1,941
Views
11
CrossRef citations to date
0
Altmetric
Editorial

Personalized medicine and economic evaluation in oncology: all theory and no practice?

, &

Abstract

The clinical definition of personalized medicine (PM) is closely related to that of pharmacogenomics. Ideally, PM could lead the pharmaceutical industry to differentiate products by subgroups of patients with the same pathology and find new gene targets for drug discovery. Here, we focus on the potential impact of PM on the design of clinical trials and economic evaluations limited to oncology (its first and main field of application). Then, we assess the European economic evaluations focused on trastuzumab and cetuximab, the two drugs usually mentioned as emblematic examples of targeted therapies. Clinical results of PM in oncology have not been as encouraging as hoped so far. Of course, economic evaluations on targeted therapies cannot help overcome the lack of clinical evidence for most of them. The two paradigmatic examples of cetuximab and trastuzumab indicate that the methodological implications on economic evaluations debated in the literature are more theoretical than practical.

Personalized medicine (PM) is a much discussed topic at present and serves as an aspiration to industry to support the development of innovative drugs that will be more effective, safer and, of course, command premium prices. But what is PM? What opportunities and challenges does it offer in clinical development and economic evaluation?

Defining PM

Increasing numbers of articles on PM have been published. Four dedicated journals were launched recently Citation[1–4], a European Association was founded in 2009 Citation[5] and a leading US academic institution ‘curates knowledge about the impact of genetic variation on drug response for clinicians and researchers’ Citation[6].

Although a bibliographic attempt to support a unique definition of PM has been published Citation[7], various competing definitions persist in the literature Citation[8], using somewhat different nomenclature (e.g., ‘stratified’/’individualized’ instead of ‘personalized’). All definitions, however, share the use of some form of genetic testing aiming to identify and target specific characteristics of the patient being treated, to give ‘the right drug to the right patient’ to maximize the effectiveness and safety of the treatment Citation[9]. Thus, ‘personalization’, based on molecular investigation, is conceptually different from ‘personalization’ tailored to the patient’s preferences and choices, which is a common feature of good quality healthcare services Citation[10]. The clinical definition of PM is closely related to that of pharmacogenomics, that is, the effects of an individual’s genetic variability on drug response Citation[11]. The desired result of pharmacogenomics testing is to maximize efficacy and/or safety by matching the best available drug or dose to an individual’s genomic profile. The EMA has issued various scientific guidance documents on pharmacogenomics since 2002 and appointed a specific working party that gives recommendations on this subject Citation[12].

The first notion of the importance of pharmacogenetic variance dates back to the Second World War and referred to American soldiers who were more or less prone to malaria depending on their ethnicity Citation[13]. However, the first identified use of the term PM in the context of ‘the right drug to the right patient’ was in an article on patients with cancer in 1999 Citation[9]. Many publications followed Citation[14], focusing on the advances that the human genome project and its technology spin-offs could bring in drugs discovery and development for the pharmaceutical industry Citation[15]. Pharmacogenomics claimed that major new drug advances would come from understanding the genetic basis of diseases. Although there is still a chasm between identifying a genetic susceptibility and developing safe and effective medicines Citation[16], the pharmaceutical industry increasingly recognized that ‘one-size-fits-all’ is not the only strategy, particularly when it is at least in part broken with many failures in late-stage development, and it had instead to consider the potential for ‘tailored’ medicines, which will cure patients according to their individual genetic makeup. This may have been fuelled by the very high prices that can be achieved for effective medicines in oncology. In marketing terms, research-oriented companies started switching from simply attempting to identify ‘block-busters’ to recognizing the value of ‘niche-busters’ Citation[17].

Understanding the role of genes is the PM cornerstone. The exploration of genes involved in different pharmacological responses may potentially be used to identify responders, nonresponders and patients at higher risk of side effects, which qualitatively affect the balance of benefits and risks. Accordingly, PM has the potential to play an important role in several areas Citation[18,19], particularly for a better definition of a disease and consequently the identification through genetic markers of subtypes of patients as specific targets. The basic idea with this kind of investigation is the possibility of diagnostic, prognostic and therapeutic strategies precisely tailored to each patient’s requirements Citation[20]. Hence, the latter-day shift from the expression ‘personalized’ to ‘precision’ medicine announced by the US President as one of the main challenges that scientific research will have to face. Ideally, PM could lead the pharmaceutical industry to: differentiate products by subgroup of patients with the same pathology, and find new gene targets for drug discovery. Patients should benefit from better health through: the higher probability of the desired outcome with a drug, and the lower probability of untoward side effects Citation[21].

Evaluating PM

Here, we focus on PM limited to oncology (its first and main field of application) Citation[16,20], that is, prospective genotyping with the aim of identifying patients on the basis of their genetic profile who, when prescribed anticancer medicines, have a substantially better ratio of benefits to risks Citation[22]. After a general background of PM, we review its potential impact on the design of clinical trials and economic evaluations, mainly referring to Europe as a setting. Then, we assess the ‘state of the art’ of the economic evaluations focused on the two drugs that are usually mentioned as emblematic examples of targeted therapies, that is, trastuzumab (TR) and cetuximab (CX), in their first indications (respectively, metastatic breast and colorectal cancers).

PM & clinical trials

Clinical trials on targeted therapies become more complicated, since the choice is no longer between a new medicine and an existing one but rather between ‘treat-all’ and ‘test-and-treat’ strategies. Some have argued that, before randomizing patients to the new drug, they should be randomized to the diagnostic test too, to prove its efficacy Citation[23]. But, more realistically, this implies a two-stage process in which we evaluate first the screening test, and then seek to confirm the effectiveness of the treatment in the identified population. Ideally, epidemiological data should be available to estimate the prevalence of the biomarker and the predictive value of the test. In general, PM should result in the need for more and different data, to reflect accurately the impact of a diagnostic test on a patient care pathway Citation[10]. These results should continue to be estimated from clinical trials, supported by well-designed prospective observational cohorts analyzed with appropriate methods and adjustments for confounding variables.

Another important issue is the timing of the diagnostic test, either together with the drug (e.g., TR) or after drug approval and thus requiring a post hoc retrospective analysis (e.g., CX). In this second case, if pharmacogenomic data were collected alongside a clinical trial, post hoc subanalyses could detect an interaction between drug response and a particular genotype in subgroups of patients, although such analyses would face challenges of multiplicity and false-positive findings. Finally, the results from companion tests are usually continuous rather than binary, so dichotomizing their scores (negative/positive) to set a cutoff for treating/not treating patients could be questionable Citation[10].

PM & economic evaluations

In the recent pharmacoeconomic literature, it has often been noted that PM needs particular guidelines to assess the costs and benefits of alternatives correctly Citation[23–25], again mainly due to the genetic tests, which drive targeted therapies and thus increase the number of potential alternatives. In principle, the cost of testing the whole population of patients should be posted only to the targeted therapy if the test was not routinely adopted before the therapy was introduced Citation[26].

Economic evaluations on targeted therapies should require not only a more complex decision space but also a clear understanding on what tests will be done after the initial one and what treatment will be given after the tests Citation[23]. In practice, even when a biomarker has been mentioned as a part of the product approval to target the eligible population by EMA, the technological procedure to be used will generally remain unspecified. To assess diagnostic performance (in terms of sensitivity and specificity) Citation[23] and its costs, one should know what test will actually be used in clinical practice – a laboratory-made test or a specific test marketed by a diagnostic company (or the pharmaceutical company itself) with a self-certified CE mark – since the way patients are managed could differ widely among centers Citation[26]. The choice of a particular test should also depend on the prevalence of the biomarker, the cost of testing and the payer’s strategy to treat all patients or not with stratified therapies depending on budgetary constraints. Finally, the different funding options in the healthcare systems should be taken into account when monetizing the costs of the targeted therapy and its ‘companion test’. For instance, the two main types of funding in the EU countries for TR in breast cancer are test and drug either through a hospital or a combination between the hospital and the third-party payer Citation[27]. Finally, the number of biomarkers for targeting care is likely to keep on increasing, so many genomic tests could become ‘obsolete’ very fast Citation[10], undermining the credibility of economic evaluations conducted according to long-term horizons.

Accordingly, the uncertainty of economic evaluations in PM is clear, both about their frame and the inputs concerning the two technologies (the drug and its companion test) Citation[23], calling for thorough sensitivity analysis. This might imply reconsidering their cost–effectiveness later in the technological lifecycle, particularly if new biomarkers for targeting care become available in the meantime Citation[10].

Two emblematic examples of targeted therapies

Two separate literature searches were conducted on the PubMed international database to select full economic evaluations conducted in Europe and published in English from January 2000 for TR and January 2008 for CX (post hoc evidence dates from that year) till May 2015. The search terms were ‘trastuzumab’ and ‘metastatic breast cancer’ or ‘cetuximab’ and ‘metastatic colorectal cancer’ combined with ‘cost’ or ‘cost–effectiveness’ or ‘economic evaluation’.

We retrieved 93 articles for TR: 83 were discarded because they did not include a full economic evaluation on TR as first-line innovative therapy in metastatic breast cancerFootnote1. Since five full economic evaluations did not concern the European setting, we selected five articles eventually Citation[28–32]. We retrieved 58 articles for CX and 47 were discardedFootnote2. Since seven full economic evaluations were not conducted in European countries, we selected four studies finally Citation[33–36].

We reviewed all nine selected studies according to a restricted checklist focused on the items relevant to a PM scenario: source of efficacy; number of alternatives; allocation and assessment of the companion test cost; proportion of companion test and targeted therapy costs on total costs; and most influential variables in sensitivity analysis.

The nine full EEs concerned six European jurisdictions , six of them concluding in favor of targeted therapies Citation[29–31,33–35] and the other three against Citation[28,32,36]. While CX efficacy was entirely derived from post hoc analyses – in one study Citation[34] adjusted through an expert panel – TR efficacy was assessed from randomized clinical trials in all but the two French studies Citation[30,31], which instead used observational studies based on uncontrolled, small samples of patients Citation[37]. All the clinical trials included had high proportions of cross-over patients.

Table 1. CX & TR studies reviewed according to our checklist focused on PM.

Four studies had more than two alternatives through the inclusion of companion tests Citation[29,33,35,36]. The cost of testing was not considered at all in three studies Citation[31,32,34] and only two of the remaining six Citation[28,36] fully attributed it to the targeted therapy alternative. The source of the companion tests’ unit cost was weak in half of these six studies Citation[28,30,35], and the proportion of the test cost on the total was negligible (<1.5%) in all of them, while that of the targeted therapy was substantial in all nine studies. As a consequence, while the targeted therapy cost was an influential variable in seven of the eight studies that conducted a sensitivity analysis, this was never the case for the companion test cost, even though its unit cost was quite high (>€200) in four studies.

Policy implications

PM is a fascinating subject in principle; however, clinical results have not been as encouraging as hoped so far, particularly in oncology, which was expected to be its major field of application and where targeted therapies have not yet been able to replace classical chemotherapy Citation[22]. Of course, there may be many reasons behind these disappointing results and some caution should be exercised about calling genes ‘cancer-related’ since not all somatic mutations are tumor driving and need to be seen in the broader context of tumor heterogeneity Citation[16,22]. In general, although target prioritization is a major issue, targeted therapy discovery and development are still the main bottlenecks Citation[22], and the growing number of companion tests might also be a marketing strategy followed by manufacturers to differentiate their bioagents Citation[26]. Somewhat cynically, post hoc analyses might be useful to find a subset of patients as a ‘market niche’ for drugs that failed to show efficacy in clinical trials for approval.

Of course, economic evaluations on targeted therapies cannot help overcome the lack of clinical evidence for most of them and improve the still confusing PM ‘state of the art’ Citation[24,25]. The two paradigmatic examples of CX and TR indicate that the methodological implications on economic evaluations debated in the literature Citation[10,23] are more theoretical than practical, since the cost of companion tests was negligible in all the European studies reviewed, so this item could in fact have been excluded from calculations, according to economic evaluation theory Citation[38]. From an economic point of view, the real ‘crux of the matter’ lies in targeted therapies, which are very expensive add-ons whose additional cost can hardly be compensated by ‘trade-offs’ with other healthcare items, particularly in end-of-life situations Citation[37,39]. Public decision makers in European countries where economic evaluations are required for pricing and reimbursement procedures should be fully aware of these limits when coping with dossiers submitted by targeted therapy manufacturers.

Financial & competing interests disclosure

N Freemantle has received funding for research and consulting from a number of companies that may be currently developing personalized medicine strategies. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Notes

183 articles were excluded, being: focused on clinical issues or other subjects not related to our aim(50); reviews or comments (13); partial economic evaluations (11); and full economic evaluations not focused on TR for treatment in metastatic cancer (9).

247 articles were excluded, being: focused on clinical issues or other subjects not related to our aim (27); reviews or comments (11); partial economic evaluations (8) and one full economic evaluation not focused on CX.

References

  • Personalized medicine. Available from: www.futuremedicine.com/loi/pme
  • Journal of personalized medicine. Available from: www.mdpi.com/2075-4426/5/1
  • Pharmacogenomics and Personalized Medicine. Available from: www.dovepress.com/pharmacogenomics-and-personalized-medicine-journal
  • Current pharmacogenomics and personalized medicine. Available from: http://benthamscience.com/journal/index.php?journalID=cppm
  • Available from: www.epemed.org/online/www/content/ENG/index.htm
  • Available from: www.pharmgkb.org
  • Schleidgen S, Klingler C, Bertram T, et al. What is personalized medicine: sharpening a vague term based on a systematic literature review. BMC Med Ethics 2013;14:55
  • Redekop WK, Mladsi D. The faces of personalized medicine: a framework for understanding its meaning and scope. ViH 2013;16(6 Suppl):S4-9
  • Langreth R, Waldholz M. New era of personalized medicine: targeting drugs for each unique genetic profile. Oncologist 1999;4(5):426-7
  • Rogowski W, Payne K, Schnell-Inderst P, et al. Concepts of ‘Personalization’ in Personalized Medicine: Implications for Economic Evaluation. Pharmacoeconomics 2015;33(1):49-59
  • Wong WB, Carlson JJ, Thariani R, Veenstra DL. Cost effectiveness of pharmacogenomics: a critical and systematic review. Pharmacoeconomics 2010;28(11):1001-13
  • Available from: www.ema.europa.eu/ema/index.jsp?curl=pages/special_topics/general/general_content_000347.jsp&mid=WC0b01ac05800baeda
  • Mancinelli L, Cronin M, Sadée W. Pharmacogenomics: The Promise of Personalized Medicine. AAPS PharmSci 2000;2(1): Article 4
  • Jorgensen JT. New Era of Personalized Medicine: A 10-Year Anniversary. Oncologist 2009;14:557-8
  • Ginsburg GS, Konstance RP, Allsbrook JS, Schulman KA. Implications of pharmacogenomics for drug development and clinical practice. Arch Intern Med 2005;165(20):2331-6
  • Andre F, Mardis E, Salm M, et al. Prioritizing targets for precision cancer medicine. Ann Oncol 2014;25:2295-303
  • Haycox A, Pirmohamed M, McLeod C, et al. Through a Glass Darkly: Economics and Personalized Medicine. Pharmacoeconomics 2014;32:1055-61
  • EMA/446337/2011 9 June 2011
  • March R. Pharmacogenomics: the genomics of drug response. Yeast 2000;17(1):16-21
  • Collins FS, Varmus H. A new initiative on precision medicine. N Eng J Med 2015;372:793-5
  • Ginsburg GS, McCarthy J. Personalized medicine: revolutionizing drug discovery and patient care. Trends Biotechnol 2001;12(19):491-5
  • Damia G, Garattini S. The pharmacological point of view of resistance to therapy in tumors. Cancer Treat Rev 2014;40(8):909-16
  • Annemans L, Redekop K, Payne K. Current Methodological Issues in the Economic Assessment of Personalized Medicine. Value Health 2013;16:S20-6
  • Chalkidou K, Rawlins M. Pharmacogenetics and cost-effectiveness analysis: a two-way street. Drug Discov Today 2011;16(19-20):873-7
  • Payne K, Shabaruddin FH. Cost-effectiveness analysis in pharmacogenomics. Pharmacogenomics 2010;11(5):643-6
  • Antonanzas F, Juarez-Castello CA, Rodriguez-Ibeas R. Is personalized medicine a panacea for health management? Some thoughts on its desirability. Eur J Health Econ 2015;16:455-7
  • Leopold C, Vogler S, Habl C, et al. Personalised medicine as a challenge for public pricing and reimbursement authorities – A survey among 27 European countries on the example of trastuzumab. Health Policy 2013;113:313-22
  • Norum J, Risberg T, Olsen JA. A monoclonal antibody against HER-2 (trastuzumab) for metastatic breast cancer: a model-based cost-effectiveness analysis. Ann Oncol 2005;16:909-14
  • Lidgren M, Wilking N, Jönsson B, Rehnberg C. Cost-effectiveness of HER2 testing and trastuzumab therapy for metastatic breast cancer. Acta Oncol 2008;47:1018-28
  • Poncet B, Bachelot T, Colin C, et al. Use of the monoclonal antibody anti-HER2 trastuzumab in the treatment of metastatic breast cancer: a cost-effectiveness analysis. Am J Clin Oncol 2008;31:363-8
  • Perez-Ellis C, Goncalves A, Jacquemier J, et al. Cost-Effectiveness Analysis of Trastuzumab (Herceptin) in HER2-Overexpressed Metastatic Breast Cancer. Am J Clin Oncol 2009;32:492-8
  • Matter-Walstra KW, Dedes KJ, Schwenkglenks M, et al. Trastuzumab beyond progression: a cost-utility analysis. Ann Oncol 2010;21:2161-8
  • Blank PR, Moch H, Szucs TD, et al. KRAS and BRAF Mutation Analysis in Metastatic Colorectal Cancer: A Cost-effectiveness Analysis from a Swiss Perspective. Clin Cancer Res 2011;17(19):6338-46
  • Asseburg C, Frank M, Köhne CH, et al. Cost-effectiveness of targeted therapy with cetuximab in patients with K-ras wild-type colorectal cancer presenting with initially unresectable metastases limited to the liver in a German setting. Clin Ther 2011;33(4):482-97
  • Vijayaraghavan A, Efrusy MB, Goke B, et al. Cost-effectiveness of KRAS testing in metastatic colorectal cancer patients in the United States and Germany. Int J Cancer 2012;131:438-45
  • Hoyle M, Peters J, Crathorne L, et al. Cost-Effectiveness of Cetuximab, Cetuximab Plus Irinotecan, and Panitumumab for Third and Further Lines of Treatment for KRAS Wild-Type Patients with Metastatic Colorectal Cancer. Value Health 2013;16:288-96
  • Garattini L, van de Vooren K, Curto A. Cost-effectiveness of trastuzumab in metastatic breast cancer: Mainly a matter of price in the EU? Health Policy 2015;119:212-16
  • Drummond M, Stoddart G, Torrance G. Methods for the economic evaluation of health care programmes. Oxford University Press, Oxford; 1988
  • Garattini L, van de Vooren K, Zaniboni A. Ethics for end-of-life treatments: Metastatic colorectal cancer is one example. Health Policy 2013;109:97-103

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.