You are on page 1of 8

Provided for non-commercial research and educational use only.

Not for reproduction, distribution or commercial use.

This article was originally published in Encyclopedia of Health Economics published by Elsevier, and the copy
attached is provided by Elsevier for the author’s benefit and for the benefit of the author’s institution, for non-
commercial research and educational use including without limitation use in instruction at your institution, sending it
to specific colleagues who you know, and providing a copy to your institution’s administrator.

All other uses, reproduction and distribution, including without limitation commercial reprints, selling or licensing
copies or access, or posting on open internet sites, your personal or institution’s website or repository, are prohibited.
For exceptions, permission may be sought for such use through Elsevier’s permissions site at:

http://www.elsevier.com/locate/permissionusematerial

Garrison L.P., and Towse A. Personalized Medicine: Pricing and Reimbursement Policies as a Potential Barrier to
Development and Adoption, Economics of. In: Anthony J. Culyer (ed.), Encyclopedia of Health Economics, Vol 2.
San Diego: Elsevier; 2014. pp. 484-490.

© 2014 Elsevier Inc. All rights reserved.


Author's personal copy

Personalized Medicine: Pricing and Reimbursement Policies as a Potential


Barrier to Development and Adoption, Economics of
LP Garrison, University of Washington, Seattle, WA, USA
A Towse, Office of Health Economics, London, UK
r 2014 Elsevier Inc. All rights reserved.

Background: Barriers to Personalized Medicine expectancy) based on patient characteristics, whereas a


predictive biomarker predicts response to an intervention
More than 10 years have passed since the completion of the first (e.g., a drug).
sequencing of the human genome in 2001. Since then, there
The slower-than-expected progress over the past decade
has been continued and growing interest in the potential to use
could be for a number of reasons – scientific, regulatory, and
this new genetic information to better predict patient response
economic. It does seem clear that the science is more difficult
to therapy. A variety of terms have been coined to describe this
than first hoped. First, it is always important to remember that
potential: personalized medicine (PM), stratified medicine,
the science behind drug development is complex and un-
tailored medicine, and individualized therapy, among others.
certain. Almost 9 out of 10 new medicines under development
This article will adopt PM as the descriptor, more because of its
fail between Phase 1 and Phase 3. Despite increasing amounts
currency and popularity than because of its more accuracy than
spent on drug development, industry productivity, as meas-
other terms. PM has been defined many a times as ‘‘providing
ured by approved new molecular entities, has been stagnant in
the right treatment to the right patient at the right time.’’
recent years. The most recent estimate from the UK Office of
Arguably, that is the aim of all medical therapy: the important
Health Economics is that the average cost of drug develop-
difference in PM is the use of a new biomarker-based diagnostic
ment has grown to approximately $1.5 billion US$2011 per
test to further define and identify a subgroup of patients (called
new compound. Clearly, the scientific unknowns and chal-
‘stratification’) for whom the treatment performs better – in
lenges are substantial.
terms of either cost-effectiveness or benefit–risk balance.
Adding the parallel development of a predictive, bio-
Incentives for the development and use of PM raise a number
marker-based test may reduce one scientific challenge but adds
of interesting economic issues.
another that involves its own uncertainties. Furthermore,
In the year 2000, Francis Collins, the current head of the
prediction based on genetic makeup is generally imprecise.
US National Institutes of Health, said: ‘‘In the next five to
Although a small number of single mutations (monogenic
seven years, we should identify the genetic susceptibility fac-
diseases) lead to specific health conditions (e.g., Huntington’s
tors for virtually all common diseases – cancer, diabetes, heart
disease), most complex diseases (such as diabetes) are affected
disease, the major mental illnesses – on down that list.’’
by a large number of genes. Although an entire genome can be
Clearly, this has not come to pass. In 2005, a more guarded
sequenced, it is only the beginning of understanding the
assessment in the report ‘Personalized Medicine: Hopes and
biological function of most genes. Although some traits, such
Realities’ from The Royal Society cautioned: ‘‘Pharmacoge-
as height, are highly heritable, others are not: indeed, twin
netics is unlikely to revolutionize or personalize medical
studies indicate that genes account for only approximately a
practice in the immediate future.’’
quarter of the variation in lifespan – perhaps the ultimate
Before considering potential reasons for this lack of ex-
measure of health. Clearly, gene–environment interaction is a
pected progress, it is useful to define some relevant biological,
very important influence. Regulation of drugs and devices may
epidemiological, and clinical concepts and terms:
also be a barrier: the approval pathway for new pharmacoge-
• Pharmacogenetics versus pharmacogenomics – the former nomic tests has not been defined until recently and the
is the study of how people’s genetic makeup affects their standards of evidence for clinical utility for combination
response to medicines, whereas the latter is the application products (i.e., drug and test) are still being debated. Further-
of genomic concepts to the development and clinical more, there is not a level playing field between in vitro diag-
application of pharmaceuticals. nostic tests (IVDs), which need regulatory approval for
• Genotype versus phenotype – the former represents a marketing, and laboratory-developed tests (LDTs) also called
person’s genetic makeup, as reflected by his or her deoxy- in-house tests (IHTs). The evidentiary requirements and
ribonucleic acid (DNA) sequence, whereas the latter is an quality controls for IVDs are much greater. In Europe, public
observable trait or characteristic of an organism (that may health providers face a different regulatory regime that does
or may not be inherited). not require each test to be approved. Yet, these different tests
• Germline versus somatic mutations – the former are and providers compete in some PM applications.
heritable variations, whereas the latter are acquired (e.g., in But there are also some potential economic barriers that
cancer). could increasingly be a factor given growing efforts to control
• Biomarkers – they are a broad array of biological indi- medical spending in most developed countries. The incentive
cators, including genetic variants, proteins, and endogen- issue for existing marketed drugs is fairly obvious. Once a new,
ous metabolites, among others. patented medicine is on the market with prices and re-
• Predictive versus prognostic biomarker – a prognostic imbursement established around the world, the manufacturer
biomarker is used to project patient health (e.g., life has a very limited economic incentive to discover the subset of

484 Encyclopedia of Health Economics, Volume 2 doi:10.1016/B978-0-12-375678-7.01210-4


Author's personal copy
Personalized Medicine 485

patients in whom the drug works best if reimbursement will theoretical analysis of economic incentives and their policy
then be restricted to the subset. The problem is that re- implications for PM R&D.
imbursement prices for drugs – especially outside the US – This work has addressed two major questions. First, is it
tend to be rigid and inflexible during the period of the patent. likely that current market structures for innovative PMs and
Thus, even if most of the aggregate health benefit – and hence their companion diagnostics will produce the optimal amount
economic value – is concentrated in a subset of patients, the of PM development and use? Second, if the current situation is
manufacturer will receive or capture a much smaller share of likely to be suboptimal, what could be done to improve it? It
that aggregate value if reimbursement is restricted to that is important to recognize that these two questions include not
subset but the reimbursed price does not rise to reflect their only short-term questions about static efficiency but also long-
higher average health gain. From a longer term dynamic per- term questions about dynamic efficiency that affect the sus-
spective, if price inflexibility holds and there is a strong pos- tainability of both PM and the patented medicines industry as
sibility that the targeting test would be forthcoming, then drug a whole, especially because PM is often cited as the ‘new
manufacturers may have much less incentive to develop the paradigm’ for drug development.
drug in the first place. This has long-term, dynamic impli- In an article published in 2002, Danzon and Towse de-
cations for the incentives for R&D investments in PMs. veloped a formal model of pharmacogenetic testing to exam-
However, some targeting using tests might actually facilitate ine the conditions under which the development of these
R&D and the demonstration of efficacy and safety, although targeted interventions was likely to be socially optimal. The
this is difficult to predict a priori unless the mechanism of model uses the common assumption of a societal willingness
action of the drug is closely linked to a biomarker, for ex- to pay for health gains (i.e., a threshold amount), as is often
ample, as was the case for imatinib (Gleevecs, Novartis) for used in pharmacoeconomic CEA. However, they let the share
chronic myelogenous leukemia (CML). of the gain captured by the drug manufacturer vary. The es-
The rigidity of diagnostic reimbursement could be an even sential elements of their model are intuitively straightforward.
larger economic barrier – especially for companion diag- Imagine that a diagnostic test can stratify a patient population
nostics for drugs that are already on the market. In the US and that has previously received a drug into responders (R), those
most EU health-care systems, reimbursement for diagnostics is who benefit from the drug, and nonresponders (N), those
based on an administered pricing system linked imperfectly to who do not benefit. Before the test is available, both re-
the expected marginal cost of production and distribution, sponders and nonresponders received the drug at price P. In
meaning that some diagnostics might garner profits, whereas Danzon and Towse’s model, testing provides social value in
others might just break even or even lose money (but persist several ways, including (1) avoiding drug spending on the
for other business reasons). Thus, there is a limited incentive nonresponders and (2) avoiding the costs and adverse health
to incur the substantial fixed costs of evidence generation that effects of adverse events in nonresponders. Of course, the
would be necessary to demonstrate the clinical utility of the value of the product among responders exists in either scen-
biomarker-based test in combination with the drug. ario. The key drivers of the social value of testing are the
This article focuses on economic issues related to pricing averted costs of adverse events times the share of N, and the
and reimbursement policies as a potential barrier to the de- cost of testing both R and N groups. Testing will generally be
velopment and adoption of new, innovative PM technology. socially beneficial if the aggregate cost of testing is less than
There are other important economic issues in PM, such as the costs savings from not using the drug plus avoiding adverse
impact on drug development costs, the relation to physician events in N.
incentives to test and to follow test results, and issues related Their model identifies the key determinants of the in-
to targeting as a strategy for late entrants into a drug class. The centives for drug manufacturers and payers to embrace phar-
next section summarizes the key theoretical issues. This is macogenetic testing. They also note implications for test
followed by a discussion of some key examples of PMs that are developers and drug development more generally. Clearly, it
in use. The relevance of literature on the cost-effectiveness of will be critical for drug manufacturers to be able to capture a
PMs is discussed next. The article ends by identifying six major large share of the aggregate value created after the test is
policy challenges. The general conclusion is that pricing and introduced. This generally means that the price paid for re-
reimbursement systems will need to implement more flexible sponders in the testing scenario must rise roughly in pro-
value-based rewards for PMs if the appropriate amount of portion to the rise in average patient benefit (due to
R&D and evidence generation is going to be supported. eliminating adverse events and/or nonresponders), compared
with the no-testing scenario, and also the price of test must be
modest, relative to the cost of treatment. Although payers
Economic Incentives for Personalized Medicine would generally prefer to pay less, in theory, they would be
Development: A Framework willing to accept a situation where the total amount paid out is
the same if the net health benefits (i.e., among the responders)
To this point, only 10–15 PM tests enjoy a significant volume are the same as long as the costs of testing and the savings
of use, and the amount of evidence about their health and from avoiding adverse events in nonresponders are factored
economic impact is limited. But many have been the subject in. Danzon and Towse conclude: ‘‘The willingness of payers to
of a cost-effectiveness analysis (CEA) (Wong et al., 2010), and award higher prices for targeted benefits ... will be essential to
most are considered cost-effective by usual standards – or they retaining neutrality in investment incentives’’ (p. 10).
would not be in use in health systems. For the purpose of Danzon and Towse (2002) also note that it is possible that
this article, the more interesting and relevant work is the the testing scenario could produce an eventual market size
Author's personal copy
486 Personalized Medicine

that is so small (in terms of total revenues to the drug average benefit (including the nonresponders). In their base
manufacturer) that it will not be sufficient to justify the costs case scenario, with no Dx available, the (patented) Tx captures
of drug development if that cost is fixed (that is, independent all of the value created (given the willingness-to-pay thresh-
of the size of the patient population). In practice, however, the old). The five other scenarios explore variations on the four
US Food and Drug Administration (FDA) permits fewer and assumptions listed in this article while recognizing that the
much smaller trials for orphan drugs. And government sub- total value created is now greater due to the value of knowing.
sidies and other incentives may be needed in these situations It does, then, become ex post a different zero-sum game, i.e.,
for socially optimal investment. They also suggest that after the diagnostic becomes available – there is now a slightly
with free entry into the business of developing tests, manu- bigger pie to divide up. The implications of the six scenarios
facturers – with this new genetic knowledge – will have are intuitive and straightforward.
an incentive to incorporate testing into the drug development If the price of the Tx is fixed, and the Dx enters the market
paradigm, which might also reduce the costs of that ex post, the revenues going to the Tx manufacturer would fall
development. This is because in most markets (with the dramatically. The Dx may capture this or not, depending on
notable exception of the US), it is not possible to increase whether its pricing is regulated and at what level. The Tx
prices once a drug is launched because of rigid government manufacturer would suffer a severe revenue loss and has a very
price regulation. Thus, ex post targeting leads to lower volume limited incentive to encourage Dx entry – unless the value
but does not guarantee an increased price to reflect the greater could be recaptured by the Tx manufacturer by owning the Dx.
health gain per patient in the smaller population. To date, Indeed, this is the circumstance for most drugs already on the
although it is clear that manufacturers are increasingly con- market: neither Tx manufacturers nor Dx developers have a
sidering testing as part of drug development the aggregate strong incentive to develop a test that identifies responders.
impact has not been large over the past 10 years. Oncotype Dx for predicting breast cancer recurrence, which is
Danzon and Towse assume the price and availability of the discussed below, is one exception: the manufacturer avoided
test is a given. They focus on the social benefits of health gains the cost-based reimbursement system in the US and charged
and of any reductions in cost. Considering many of these same approximately $3500 for the test. It is true that some panels of
issues in a less formal analysis, Garrison and Austin (2007) build diagnostic tests might be lucrative under this system, but this
on this analytical approach by analyzing the incentives for both would seem to be less likely true for the novel complex
the diagnostic company and the drug company for the codeve- diagnostics needed for PM if their payment is based on the
lopment of companion diagnostic test (Dx) and a first-in-class summation of the expected costs of the analytic steps as op-
drug therapy (Tx). They also explore an additional potential posed to a measure of the incremental health gain.
benefit of PM – the ‘value of knowing.’ They consider six scen- If the Tx manufacturer’s drug comes to market in combin-
arios that represent combinations of the following four factors: ation with a companion test, then there are several different
possibilities. If the Dx is subject to marginal cost-based re-
1. whether Tx and Dx pricing reimbursement are value based
imbursement, then the manufacturer will want to capture as
or cost based, and whether they are flexible over time;
much value as possible through the drug price, which has much
2. timing – whether Tx is already on the market, i.e., ex post
more flexibility at launch in most countries. If the Tx manu-
versus ex ante;
facturer also owns the Dx, then, in theory, the value capture
3. whether intellectual property protection – to prevent
could be split arbitrarily between the two. But, in practice, given
copycats – is a barrier to entry;
administered pricing for the Dx and strong intellectual property
4. the competitiveness of insurance market over short versus
protection for the drug, the incentive probably remains to
long term.
capture as much value through the drug price as possible.
Although the overall result in terms of the importance of These scenarios illustrate several key points about the
flexible drug pricing and reimbursement is the same, Garrison economic barriers that companion Dx–Tx products face. First
and Austin extend the model by adding some value creation for and foremost is that inflexible pricing and reimbursement,
the ‘value of knowing,’ i.e., that the test–drug combination will which does not adjust to reflect value created, could under-
be of higher value to the responders as they will know they will mine the rewards for developing PMs. Second, manufacturers
benefit. Assuming that they are risk averse, this reduction in would ideally enter the market with a combination product
uncertainty should give them greater peace of mind. This makes that has been clinically tested and validated ex ante as a
the total pool of value created larger: how this aggregate value is combination. Flexible, value-based pricing and reimburse-
divided among patients, payer/insurers, drug manufacturers, ment would appear to be a necessary condition for en-
and test developers is an important question. They also em- couraging optimal investments to produce more PM; of
phasize that although pricing and reimbursement for new drugs course, it cannot guarantee a large volume of PM development
could be considered ‘value-based’ in the US and the key mar- because scientific, regulatory, and drug development realities
kets in the EU during the patent life, reimbursement for diag- represent constraints.
nostic tests tends to be a more rigid, administered pricing
system, which could be called ‘cost based.’
Their illustrative model considers a case where 20% of the Personalized Medicine Products
users are responders and 80% are nonresponders. In the ab-
sence of a test, the value created in the 20% is essentially Although some have been disappointed by the slow progress
spread over the 100% (subject to adjustment for adverse in PM over the past 12 years, there has been a gradual accu-
events in the nonresponders) by setting the price based on the mulation of PM products, so that now between 10 and 20
Author's personal copy
Personalized Medicine 487

Table 1 Companion diagnostic testing in PM

Technology Economic and testing features

HER2 testing for breast cancer A low-cost immunohistochemistry (IHC) (approx. $100–$200) test for human epidermal growth factor receptor
2 (HER2)-positivity was used in the initial clinical trial program and was provided by diagnostic companies.
Subsequently, a higher cost and more accurate test ($300–$500) was developed (called ‘FISH’) and is in use.
Approximately 80% of initial testing is done with IHC, with FISH retesting for patient with equivocal results.
The drug manufacturer receives nearly all of the economic value created by the combination from the drug
trastuzumab (Herceptins, Roche)
BCR-ABL testing for chronic An example of an ex ante test (breakpoint cluster region-Abelson (BCR-ABL) gene) closely tied to the
myelogenous leukemia (CML) development of the drug: large majority of value capture by the drug imatinib (Gleevecs, Novartis). A second,
BCR-ABL test is used to monitor for resistance and assignment to second-line therapies
Oncotype Dxs (Genomic Health) An example of a relatively high-cost, value-capturing test aimed at avoiding unproductive chemotherapy
for breast cancer recurrence
EGFR mutation testing in An example where the stratifying mutation (epidermal growth factor receptor (EGFR)) was identified in trials
nonsmall-cell lung cancer that also included test-negative patients
(NSCLC)
HLA-B5701 allele testing for Example of a test to identify patients who are more likely to suffer a severe adverse reaction to the HIV drug
abacavir in HIV abacavir (Ziagens, ViiV Healthcare)
KRAS testing in colorectal cancer The KRAS mutation predicts which patients will not respond to two different monoclonal antibody treatments
for colorectal cancer. The biomarker was identified after the products were on the market
PreDxs (Tethys Biosciences) This multimarker test identifies which prediabetic patients are at high risk of progressing to Type 2 diabetes: it
diabetes risk test indicates whether to begin prophylactic treatment with metformin
ALK mutation testing in NSCLC Example of the drug crizotonib (Xalkoris, Pfizer) that targets a small subset (approximately 4%) of patients in
disease condition with significant unmet medical need. It offers substantial survival gains in the subset, but
with high testing cost per identified responder that must be factored in

notable PM products are available. Many of the PM products immunohistochemistry (IHC) test and a more expensive and
have been in oncology where it has been possible to link more accurate fluorescence in situ hybridization test (FISH).
somatic mutations to chemotherapeutic response. Table 1 IHC is the more common test and is used in 80% of all initial
presents some examples of PM products that illustrate the tests in the US. Even after all these years of experience, the
range of issues that arise in combining companion diagnostics optimal companion testing strategy is still under debate due
with drugs to achieve PM. More details on three of these ex- to uncertainty about real-world test performance: approxi-
amples are provided in the next three paragraphs. mately 80% of women with breast cancer receive the IHC test
Trastuzumab (Herceptins, Roche), a biological compound initially and some are retested with FISH.
for the treatment of human epidermal growth factor receptor 2 Imatinib (Gleevecs, Novartis) is the unusual example of a
(HER2)-positive breast cancer, has been called the first ‘poster PM combination product developed through rational drug
child’ for PM. It was first approved in the US for the treatment design. It has been heralded as a virtual cure for many patients
of metastatic breast cancer (MBC) in 1998. Longer term (3-year with CML. It has the distinction of having one of the fastest
follow-up) trials were initiated in early-stage breast cancer approval times by the FDA. The breakpoint cluster region-
(EBC) and marketing approval was received in 2004. It provides Abelson (BCR-ABL) enzyme that promotes cancer cell devel-
a number of useful lessons as a case study in PM. First, these opment appears only in cancer cells, can be identified by a
combination products can have a long gestation and life cycle. test, and can be blocked. Once again, the large majority of the
The potential of the HER mechanism was discovered in the value created is captured by the drug, which has been esti-
early 1980s, but it took approximately 15 years to yield a viable mated to be cost-effective in the US with a cost per QALY of
compound combined with a predictive test. Second, the EBC approximately $50 000.
indication, which was approved 6 years later than the MBC Oncotype Dxs for predicting breast cancer recurrence
indication, produced much larger per-patient health gains could be considered the poster child for a value-capturing PM
and benefited many more women in the aggregate. Because test at approximately $3500 per test in the US. It is based on a
the initial price of trastuzumab for treatment of MBC was set ‘21-gene signature’ that was constructed through retrospective
closer to the implicit willingness-to-pay threshold for cost- analysis of historical tumor samples to generate a patented
effectiveness (4US$100 000 per quality-adjusted life-year index to predict the likelihood of recurrence. The major eco-
(QALY)) in the US, treatment in the EBC indication was much nomic benefit is that it avoids chemotherapy costs and side
more cost-effective (oUS$30 000 per QALY) (Garrison et al., effects (including the risk of death) in women with EBC. The
2007). So what appeared to be high-cost medicine for its manufacturer was able to circumvent usual coding and pricing
initial approved indication can be reasonably cost-effective practices in obtaining the US Medicare reimbursement. The
over the entire product life cycle. The company might well alternative would have been to use ‘code stacking’ of analytic
have priced it higher if the favorable results in EBC would steps, such as RNA extraction, reverse transcription, gene
have been anticipated. Another complexity of the trastuzu- amplification, and interpretation and report. But this would
mab story is the persistence of basic issues about testing have resulted in a payment level of only approximately
strategy. Two general types of tests are in use – a cheaper US$540 (Gustavsen et al., 2010).
Author's personal copy
488 Personalized Medicine

Personalized Medicine, Cost-Effectiveness Analysis, mechanism of action will work, the challenge of parallel de-
and Pharmacoeconomics velopment of a new companion diagnostic, and the un-
certainty of genetic prediction of complex traits. Clearly, the
In general, the methods of economic evaluations in PM are no scientific unknowns and challenges are substantial. None-
different than standard CEA. In the usual case in pharmacoe- theless, it is important to ask what can be done to optimize
conomics, the cost-effectiveness of a new medicine is assessed the economic incentives. The following six potential chal-
in a population with a particular disease (such as diabetes or lenges need to be researched, and, if appropriate, policy
rheumatoid arthritis) for the subset of patients who use the changes made.
drug. A standard pharmaceutical CEA compares the impact of
the use of a new medicine on health outcomes (usually
measured in terms of QALYs gained) and on medical costs. The Flexible Value-Based Pricing for Drugs
impact of PM further segments this subpopulation through the
Economic theory would suggest that payers and pricing and
use of the biomarker-based test. The CEA, therefore, changes to
reimbursement authorities should allow for flexible pricing
the question of the economic impact of the use of the com-
(both up and down) for drugs at launch and postlaunch if the
bination of the test and the treatment versus using the drug (or
evidence suggests that they can be targeted in a narrower pa-
another treatment) without testing in the full population. The
tient group or used in a number of different indications or
test is often called a companion diagnostic, and the pair has
subgroups of different value. The UK 2009 Pharmaceutical
been called a ‘codependent technology’ (e.g., by regulators in
Price Regulation Scheme, for example, included (1) provisions
Australia). The overall standard analysis in the field of phar-
for ‘flexible pricing’ that allowed drug developers to seek ap-
macoeconomics remains ‘cost-utility analysis’ with the primary
proval from the National Institute for Health and Clinical
metric being the incremental cost-effectiveness ratio (ICER) in
Excellence for higher prices when evidence of value increased
terms of cost per QALY gained, although it is not used by payers
and (2) provisions for new indications to be launched at
in all jurisdictions, for example, Germany.
different prices (higher or lower) than existing indications.
Given the cost of the test and the cost of the treatment, the
However, neither of these provisions has been used to date in
usual principles of CEA apply, except that the cost of the
the UK. This may reflect uncertainty on the part of companies
testing and outcomes for all test recipients must be included in
as to how the policy would be applied or the linkage of the UK
the analysis. It has been argued that the QALY may not capture
market via parallel trade and reference pricing to other EU
the utility gain that patients may receive from the value of
markets where such provisions do not apply.
knowing, i.e., the reduction in uncertainty in terms of whether
the medicine will work well for them. From a modeling ap-
proach, this reduction amounts to less uncertainty about the
Regulatory Flexibility in Drug–Test Combination
value of the drug (assuming the test has high accuracy). If this
Development
were to be included, some add-on or adjustment to the QALY
gain would be needed: there are various preference elicitation Given the evidence on drug development failure rates,
techniques in economics that can be used to elicit willingness pharmaceutical companies can be expected to be resistant to
to pay for such product attributes. increases in development costs caused by adding a test into the
Besides creating value by reducing uncertainty, a targeted development program (notwithstanding the potential advan-
Dx–Tx combination could create further value in a population tages of ex ante vs. ex post stratification for the ability of the
beyond what is captured in the QALY and ICER in, at least, two pharmaceutical company to extract rents). In the case of the
other ways. First, suppose that some responders were non- drug–test companion development, one might, therefore, ex-
compliant because they were not sure whether or not they were pect that it is important initially that the delivery of a proto-
responding. A companion Dx could increase their confidence type assay for use in Phase 3 development does not call for
about response and hence compliance with Tx, producing significant investment in advance of being in position to rec-
better outcomes in those patients, and thus creating more value ognize the efficacy or otherwise of the drug itself in Phase 2. To
at the aggregate level. Second, in the situation where the Dx is achieve this would require flexibility in the regulatory hurdles
not available, some patients who would be responders might for tests as part of drug development. (And this links to the
not actually choose to go to the doctor for the broader diag- importance for achieving socially optimal outcomes of having
nosis. With an available Dx, however, they might seek this care. an environment in which better quality follow-on tests are
Thus, overall uptake would increase. Measuring this aggregate incentivized to enter the market.) This flexibility in the drug
value gain would require a somewhat more sophisticated development process may require the payer’s pricing and re-
model than is typically used in CEA for a typical patient: a imbursement arrangements for drug–test combinations to
population-level uptake and use model would be needed, more concentrate on the evidence from the Phase 3 drug develop-
akin to a budget impact model. But it is feasible to model and ment trial and not require a patient randomization to use
estimate these additional sources of value. the test (e.g., the ‘double randomized trial’ proposed in the
Australian government’s guidelines for codependent technol-
ogies.) As the model changes over time, drug developers may
Regulatory and Policy Issues and Implications not have to codevelop new tests and biomarkers but can draw
on existing ones. In this context, the key issue will not be for
There are several major scientific challenges in the develop- the drug developer: instead, it will be necessary to ensure that
ment of PM, including the low probability that a new diagnostic developers have an incentive to bring improved
Author's personal copy
Personalized Medicine 489

tests to the market and will obtain value-based remuneration manufacturer thought in advance that this might happen, the
for them. prospect of lower revenues would have reduced the likelihood
of them making the initial investment. One could argue,
however, that it was the drug manufacturer’s invention that
Flexible Value-Based Reimbursement for Diagnostics created the health gain in the responders. Then, the extra value
created by the Dx manufacturer consists of (1) avoiding any
Testing that enables better choice of treatment or improved
adverse event treatment costs and any related health losses in
disease management may generate downstream health effects
the nonresponders plus (2) the ‘value-of-knowing premium.’
of extended life and/or improved quality of life. In addition, a
(Note that if the premium were, say, 5% of total health gain, it
diagnostic test can enhance the level of information about a
would be worth $50 million: an amount that could support
specific clinical condition or health state, and so reduce or
substantial evidence generation.) With flexible value-based
eliminate uncertainty. This may have value independent of any
pricing, this logic would argue that the drug innovator’s price
ability to improve treatment choice – although insurers’ will-
would be roughly doubled (i.e., to $1 billion in revenues) and
ingness to pay for this may be unclear. Historically, pricing
the Dx innovator would receive a reward in relation to cost
and reimbursement systems for diagnostics have been focused
savings and QALY gains in the nonresponders plus a premium
on the expected cost of making and conducting the test (which
for the value of knowing (i.e., $50 million). This split is ar-
may depend on the technology platform used) and not
bitrary in a static sense, but it can be argued that it reinforces
the value delivered. This has meant that the price of a new
dynamic efficiency by considering the relative size of the
diagnostic is often based on the price of existing tests (‘cross-
investments needed to develop a drug versus a diagnostic.
walking’) with similar clinical use or with similar character-
Clearly, this issue deserves further research.
istics or on production cost based on analytic steps. For
example, in the US, a number of diagnostics are reimbursed
via code stacking by reporting a combination of reimburse-
‘Follower’ Diagnostic Tests ‘Piggyback’ on Clinical Utility
ment Current Procedural Terminology codes describing
Evidence
laboratory protocol stages. Theory would suggest that both
diagnostic-dedicated and drug health technology assessment Evidence on drug effectiveness and value is generated by the
(HTA) processes should use a common, consistent, and drug developer in order to obtain regulatory and pricing ap-
comprehensive approach to assessing value to enable the proval. The drug company has a patent (no one can copy the
development of pricing and reimbursement arrangements product) and also regulatory exclusivity on the data they
capturing the full incremental value offered by those tech- generate (no one can reference the data to get regulatory ap-
nologies. Policymakers also need to consider the incentive for proval by claiming that their product does the same). The
companies to invest in evidence collection to raise the stand- situation is more complicated in diagnostics: patents may be
ard of clinical utility data available to support the case for less robust, regulatory hurdles are lower, and so it is more
using a companion test. Such incentives might come from difficult to prevent others appropriating the benefits of evi-
rewarding competing tests that supply evidence that they dence from studies the diagnostic manufacturer has paid for.
bring improved health gain and information through greater As a consequence, there may be underinvestment in evidence
accuracy. generation. Data exclusivity could be given to evidence for
diagnostics. This would require ‘follow-on’ tests to replicate
the evidence generated by the ‘first-in-class’ test (as is the case
Dividing the Combination Value between the Drug and the for drugs). However, under current diagnostic regulations in
Diagnostic the US and EU countries, LDTs (and in the EU any tests
provided by public health providers) have to meet lower
Accepting for now the notion that flexible value-based pricing
regulatory hurdles and so could not be prevented from pig-
is desirable for both an innovative PM and an innovative
gybacking on clinical utility evidence. In addition to the data
companion diagnostic raises the question, how can the
exclusivity requirement, there may, therefore, need to be an
problem of synergistic/codependent technologies noted above
expectation that payers will pay more for the test with the
be best resolved? As a starting point, it may be easier to start
stronger evidence base (i.e., that they will ignore tests without
with the case of the ex post, ‘standalone’ test. Recall that the
an evidence base when making HTA). Of course, a balance has
average development cost of a new drug is $1.5 billion
to be struck, as with patenting. The objective is not to delay
US$2011, and the productivity of drug R&D has been de-
competition to provide innovative tests but to ensure initial
clining over time. In contrast, the cost to develop a new bio-
providers have the potential to earn a return if they have
marker-based assay is in the order of $10 million to $50
evidence to support claims of value. Research is clearly needed
million – at the high end if additional clinical trials are needed
on both effective incentives to prevent piggybacking for a least
to validate the biomarker-based test. Consider a hypothetical
a period of time and how long they should be put in place.
case where an existing biopharmaceutical is on the market and
earning $1 billion in annual revenues despite having only a
50% response rate. Suppose a reliable biomarker-based test is
Flexibility in Diagnostic Test Evidence for Reimbursement
invented that can predict the responders. In theory, the payer
would be indifferent to giving them $500 million and perhaps Given the US and EU regulatory environments, a pragmatic
a premium for the value of knowing (i.e., reduced uncertainty approach could be taken to collecting evidence on the clinical
for both the payer and the patient). Clearly, if the drug utility of diagnostics, for example, using small randomized
Author's personal copy
490 Personalized Medicine

studies (if not built into Phase 3 of drug development), which


lead to conditional reimbursement approval and evidence- Pharmaceutical Pricing and Reimbursement Regulation in Europe.
based reimbursement rate to incentivize manufacturers plus Policy Responses to Uncertainty in Healthcare Resource Allocation
real-world, postlaunch data collection. To facilitate real-world Decision Processes. Pricing and Reimbursement of
data collection, increased investment in national e-health re- Biopharmaceuticals and Medical Devices in the USA. Research and
cords would, certainly, help, although the overall investment Development Costs and Productivity in Biopharmaceuticals. Value of
case for such records is much greater than simply facilitating Information Methods to Prioritize Research
assessment of the benefits (or otherwise) of developing and
validating diagnostic tests.
References

Conclusion Danzon, P. and Towse, A. (2002). The economics of gene therapy and of
pharmacogenetics. Value Health 5(1), 5–13.
Although there is a general perception that the growth of PM Garrison, L. P. and Austin, M. J. F. (2007). The economics of personalized
medicine: A model of incentives for value creation and capture. Drug
has been slower than hoped, it is not clear how much of this
Information Journal 41, 501–509.
perceived shortfall is due to scientific and regulatory con- Garrison, L. P., Veenstra, D. L., Carlson, R. J., Carlson, J. J. and Meckley, L. M.
straints versus economic incentives. Many of the PM products (2007). Backgrounder on pharmacogenomics for the pharmaceutical and
have been in oncology, where it has been possible to link biotechnology industries: Basic science, future scenarios, policy directions.
somatic mutations to drug response. The science is more dif- Report of the Pharmaceutical Outcomes Research Policy and Program.
Department of Pharmacy, University of Washington. Available at: http://
ficult outside of oncology. There are, clearly, problems with sop.washington.edu/porpp/general/reports.html (accessed 08.10.13).
economic incentives in this area as well. However, the em- Gustavsen, G., Phillips, K. and Pothier, K. (2010). The reimbursement landscape for
pirical evidence on this is limited by the challenge of con- novel diagnostics. Weston, MA: Health Advances. Available at: http://
structing the counterfactual. These may reflect the scientific www.healthadvances.com/pdf/novel_diag_reimbursement.pdf (accessed
08.10.13).
challenge: not enough drug-diagnostic combinations have
Wong, W. B., Carlson, J. J., Thariani, R. and Veenstra, D. L. (2010). Cost
received regulatory approval to allow exploration of the issues effectiveness of pharmacogenomics: A critical and systematic review.
around commercial success. However, the relatively small Pharmacoeconomics 28(11), 1001–1013.
number of regulatory approvals may also reflect perceived
commercial limitations as well as scientific challenges.
Over the past dozen years, the thinking about PM pre- Further Reading
dictive tests has evolved from thinking primarily about using
genetic mutations or combinations as predictors of clinical Blair, E. D., Stratton, E. K. and Kaufmann, M. (2012). The economic value of
response to a wider range of ‘biomarkers,’ and even to chan- companion diagnostics and stratified medicines. Expert Review of Molecular
Diagnostics 12(8), 791–794.
ging the name of the whole area to ‘molecular diagnostics.’ Davis, J. C., Furstenthal, L., Desai, A. A., et al. (2009). The microeconomics of
Owing to the rapidly falling cost of sequencing an individual’s personalized medicine: Today’s challenge and tomorrow’s promise. Nature
entire genome (e.g., projected to be less than $1000 in the near Reviews Drug Discovery 8(4), 279–286.
future), a new set of economic issues are arising around the Faulkner, E., Annemans, L., Garrison, L., et al. Personalized Medicine Development and
Reimbursement Working Group (2012). Challenges in the development and
question of how to use this plethora of data from whole
reimbursement of personalized medicine – payer and manufacturer perspectives and
genome scans. These new economic issues have not been implications for health economics and outcomes research: A report of the ISPOR
addressed here but will need to be better understood in future Personalized Medicine Special Interest Group. Value Health 15(8), 1162–1171.
discussions of the economics of PM. In the extreme, the Garrison, L. P. and Austin, M. J. F. (2006). Linking pharmacogenetics-based
emerging pharmacogenomics diagnostic industry will change diagnostics and pharmaceuticals for personalized medicine: Scientific and
economic challenges. Health Affairs 25(5), 1281–1290.
drastically, making it possible for a whole genome test to be Keeling, P., Roth, M. and Zietlow, T. (2012). The economics of personalized
performed at birth on an individual. Still, testing for somatic medicine: Commercialization as a driver of return on investment.
mutations would be needed and incentives would be required New Biotechnology 29(6), 720–731.
to invest in providing evidence that a particular set of bio- Meckley, L. M. and Neumann, P. J. (2010). Personalized medicine: Factors
influencing reimbursement. Health Policy 94(2), 91–100.
markers (including genes) is predictive of disease.
Ramsey, S. D., Veenstra, D. L., Garrison, L. P., et al. (2006). Toward evidence-
based assessment of laboratory-based diagnostics and genetic test for health
plan reimbursement. American Journal of Managed Care 12, 197–202.
Trusheim, M. R., Berndt, E. R. and Douglas, F. L. (2007). Stratified medicine:
See also: Adoption of New Technologies, Using Economic
Strategic and economic implications of combining drugs and clinical
Evaluation. Biopharmaceutical and Medical Equipment Industries, biomarkers. Nature Reviews Drug Discovery 6(4), 287–293.
Economics of. Cost-Effectiveness Modeling Using Health State Utility Veenstra, D. L. (2009). The economic value of innovative treatments over the
Values. Economic Evaluation, Uncertainty in. Information Analysis, product life cycle: The case of targeted trastuzumab chemotherapy for breast
Value of. Patents and Other Incentives for Pharmaceutical Innovation. cancer. Value in Health 12(8), 1118–1123.

You might also like