Cutsem, E. et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J. Clin. Oncol. 30, 3499–3506 (2012).

    Article  PubMed  Google Scholar 

  82. Grothey, A. et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 381, 303–312 (2013).

    Article  CAS  PubMed  Google Scholar 

  83. Fuchs, C. et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 383, 31–39 (2014).

    Article  CAS  PubMed  Google Scholar 

  84. European Commission. Core principles on relative effectiveness (Pharmaceutical forum– Working Group on Relative Effectiveness). https://ec.europa.eu/docsroom/documents/7581?locale=en (2014).

  85. European Parliament: Directorate-General for Internal Policies. Differences in costs of and access to pharmaceutical products in the EU. https://www.europarl.europa.eu/RegData/etudes/etudes/join/2011/451481/IPOL-ENVI_ET(2011)451481_EN.pdf (2011).

Download references

Author information

Authors and Affiliations

Authors

Contributions

F.P., U.W., D.P., N.W. and J.D. researched data for the article; all the authors discussed the manuscript contents; F.P., U.W. and J.D. wrote the article; and all the authors reviewed it before submission.

Corresponding author

Correspondence to Julio Delgado.

Ethics declarations

Competing interests

The authors declare no competing interests.

Disclaimer

The views expressed in this article are the personal views of the authors and may not be understood or quoted as being made on behalf of or reflecting the position of the agencies and organizations with which the authors are employed or affiliated.

Additional information

Dedication

We dedicate this paper to Bertil Jonsson, clinical assessor, Medical Products Agency, Sweden, and past vice-chairman of the EMA Scientific Advice Working Party and of the Oncology Working Party.

Peer review information

Nature Reviews Clinical Oncology thanks J. Del Paggio, C. A. Uyl-de Groot and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

Benefit–risk evaluations

Together with evaluation of the ‘quality’, ‘safety’ and ‘efficacy’ of a new drug, the evaluation of the benefit−risk balance is the cornerstone of the scientific opinions of regulatory agencies (including the EMA) when assessing new drug applications. This evaluation is based on the balance between the favourable effects (benefits) of a medicine against its unfavourable effects (harms, commonly referred to as ‘risks’). Regulatory agencies can only recommend authorization of medicines with a positive benefit–risk balance. In conventional marketing authorizations, regulatory agencies do not evaluate the benefit–risk balance of medicines in the context of all approved drugs for the same indication, but instead base their assessments on the ‘absolute’ benefit–risk (exclusively the benefits versus the harms from the drug).

Cost-effectiveness

Cost-effectiveness analysis compares the relative costs and relative effects of two or more courses of action. Effects can be measured, for example, in years of life gained from the intervention or number of surgical procedures avoided85.

Effectiveness

This term refers to the extent to which an intervention is able to cause the intended pharmacological effects when provided under the usual circumstances of health-care practice, also referred to as ‘in real life’. Effectiveness is distinguished from efficacy to refer to the smaller magnitude of effects often assumed or observed at the population level when the medicine is provided under the usual circumstances compared with the ideal circumstances of a controlled clinical trial setting (owing, among other things, to patient selection or monitoring)84. This concept is mainly used in the context of health technology assessment and is not a regulatory requirement for a marketing authorization of new medicines.

Efficacy

This term generally refers to the ability of a medicine to cause the intended pharmacological effects (referred to as ‘benefits’ or ‘favourable effects’, as opposed to ‘harms’, ‘risks’ or ‘unfavourable effects’) under ideal circumstances. In oncology, efficacy is often measured directly using clinical outcomes such as overall survival in randomized controlled trials. Efficacy is often distinguished from ‘activity’, typically measured in single-arm trials and evaluating a pharmacodynamic effect that is not necessarily associated with a clinical effect, such as tumour shrinkage on imaging. Efficacy is also often distinguished from ‘effectiveness’84.

Health technology assessment

(HTA). Systematic evaluation of the properties and effects of a health technology, addressing its intended and unintended consequences, and aimed at informing decision-making. HTAs involve evaluation of clinical effectiveness, safety, cost-effectiveness and, when broadly applied, societal, ethical and legal aspects. A major application of HTAs is in informing reimbursement and coverage decisions by insurers and national health-care systems.

Relative effectiveness

Comparison of an intervention with available treatments — that is, this term refers to the extent to which an intervention does more good than harm compared with one or more alternative interventions under the usual circumstances of health-care practice. The concept differs from ‘effectiveness’ owing to its comparative nature84. A similar distinction exists for efficacy and relative efficacy. This concept is mainly used in the context of health technology assessment and is not a regulatory requirement for a marketing authorization of new medicines.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pignatti, F., Wilking, U., Postmus, D. et al. The value of anticancer drugs — a regulatory view. Nat Rev Clin Oncol 19, 207–215 (2022). https://doi.org/10.1038/s41571-021-00584-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41571-021-00584-z

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research