Economic Evaluation in Genomic Medicine
Economic Evaluation in Genomic Medicine
Economic Evaluation in Genomic Medicine
GENOMIC MEDICINE
Economic Evaluation in
GENOMIC MEDICINE
VASILIOS FRAGOULAKIS
University of Patras School of Health Sciences,
Department of Pharmacy, Patras, Greece;
National School of Public Health,
Athens, Greece
CHRISTINA MITROPOULOU
Erasmus University Medical Center,
Department of Clinical Chemistry,
Rotterdam, The Netherlands
MARC S. WILLIAMS
Geisinger Health System, Danville,
Pennsylvania, USA
GEORGE P. PATRINOS
University of Patras School of Health Sciences,
Department of Pharmacy, Patras, Greece
Notices
Knowledge and best practice in this field are constantly changing. As new research
and experience broaden our understanding, changes in research methods, professional
practices, or medical treatment may become necessary.
Practitioners and researchers may always rely on their own experience and knowledge
in evaluating and using any information, methods, compounds, or experiments described herein. In
using such information or methods they should be mindful of their own safety and the safety of
others, including parties for whom they have a professional responsibility.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume
any liability for any injury and/or damage to persons or property as a matter of products liability,
negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas
contained in the material herein.
ISBN: 978-0-12-801497-4
British Library Cataloguing-in-Publication Data
A catalogue record for this book is available from the British Library
Library of Congress Cataloging-in-Publication Data
A catalog record for this book is available from the Library of Congress
ix
x Preface
however, we have kept the researcher’s point of view and have only
superficially dealt with certain political issues to contribute impartially, we
hope, to the reader’s critical thinking.
With regard to the text itself, we offer this suggestion: while reading,
do not skip any words, symbols, or gene and biomarker names you do
not understand. The reason why most people abandon the study of any
subject is because they skip over a word or a symbol they cannot decipher
and thus become unable to apply or understand what they are reading.1 If
you find that you are having trouble understanding the text, go back, find
a point where you had complete conceptual understanding, and look for
a word whose definition you do not know. As soon as you locate it, use a
suitable dictionary (either an economics dictionary, if the word is a term
of the nomenclature; a biologic or medical dictionary,2 if the term relates
to genetics; or even a conventional dictionary for an “ordinary” word)
and clarify it. Both economic evaluation and genomic medicine use a few
specialized terms that need to be genuinely understood and not just
memorized. You should also keep in mind the fact that evaluating the
data presented here is just as important, if not more, than simply learning
it. As you study this text, we hope that you will remain critical, that you
will remember all that you consider to be true or useful, and that
you will compare the information you read with your own personal
observations.
We hope that the time you spend reading the text will be worth it,
and that it will also give you some ideas for further study or application
of the subject. If you achieve this, then we might say your effort had high
personal utility and was a cost-effective choice for you and for us as well.
The Authors
January 2015
1
http://www.appliedscholastics.org. Applied Scholastics International, a nonprofit educa-
tional organization based in Missouri, was founded by a consortium of American educa-
tors in 1972. Administered by the Association for Better Living and Education, it is
dedicated to the broad implementation of learning tools researched and developed by
American author and educator L. Ron Hubbard.
2
Genetics Home Reference (http://www.ghr.nlm.nih.gov/glossary) and National Human
Genome Research Institute (http://www.genome.gov/glossary).
CHAPTER 1
Improved
Unchanged
Worsened
philosophy of evaluation, supports the notion that EBM should draw its
conclusions from outcome research (Braslow, 1999; Claridge and Fabian,
2005; Sur and Dahm, 2011).
In this way, EBM attempts to link its indications with medical practice to
improve health care quality and effectiveness at a personalized level. The imple-
mentation of its principles requires the integration of personal clinical experi-
ence with the most reliable scientific clinical data. Historically, the method of
physician self-education in practicing EBM was developed at the Canadian
McMaster University, and in Europe important work is being done at
Oxford, at the NHS Research and Development Centre for EBM, which is
its counterpart in the United Kingdom (Cohen, 1996; Hadley et al., 2007).
In contrast, the United States does not have a single center for EBM. Instead,
a number of stakeholders have assumed roles, including the government (e.g.,
Agency for Healthcare Research and Quality [AHRQ], http://www.ahrq.
gov; Health Resources and Services Administration [HRSA], http://www.
hrsa.gov), public private partnerships (Patient Centered Outcomes Research
Institute [PCORI], http://www.pcori.org; United States Preventive Services
Task Force [USPSTF], http://www.uspreventiveservicestaskforce.org), and
professional societies.
In many cases, however, the distinction between “good clinical practice”
and “available economic resources” is controversial and often provides results
6 Economic Evaluation in Genomic Medicine
in web version of Figure 1.1). If the ultimate goal were to reduce expen-
ditures, then the state would simply cease to provide health care services
to certain citizens, which would achieve immediate savings but would
inflame the public sense of justice and would strain social cohesion. Such
expenditure restrictions are socially justified and financially effective only
if they include a substantial restructuring of the system to save resources
without reducing benefits.
In conclusion, economic evaluation is more of a guideline for a
systematic and strict way of thinking than a dogmatic set of quantification
methods that will mechanically determine the therapeutic options to
which society should direct its resources.
REFERENCES
Banta, D., 2003. The development of health technology assessment. Health Policy 63 (2),
121 132.
Braslow, J.T., 1999. History and evidence-based medicine: lessons from the history of
somatic treatments from the 1900s to the 1950s. Ment. Health Serv. Res. 1 (4),
231 240.
Claridge, J.A., Fabian, T.C., 2005. History and development of evidence-based medicine.
World J. Surg. 29 (5), 547 553.
Cohen, L., 1996. McMaster’s pioneer in evidence-based medicine now spreading his mes-
sage in England. Can. Med. Assoc. J. 154 (3), 388 390.
Hadley, J.A., Davis, J., Khan, K.S., 2007. Teaching and learning evidence-based medicine
in complementary, allied, and alternative health care: an integrated tailor-made course.
J. Altern Complement Med. 13 (10), 1151 1155.
Sur, R.L., Dahm, P., 2011. History of evidence-based medicine. Ind. J. Urol. 27 (4),
487 489.
Van Den Bos, J., Rustagi, K., Gray, T., Halford, M., Ziemkiewicz, E., Shreve, J., 2011.
The $17.1 billion problem: the annual cost of measurable medical errors. Health Aff.
(Millwood) 30 (4), 596 603.
Williams, M.S., 2014. Genomic medicine implementation: learning by example. Am. J.
Med. Genet. C Semin. Med. Genet. 166, 8 14.
CHAPTER 2
INTRODUCTION
The central dogma of genomic medicine is to exploit the individual’s
genomic sequence in combination with other clinical information to
enhance the clinical decision-making process. In recent years, significant
advances have enriched our knowledge regarding the molecular etiology
of a wide range of human genetic diseases, allowing for better disease
prevention, prognosis, and treatment. In parallel, genomic technology
has progressed rapidly and, as a result, genomics research has the poten-
tial to aid clinicians in their task of estimating disease risk and individu-
alizing treatment modalities. This constitutes the basis of genomic
medicine, a new discipline that promises to enhance opportunities for
the customization of patient care and the personalization of conventional
therapeutic interventions.
The concept of genomic or personalized medicine stems from as early as
400 B.C., at which time Hippocrates of Kos (460 370 B.C.E.) stated that
“. . .it is more important to know what kind of person suffers from a disease
than to know the disease a person suffers.” This ancient statement encapsulates
the essence of modern personalized genomic medicine. The first application
of individualized care based on presumed genetic information is documented
in the Talmud (Yevamot 64b). Rabbi Judah the Prince (135 217 C.E.) ruled
that if a woman’s first two children died from blood loss after circumcision,
the third son should not be circumcised. A second disagreed and ruled that
the third son may be circumcised; however, if this infant died, the fourth child
should not be circumcised. There was agreement that abnormal bleeding was
hereditary but disagreement regarding how many events were required to
establish a pattern and therefore exempt a child from circumcision
APPLICATIONS OF PHARMACOGENOMICS
At present, the US Food and Drug Administration (FDA) and the European
Medicines Agency (EMA) provide pharmacogenomic information on the
label of more than 150 drugs. Although only a few recommend that
pharmacogenomic tests must be performed prior to prescribing these drugs,
the inclusion of the information for providers is indicative of the importance
these agencies place on this emerging field. The Clinical Pharmacogenetics
Implementation Consortium has published a series of evidence-based clinical
guidelines for use of pharmacogenetic information to guide therapy
Genomic Medicine Today: An Introduction for Health Economists 11
Oncology Examples
Trastuzumab, a monoclonal antibody blocking v-erb-b2 erythroblastic
leukemia viral oncogene homolog 2 (HER2, also ERBB2) receptor
protein, is one of the prime examples for which pharmacogenomic test-
ing is routinely used. It has been shown that variable expression of the
HER2 receptor gene determines the likelihood that a patient will respond
to trastuzumab. HER2 overexpression is observed in approximately one-
fourth of breast cancer patients. It is correlated with poor prognosis,
increased tumor formation, and metastasis, as well as resistance to chemo-
therapeutic agents. As such, HER2 testing either through genetic testing
or through analysis of the HER2 protein product identifies patients who
will likely respond to trastuzumab. Other examples are the tyrosine kinase
inhibitors erlotinib and gefitinib, which are designed to target the epider-
mal growth factor receptor (EGFR). EGFR has been shown to play a
role in predisposition to lung cancer. EGFR variants are now used to pre-
dict improved progression-free survival with gefitinib in a comparison
with carboplatin paclitaxel (Mok et al., 2009).
Irinotecan is yet another example of a drug for which a pharmacoge-
nomic test can help to identify colorectal cancer patients who are likely
to experience adverse reactions during treatment, such as diarrhea and
severe neutropenia. The UGT1A1 28 variant is associated with increased
toxicity to irinotecan; as such, patients homozygous for the UGT1A1 28
allele are at higher risk for development of irinotecan-associated neutro-
penia and diarrhoea. The FDA recommended an addition to the irinote-
can package insert to include UGT1A1 28 genotype as a risk factor for
severe neutropenia. However, it is important to note that in the
Evaluation of Genomic Applications in Practice and Prevention (EGAPP)
working group’s recommendation regarding irinotecan, it was noted that
there is evidence that presence of this genotype results in improved
response to irinotecan, that is, improved efficacy (Berg et al., 2009). This
means that when evaluating a pharmacogenomic test, care must be taken
to identify impact on both efficacy and adverse events. The following
example also illustrates this principle.
12 Economic Evaluation in Genomic Medicine
Cardiology Examples
In recent years, cardiology became the other medical specialty in which
pharmacogenomics applications are emerging into practice, particularly
related to warfarin, acenocoumarol, and clopidogrel. For more than
Genomic Medicine Today: An Introduction for Health Economists 13
Psychiatric Examples
Unlike oncology and cardiology, pharmacogenomics and personalized medi-
cine are still far from being achieved in psychiatry (Alda, 2013).
Pharmacokinetic pathways in which cytochrome P450 isoenzymes, such as
CYP2D6, CYP1A1, CYP3A4, CYP2C9, and CYP2C19, are involved may
predict serum levels of antidepressants and antipsychotics. Numerous studies
14 Economic Evaluation in Genomic Medicine
Public Economic
health Genome evaluation
genomics informatics in genomic
medicine
Health care
Genomics
Pharmacogenomics professionals
research
genomics
education
Stakeholder
analysis General
Genethics public
in genomic
medicine genomics
awareness
Figure 2.1 Schematic depiction, as a jigsaw puzzle, of the various disciplines that
constitute the multidisciplinary field of genomic medicine. Note the central role of
genomics research, which is depicted as the central piece in the puzzle that needs
to be complemented by the other pieces to fulfill the promise of genomic medicine.
REFERENCES
Abrahams, E., Ginsburg, G.S., Silver, M., 2005. The personalized medicine coalition:
goals and strategies. Am. J. Pharmacogenomics 5, 345 355.
Alda, M., 2013. Personalized psychiatry: many questions, fewer answers. J. Psychiatry
Neurosci. 38, 363 365.
Arranz, M.J., de Leon, J., 2007. Pharmacogenetics and pharmacogenomics of schizophrenia:
a review of last decade of research. Mol. Psychiatry 12, 707 747.
Berg, A.O., Armstrong, K., Botkin, J., Calonge, N., Haddow, J., Hayes, M., et al., 2009.
Recommendations from the EGAPP working group: can UGT1A1 genotyping
reduce morbidity and mortality in patients with metastatic colorectal cancer treated
with irinotecan? Genet. Med. 11 (1), 15 20.
Bodin, L., Verstuyft, C., Tregouet, D.A., Robert, A., Dubert, L., Funck-Brentano, C., et al.,
2005. Cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1)
genotypes as determinants of acenocoumarol sensitivity. Blood 106, 135 140.
Bota, S., Sporea, I., Şirli, R., Neghină, A.M., Popescu, A., Străin, M., 2013. Role of
interleukin-28B polymorphism as a predictor of sustained virological response in
patients with chronic hepatitis C treated with triple therapy: a systematic review and
meta-analysis. Clin. Drug Investig. 33 (5), 325 331.
Carlson, J.J., Garrison, L.P., Ramsey, S.D., Veenstra, D.L., 2009. The potential clinical and
economic outcomes of pharmacogenomic approaches to EGFR-tyrosine kinase
inhibitor therapy in non-small-cell lung cancer. Value Health 12, 20 27.
Chouchana, L., Narjoz, C., Beaune, P., Loriot, M.A., Roblin, X., 2012. Review article:
the benefits of pharmacogenetics for improving thiopurine therapy in inflammatory
bowel disease. Aliment. Pharmacol. Ther. 35 (1), 15 36.
Cooper, D.N., Brand, A., Dolzan, V., Fortina, P., Innocenti, F., Lee, M.T., et al., 2014.
Bridging genomics research between developed and developing countries: the geno-
mic medicine alliance. Pers Med. 11, 615 623.
Deverka, P.A., McLeod, H.L., 2008. Harnessing economic drivers for successful clinical
implementation of pharmacogenetic testing. Clin. Pharmacol. Ther. 84, 191 193.
Deverka, P.A., Vernon, J., McLeod, H.L., 2010. Economic opportunities and challenges
for pharmacogenomics. Annu. Rev. Pharmacol. Toxicol. 50, 423 437.
Evaluation of Genomic Applications in Practice and Prevention (EGAPP) Working
Group, 2007. Recommendations from the EGAPP working group: testing for
cytochrome P450 polymorphisms in adults with nonpsychotic depression treated with
selective serotonin reuptake inhibitors. Genet. Med. 9 (12), 819 825.
Flowers, C.R., Veenstra, D., 2004. The role of cost-effectiveness analysis in the era of
pharmacogenomics. Pharmacoeconomics 22, 481 493.
Frueh, F.W., Gurwitz, D., 2004. From pharmacogenetics to personalized medicine: a vital need
for educating health professionals and the community. Pharmacogenomics 5, 571 579.
Gage, B.F., Eby, C., Johnson, J.A., Deych, E., Rieder, M.J., Ridker, P.M., et al., 2008.
Use of pharmacogenetic and clinical factors to predict the therapeutic dose of warfa-
rin. Clin. Pharmacol. Ther. 84, 326 331.
Geisler, T., Schaeffele, E., Dippon, J., Winter, S., Buse, V., Bischofs, C., et al., 2008. CYP2C19
and nongenetic factors predict poor responsiveness to clopidogrel loading dose after
coronary stent implantation. Pharmacogenomics 9, 1251 1259.
Ginsburg, G.S., Willard, H.F., 2009. Genomic and personalized medicine: foundations
and applications. Transl. Res. 154, 277 287.
Girnita, D.M., Burckart, G., Zeevi, A., 2008. Effect of cytokine and pharmacogenomic
genetic polymorphisms in transplantation. Curr. Opin. Immunol. 20, 614 625.
Gladding, P., Webster, M., Ormiston, J., Olsen, S., White, H., 2008. Antiplatelet drug
nonresponsiveness. Am. Heart J. 155, 591 599.
Genomic Medicine Today: An Introduction for Health Economists 23
Gurwitz, D., Weizman, A., Rehavi, M., 2003. Education: teaching pharmacogenomics to
prepare future physicians and researchers for personalized medicine. Trends Pharmacol.
Sci. 24, 122 125.
Holtzman, N.A., Watson, M.S. (Eds.), 1997. Promoting safe and effective genetic testing
in the United States. Final report of the task force on genetic testing. ,http://www.
genome.gov/10001733..
Huizenga, C.R., Lowstuter, K., Banks, K.C., Lagos, V.I., Vandergon, V.O., Weitzel, J.N.,
2010. Evolving perspectives on genetic discrimination in health insurance among
health care providers. Fam. Cancer 9 (2), 253 260.
Issa, A.M., 2002. Ethical perspectives on pharmacogenomic profiling in the drug develop-
ment process. Nat. Rev. Drug Discov. 1, 300 308.
Kampourakis, K., Vayena, E., Mitropoulou, C., Borg, J., van Schaik, R.H., Cooper, D.N.,
et al., 2014. Key challenges for next generation pharmacogenomics. EMBO Rep. 15
(5), 472 476.
Klein, T.E., Altman, R.B., Eriksson, N., Gage, B.F., Kimmel, S.E., Lee, M.T., et al.,
2009. Estimation of the warfarin dose with clinical and pharmacogenetic data. N.
Engl. J. Med. 360, 753 764.
Lee, S.S., Mudaliar, A., 2009. Medicine. Racing forward: the genomics and personalized
medicine act. Science 323, 342.
Mai, Y., Koromila, T., Sagia, A., Cooper, D.N., Vlachopoulos, G., Lagoumintzis, G., et al.,
2011. A critical view of the general public’s awareness and physicians’ opinion of the
trends and potential pitfalls of genetic testing in Greece. Pers. Med. 8, 551 561.
Mai, Y., Mitropoulou, C., Papadopoulou, X.E., Vozikis, A., Cooper, D.N., van Schaik,
R.H., et al., 2014. Critical appraisal of the views of healthcare professionals with
respect to pharmacogenomics and personalized medicine in Greece. Pers. Med. 11
(1), 15 26.
Mallal, S., et al., 2008. HLA-B 5701 screening for hypersensitivity to abacavir. N. Engl. J.
Med. 358, 568 579.
Matloff, E.T., Shappell, H., Brierley, K., Bernhardt, B.A., McKinnon, W., Peshkin, B.N.,
2000. What would you do? Specialists’ perspectives on cancer genetic testing, prophy-
lactic surgery, and insurance discrimination. J. Clin. Oncol. 18, 2484 2492.
Mega, J.L., Close, S.L., Wiviott, S.D., Shen, L., Hockett, R.D., Brandt, J.T., et al., 2009.
Cytochrome p-450 polymorphisms and response to clopidogrel. N. Engl. J. Med.
360, 354 362.
Mette, L., Mitropoulos, K., Vozikis, A., Patrinos, G.P., 2012. Pharmacogenomics and
public health: implementing populationalized medicine. Pharmacogenomics 13 (7),
803 813.
Mitropoulos, K., Johnson, L., Vozikis, A., Patrinos, G.P., 2011. Relevance of pharmacoge-
nomics for developing countries in Europe. Drug Metabol. Drug Interact. 26, 143 146.
Mitropoulou, C., Mai, Y., van Schaik, R.H., Vozikis, A., Patrinos, G.P., 2015.
Documentation and analysis of the policy environment and key stakeholders in phar-
macogenomics and genomic medicine in Greece. Public Health Genomics 17,
280 286.
Mizzi, C., Mitropoulou, C., Mitropoulos, K., Peters, B., Agarwal, M.R., van Schaik, R.H.,
et al., 2014. Personalized pharmacogenomics profiling using whole genome sequencing.
Pharmacogenomics 15 (9), 1223 1234.
Mok, T.S., Wu, Y.L., Thongprasert, S., Yang, C.H., Chu, D.T., Saijo, N., et al., 2009.
Gefitinib or carboplatin paclitaxel in pulmonary adenocarcinoma. N. Engl. J. Med.
361, 947 957.
Pacheu-Grau, D., Gomez-Duran, A., Lopez-Perez, M.J., Montoya, J., Ruiz-Pesini, E.,
2010. Mitochondrial pharmacogenomics: barcode for antibiotic therapy. Drug Discov.
Today 15, 33 39.
24 Economic Evaluation in Genomic Medicine
Patrinos, G.P., 2010. General considerations for integrating pharmacogenomics into main-
stream medical practice. Hum. Genomics 4, 371 374.
Pauker, S.G., Kassirer, J.P., 1987. Decision analysis. N. Engl. J. Med. 316 (5), 250 258.
Perlis, R.H., Smoller, J.W., Ferreira, M.A., McQuillin, A., Bass, N., Lawrence, J., et al.,
2009a. A genome-wide association study of response to lithium for prevention of
recurrence in bipolar disorder. Am. J. Psychiatry 166, 718 725.
Perlis, R.H., Patrick, A., Smoller, J.W., Wang, P.S., 2009b. When is pharmacogenetic
testing for antidepressant response ready for the clinic? A cost-effectiveness analysis
based on data from the STAR D study. Neuropsychopharmacology 34,
2227 2236.
Picard, F.J., Bergeron, M.G., 2002. Rapid molecular theranostics in infectious diseases.
Drug Discov. Today 7, 1092 1101.
Pierce, B.L., Ahsan, H., 2010. Case-only genome-wide interaction study of disease risk,
prognosis and treatment. Genet. Epidemiol. 34, 7 15.
Piquette-Miller, M., Grant, D.M., 2007. The art and science of personalized medicine.
Clin. Pharmacol. Ther. 81, 311 315.
Pirmohamed, M., 2006. Warfarin: almost 60 years old and still causing problems. Br. J.
Clin. Pharmacol. 62, 509 511.
Pisanu, C., Tsermpini, E.E., Mavroidi, E., Katsila, T., Patrinos, G.P., Squassina, A., 2014.
Assessment of the Pharmacogenomics Educational Environment in Southeast Europe.
Public Health Genomics 17, 272, 279.
Potamias, G., Lakiotaki, K., Katsila, T., Lee, M., Topouzis, S., Cooper, D.N., et al., 2014.
Deciphering next-generation pharmacogenomics: an information technology perspec-
tive. OPEN Biol. 4 (7), 140071.
Priest, V.L., Begg, E.J., Gardiner, S.J., Frampton, C.M., Gearry, R.B., Barclay, M.L.,
et al., 2006. Pharmacoeconomic analyses of azathioprine, methotrexate and prospec-
tive pharmacogenetic testing for the management of inflammatory bowel disease.
Pharmacoeconomics 24, 767 781.
Relling, M.V., Hancock, M.L., Rivera, G.K., Sandlund, J.T., Ribeiro, R.C., Krynetski,
E.Y., et al., 1999. Mercaptopurine therapy intolerance and heterozygosity at the thio-
purine S-methyltransferase gene locus. J. Natl. Cancer Inst. 91, 2001 2008.
Reydon, T.A., Kampourakis, K., Patrinos, G.P., 2012. Genetics, genomics and society: the
responsibilities of scientists for science communication and education. Pers. Med. 9,
633 643.
Robertson, J.A., 2001. Consent and privacy in pharmacogenetic testing. Nat. Genet. 28,
207 209.
Rothstein, M.A., Epps, P.G., 2001. Ethical and legal implications of pharmacogenomics.
Nat. Rev. Genet. 2, 228 231.
Schalekamp, T., Brassé, B.P., Roijers, J.F., van Meegen, E., van der Meer, F.J., van Wijk,
E.M., et al., 2007. VKORC1 and CYP2C9 genotypes and phenprocoumon anticoa-
gulation status: interaction between both genotypes affects dose requirement. Clin.
Pharmacol. Ther. 81, 185 193.
Smart, A., Martin, P., Parker, M., 2004. Tailored medicine: whom will it fit? The ethics
of patient and disease stratification. Bioethics 18, 322 342.
Spear, B.B., Heath-Chiozzi, M., Huff, J., 2001. Clinical application of pharmacogenetics.
Trends Mol. Med. 7, 201 204.
Squassina, A., Manchia, M., Manolopoulos, V.G., Artac, M., Lappa-Manakou, C.,
Karkabouna, S., et al., 2010. Realities and expectations of pharmacogenomics and per-
sonalized medicine: impact of translating genetic knowledge into clinical practice.
Pharmacogenomics 11, 1149 1167.
Genomic Medicine Today: An Introduction for Health Economists 25
Squassina, A., Manchia, M., Borg, J., Congiu, D., Costa, M., Georgitsi, M., et al., 2011.
Evidence for association of an ACCN1 gene variant with response to lithium treat-
ment in Sardinian patients with bipolar disorder. Pharmacogenomics 12, 1559 1569.
Stanulla, M., Schaeffeler, E., Flohr, T., Cario, G., Schrauder, A., Zimmermann, M., et al.,
2005. Thiopurine methyltransferase (TPMT) genotype and early treatment response
to mercaptopurine in childhood acute lymphoblastic leukemia. JAMA 293 (12),
1485 1489.
Tamaoki, M., Gushima, H., Tsutani, K., 2007. Awareness survey of parties involved in
pharmacogenomics in Japan. Pharmacogenomics 8, 275 286.
Tsermpini, E.E., Assimakopoulos, K., Bartsakoulia, M., Economou, G., Papadima, E.,
Mitropoulos, K., et al., 2014. Individualizing clozapine and risperidone treatment for
schizophrenia patients. Pharmacogenomics 15 (1), 95 110.
Vaszar, L.T., Cho, M.K., Raffin, T.A., 2003. Privacy issues in personalized medicine.
Pharmacogenomics 4, 107 112.
Williams, M.S., 2007. Insurance coverage for pharmacogenomic testing in the United
States. Pers. Med. 4 (4), 479 487.
CHAPTER 3
INTRODUCTION
The field of health economics is more about understanding utility and
effectiveness of medical interventions rather than focusing strictly on costs.
In its ideal application, health economics helps inform the decision-making
process to improve the quality of health outcomes while optimizing health
care expenditures.
There are three key features in health economics analyses as currently
applied: (i) they are more focused on the benefits received by the health care
system and society as a whole rather than the individual/patient, namely
price, health state, improvements in quality of life, expansion of life expec-
tancy, and resources saved, to name a few; (ii) the recipient of the medical
intervention is, in the majority of cases, not the most informed medical
decision-maker and, as such, does not have a complete picture of the poten-
tial benefits and harms of a given medical intervention or decision; and
(iii) most of the time, the recipients of medical interventions do not directly
pay for these treatments; rather, payment is received from a third party that
the recipient supports through taxes, medical premiums, or a shared model
of employer/employee contributions. As such, health economics aims to
better understand the value and costs of a certain medical intervention
compared with another by taking into assumption all the factors that impact
on patients, health care providers, the health care system in general, and,
ultimately, society.
Health economics uses various economic evaluation tools to evaluate
medical interventions, including:
• Cost-effectiveness analysis (CEA): This sort of analysis aims to deter-
mine whether a medical intervention for disease diagnosis, prevention,
and/or treatment improves clinical outcomes enough to justify the
considering the fact that results may differ among different patient
groups on the basis of genome characteristics, which can directly
impact risk stratification as well as medical interventions.
REFERENCES
Basu, A., Meltzer, D., 2007. Value of information on preference heterogeneity and indi-
vidualized care. Med. Decis. Making 27 (2), 112 127.
Bock, J.A., Fairley, K.J., Smith, R.E., Maeng, D.D., Pitcavage, J.M., Inverso, N.A., et al.,
2014. Cost-effectiveness of IL28 genotype-guided protease inhibitor triple therapy
Economic Evaluation and Genomic Medicine: What Can They Learn from Each Other? 35
INTRODUCTION
Cost-effectiveness analysis (CEA) is defined as an analytical technique
intended for the systematic comparative evaluation of the overall cost and
benefit generated by alternative therapeutic interventions for the manage-
ment of a disease (WHO Guide to Cost-effectiveness Analysis, 2003). The
cost associated with the different treatments is measured in monetary units,
whereas the benefit from the interventions is estimated in similar physical
units that can be determined objectively by suitable measurements.
Therapeutic interventions usually diminish the impact of the disease, elimi-
nate its symptoms, improve quality of life, and also prolong survival when
this is possible. However, society’s health care needs increase over time; the
associated costs are too great and the available resources are too limited to
meet current needs.
The idea of comparative evaluation of alternative interventions is fun-
damental in economics and is related to the way in which economists
assess the value of the relevant options. Economic evaluation, a major
part of which is CEA, is a standard approach to the problem of evaluating
alternative interventions through their opportunity cost. This term is used
to describe the process of evaluating different health technologies by con-
sidering the opportunity cost, meaning the best possible alternative
options at the societal and the individual levels (Palmer and Raftery,
1999). This cost should not be confused with the explicit cost of acquir-
ing a technology, which is constant and determined based on historical
information; instead, it should be evaluated periodically because it is a
function of current practices and changes over time as new treatments
become widely available to the general population.
The application of CEA allows such a comparison to be made, and
the limited resources that the health systems can allocate can be used
effectively based on optimization behaviors grounded in the tenets of
Cost
C
A
Benefit
Figure 4.1 Mean versus incremental cost and the “do nothing” option.
mean effectiveness of the new treatment, the mean benefit of the standard
treatment, the mean cost of the new treatment, and the mean cost of the
standard treatment, whereas ΔE 5 ET ES and ΔC 5 CT CS are the
definitions of the differences in cost and effectiveness, respectively.
Comparative evaluation will give the following four possible scenarios
(Drummond et al., 2005):
A. ΔE 5 ET 2 ES . 0 and ΔC 5 CT 2 CS . 0
This is the most common case. Usually, an increase in mean sur-
vival with the innovative treatment and a corresponding increase in
overall cost associated with its administration are observed. In recent
years, new discoveries have been made continuously as new drugs,
new biological and gene therapies, new diagnostic methods, targeted
treatments, or new procedures have been developed. Technology, in
most cases, significantly increases the cost of services because of the
great developing, purchasing, and operating expenses; the price of
technology (which is a fraction of the overall cost of the intervention)
tends to incorporate these costly processes. In such cases, there should
be a criterion by which to assess whether the increased cost is justi-
fied by the additional effectiveness (Figure 4.2, Box A).
B. ΔE 5 ET ES . 0 and ΔC 5 CT CS , 0
In this scenario, the new treatment dominates because it provides
more effectiveness and is associated with lower cost than the standard
treatment. In this case, the previous technology must be completely
discarded and the new treatment must be fully adopted by the health
system (Figure 4.2, Box B).
Improved B A
Unchanged
Worsened D C
Figure 4.2 The value framework, adjusted. Boxes AD depict various scenarios
(see text for details).
Introduction to the Technical Issues of Economic Evaluation 43
C. ΔE 5 ET ES , 0 and ΔC 5 CT CS . 0
This unpromising scenario for the new technology includes
increased cost but lower effectiveness from its use compared with the
standard treatment. According to the applicable terminology, the new
treatment is dominated by the older one and should be discarded
because it is not in the society’s best interest and is not a good health
investment (Figure 4.2, Box C).
D. ΔE 5 ET ES , 0 and ΔC 5 CT CS , 0
In this fourth scenario, the difference in survival is for the standard
treatment, even though the use of the new treatment is associated
with resource savings. In scenario D (as in scenario A), the ultimate
decision to adopt or reject the new technology will be based on
weighing the savings (which is the desirable outcome) and the
reduced effectiveness (which is a negative consequence) when com-
paring treatments. This criterion is essentially the same as in the first
scenario, but the level of the indicator may differ (Figure 4.2, Box D).
A special case of these scenarios is the comparison of costs only when
clinical practice has failed to show a difference in favor of one of the
compared technologies. In this case, the treatment associated with the
lowest overall cost will be selected. This type of analysis is called cost-
minimization analysis and is not considered a complete economic analysis
because it does not examine the relative benefit of the treatments.
Nevertheless, we should note that failure to demonstrate a statistically sig-
nificant difference between treatments in a study does not necessarily
mean that the interventions are equivalent. “Statistical significance” is a
widely used concept but is based on an arbitrary level of significance
determined from the literature. Economic evaluation, however, is inter-
ested in the mean estimate of quantities (i.e., the mean expected differ-
ence between interventions) (Claxton, 1999). Let us imagine a very
simplistic example. Assume that we live in a world without costs; in
which case, the only thing we would want to compare between two ther-
apeutic interventions would be their effectiveness measured in “years of
life gained.” As in standard statistical practice, if the confidence interval of
the difference in survival between interventions includes zero, then we
may say that there is no statistically significant difference between treat-
ments, at a possibility of 95%, for the population from which the samples
were taken. In practice, however, we are not interested in inductive con-
clusions; rather, we need to decide whether to administer one treatment
or the other, disjunctively (i.e., with no intermediate alternative). In such
44 Economic Evaluation in Genomic Medicine
a case, we would select the treatment with the greatest expected mean
value, regardless of the confidence interval. Therefore, it is possible to
perform CEA even without a statistically significant difference in survival
between interventions (Briggs and O’Brien, 2001).
If, however, the equivalence of the interventions is assured, then com-
paring costs will reveal the maximizing option and will lead to the adop-
tion of the least expensive technology. In the marginal case where neither
cost nor benefit differs, although the patients’ quality of life is—or may
be reasonably assumed to be—equal, the economic analysis is unable to
provide an answer and other clinical and societal criteria must be consid-
ered to reach a decision. The cases described here are presented graphi-
cally in Figure 4.3 (Black, 1990).
The axes show the difference between cost and benefit for interven-
tions T and S; the differences are indicated with the Greek delta (Δ).
Quadrant I represents the first scenario. The new technology increases
survival as well as cost compared with the standard treatment. If the addi-
tional cost is not forbidding, then the new technology will be adopted;
however, if the cost is considered excessive, then it will be rejected.
There is also a “gray zone” where the additional benefit is associated with
higher cost and the result will be uncertain until the expense considered
II + New I
Δ- Cost
technology
rejected
?
Δ- Effectiveness – Δ- Effectiveness +
New technology
rejected
New technology
? accepted
New
Δ- Cost –
technology
accepted
III IV
+
II I
Δ- Cost
New technology
rejected
New technology
rejected
New technology
accepted
Δ- Effectiveness – Δ- Effectiveness +
New technology
rejected
New
technology New technology
rejected accepted
New
technology
Δ- Cost –
accepted
III IV
ΔC
λ . ICERηλ .
ηλχΔE . ΔC or λχΔE 2 ΔC . 0
ΔE
Introduction to the Technical Issues of Economic Evaluation 47
The increase in benefit is greater than the increase in cost for points
within the first quadrant, given that λ is by definition a positive number,
whereas if ΔE , 0 and ΔC , 0, then the term λχΔE is a negative num-
ber and the term 2ΔC is a greater positive number; therefore, the cost
(savings) is greater than the benefit (obtained by S) and treatment T is
again a cost-effective option.
In reality, ΔE and ΔC are not simply two data points (values); they
are actually distributions with an estimated mean and a standard devia-
tion (just like the ICER). Consequently, in some cases it is convenient
to alter the ICER equation with a suitable linear transformation.
Because the ICER is an index (ratio) of two other statistical measures
(ΔC and ΔE), it does not follow any known statistical distribution;
instead, it presents certain assessment difficulties when the difference
between treatments is small. In this case, the limits become infinite,
which complicates the statistical manipulation of the results. If S has a
marginally greater benefit, then the limit of ICER tends to negative
infinity; however, if T has slightly greater survival, then the ICER tends
to positive infinity. Let us assume, for example, a value of
ΔC 5 h10,000 and of ΔE 5 0.001 or ΔE 5 20.001. At these values,
the ICER obviously tends to infinity. The ICER presents one additional
difficulty: it does not provide clear information about which of the
treatments being compared should be adopted if we do not already
know the quadrant we are in, such as if the signs of ΔE and ΔC are
unknown. In this case, problems in estimating 95% confidence intervals
would arise if we were to classify negative ICER ratios together with
positive ones.
To overcome such difficulties in difference ratios, the following trans-
formation has been proposed:
λχΔE 2 ΔC 5 NMB
where NMB is the net monetary benefit arising from the difference in benefit
and in cost for the treatments being compared and for a given value of λ.
A therapeutic option in this case is considered acceptable if (and only if)
NMB . 0, regardless of the sign of ΔE.
A diagrammatic description for NMB and λ compared with the
ICER is given in the cost-effectiveness diagram (Figure 4.5, where bλ is
the NMB).
The diagram shows that the slope of the WTP (i.e., the λ) is greater
than the slope of the ICER, which proves that option A is more
48 Economic Evaluation in Genomic Medicine
+ New technology
II I
Δ- Cost
rejected B
WTP
bλ
New technology
rejected
λ A
ICER = ΔC / ΔE
Δ- Effectiveness – Δ- Effectiveness +
New technology
rejected
New technology
New technology accepted
accepted
Δ- Cost –
III IV
Treatment comparison
High cost
Drug E
Time 3: Drug C
Drug D
LC Expensive?
Time 2: Drug B
Time 1: Drug A
A 12 12,500
B 13 13,000
C 11 23,000
D 10 24,000
E 8 8,000
Result:
Option Benefit (years) Cost (h)
E 8 8,000
D 10 24,000
C 11 23,000
A 12 12,500
B 13 13,000
ICER 5 ð11; 000 10; 000Þ=ð2:5 2:4Þ 5 h10; 000 per QALY
0 10 20 30 40 50 60 70 80 90 100
Patient
P
Perfect health
Treatment B
1–P
Death
Quality of life
Complete health
a b Time
Quality of life = a/b
Figure 4.9 The time trade-off.
even to only one question, then that patient loses an additional 20.269
points. The final estimate will be the patient’s quality of life (which will be
a number between 0 and 1). We should note that because such analyses are
based on statistical methods, the relevant scores will incorporate the flaws
of these methods. For example, there may be cases with scores less than
zero, cases where the loss of welfare due to the disease is nonlinear, and
other cases. A detailed presentation of the advantages and disadvantages of
such instruments is beyond the scope of this work.
In conclusion, if one wants to estimate QALYs using a clinical ques-
tionnaire (e.g., PASI for psoriasis or DAS28 for rheumatoid arthritis),
then statistical methods should be used to convert clinical estimates to
QALYs based on the literature. To do this, one would enter the clinical
estimates into an equation that includes utility and therefore calculate the
QALYs (e.g., an equation like QALY 5a b 3 PASI, where a and b are
calculated for specific samples through statistical regression).
groups, between special-interest groups and the rest of the society, and
so on, so the unquestioning acceptance of additivity should be evalu-
ated according to the problem to be solved and with consideration to
the available data in each case.
9. A QALY is a QALY regardless of who gains or loses it. The last three
axioms are obviously simplistic and research is being done to rebut
some of their features so that they will more clearly reflect the real
world and the decision of the decision-makers. For example, an ambi-
tious approach would be to establish the “weight” of a QALY
obtained from cardiology, oncology, or other field and then additively
maximize some type of social welfare equation. In any case, such
issues are rather delicate and require broad participation of many soci-
etal partners to make decisions based on mathematical models.
In concluding this practical summary, there is another “narrower”
approach to the QALY in applied research that is more commonly used
but that is less interesting from a theoretical viewpoint. This second
approach involves the investigation of differences in the quality of life of
patients with comparative treatments for the same condition. This type of
approach is quite reasonable because many new innovative treatments are
not different from older ones with regard to survival but are easier to
administer and have fewer side effects. These are all important factors and
should be taken into account.
4. If estimates came from a subgroup analysis, were the groups prespecified at the beginning
of the study? 1
5. Was uncertainty handled by (1) statistical analysis to address random events and (2) sensitivity
analysis to cover a range of assumptions? 9
6. Was incremental analysis performed between alternatives for resources and costs? 6
7. Was the methodology for data abstraction (including the value of health states and other
benefits) staled? 5
8. Did the analytic horizon allow time for all relevant and important outcomes? Were benefits
and costs that went beyond 1 year discounted (3%–5%) and justification given for the
discount rate? 7
9. Was the measurement of costs appropriate and the methodology for the estimation of
quantities and unit costs clearly described? 8
10. Were the primary outcome measure(s) for the economic evaluation clearly stated and did they 6
include the major short-term was justification given for the measures/scales used?
11. Were the health outcomes measures/scales valid and reliable? If previously tested valid and
reliable measures were not available, was justification given for the measures/scales used? 7
12. Were the economic model (including structure), study methods and analysis, and the
components of the numerator and denominator displayed in a clear, transparent manner? 8
13. Were the choice of economic model, main assumptions, and limitations of the study stated
and justified? 7
14. Did the author(s) explicitly discuss direction and magnitude of potential biases? 6
15. Were the conclusions/recommendations of the study justified and based on the study results? 8
16. Was there a statement disclosing the source of funding for the study? 3
TOTAL POINTS 100
construct validity (Ofman et al., 2003). This led Chiou et al. (2003) to
develop and validate a Quality of Health Economic Studies (QHES)
instrument that the authors claim will simplify the assessment of quality of
health economic evaluations. The QHES uses 16 explicit elements that
are answerable using a yes/no approach combined with a weighted scor-
ing system with a total possible score of 100. A full evaluation of the
QHES instrument is beyond the scope of the chapter; however, a copy of
the tool is provided here (Figure 4.10).
CONCLUSIONS
The reader will note that the elements included in Figure 4.10 reflect the
content of this chapter very well. This tool not only is useful in the critical
appraisal of published economic studies but also can serve as a guide in the
design of economic studies. Careful attention to the methods outlined in
this chapter and the elements from the QHES tool will improve the valid-
ity and reduce the bias of economic analyses. This approach will yield
results that are more relevant to and trusted by the intended audience.
62 Economic Evaluation in Genomic Medicine
REFERENCES
Barber, J.A., Thompson, A., 1998. Analysis and interpretation of cost data in randomised
controlled trials: review of published studies. BMJ 317, 11951200.
Black, W.C., 1990. The CE plane: a graphic representation of cost-effectiveness. Med.
Decis. Making 10, 212214.
Briggs, A.H., O’Brien, B.J., 2001. The death of cost-minimization analysis? Health Econ.
10 (2), 179184.
Canning, D., 2009. Axiomatic foundations of cost effectiveness analysis. Working Paper Series.
PGDA Working Paper No.51 ,http://www.hsph.harvard.edu/pgda/working.htm..
Cantor, S., 1994. Cost-effectiveness analysis, extended dominance, and ethics. A quantita-
tive assessment. Med. Decis. Making 14, 259265.
Chiou, C.F., Hay, J.W., Wallace, J.F., Bloom, B.S., Neumann, P.J., Sullivan, S.D., et al.,
2003. Development and validation of a grading system for the quality of cost-
effectiveness studies. Med. Care 41 (1), 3244.
Claxton, K., 1999. The irrelevance of inference: a decision-making approach to the
stochastic evaluation of health care technologies. J. Health Econ. 18 (3), 341364.
Collett, D. (Ed.), 2003. Modelling Survival Data in Medical Research, second ed.
Chapman & Hall/CRC Press, Boca Raton, FL.
Detsky, A.S., Naglie, I.G., 1990. A clinician’s guide to cost-effectiveness analysis. Ann.
Intern. Med. 113 (2), 147154.
Drummond, M., Sculpher, M., Torrance, G., 2005. Methods for the Economic
Evaluation of Health Care Programmes. Oxford University Press, Oxford.
Efron, B., Tibshirani, R.J., 1993. An Introduction to the Bootstrap. Chapman & Hall/
CRC Press, Boca Raton, FL.
EuroQol Group Association, EuroQol EQ-5Dt. ,http://www.euroqol.org/fileadmin/
user_upload/Documenten/PDF/Products/Sample_UK__English__EQ-5D-3L_Paper_
Self_complete_v1.0__ID_23963_.pdf..
Goetghebeur, M.M., Rindress, D., 1999. Towards a European consensus on conducting
and reporting health economic evaluations—a report from the ISPOR inaugural
European conference. Value Health 2, 281287.
Gray, A., Clarke, P., Wolstenholme, J., 2011. Applied Methods of Cost-Effectiveness
Analysis in Healthcare. Oxford University Press, Oxford.
Morrison, G.C., 1998. Understanding the disparity between WTP and WTA:
endowment effect, substitutability, or imprecise preferences? Econ. Lett. 59,
189.
National Institute for Health and Clinical Excellence. Guide to the Methods of
Technology Appraisal, March 2011. ,http://www.nice.org.uk/media/B52/A7/
TAMethodsGuideUpdatedJune2008.pdf..
O’Brien, B.J., Gafni, A., 1996. When do the dollars make sense? Toward a conceptual
framework for contingent valuation studies in health care. Med. Decis. Making 16,
288299.
O’Brien, B.J., Gertsen, K., Willan, A.R., Faulkner, L.A., 2002. Is there a kink in consu-
mers’ threshold value for cost-effectiveness in health care? Health Econ. 11 (2),
175180.
Ofman, J.J., Sullivan, S.D., Neumann, P.J., Chiou, C.F., Henning, J.M., Wade, S.W.,
et al., 2003. Examining the value and quality of health economic analyses: implica-
tions of utilizing the QHES. J. Manag. Care Pharm. 9 (1), 5361.
Palmer, S., Raftery, J., 1999. Opportunity cost. BMJ 318 (7197), 15511552.
Pauly, M.V., 1995. Valuing Health Care Benefits in Money Terms. Cambridge University
Press, Cambridge.
Introduction to the Technical Issues of Economic Evaluation 63
Ramsey, S., Willke, R., Briggs, A., Brown, R., Buxton, M., Chawla, A., et al., 2005.
Good research practices for cost-effectiveness analysis alongside clinical trials: the
ISPOR RCT-CEA task force report. Value Health 8 (5), 521530.
Sullivan, R., Peppercorn, J., Sikora, K., et al., 2011. Delivering affordable cancer care in
high-income countries. Lancet Oncol. 12 (10), 933980.
Torrance, G.W., 1986. Measurement of health state utilities for economic appraisals:
a review. J. Health Econ. 5, 130.
WHO guide to cost-effectiveness analysis, 2003. ,www.who.int/choice/publications/
p_2003_generalised_cea.pdf..
Willan, A., Lin, D.Y., 2001. Incremental net benefit in randomized clinical trials. Stat.
Med. 20, 15631574.
CHAPTER 5
INTRODUCTION
In the previous chapter we have provided an overview of various technical
issues of economic evaluation. In this chapter, we describe some methodo-
logical issues involved in the economic evaluation of health services. The
handling of each of these factors may be extremely important because it can
lead to entirely different evaluation results for similar health technologies. The
decision-makers must be aware of the assumptions used in each economic
model and understand how the assumptions can impact the results of the
model. Only then will they be able to use the analysis results as a practical
tool for drafting and implementing political decisions.
MODEL TYPES
Generally speaking, there are two types of analysis used in economic evalua-
tion. The first uses patient data (raw data), when the relevant records are
available, to calculate all basic economic quantities. In this case, we use
techniques borrowed from statistics (which will be explained in the next
chapter). A typical case is economic evaluation performed in parallel with a
clinical trial. In the second type, we create models simulating the course of
the disease with data from the literature. In certain cases, we might use data
both from the literature and from the available clinical trial results. For
example, in certain cases of chronic diseases, we would be interested in the
economic evaluation performed in parallel with a clinical trial (short-term),
but we would also be interested in economic evaluation in the longer-term
for the same patients; this would necessitate combining data from clinical
studies with data from available databases (registries). We also need to deter-
mine how our results would be affected if we combined our clinical trial
data with cost data from different countries.
time (at least) generally increases the patient’s odds of moving more
rapidly to a less favorable state of health (e.g., death by old age), regardless
of whether there have been any other adverse developments in the parti-
cular disease being examined.
According to this model, all the patients with the lowest ICER (colorectal
cancer patients) would be financed first, then breast cancer patients, and so
on. In such a case, certain expensive treatments would be left without
reimbursement because they would be significantly more expensive than
others and therefore uneconomical for the society, and resources would be
insufficient to cover all needs.
A full utilization of the model would require the existence and awareness
of the society’s welfare equation, a desire by all citizens to maximize this
equation, and the establishment of an independent authority that would
implement the citizens’ wishes through this fully mathematicized equation.
It is, however, intuitively obvious that all of these represent a theoretical
construct that does not match the reality or the needs of health care
decision-makers, nor does it account for political and special interest
influences. Furthermore, it is difficult (and possibly pointless) to compare dif-
ferent forms of cancer, let alone completely different disease types that require
different patient treatments, that are associated with different ICER values,
and also that have different severities, as mentioned in the previous chapter.
Furthermore, the development of the model assumes that the society
is using a fixed budget, that a fixed efficiency of scale in health services,
which is not affecting the ICER, may be assumed when a program is
scaled up or down, that full information is disclosed for all available treat-
ments, and that the individual programs are completely independent.
In many cases, the assumption of a fixed budget may not be true
because governments may invest more resources into an intervention
program that can appreciably improve the level of health for the popula-
tion. If the budget is indeed fixed, then the acceptable ICER will change
over time as new treatments are developed, which will alter the relative
position of the respective diseases in the league table. According to a
different version of the model, the ICER could remain fixed provided
the budget is variable. In that case, the introduction of new treatments
would have no effect on the ICER because the budget would be adjusted
accordingly by the equivalent percentage of the additional burden caused
by the introduction of the new interventions.
Furthermore, the assumption of fixed economies of scale may be stip-
ulated only if the fixed cost of a program is zero or negligible, so that any
changes in its implementation scale will lead to similar health results. The
assumption of full disclosure is a simplification because no health system,
insurance carrier, or private enterprise can ever be fully informed. In
almost every case, it is not certain what benefits will arise and what will
Advanced Methodological Aspects in the Economic Evaluation 69
1
Probability of rejection
Model 1
Model 2
0.5 Model 3
Model 4
0
0 10,000 20,000 30,000 40,000 50,000 60,000 70,000 80,000 90,000 100,000
Cost-effectiveness ratios
Source: Devlin and Parkin. Health economics 2004; 13: 437–452
Figure 5.1 Probability of acceptance depending on the ICER.
Advanced Methodological Aspects in the Economic Evaluation 71
returns on scale and full divisibility. Constant returns mean that the λ is
unrelated to the size of the implemented program. In its simplified form,
full divisibility means that we can purchase any health care program on a
scale as small as needed; this is a simplified form and full-divisibility for-
mulation. Unfortunately, the conditions described here for the correct
calculation of the λ are not yet met by any health system in the world.
ESTIMATES OF EFFECTIVENESS
For economic evaluation, one needs to accurately and reliably estimate
the effectiveness of each median intervention. In many cases one can refer
to various sources to estimate the effectiveness of a treatment, and each of
these sources may have different advantages and disadvantages. The rank-
ing of such sources reflects, in part, the reliability of the data collected
and, therefore, the quality of the analysis results.
The best known source is the clinical trial, which belongs to a wider class
of studies called “controlled experiments.” Clinical trials are scientific experi-
ments involving people who suffer from a disease that assess the difference in
response between a new treatment and an alternative, for example, different
therapy regimens are selected and compared with each other (http://www.
who.int). Clinical trials are conducted in four phases with each one assessing
different aspects of the technology undergoing investigation, such as safety,
maximum tolerated dose of a drug, effectiveness, and comparison of the drug
with other alternative treatments in a large population sample. They are
considered the most reliable source of data and are currently an obligatory
crucible through which each treatment must pass to be used in the general
population. They are characterized by high internal reliability thanks to the
very strict patient inclusion criteria and the standardized documentation of
the results, they may apply double-blind randomization, and they have high
validity thanks to support by modern statistical techniques that minimize
sampling errors and bias. Their disadvantage is their low external reliability
because general population patients have lower compliance to treatment, may
suffer from other concomitant diseases, and may present different charac-
teristics than the study reference population.
The second source of data is meta-analysis. Meta-analysis is a statistical
technique aimed at summarizing results obtained from clinical trials
(Petiti, 1994). It does not constitute original research but draws its infor-
mation from the clinical trials included in the analysis. The most impor-
tant advantage of meta-analysis is that it can draw conclusions at greater
Advanced Methodological Aspects in the Economic Evaluation 73
valued (Muennig, 2008). The study perspective is a key factor when deter-
mining the cost categories that will ultimately be involved in the analysis.
The approach selected will not always be the same; it will differ
depending on the purpose of the analysis. For example, if the issue
involves distributing resources between various sectors of the economy
(such as education, health, defense), then the analysis to be used will be
the cost-benefit analysis to determine all the consequences of the rele-
vant options. If, instead, the analysis focuses on distributing resources
between different sectors of health care (prevention, treatment, etc.) or
between different interventions for the treatment of a specific condi-
tion, then the consequences to be measured will be more limited. The
selection of analysis method is also affected by the person or institution
performing it (patient, hospital, insurance carrier, etc.) and by the avail-
ability of relevant data. In practice, economic evaluation is used exten-
sively for “narrow-type” analyses (e.g., alternative interventions within
a disease), and less so for interventions involving different sectors of the
economy. In this case, economic theory assumptions do not fully reflect
the individuals’ selections and economic evaluation is commonly used
as a tool and not as a complete system of values with which to make
the relevant decisions.
For example, if the study concerns an insurance carrier, then the anal-
ysis is performed from the perspective of the insurance fund and supplier
charges are examined; however, if the study focuses on all possible conse-
quences, then the perspective is social (Table 5.2). The social perspective
includes all possible consequences with no regard for who pays for them.
If the analysis concerns an employer, then it would include the charges
for insurance costs and loss of employee productivity. In this case, costs
such as transfer to the hospital, feeding costs, and others, which are
covered by the employee, will not be included in the analysis, whereas
costs related to loss of productivity or temporary personnel would be
included. Currently, the social perspective is considered to be the most
appropriate for economic analyses because it includes all of the relevant
economic flows within a system. However, this should not be interpreted
to mean that the tools of economic analysis are not useful for decision-
making from other stakeholder perspectives.
When examining the inclusion of costs, we use a process through
which the resources expended for the production of medical interventions,
the human-hours consumed for providing health care, and the loss to the
productive process due to the patient’s inability to work and contribute to
the national product are calculated.
Specifically, the concept of cost includes the following (Phillips, 2005):
• Direct costs: The actual cost consumed for the intervention
• Direct health care costs: The cost caused by health care suppliers (the
total expenditures for monitoring, treatment, diagnostic tests, medi-
cation, etc., which result from the treatment)
• Direct nonhealth care costs: Expenditures arising for the patient as a
result of the disease as well as the treatment-seeking process (home
help costs, travel expenses, special diet expenses, etc.)
• Indirect cost: Financial losses that are a result of the disease and do not
include the costs for providing treatment
• Indirect cost essentially refers to the loss of productivity because of
the disease, either because of work absenteeism or because of reduced
productivity (presenteeism)
• It reflects the value of the goods that the patient could have pro-
duced if he/she had not become sick
• It usually includes lost productivity, free time, time expended by
relatives providing assistance, and other time lost
• Finally, this type of cost includes lost productivity due to premature
death
• Invisible (intangible) cost: A term that describes difficult-to-measure
consequences of the disease and its treatment
• It is due to the pain, discomfort, reduced quality of life, or other
social or moral consequences of the disease (or its treatment)
Advanced Methodological Aspects in the Economic Evaluation 77
Prices will change every year; therefore, any health expenditures made
today will have greater value compared with future expenditures. From a
different viewpoint, one could claim that as the national product grows
over time, a fixed flow of expenditure will account for a smaller percent-
age of the total income and therefore reflect a smaller burden, which
should be calculated by the discount method. In this case, the discount
rate reflects the rate of economic growth.
Furthermore, even in a world without inflation, a person would rather
have an amount of money today than the same amount at any future
time. In other words, they would rather enjoy benefits today than in the
future. This approach does not reflect any specific economic theories, but
rather it reveals the effect of psychological and social factors outside
conventional limits of rational thought in individuals or patients. A person
will consider future cash flows uncertain; therefore, short-term cash flows
are more significant than long-term flows.
To define in practice the preferred discount rate, one possible
approach would take into account the interest rate of a financial product
free from market risk, such as long-term Treasury bonds, based on the
rates of individual time preferences. A second alternative would be to
determine the funds’ shadow price, which takes into account the social
time preference rate. With this analysis, the social time preference interest
rate is determined by the total loss of current private consumption in
relation to the long-term flow of benefit that will be obtained by under-
taking a public program, specifically the health care intervention program
under study. The two methods are associated with advantages and disad-
vantages. A full theoretical presentation of the respective arguments is
beyond the scope of this chapter.
Similar arguments are presented for benefit discounting, which has
also caused disputes among economists (Drummond and Torrance, 2005).
Those opposed to benefit discounting argue that it is difficult to conceive
of a “long-term investment in health” or an exchange of cash flows by
individuals or society. They also maintain that flow discount implies that
less weight is given to future health care benefits, which means that the
current generation is given priority over future generations. The final
argument is that people have different understanding and approaches to
the monetary cost consumed at different time periods and to the health
benefits obtained by health care intervention programs.
On the opposing side, the supporters of the discount technique
claim that, after an intervention, the patient will “consume” health care
benefits over the long-term; therefore, a discount of future flows appears
80 Economic Evaluation in Genomic Medicine
C
PDV 5
ð11rÞn
Table 5.4 Comparison of discount rates for cost and benefits in various countries
Cost Benefit
Baltic countries 5% 5%
Belgium 3% 1.5%
Canada 5%; SA: 03% 5%; SA: 03%
Finland 3% and 0% 3% and 0%
France 0%, 3% and 5% 0%, 3% and 5%
Germany 5%, SA (0%, 3%, 10%) 5%, SA (0%, 3%, 10%)
Hungary 5%; SA: 36% 5%; SA: 06%
Ireland YES YES
Italy 3%; SA: 08% 3%; SA: 08%
New Zealand 3.5%, SA: (0%, 5%, 10%) 3.5%, SA: (0%, 5%, 10%)
Norway 2.5%, SA: 08% 2.55%, SA: 08%
Poland 0% and 5% 0% and 5%
Portugal 5% 5%, 0%
Scotland 6%; SA: 0%—full 1.5%; SA: 0%—full
treasury discount rate Treasury discount rate
Spain 6% 6%
Sweden 3%; SA: 05% 3%; SA: 05%
The Netherlands 4% 1.5%
United Kingdom 3.5%; SA: 06% 3.5%; SA: 06%
United States Expert recommendation Expert recommendation
Switzerland 2.5%, 5%, 10% 2.5%, 5%, 10%
China Yes and no, with Yes and no, with
justification justification
Austria 5%, SA: 310% 5%, SA: 310%
Brazil 5%, SA: 010% 5%, SA: 010%
Cuba Yes, as recommended by Yes, with justification
state experts
Slovakia 7% 7%
Thailand 3% 3%
Mexico 5%, SA: 37% 5%, SA: 37%
SA, sensitivity analysis.
Source: Data adapted from http://www.ispor.com, International Society for Pharmacoeconomics and
Outcome Research (ISPOR, 2010).
If the cost of the intervention is spread equally over n years, then the
formula is generalized as follows:
C C C C
PDV 5 1 1 2 1 3 1?1
ð11rÞ ð11rÞ ð11rÞ ð11rÞn
82 Economic Evaluation in Genomic Medicine
or
1 1 1 1
PDV 5 C 1 1 2 1 3 1?1
ð11rÞ ð11rÞ ð11rÞ ð11rÞn
or
1 2 ð11rÞ2n
PDV 5 C
r
An example of the technique’s application for a 10-year investment at
a rate of 5%, beginning at year 1, is given in Table 5.5.
In this example, an amount of h10,000 invested annually over 10 years is
equivalent to h7,722 in present values. The result can be directly calculated
using the formula as follows:
1 2 ð11rÞ2n 1 2 ð110:05Þ210
PDV 5 C 5 h1;000 5 h7;722
r 0:05
For a technical manipulation of inflation and time preferences for a
health program, we can either appreciate all future flows using a single,
projected percentage of the inflation (provided it is the same for all
elements of the cost), or we can appreciate none of the flows and use a
lower single interest rate that will express time preferences only. However,
if the elements composing the cost have different rates of price change,
then either we appreciate them at the appropriate inflation rate and then
discount them at a generic rate (inflation 1 time preference) or we do not
Table 5.5 Calculation of the PDV for a 10-year health investment projecta
Year Interest rate Cost (h) Discounted cost (h)
1 0.952 1,000 952
2 0.907 1,000 907
3 0.864 1,000 864
4 0.823 1,000 823
5 0.784 1,000 784
6 0.746 1,000 746
7 0.711 1,000 711
8 0.677 1,000 677
9 0.645 1,000 645
10 0.614 1,000 614
Total 7.722/10 years 5 0.722 10,000 7,722
a
(r 5 5%).
Advanced Methodological Aspects in the Economic Evaluation 83
appreciate and we instead use a weighted total discount rate that expresses
only time preference. In practice, for reasons of simplicity, prices are taken
as fixed (without inflation) and only one rate, expressing time preference
between alternative treatments, is used. In many cases, this assumption is
methodologically consistent because many interventions have brief time
scales and their price changes are negligible. Alternatively, we might say
that the discount rate reflects the combined effect of inflation, growth of
the economy, and social time preference.
SENSITIVITY ANALYSIS
The term “sensitivity” essentially refers to the way in which our results
change when we change our model’s assumptions. If sensitivity is high, then
the results vary greatly when we change certain assumptions; these assump-
tions must be very robustly established for our model to have any validity.
To put it concisely and intuitively, we could say that economic evalua-
tion is beset by many kinds of uncertainties. First, there is uncertainty about
the structure of the model we have created (structural uncertainty). For
example, is our approach to the issue the correct one? Does our analysis
reflect reality? Is our model of clinical practice incorrect? To answer such
questions, we need to critically evaluate our model and perform various
sensitivity analyses, changing its structure to judge its value.
Another source of uncertainty is heterogeneity between the various
subgroups to which the model refers (variability due to heterogeneity). If
we want to compare the cost and effectiveness of a new cardiology drug
with the standard treatment for the management of acute myocardial
infarction, then perhaps we should perform separate analyses for men and
women, young and elderly patients, patients with previous infarctions, and
so on, because these groups will give different results. Because heterogene-
ity is important for economic models, it should be included in economic
evaluation. Such subanalyses (men vs. women, etc.) are sensitivity analyses
for heterogeneity.
Uncertainty also exists between patients. We cannot be sure how long
a patient will actually live after a procedure, and even “similar” patients
do not survive for the same length of time, nor does their care cost the
same. This type of uncertainty is called first-order uncertainty. This would
theoretically decrease if data from studies with large samples or valid
meta-analyses were available. Realistically, there will always be some first-
order uncertainty because nature is inherently stochastic.
84 Economic Evaluation in Genomic Medicine
ET = 10 months ES = 9 months
CT = €10,000 CS = €9,000
– +
ΔE
–
Figure 5.4 Point representation of the ICER.
Advanced Methodological Aspects in the Economic Evaluation 87
Experiment 1
CT = €12,000 CS = €6,000
Experiment 2
ET = 14 months ES = 10 months
CT = €14,000 CS = €10,000
Note that in the two experiments we ran, the ΔC, the ΔE, and the
ICER are different each time, which means that we have moved on from
deterministic results to stochastic analysis (i.e., analysis that includes uncer-
tainty). We should, however, note that in this simple example we did not
take into account any correlation in the data, but we arbitrarily assumed that
there is no correlation. For that reason, we allow variables to move indepen-
dently along their entire distribution. In reality, correlation often appears in
the data; for example, if the cost is high, then survival may also be high.
This concept of correlation is represented schematically in Figure 5.6.
Regardless of correlation (positive or negative), by this process we
create many ICERs (as many as 5,000 or more) and plot them in a diagram
as shown in Figure 5.7, where each point represents one experiment
and, therefore, one calculation of the ICER (the diagram is based on hypo-
thetical results).
This diagram prompts the following question: How can I handle all of
this information and represent it concisely? The last step of this analysis is
to assume various values of λ within a reasonable range and use these to
find the percentage of points that are cost-effective. In this way, instead of
relying on an unrealistic approach in which the intervention would be
considered 100% cost-effective at exactly h1 above the ICER we calcu-
lated and not cost-effective at h1 below it, again with a probability of
100%, the analysis will now take a probabilistic nature. Let A, B, and C
symbolize various values of λ, which we assume (in applied analysis the
Advanced Methodological Aspects in the Economic Evaluation 89
Incremental cost
800
600 €
400
ys
200
–200
–400
–0.40 –0.20 0 0.20 0.40 0.60 0.80 1.00 1.20
Incremental effect
Figure 5.7 Dot plot of ICER representation.
600 €
400
ys
200
–200
–400
–0.40 –0.20 0 0.20 0.40 0.60 0.80 1.00 1.20
Incremental effect
Figure 5.8 Derivation of the acceptability curve.
0.8
0.7
0.6
0.5
0.4
0.3
0.2
0.1
CONCLUSIONS
This chapter, like the previous one, has presented the theory and metho-
dology behind high-quality economic analysis in health care. In the sub-
sequent chapters, we illustrate the application of economic analysis to
genomic medicine interventions that are emerging into clinical practice,
using some key examples from the published literature. Furthermore, we
describe the special provisions required to accurately model the evaluation
of the analysis costs and resource use, and the analysis outcomes and effec-
tiveness in genomic medicine applications.
REFERENCES
Arrow, K.J., 1963. Social Choice and Individual Values, second ed. Wiley, New York, NY.
Barton, G.R., Briggs, A.H., Fenwick, E.A., 2008. Optimal cost-effectiveness decisions:
the role of the cost-effectiveness acceptability curve (CEAC), the cost-effectiveness
acceptability frontier (CEAF), and the expected value of perfection information
(EVPI). Value Health 11 (5), 886897.
Briggs, A.H., Fenn, P., 1998. Confidence intervals or surfaces? uncertainty on the cost-
effectiveness plane. Health Econ. 7, 723740.
Casali, P.G., 2007. The off-label use of drugs in oncology: a position paper by the
European Society for Medical Oncology (ESMO). Ann. Oncol. 18 (12), 19231925.
Choi, I.Y., Kim, T.M., Kim, M.S., Mun, S.K., Chung, Y.J., 2013. Perspectives on clinical
informatics: integrating large-scale clinical, genomic, and health information for clini-
cal care. Genomics Inform. 11 (4), 186190.
Cialkowska, M., Adamowski, T., Piotrowski, P., Kiejna, A., 2008. What is the Delphi
method? Strengths and shortcomings. Psychiatr. Pol. 42 (1), 515.
Claxton, K., Sculpher, M., McCabe, C., et al., 2005. Probabilistic sensitivity analysis for
NICE technology assessment: not an optional extra. Health Econ. 14 (4), 339347.
Cohen, B.J., 2003. Discounting in costutility analysis of healthcare interventions:
reassessing current practice. Pharmacoeconomics 21 (2), 7587.
Daggy, J.K., Thomas III, J., Craig, B.A., 2011. Modeling correlated healthcare costs.
Expert Rev. Pharmacoecon. Outcomes Res. 11 (1), 101111.
Devlin, N., Parkin, D., 2004. Does NICE have a cost-effectiveness threshold and what
other factors influence its decisions? a binary choice analysis. Health Econ. 13 (5),
437452.
Drummond, M.F., Torrance, G.W., 2005. Methods for the Economic Evaluation of
Health Care Programmes. Oxford University Press, Oxford.
Eichler, H.G., Kong, S.X., Gerth, W.C., Mavros, P., Jonsson, B., 2004. Use of cost-
effectiveness analysis in health-care resource allocation decision-making: how are cost-
effectiveness thresholds expected to emerge? Value Health 7 (5), 518528.
Fenwick, E., O’Brien, B.J., Briggs, A.H., 2004. Cost-effectiveness acceptability curves—
facts, fallacies and frequently asked questions. Health Econ. 13, 405415.
Friedman, M., 1953. Essays in Positive Economics. University of Chicago Press, Chicago, IL.
Greenberg, D., Earle, C., Fang, C.H., Eldar-Lissai, A., Neumann, P.J., 2010. When is
cancer care cost-effective? a systematic overview of cost-utility analyses in oncology.
J. Natl. Cancer Inst. 102 (2), 8288.
96 Economic Evaluation in Genomic Medicine
Greenfield, S., Steinberg, E., 2011. Clinical Practice Guidelines We Can Trust. Institute
of Medicine, National Academies Press, Washington, DC.
Hillman, A.L., Kim, M.S., 1995. Economic decision making in healthcare. A standard
approach to discounting health outcomes. Pharmacoeconomics 7 (3), 198205.
Jain, R., Grabner, M., Onukwugha, E., 2011. Sensitivity analysis in cost-effectiveness
studies: from guidelines to practice. Pharmacoeconomics 29 (4), 297314.
Katz, D.A., Welch, H.G., 1993. Discounting in cost-effectiveness analysis of healthcare
programmes. Pharmacoeconomics 3 (4), 276285.
Meineke, F.A., Stäubert, S., Löbe, M., Winter, A.A., 2014. Comprehensive clinical
research database based on CDISC ODM and i2b2. Stud. Health Technol. Inform.
205, 11151119.
Meltzer, D., 2001. Addressing uncertainty in medical cost-effectiveness analysis implications
of expected utility maximization for methods to perform sensitivity analysis and the use
of cost-effectiveness analysis to set priorities for medical research. J. Health Econ. 20 (1),
109129.
Muennig, P., 2008. Cost Effectiveness Analysis in Health Care, second ed. Jossey-Bass,
San Francisco, CA.
O’Hagan, A., Stevens, J.W., Montmartin, J., 2000. Inference for the cost-effectiveness
acceptability curve and cost-effectiveness ratio. Pharmacoeconomics 17 (4), 339349.
Petiti, D., 1994. Meta-Analysis, Desicion Analysis, and Cost-Effectiveness Analysis.
Oxford University Press, New York, NY.
Phillips, C.G., 2005. Health Economics. An introduction for Health Professionals.
Blackwell Publishing Ltd, Massachusetts.
Prokosch, H.U., Ganslandt, T., 2009. Perspectives for medical informatics. Reusing the
electronic medical record for clinical research. Methods Inf. Med. 48 (1), 3844.
Shane, F., Loewenstein, G., O’Donoghue, T., 2002. Time discounting and time preference:
a critical review. J. Econ. Lit. 40 (2), 351401.
Smith, D.H., Gravelle, H., 2001. The practice of discounting in economic evaluations of
healthcare interventions. Int. J. Technol. Assess. Health Care 17 (2), 236243.
Towse, A., 2009. Should NICE’s threshold range for cost per QALY be raised? Yes. BMJ
338 (b181).
Weinstein, M., Zeckhauser, R., 1973. Critical ratios and efficient allocation. J. Public Econ.
2, 147157.
Yoder, J.L., 2008. The importance of cost inputs and sensitivity analyses in a cost-
effectiveness analysis. Clin. Drug Investig. 28 (7), 461462.
CHAPTER 6
INTRODUCTION
The preceding chapters have explored the theory and methods behind
high-quality economic analysis in health care. Although some examples
from genomic medicine have been used to illustrate theoretical aspects of
the methods, this chapter uses examples from the published literature to
examine the application of economic analysis to genomic medicine inter-
ventions that are emerging into clinical practice. As has been noted previ-
ously in the book, these are early days for the field, so the number of
published studies is still relatively small. Nonetheless, the studies that have
been published represent a diverse approach to the application of eco-
nomic analysis to genomic medicine instances that serve the purposes of
this chapter well.
This chapter is organized to illustrate some of the opportunities and
challenges introduced in the prior chapters. The examples emphasize
examination of the critical aspects of the analysis that can impact the
validity of the analysis. The chapter finishes with an exploration of the
opportunity economic analysis presents in emerging countries looking to
implement genomic medicine. The reader should bear in mind that this
chapter is not an exhaustive review of the few existing studies of eco-
nomic evaluation in genomic medicine, but rather a collection of the
most representative studies to demonstrate the importance of economic
evaluation in genomic-guided therapies.
Appropriate price indexing was used and a discount rate for future costs
was applied. The authors appropriately considered differences in allele fre-
quency based on ethnicity and included realistic ethnic distributions in
the model. The authors made clear that the only complication of treat-
ment considered was the most severe adverse event, myelosuppression.
The estimates of the frequency were derived primarily from adult studies
in which 6-MP was used for inflammatory bowel disease or other inflam-
matory conditions. Applying adult experience to pediatric patients is a
problem. In addition, the indication for treatment can make a huge dif-
ference in the balance of risk and benefit, as is illustrated in the following
section of this chapter. A key parameter for the model is the cost of an
episode of myelosuppression. The authors identified a number of sources
for costs associated with inpatient and outpatient treatments, developing
an average cost of the treatment from the perspective of three different
national health systems and then making a determination of the average
cost to be used for purposes of the model. The sources were cited and
the calculations used were provided. Another challenging parameter to
determine is how many of the adverse events can be attributed to the
presence of a TPMT variant. Although presence of decreased TPMT
activity is known to be associated with increased risk of myelosuppression,
the nature of the treatment means that even those with normal activity
are at risk for an adverse event. In fact, given that the majority of patients
have normal TPMT activity, the absolute number of adverse events in
this population is higher than in the population with decreased activity.
The authors concluded that, for the purposes of the model, they would
attribute 32% of all cases of myelosuppression to a TPMT variant. Stated
another way, for every ADE occurring in a patient with decreased
TPMT activity, two would occur in patients with normal activity. It is
important to state again that the estimates of ADE risk were obtained in
studies of adults for disorders other than ALL. Finally, the authors noted that
severe ADEs have the potential to result in loss of life. As could be
expected in a study that focuses on children, the prevention of a child-
hood death leads to a large number of life-years gained. This must be
tempered by the fact that ALL can also lead to premature death, so realis-
tically estimating the average life expectancy of all treated patients com-
pared with peers without ALL is very important. We return to this point
because it is critical in the balance of risk and efficacy. Given the uncer-
tainty in many of the model parameters, the authors used sensitivity
analyses to test the impact of the assumptions made.
Economic Evaluation in the Genomic Era: Some Examples from the Field 103
is decreased by 10%, then the mortality rate would increase from 10% to
11%. This means that the number of deaths in the genotyped group
would now be 10,900 (11,000 deaths—100 deaths prevented by genotyp-
ing), 900 more deaths than in the nongenotyped group. The impact on
efficacy would have to be reduced to a mere 1% difference for the num-
ber of deaths to be equal in the two groups, a number that seems unreal-
istically low given the potential impact on MRD. Clearly, the truth is
“still out there;” however, this should illustrate the importance of consid-
ering both efficacy and harm for the purposes of economic analyses.
The model did not identify a significant difference related to adverse events,
which the authors attribute to a low event rate. Interestingly, the efficacy
was similar for all three of the alternative therapies, which was unexpected
given that the monitoring and TMPT genotyping plus monitoring arms
allowed response-based dosing of AZA, which would be expected to result
in higher efficacy. It was not clear from the article if the assumptions for
efficacy were based on published data (ideally from prospective clinical
trials) or relied on expert opinion. These two results raise some questions
about the assumptions used in the model, an issue that the authors
acknowledge as a weakness given the reliance on expert opinion as opposed
to clinical trial data. The lack of robust clinical trial evidence represents an
ongoing challenge for economic analysis, as evidenced by the fact that mul-
tiple analyses over nearly 15 years still have not yielded a definitive answer
regarding the cost-effectiveness of TPMT genotyping prior to the use of
thiopurines for any indication.
PERSPECTIVES ON PERSPECTIVE
Economic analyses must specify from what perspective they are being
performed. The perspective used is important if the result is to be of use
to the intended stakeholder. To illustrate this, we use the example of
tumor-based screening for Lynch syndrome.
Colorectal cancer (CRC) is one of the most common cancers world-
wide. Approximately 2 5% of CRC is due to germline mutations in one
of several DNA mismatch repair genes, a condition called Lynch syn-
drome. Individuals who carry such a mutation have a lifetime risk of devel-
oping CRC that approaches 80%, and they are at higher risk for several
other cancers. Identification of a mutation carrier allows initiation of ear-
lier and more frequent colonoscopy and consideration of prophylactic sur-
gery, which has been shown to reduce the risk of developing certain types
of cancer associated with Lynch syndrome. Identification of an index case
allows identification of at-risk family members who have not yet devel-
oped cancer. The most effective way to identify Lynch syndrome in
a patient presenting with CRC is to screen the tumor for changes
that are indicative of a mutation (e.g., absence of staining for one of
the mismatch repair proteins or DNA microsatellite instability). Tumor-
based screening was recommended based on a systematic evidence review
and synthesis by the Evaluation of Genomic Applications in Practice and
Prevention Working Group (EGAPP, 2009). Soon thereafter, economic
106 Economic Evaluation in Genomic Medicine
including the number of CRC patients treated annually within our deliv-
ery system and the number for which we expect to have tumor tissue
available for screening. Our models allowed us to estimate the percent of
total test costs that would fall under bundled hospital payments (approxi-
mately 75%) and percent of screened patients who would be eligible for
sequencing, thus requiring consent and genetic counseling (5 6%). After
implementation of screening, a question arose as to whether screening
should be restricted to patients aged 50 years or younger, given that
Lynch syndrome usually results in tumors appearing at a younger age.
Models were used to assess the sensitivity of screening and cost per case
detected using a variety of age cutoffs. Applying cutoffs reduced the cost
of the screening program with a concomitant decrease in sensitivity.
Presentation of the data allowed leadership to consider both costs and
clinical impact, and it was ultimately decided not to impose an age cutoff
for screening (Gudgeon et al., 2013). Finally, the health care system col-
lects data from the screening of all of the key parameters of the model,
allowing testing of the performance of the screening program, allowing
for continuous improvement.
CONCLUSIONS
Genomic cost-effectiveness analysis has the potential to inform assess-
ments about the value of current and emerging technologies and priori-
tize value-based decisions about adoption and investment. Efforts to close
evidence gaps can strategically target areas of greatest need and potential
health and cost impacts. Economic evaluations from the perspective of
the relevant stakeholder can provide information and guidance for
decision-making and policy-making. Results from these evaluations can
include estimated ranges and threshold levels for key outcome variables to
achieve desirable real-world results. Economic analyses should adhere to
quality standards such as the QHES tool. Most importantly, given the rap-
idly changing nature of the evidence in genomics, economic models must
be developed that are flexible and adaptable. Ultimately, high-quality and
robust models must be developed that can be utilized by experienced sta-
keholders without high-level training in economics to encourage routine
use of these models to assist in decision-making.
REFERENCES
Bock, J.A., Fairley, K.J., Smith, R.E., Maeng, D.D., Pitcavage, J.M., Inverso, N.A., et al.,
2014. Cost-effectiveness of IL28B genotype-guided protease inhibitor triple therapy
versus standard of care treatment in patients with hepatitis C genotypes 2 or 3 infection.
Public Health Genomics 17, 306 319.
Classen, D.C., Pestotnik, S.L., Evans, R.S., Lloyd, J.F., Burke, J.P., 1997. Adverse drug events
in hospitalized patients. Excess length of stay, extra costs, and attributable mortality.
JAMA 22 (277), 301 306.
Classen, D.C., Jaser, L., Budnitz, D.S., 2010. Adverse drug events among hospitalized
medicare patients: epidemiology and national estimates from a new approach to
surveillance. Joint Comm. J. Qual. Patient Saf. 36 (1), 12 21.
Dong, D., Sung, C., Finkelstein, E.A., 2012. Cost-effectiveness of HLA-B 1502 genotyp-
ing in adult patients with newly diagnosed epilepsy in Singapore. Neurology 79,
1259 1267.
Dubinsky, M.C., Reyes, E., Ofman, J., Chiou, C.F., Wade, S., Sandborn, W.J., 2005. A
cost-effectiveness analysis of alternative disease management strategies in patients with
Crohn’s disease treated with azathioprine or 6-mercaptopurine. Am. J. Gastroenterol.
100, 2239 2247.
Evaluation of Genomic Applications in Practice and Prevention (EGAPP) Working
Group, 2009. Recommendations from the EGAPP working group: genetic testing
strategies in newly diagnosed individuals with colorectal cancer aimed at reducing
morbidity and mortality from lynch syndrome in relatives. Genet. Med. 11, 35 41.
FDA Information for Healthcare Professionals: Abacavir (marketed as Ziagen)
and Abacavir-Containing Medications. ,http://www.fda.gov/Drugs/DrugSafety/
PostmarketDrugSafetyInformationforPatientsandProviders/ucm123927.htm . (accessed
31.07.14).
112 Economic Evaluation in Genomic Medicine
Gudgeon, J.M., Williams, J.L., Burt, R.W., Samowitz, W.S., Snow, G.L., Williams, M.S.,
2011. Lynch syndrome screening implementation: business analysis by a healthcare
system. Am. J. Manag. Care 17, e288 e300.
Gudgeon, J.M., Belnap, T.W., Williams, J.L., Williams, M.S., 2013. Impact of age cutoffs
on a Lynch syndrome screening program. J. Oncol. Pract. 9, 175 179.
Hughes, C.A., Foisy, M.M., Dewhurst, N., Higgins, N., Robinson, L., Kelly, D.V., et al.,
2008. Abacavir hypersensitivity reaction: an update. Ann. Pharmacother. 42, 387 396.
Hughes, D.A., Vilar, F.J., Ward, C.C., Alfirevic, A., Park, B.K., Pirmohamed, M., 2004.
Cost-effectiveness analysis of HLA B 5701 genotyping in preventing abacavir hyper-
sensitivity. Pharmacogenetics 14, 335 342.
Kauf, T.L., Farkouh, R.A., Earnshaw, S.R., Watson, M.E., Maroudas, P., Chambers, M.
G., 2010. Economic efficiency of genetic screening to inform the use of abacavir sul-
fate in the treatment of HIV. Pharmacoeconomics 28, 1025 1039.
Mallal, S., Phillips, E., Carosi, G., Molina, J.M., Workman, C., Tomazic, J., et al., 2008.
PREDICT-1 study team. HLA-B 5701 screening for hypersensitivity to abacavir. N.
Engl. J. Med. 358, 568 579.
Mitropoulos, K., Johnson, L., Vozikis, A., Patrinos, G.P., 2011. Relevance of pharmacoge-
nomics for developing countries in Europe. Drug Metabol. Drug Int. 26, 143 146.
Mitropoulou, C., Fragoulakis, V., Bozina, N., Vozikis, A., Supe, S., Bozina, T, et al.,
Economic evaluation for pharmacogenomic-guided warfarin treatment for elderly
croatian patients with atrial fibrillation. In Press 2015.
Mizzi, C., Mitropoulou, C., Mitropoulos, K., Peters, B., Agarwal, M.R., van Schaik, R.H.,
et al., 2014. Personalized pharmacogenomics profiling using whole genome sequencing.
Pharmacogenomics 15, 1223 1234.
Mvundura, M., Grosse, S.D., Hampel, H., Palomaki, G.E., 2010. The costeffectiveness of
genetic testing strategies for lynch syndrome among newly diagnosed patients with
colorectal cancer. Genet. Med. 12, 93 104.
Ofman, J.J., Sullivan, S.D., Neumann, P.J., Chiou, C.F., Henning, J.M., Wade, S.W.,
et al., 2003. Examining the value and quality of health economic analyses: implica-
tions of utilizing the QHES. J. Manag. Care Pharm. 9, 53 61.
Relling, M.V., Gardner, E.E., Sandborn, W.J., Schmiegelow, K., Pui, C.H., Yee, S.W.,
et al., 2011. Clinical Pharmacogenetics Implementation Consortium. Clinical phar-
macogenetics implementation consortium guidelines for thiopurine methyltransferase
genotype and thiopurine dosing. Clin. Pharmacol. Ther. 89, 387 391.
Relling, M.V., Gardner, E.E., Sandborn, W.J., Schmiegelow, K., Pui, C.H., Yee, S.W.,
et al., 2013. Clinical Pharmacogenetics Implementation Consortium guidelines for
thiopurine methyltransferase genotype and thiopurine dosing. Clin. Pharmacol. Ther.
Advance online publication 20 February 2013. http://dx.doi.org/10.1038/clpt.2013.4.
Saag, M., Balu, R., Phillips, E., Brachman, P., Martorell, C., Burman, W., et al., 2008.
Study of hypersensitivity to abacavir and pharmacogenetic evaluation study team.
High sensitivity of human leukocyte antigen-B 5701 as a marker for immunologically
confirmed abacavir hypersensitivity in white and black patients. Clin. Infect. Dis. 46,
1111 1118.
San Miguel, R., Gimeno-Ballester, V., Mar, J., 2014. Cost-effectiveness of protease inhibi-
tor based regimens for chronic hepatitis C: a systematic review of published literature.
Expert Rev. Pharmacoecon Outcomes Res. 14, 387 402.
Schackman, B.R., Scott, C.A., Walensky, R.P., Losina, E., Freedberg, K.A., Sax, P.E.,
2008. The cost-effectiveness of HLA-B 5701 genetic screening to guide initial antire-
troviral therapy for HIV. AIDS 22, 2025 2033.
Snyder, S.R., Mitropoulou, C., Patrinos, G.P., Williams, M.S., 2014. Economic evaluation of
pharmacogenomics: a value-based approach to pragmatic decision-making in the face of
complexity. Public Health Genomics 17, 256 264.
Economic Evaluation in the Genomic Era: Some Examples from the Field 113
Stanulla, M.(1), Schaeffeler, E., Flohr, T., Cario, G., Schrauder, A., Zimmermann, M.,
et al., 2005. Thiopurine methyltransferase (TPMT) genotype and early treatment
response to mercaptopurine in childhood acute lymphoblastic leukemia. JAMA 293,
1485 1489.
Tangamornsuksan, W., Chaiyakunapruk, N., Somkrua, R., Lohitnavy, M., Tassaneeyakul, W.,
2013. Relationship between the HLA-B 1502 allele and carbamazepine-induced Stevens-
Johnson syndrome and toxic epidermal necrolysis: a systematic review and meta-analysis.
JAMA Dermatol. 149, 1025 1032.
van den Akker-van Marle, M.E., Gurwitz, D., Detmar, S.B., Enzing, C.M., Hopkins, M.M.,
Gutierrez de Mesa, E., et al., 2006. Cost-effectiveness of pharmacogenomics in clinical
practice: a case study of thiopurine methyltransferase genotyping in acute lymphoblastic
leukemia in Europe. Pharmacogenomics 7, 783 792.
CHAPTER 7
Special Requirements
for Economic Evaluation
and Health Technology
Assessment in Genomic Medicine
INTRODUCTION
From the previous chapter, it is obvious that genomic interventions could
enable improved patient stratification and tailor-made treatment interven-
tions. To date, however, most genetic tests focus on a gene-by-gene
approach, rarely detecting multiple genomic variants at high resolution
and accuracy. Technological breakthroughs have led to the development
and early adoption of genomic tests with the potential to meet these crite-
ria, namely whole-exome and whole-genome sequencing. The latter
technologies can simultaneously detect at a genome-wide scale all of an
individual’s genomic variants and, as such, have huge potential in genomic
medicine (Mardis, 2008; Mizzi et al., 2014). Hybrid technologies such as
next-generation sequencing panels sequence multiple genes known to be
associated with a given condition, such as hereditary cancer (Laduca et al.,
2014) or intellectual disability (Flore and Milunsky, 2012). These have the
advantages of improved sequencing quality and depth compared with
whole-exome and genome sequencing while minimizing issues related to
incidental findings (ACMG IF report). As an example for pharmacoge-
nomic testing, PGRNSeq (designed by the Pharmacogenomics Research
Network) is a next-generation sequencing platform that assesses sequence
variation in 84 proposed pharmacogenes (Gordon et al., 2012).
To date, only a small fraction of drugs have pharmacogenomic infor-
mation on their labels and are approved by the US Food and Drug
Administration (http://www.fda.gov) and the European Medicines
Agency (http://www.ema.europa.eu). Also, there is a variable rate of
adoption of genomics in various countries worldwide, and this rate is
ANALYSIS COSTS
The costs of each medical intervention constitute the cornerstone of all
economic evaluations. In contrast to cost data used in classic economic
evaluation, in genomic medicine there is often uncertainty regarding
which costs should be collected, the time that they should be collected,
Special Requirements for Economic Evaluation in Genomic Medicine 117
and how costs vary between different laboratories and health care systems.
There are a variety of factors that lead to this problem. For example, in
the United States, reimbursement for genetic tests has been based on a
“stack” of procedure-based codes (e.g., DNA extraction and purification,
amplification, sequencing, etc.). Different laboratories could use different
procedures to generate results, meaning that the same genetic test can
have markedly different costs and reimbursement due to the specific pro-
cedure that was used. Another issue is that a germline genetic test result
does not change over the course of a person’s lifetime and thus could be
used multiple times for care decisions. An example of this is the pharma-
cogene CYP2D6, which is estimated to be involved in the oxidative
metabolism of 25% of the drugs used in patient care (Samer et al., 2013).
How would one perform a cost-effectiveness analysis for CYP2D6 if the
horizon is the patient’s life span?
In genomic medicine, there are direct and indirect costs that should
be taken into consideration for economically evaluating a genomics-
related intervention (Table 7.1). This implies that health economists
involved in such economic evaluation should ensure that all relevant
costs are included in their analysis. Such costs would include patient
recruitment (Giacomini et al., 2003; Rogowski et al., 2010), sample
Table 7.1 Main costs items involved in economic evaluation in Genomic Medicine
Direct costs
• Patient recruitment, sample collection (blood, saliva, buccal swabs, etc.)
• Nucleic acid isolation
• Genetic testing, including amplification and purification
• Nature of genomic variant tested (germline vs. somatic)
• Data analysis, including data storage and analysis using informatics solutions
and genomic databases
• Frequency of data analysis (on the basis of novel genomics research findings)
• Accreditation, Quality Control of genetic testing platforms and assays
• Genetic counseling and results communication
• Post-testing actions (including treatment options, follow-up testing,
monitoring, etc.)
• Training of personnel, health care professionals
• Infrastructure acquisition and maintenance
Indirect costs
• Patient’s productivity loss
• Family costs (travel, accommodation, productivity loss)
118 Economic Evaluation in Genomic Medicine
collection (blood, saliva, buccal swabs, etc.) and delivery, genomic testing
(Giacomini et al., 2003), genetic data analysis, which includes the appro-
priate genome informatics tools and databases (Feero et al., 2013), report-
ing of genetic test results to the interested parties, namely patients and
their physicians, and genetic counseling (Veenstra et al., 2000; Payne,
2007; Faulkner et al., 2012; Singer and Watkins, 2012). Those costs
should be also accompanied by the costs accrued by the medical interven-
tions dictated by the genetic test results, such as costs of treatment
(from which the costs from the adverse drug reactions and follow-up tests
avoided due to the genetic testing should be deducted). The latter costs
can be significant. For example, a targeted resequencing or a microarray-
based approach, both of which are truly expensive tests on a stand-alone
basis, to identify the genetic basis of a rare mental disorder could be cost-
effective if the costs for follow-up testing can be avoided (Wordsworth
et al., 2007). As noted in Chapter 6, in the case of chronic hepatitis C,
even a small increment of improvement due to a genomic intervention
can be cost-effective if the consequences of treatment failure (end-stage
liver disease hepatocellular carcinoma) are very costly (Bock et al., 2014).
Another important cost item involves drug response and/or disease pro-
gression monitoring. In the case of warfarin treatment in which monitor-
ing, for example, CYP2C9 genotyping, is relatively cheap (Veenstra et al.,
2000), genomic interventions to individualize treatment may not be cost-
effective. At this point, it must be noted that this might not be the case in
developing countries, where genotyping and follow-up treatment costs
may vary (Mitropoulou et al., 2015). As such, genomic tests that are more
likely to be cost-effective are those for conditions for which monitoring is
expensive and cumbersome (Veenstra et al., 2000) or when adverse events
and treatment failures that could be prevented by the use of genomic test-
ing are very expensive (Kauf et al., 2010; Bock et al., 2014). Unfortunately,
there are not yet established guidelines and reimbursement rates for
genomic-based interventions and testing because the genomic technologies
either are too new for reimbursement rates to have been established, such
as whole-genome sequencing, or are integrated as part of a cost item for an
entire treatment modality, in which case costs may vary considerably
depending on the genetic testing laboratory cost policy (Payne, 2009; Van
Rooij et al., 2012; Singer and Watkins, 2012; Deverka et al., 2012;
Malhotra et al., 2012). The latter differences may be the result of the use of
either laboratory-developed assays versus commercially available and
quality-certified genotyping kits (De Leon, 2009; Djalalov et al., 2011) or
Special Requirements for Economic Evaluation in Genomic Medicine 119
the variable prices among different countries (van den Akker-van Marle
et al., 2006). In the latter case, it is noteworthy that genome-guided warfa-
rin treatment may be cost-effective in developing countries (Mitropoulos
et al., 2011) that do not have dedicated anticoagulation monitoring clinics
but not cost-effective in developed countries (Mitropoulou et al., 2015),
which makes it difficult if not impossible to generalize economic evaluation
results between different health care systems.
The nature of the genomic variant being tested is also an important
parameter that should be taken into consideration in economic evaluation
studies pertaining to genomic medicine. Genomic variants may be either
germline, in which case genetic testing should only be performed once
during a patient’s lifetime (De Leon, 2009), or acquired/somatic, which in
some cases requires recurrent genetic analyses, like in the case of chronic
lymphocytic leukemia (Knight et al., 2012). In addition, for new technolo-
gies such as next-generation sequencing, data analysis with its attendant
costs may need to be repeated more than once because genomic data
interpretation is frequently updated with new genomics research results,
allowing for novel genotype phenotype correlations on data re-evaluation
(Gurwitz et al., 2009; Rogowski et al., 2010).
Apart from the direct costs indicated previously, there are also indirect
costs such as the productivity costs (e.g., time lost from work) or the costs
for a patient to seek the proper treatment, which reflects the time that will
be lost from work. To return to the warfarin cost-effectiveness example,
although the cost-effectiveness may be marginal, as noted by Meckley et al.
(2010), from the patient perspective having two to three fewer INR
measurements represents a significant reduction in life disruption that, if
quantified, could impact the cost-effectiveness conclusion. Another
example would be the use of a gene expression panel to stratify risk of
recurrence in endocrine receptor positive, node-negative breast cancer
patients to determine which patients are less likely to benefit from adjunct
chemotherapy based on a low recurrence risk (Vataire et al., 2012; Carlson
and Roth, 2013). These patients could choose to forego chemotherapy
with its attendant morbidity with a modest impact on cancer-related
outcomes, supporting this as a cost-effective (or possibly even cost-saving)
intervention (Rouzier et al., 2013).
Finally, education and training costs are additional cost items in genomic
medicine economic evaluation studies (Reydon et al., 2012; Kampourakis
et al., 2014), although these costs may be difficult to calculate and/or model
accurately because of the complexity of genetic tests performed in a single
120 Economic Evaluation in Genomic Medicine
ANALYSIS METHOD
According to the few available economic evaluation studies in genomic
medicine, it seems that no golden rule is currently available, contrary to
the majority of economic evaluation studies of classical (nongenomic)
interventions that take into consideration the direct impact of a medical
intervention on the health care system and public health (Mette et al.,
2012). The information provided by genomic analysis, particularly whole-
genome sequencing performed very early in life, can have long-term
implications that are not taken into consideration with classical economic
evaluation studies. Consider identifying an α-synuclein gene variant
leading to Parkinson disease in young asymptomatic patients. In this case,
classical studies fail to estimate the long-term cumulative costs and effects
for such a patient (Rogowski et al., 2007, 2009; Grosse et al., 2010).
Also, the timing of an economic evaluation study may be an equally
important parameter because the genetic testing costs are rapidly decreasing,
whereas their specificity and accuracy are steadily increasing (Conti et al.,
2010; Goddard et al., 2012; Lin et al., 2012). This fact may lead to patient
subgroup stratification with direct impact on individualized treatments, or
the incorporation in the clinical practice of microarray-based genetic screen-
ing tests (e.g., AmpliChip [Roche Diagnostics], DMET1 [Affymetrix],
etc.) that were previously used for research purposes only.
Cost-effectiveness of a specific genomic technology and/or a genome-
based intervention can only be assessed in the context of a specific clinical
application involving a certain patient subpopulation. As such, when
performing economic evaluation for such a technology or intervention, it
is of utmost importance to carefully identify and select the comparators.
For example, one should assess both genetic and nongenetic testing in
addition to genetic in conjunction with nongenetic testing (Sanderson
et al., 2005; Hall et al., 2012); in the latter case, combining a cheap
conventional screening approach with an expensive genetic test may
prove to be cost-effective, like in the case of combining immunohis-
tochemistry with DNA sequencing, respectively, for the identification
of patients with Lynch syndrome among newly diagnosed patients
with colorectal cancer (Mvundura et al., 2010). In the case of warfarin,
Special Requirements for Economic Evaluation in Genomic Medicine 121
OUTCOME OF ANALYSIS
There are several methodological issues in economic evaluation studies in
genomic medicine that arise when measuring the outcomes of a genomic
intervention, such as the type of outcome measure to be used in the anal-
ysis, personal utility, and the importance of individual outcomes. The last
of these is particularly relevant to genomic medicine because, although
the probability of an individual outcome is considered in economic analy-
ses, the predictive nature of genomic information provides more detailed
information specific to individual patients. This has led some to conclude
that new types of studies are needed to truly capture the impact of
genomic medicine (Hu, 2012; Garraway, 2013). If true, then this will
inevitably impact economic analyses as well.
In economic evaluation studies for genomic medicine interventions,
preference-based outcome measures (e.g., the EuroQol five-dimensional
[EQ-5D] questionnaire) should be used because they can be more easily
and uniformly compared among different population subgroups by col-
lecting data across a broad range of health-related quality-of-life domains.
122 Economic Evaluation in Genomic Medicine
However, QALYs measured using these measures do not reflect all possi-
ble health states applicable for genomic interventions (e.g., an asymptom-
atic patient with a predisposing genomic variant).
Genomic information, especially in the case of incidental findings, may
offer significant benefits and, at the same time, harms for a person. Some
genomic information lacks clinical utility (that is, there are no medical
actions that can be taken that can prevent or treat the condition identified
by the genomic test). This type of information can affect a person’s well-
being and decision-making (Foster et al., 2009; Roth et al., 2011; Garau
et al., 2013), a concept referred to as personal utility. There are factors that
have a positive impact on personal utility, such as prognostic or diagnostic
information that explains a certain genotype phenotype correlation, ends
a diagnostic odyssey, or rationalizes a therapeutic intervention. Also, geno-
mic information can be reassuring and can relieve anxiety (Asch et al.,
1996; Caughey, 2005; Grosse et al., 2010), allowing patients and their rela-
tives to better plan their lives and make lifestyle decisions, such as in the
case of APOE E4 allele testing as a predisposing factor in Alzheimer’s dis-
ease (Grosse et al., 2009, 2010; Payne et al., 2012). At the same time, there
are factors that have a negative impact on personal utility. Anxiety may be
increased if a genetic test result indicates that a patient is a nonresponder
or is at increased risk for development of adverse reactions to a certain
medical treatment, when no treatment alternatives exist (Conti et al.,
2010; Khoury et al., 2011), or when mutations of unknown significance
or other incidental findings are false-positives results (Jarrett and Mugford,
2006; Foster et al., 2009; Payne et al., 2012; Patrinos et al., 2013). In other
words, if a patient is genomically diagnosed with a predisposition for
development of Alzheimer’s disease prior to the development of symp-
toms, this does not provide them with any clinical utility as traditionally
defined because there are no effective treatments to prevent development
of the disease. However, it considerably impacts their personal utility,
sometimes in a positive way through adoption of a healthier lifestyle or
purchase of long-term care insurance, but it can also lead to anxiety,
despair, and pursuit of ineffective therapies that incur cost but provide no
benefit (Roberts et al., 2011). Such result may also worsen family dynam-
ics, lead to fear of discrimination and stigmatization (Grosse et al., 2008),
and impact reproductive decisions (Jarrett and Mugford, 2006; Foster
et al., 2009; Rogowski et al., 2010). False-negative genetic test results can
also lead to false reassurance and encourage unhealthy behaviors, such as
smoking and drinking (Grosse et al., 2008).
Special Requirements for Economic Evaluation in Genomic Medicine 123
ANALYSIS EFFECTIVENESS
Economic evaluations also incorporate some measure of the effectiveness
of the intervention being evaluated, such as the unpredictable behavior of
patients and their physician. The complexity of genomic effectiveness
data makes the task of measuring the effectiveness of the analysis a
challenging one.
It is well-understood that the cost-effectiveness of a certain genomic
intervention is dependent on how patients and their physicians use these
interventions and how they respond to the genomic test results (e.g.,
whether patients change their lifestyle or alter their treatment modalities).
As such, information regarding behavioral responses should be included
into economic calculations to evaluate genomic technology and a geno-
mic medicine intervention (Asch et al., 1996; Caughey, 2005; Rogowski
et al., 2007; 2010; Conti et al., 2010). However, very little information
regarding how to model behavioral attitudes is available (Conti et al.,
2010). One cannot be sure whether all patients will want to have their
genome, in part or as a whole, tested, whether they will they adjust their
behavior to comply with the advice of a genetic counselor (Rogowski,
2009; Deverka et al., 2012), or whether physicians will take information
from genomic tests into account when making clinical decisions (Conti
et al., 2010). In some case, physicians confess that their genetics literacy is
very limited to make informed decisions based on genetic test results
(Mai et al., 2011, 2014). Some investigators such as those referenced in
the Oncotype DX and Alzheimer’s disease examples are using study
designs that capture patient and physician choices in response to genomic
information, thus generating data that can be used in subsequent eco-
nomic analyses. These analyses will have much more relevance to real-
world decision-making compared with studies that rely on untested
assumptions about patient and physician behaviors.
124 Economic Evaluation in Genomic Medicine
CONCLUSIONS
From the previous paragraphs, it is obvious that economic evaluation in
genomic medicine and health technology assessment has significant differ-
ences when compared with classical health economics and poses a num-
ber of challenges for health economists. To date, there are some solutions
to these challenges that have already been proposed and could be incor-
porated in future economic evaluations in genomic medicine. The goal
will be to introduce complex genomic effectiveness data into economic
analyses as soon as next-generation sequencing is incorporated into clini-
cal practice to facilitate informed decisions about implementation,
resource allocation, and investment.
Also, it is of paramount importance to incorporate personal utility of
genomic tests into the economic evaluation studies. In the United States
and in other countries where the genetic tests are reimbursed, a flat cost-
based scheme is used, leaving very little flexibility to consider personal
utility of a genetic test. However, this may be more relevant for regula-
tory bodies rather than health care providers, whose aim is to improve
public health and, as such, personal utility may not be so relevant (Grosse
et al., 2008). Early efforts to value personal utility should be increased to
develop effective solutions to this challenge.
Special Requirements for Economic Evaluation in Genomic Medicine 125
REFERENCES
Alberts, S.R., Yu, T., Behrens, R.J., et al., 2013. Real-world comparative eco-
nomics of a 12-gene assay for prognosis in stage II colon cancer. J. Clin. Oncol.
31 (4), 391.
Asch, D.A., Hershey, J.C., Pauly, M.V., Patton, J.P., Jedrziewski, M.K., Mennuti, M.T.,
1996. Genetic screening for reproductive planning: methodological and conceptual
issues in policy analysis. Am. J. Public Health 86 (5), 684 690.
Bock, J.A., Fairley, K.J., Smith, R.E., Maeng, D.D., Pitcavage, J.M., Inverso, N.A., et al.,
2014. Cost-effectiveness of IL28 genotype-guided protease inhibitor triple therapy ver-
sus standard of care treatment in patients with hepatitis C genotypes 2 or 3 infection.
Public Health Genomics 17, 306 319.
126 Economic Evaluation in Genomic Medicine
Buchanan, J., Wordsworth, S., Schuh, A., 2013. Issues surrounding the health economic
evaluation of genomic technologies. Pharmacogenomics 14 (15), 1833 1847.
Carlson, J.J., Roth, J.A., 2013. The impact of the Oncotype Dx breast cancer assay in
clinical practice: a systematic review and meta-analysis. Breast Cancer Res. Treat. 141
(1), 13 22.
Caughey, A.B., 2005. Cost-effectiveness analysis of prenatal diagnosis: methodological
issues and concerns. Gynecol. Obstet. Invest. 60 (1), 11 18.
Conti, R., Veenstra, D.L., Armstrong, K., Lesko, L.J., Grosse, S.D., 2010. Personalized
medicine and genomics: challenges and opportunities in assessing effectiveness, cost-
effectiveness, and future research priorities. Med. Decis. Making 30 (3), 328 340.
De Leon, J., 2009. Pharmacogenomics: the promise of personalized medicine for CNS
disorders. Neuropsychopharmacology 34 (1), 159 172.
Deverka, P.A., Schully, S.D., Ishibe, N., Carlson, J.J., Freedman, A., Goddard, K.A., et al.,
2012. Stakeholder assessment of the evidence for cancer genomic tests: insights from
three case studies. Genet. Med. 14 (7), 656 662.
Djalalov, S., Musa, Z., Mendelson, M., Siminovitch, K., Hoch, J., 2011. A review of eco-
nomic evaluations of genetic testing services and interventions (2004 2009). Genet.
Med. 13 (2), 89 94.
Fang, H., Liu, X., Ramı́rez, J., Choudhury, N., Kubo, M., Im, H.K., et al., 2014.
Establishment of CYP2D6 reference samples by multiple validated genotyping plat-
forms. Pharmacogenomics J. 14, 564 572.
Faulkner, E., Annemans, L., Garrison, L., Helfand, M., Holtorf, A.P., Hornberger, J.,
et al., 2012. Challenges in the development and reimbursement of personalized
medicine—payer and manufacturer perspectives and implications for health economics
and outcomes research: a report of the ISPOR personalized medicine special interest
group. Value Health 15 (8), 1162 1171.
Feero, W., Wicklund, C., Veenstra, D.L., 2013. The economics of genomic medicine:
insights from the iom roundtable on translating genomic-based research for health.
JAMA 309 (12), 1235 1236.
Flore, L.A., Milunsky, J.M., 2012. Updates in the genetic evaluation of the child with global
developmental delay or intellectual disability. Semin. Pediatr. Neurol. 19 (4), 173 180.
Foster, M.W., Mulvihill, J.J., Sharp, R.R., 2009. Evaluating the utility of personal geno-
mic information. Genet. Med. 11 (8), 570 574.
Garau, M., Towse, A., Garrison, L., Housman, L., Ossa, D., 2013. Can and should value
based pricing be applied to molecular diagnostics? Pers. Med. 10 (1), 61 72.
Garraway, L.A., 2013. Genomics-driven oncology: framework for an emerging paradigm.
J. Clin. Oncol. 31 (15), 1806 1814.
Giacomini, M., Miller, F., O’Brien, B.J., 2003. Economic considerations for health insur-
ance coverage of emerging genetic tests. Community Genet. 6 (2), 61 73.
Goddard, K.A., Knaus, W.A., Whitlock, E., Lyman, G.H., Feigelson, H.S., Schully, S.D.,
et al., 2012. Building the evidence base for decision making in cancer genomic medi-
cine using comparative effectiveness research. Genet. Med. 14 (7), 633 642.
Gordon, A.S., Smith, J.D., Xiang, Q., Metzker, ML., Gibbs, RA., Mardis, ER., et al.,
PGRNseq: a new sequencing-based platform for high-throughput pharmacoge-
nomic implementation and discovery. Abstract presented at the 62nd Annual
Meeting of the American Society of Human Genetics: November 6 8, 2012;
San Francisco, CA. Program #244. ,http://www.ashg.org/2012meeting/abstracts/
fulltext/f120122669.htm..
Grosse, S.D., Kalman, L., Khoury, M.J., 2010. Evaluation of the validity and utility of
genetic testing for rare diseases. Adv. Exp. Med. Biol. 686, 115 131.
Grosse, S.D., McBride, C.M., Evans, J.P., Khoury, M.J., 2009. Personal utility and geno-
mic information: look before you leap. Genet. Med. 11 (8), 575 576.
Special Requirements for Economic Evaluation in Genomic Medicine 127
Grosse, S.D., Rogowski, W.H., Ross, L.F., Cornel, M.C., Dondorp, W.J., Khoury, M.J.,
2010. Population screening for genetic disorders in the 21st century: evidence, eco-
nomics, and ethics. Public Health Genomics 13 (2), 106 115.
Grosse, S.D., Wordsworth, S., Payne, K., 2008. Economic methods for valuing the outcomes
of genetic testing: beyond cost-effectiveness analysis. Genet. Med. 10 (9), 648 654.
Gurwitz, D., Zika, E., Hopkins, M.M., Gaisser, S., Ibarreta, D., 2009. Pharmacogenetics
in Europe: barriers and opportunities. Public Health Genomics 12 (3), 134 141.
Guzauskas, G.F., Garrison, L.P., Stock, J., Au, S., Doyle, D.L., Veenstra, D.L., 2013.
Stakeholder perspectives on decision-analytic modeling frameworks to assess genetic
services policy. Genet. Med. 15 (1), 84 87.
Hall, P.S., McCabe, C., Stein, R.C., Cameron, D., 2012. Economic evaluation of geno-
mic test-directed chemotherapy for early-stage lymph node-positive breast cancer.
J. Natl. Cancer Inst. 104 (1), 56 66.
Holt, S., Bertelli, G., Humphreys, I., Valentine, W., Durrani, S., Pudney, D., et al., 2013.
A decision impact, decision conflict and economic assessment of routine Oncotype
DX testing of 146 women with node-negative or pNImi, ER-positive breast cancer
in the UK. Br. J. Cancer 108 (11), 2250 2258.
Hu, V.W., 2012. Subphenotype-dependent disease markers for diagnosis and personalized
treatment of autism spectrum disorders. Dis. Markers. 33 (5), 277 288.
Jarrett, J., Mugford, M., 2006. Genetic health technology and economic evaluation: a
critical review. Appl. Health Econ. Health Policy. 5 (1), 27 35.
Kampourakis, K., Vayena, E., Mitropoulou, C., Borg, J., van Schaik, R.H., Cooper, D.N.,
et al., 2014. Key challenges for next generation pharmacogenomics. EMBO Rep. 15
(5), 472 476.
Kauf, T.L., Farkouh, R.A., Earnshaw, S.R., Watson, M.E., Maroudas, P., Chambers, M.G.,
2010. Economic efficiency of genetic screening to inform the use of abacavir sulfate in
the treatment of HIV. Pharmacoeconomics 28 (11), 1025 1039.
Khoury, M.J., Gwinn, M., Dotson, W.D., Bowen, M.S., 2011. Is there a need for
PGxceptionalism? Genet. Med. 13 (10), 866 867.
Knight, S.J.L., Yau, C., Clifford, R., Timbs, A.T., Sadighi Akha, E., Dréau, H.M., et al.,
2012. Quantification of subclonal distributions of recurrent genomic aberrations in
paired pre-treatment and relapse samples from patients with B-cell chronic lympho-
cytic leukemia. Leukemia 26 (7), 1564 1575.
Kopits, I.M., Chen, C., Roberts, J.S., Uhlmann, W., Green, R.C., 2011. Willingness to
pay for genetic testing for Alzheimer’s disease: a measure of personal utility. Genet.
Test. Mol. Biomarkers 15 (12), 871 875.
Laduca, H., Stuenkel, A.J., Dolinsky, J.S., Keiles, S., Tandy, S., Pesaran, T., et al., 2014.
Utilization of multigene panels in hereditary cancer predisposition testing: analysis of
more than 2,000 patients. Genet Med. 16, 830 837.
Lin, X., Tang, W., Ahmad, S., et al., 2012. Applications of targeted gene capture and
next-generation sequencing technologies in studies of human deafness and other
genetic disabilities. Hear. Res. 288 (1 2), 67 76.
Mai, Y., Koromila, T., Sagia, A., Cooper, D.N., Vlachopoulos, G., Lagoumintzis, G., et al.,
2011. A critical view of the general public’s awareness and physicians’ opinion of the
trends and potential pitfalls of genetic testing in Greece. Pers. Med. 8 (5), 551 561.
Mai, Y., Mitropoulou, C., Papadopoulou, X.E., Vozikis, A., Cooper, D.N., van Schaik, R.H.,
et al., 2014. Critical appraisal of the views of healthcare professionals with respect to phar-
macogenomics and personalized medicine in Greece. Pers. Med. 11 (1), 15 26.
Malhotra, A.K., Zhang, J.P., Lencz, T., 2012. Pharmacogenetics in psychiatry: translating
research into clinical practice. Mol. Psychiatr 17 (8), 760 769.
Mardis, E.R., 2008. The impact of next-generation sequencing technology on genetics.
Trends Genet. 24 (3), 133 141.
128 Economic Evaluation in Genomic Medicine
Meckley, L.M., Gudgeon, J.M., Anderson, J.L., Williams, M.S., Veenstra, D.L.A., 2010.
Policy model to evaluate the benefits, risks and costs of warfarin pharmacogenomic
testing. Pharmacoeconomics 28 (1), 61 74.
Mette, L., Mitropoulos, K., Vozikis, A., Patrinos, G.P., 2012. Pharmacogenomics and
public health: implementing populationalized medicine. Pharmacogenomics 13 (7),
803 813.
Mitropoulos, K., Johnson, L., Vozikis, A., Patrinos, G.P., 2011. Relevance of pharmaco-
genomics for developing countries in Europe. Drug Metabol. Drug Interact. 26 (4),
143 146.
Mitropoulou, C., Fragoulakis, V., Bozina, N., Vozikis, A., Supe, S., Bozina, T., et al.,
2015. Economic evaluation for pharmacogenomic-guided warfarin treatment for
elderly croatian patients with atrial fibrillation. Pharmacogenomics. (in press).
Mizzi, C., Mitropoulou, C., Mitropoulos, K., Peters, B., Agarwal, M.R., van Schaik, R.H.,
et al., 2014. Personalized pharmacogenomics profiling using whole genome sequencing.
Pharmacogenomics 15, 1223 1234.
Mvundura, M., Grosse, S.D., Hampel, H., Palomaki, G.E., 2010. The cost-effectiveness
of genetic testing strategies for lynch syndrome among newly diagnosed patients with
colorectal cancer. Genet. Med. 12 (2), 93 104.
Patrinos, G.P., Baker, D.J., Al-Mulla, F., Vasiliou, V., Cooper, D.N., 2013. Genetic tests
obtainable through pharmacies: the good, the bad and the ugly. Hum. Genomics 7
(1), 17.
Payne, K., 2007. Towards an economic evidence base for pharmacogenetics: consideration
of outcomes is key. Pharmacogenomics 9 (1), 1 4.
Payne, K., 2009. Fish and chips all round? Regulation of DNA-based genetic diagnostics.
Health Econ. 18 (11), 1233 1236.
Payne, K., McAllister, M., Davies, L.M., 2012. Valuing the economic benefits of complex
interventions: when maximising health is not sufficient. Health Econ. 22 (3),
258 271.
Recommendations from the EGAPP Working Group: testing for cytochrome P450,
2007. Polymorphisms in adults with nonpsychotic depression treated with selective
serotonin reuptake inhibitors. Evaluation of genomic applications in practice and
prevention (EGAPP) working group. Genet Med. 9 (12), 819 825.
Reydon, T.A., Kampourakis, K., Patrinos, G.P., 2012. Genetics, genomics and society:
the responsibilities of scientists for science communication and education. Pers. Med.
9 (6), 633 643.
Roberts, J.S., Christensen, K.D., Green, R.C., 2011. Using Alzheimer’s disease as a model
for genetic risk disclosure: implications for personal genomics. Clin. Genet. 80 (5),
407 414.
Rogowski, W.H., 2007. Current impact of gene technology on healthcare. A map of
economic assessments. Health Policy 80 (2), 340 357.
Rogowski, W.H., 2009. The cost-effectiveness of screening for hereditary hemochroma-
tosis in Germany: a remodeling study. Med. Decis. Making 29 (2), 224 238.
Rogowski, W.H., Grosse, S.D., John, J., Kääriäinen, H., Kent, A., Kristofferson, U., et al.,
2010. Points to consider in assessing and appraising predictive genetic tests. J.
Community Genet. 1 (4), 185 194.
Rogowski, W.H., Grosse, S.D., Khoury, M.J., 2009. Challenges of translating genetic tests
into clinical and public health practice. Nat. Rev. Genet. 10 (7), 489 495.
Roth, J.A., Garrison Jr., L.P., Burke, W., Ramsey, S.D., Carlson, R., Veenstra, D.L., 2011.
Stakeholder perspectives on a risk-benefit framework for genetic testing. Public
Health Genomics 14 (2), 59 67.
Special Requirements for Economic Evaluation in Genomic Medicine 129
Rouzier, R., Pronzato, P., Chéreau, E., Carlson, J., Hunt, B., Valentine, W.J., 2013.
Multigene assays and molecular markers in breast cancer: systematic review of health
economic analyses. Breast Cancer Res. Treat. 139 (3), 621 637.
Samer, C.F., Lorenzini, K.I., Rollason, V., Daali, Y., Desmeules, J.A., 2013. Applications
of CYP450 testing in the clinical setting. Mol. Diagn. Ther. 17 (3), 165 184.
Sanderson, S., Zimmern, R., Kroese, M., Higgins, J., Patch, C., Emery, J., 2005. How
can the evaluation of genetic tests be enhanced? Lessons learned from the ACCE
framework and evaluating genetic tests in the United Kingdom. Genet. Med. 7 (7),
495 500.
Singer, D.R.J., Watkins, J., 2012. Using companion and coupled diagnostics within strat-
egy to personalize targeted medicines. Pers. Med. 9 (7), 751 761.
Snyder, S., Mitropoulou, C., Patrinos, G.P., Williams, M.S., 2014. Economic evaluation
of pharmacogenomics: a value-based approach to pragmatic decision-making in the
face of complexity. Public Health Genomics 17, 256 264.
van den Akker-van Marle, M.E., Gurwitz, D., Detmar, S.B., Enzing, C.M., Hopkins, M.M.,
Gutierrez de Mesa, E., et al., 2006. Cost-effectiveness of pharmacogenomics in clinical
practice: a case study of thiopurine methyltransferase genotyping in acute lymphoblastic
leukemia in Europe. Pharmacogenomics 7 (5), 783 792.
Van Rooij, T., Wilson, D.M., Marsh, S., 2012. Personalized medicine policy challenges:
measuring clinical utility at point of care. Expert Rev. Pharmacoecon. Outcomes
Res. 12 (3), 289 295.
Vataire, A.L., Laas, E., Aballea, S., Gligorov, J., Rouzier, R., Chereau, E., 2012. Cost-
effectiveness of a chemotherapy predictive test. Bull. Cancer 99 (10), 907 914.
Veenstra, D.L., Higashi, M.K., Phillips, K.A., 2000. Assessing the cost-effectiveness of
pharmacogenomics. AAPS PharmSci. 2 (3), E29.
Wordsworth, S., Buchanan, J., Regan, R., Davison, V., Smith, K., Dyer, S., et al., 2007.
Diagnosing idiopathic learning disability: a cost-effectiveness analysis of microarray
technology in the national health service of the United Kingdom. Genomic Med. 1
(1 2), 35 45.
CHAPTER 8
INTRODUCTION
As mentioned in Chapters 4 and 5, economic evaluation compares the
costs and health effects of an intervention aiming to maximize health
benefits from available resources (Cartwright, 1999). So far, for innovative
interventions, the most commonly used formal investigation is implemen-
ted via the determination of incremental cost-effectiveness ratio (ICER;
O’Brien and Briggs, 2002; see Chapter 4). ICER is defined as the ratio
of incremental cost (ΔC ) of an intervention divided by the incremental
effectiveness (ΔE ), measured either as additional life-years (LY) or addi-
tional quality-adjusted life-years (QALYs). Based on the classical decision-
making approach of the cost-effectiveness analysis, ICER is compared
with a fixed amount that a policymaker is willing to pay (λ) for an addi-
tional LY or QALY. In this methodology context, if ICER is below λ,
the new intervention meets the basic criterion for reimbursement by the
payers (McCabe et al., 2008). In practice, this methodology assumes that
the policymakers are willing to pay this amount, regardless of the amount
of QALYs “purchased,” and in that way are ignoring the budget con-
straint. However, certain limitations must be considered in this approach
because: (i) there is no clear linkage between λ and the budget affordabil-
ity of health services (Towse, 2009) and (ii) it assumes, silently, an instant
adjustment of budget to absorb the cost of innovation (Gafni and Birch,
2006). Thus, the adoption of the existing process has led to the adoption
of costly decisions (Sendi et al., 2002; Gyrd-Hansen, 2005; Birch and
Gafni, 2006) and consequently increased expenditures for health care
systems (Gafni and Birch, 2006). Even if the budget does not face hard
constraints, it is reasonable to assume that there is an upper budget bound,
especially today in the presence of a global economic crisis. In addition,
prerequisites were fully satisfied would the true value of λ or the desired
mix of budget across several disease-specific areas have been estimated.
Mathematical programming can incorporate some restrictions referred to
as ethical or technical issues (minimum budget spent on a specific disease,
divisibility, etc.), but an amount of exogenous information for the pattern
of budget allocation must also be provided as an input to solve the maximi-
zation problem. These are some crucial problems that face practical
research today. In this chapter, we propose a new simple way of thinking
and an alternative methodological approach for decision-making that takes
into account the budget constraint and relaxes the constant return on scale
assumption of λ.
Bt 5 CðSÞpy Es ð8:1Þ
For instance, if the state is willing to pay h10,000 for a patient during
the remaining lifetime (which is expected to be 2 years), the mean cost
per year for this patient that could be covered is limited to h5,000 per
year. Figure 8.1 depicts the relation between the average cost per patient
per year and the effectiveness of the standard intervention via the rational
function Eq. (8.1).
An increased effectiveness in a given budget Bt from the substitution
of standard intervention with a new intervention with equal total cost
must be followed by a decrease in the average cost per patient per year
and vice versa. The area under the curve is equal for each point across
the curve and expresses the total cost per patient. When a new and more
Cpy
C(T)py
Es Et E
Figure 8.1 Effectiveness and average cost per year for a given budget constraint.
Cpy: average cost per patient per year; C(S)py: average cost per patient per year for the
standard intervention; C(T)py: average cost per patient per year for the new intervention;
budget constraint refers to the available budget per patient for his/her life span; Es: effec-
tiveness of the standard intervention; Et: effectiveness of the new intervention.
A New Methodological Approach for Cost-Effectiveness Analysis in Genomic Medicine 135
The reader must keep in mind that Eq. (8.2) is satisfied even if C(T)py
is equal, lower, or higher than C(S)py.
For instance, let us assume that the total cost of a new intervention has
been determined at 20,000 against 10,000 for the standard one. So, we have:
C(T)pyEt 5 20,000 and C(S)pyEs 5 10,000. If Et 5 2 LYs and Es 5 1 LY,
then C(S)py 5 C(T)py; if Et 5 3 and Es 5 1 LY, then C(S)py . C(T)py,
and so on.
This is depicted in Figures 8.2 and 8.3. Figure 8.2 depicts the case
where the most expensive and cost-infeasible interventions due to a specific
budget have equal, lower, or higher average costs per patient per year.
Figure 8.3 depicts the budget constraint expansion required for affording
new—more costly—interventions. Every single curve that is more distant
from the axes reveals an increased expenditure or a higher budget.
Cpy
ΔCpy>0
ΔCpy=0
C(S)py Budget constraint
ΔCpy<0
Es E
Figure 8.2 Cost-infeasible interventions with lower, equal, and higher average cost
per year. Cpy: average cost per patient per year; C(S)py: average cost per patient per
year for the standard intervention; ΔCpy: the difference in the average cost per year
per patient between the standard and new intervention; budget constraint refers to
the available budget per patient for his/her life span; Es: effectiveness of the standard
intervention.
136 Economic Evaluation in Genomic Medicine
In a general form, the mean cost per patient per year for the new
intervention could be expressed as:
Ct
CðT Þpy 5 or
Et
Ct
CðT Þpy 5 or
Es 1 ΔE
ð8:3Þ
Cs 1 ΔC
CðT Þpy 5 or
Es 1 ΔE
CðSÞpy Es 1 ΔC
CðT Þpy 5
Es 1 ΔE
Cpy
Budget constraint
map
C(T2)py
C(S)py
C(T1)py
Es Et1 Et2
Figure 8.3 New interventions and budget constraint expansion path. C(S)py: average
cost per patient per year for the standard intervention; C(T1)py: average cost per
patient per year for the new T1 intervention; C(T2)py: average cost per patient per
year for the new T2 intervention; budget constraint refers to the available budget
per patient for his/her life span; Es: effectiveness of the standard intervention;
Et1: effectiveness of the new T1 intervention; Et2: effectiveness of the new
T2 intervention.
A New Methodological Approach for Cost-Effectiveness Analysis in Genomic Medicine 137
Let us assume that the threshold for a QALY is unknown but equal to
λ. In addition to that, we know that ΔC 5 λ ΔE. Thus, we have:
CðSÞpy Es 1 λΔE
CðTÞpy 5 ð8:4Þ
Es 1 ΔE
From Eq. (8.4) it must be obvious that we have connected an incre-
mental quantity (λ) with an average one (C(T )py).
Thus, we can determine the expansion path of the average cost per
patient per year when a new, more costly intervention is introduced as a
function of ΔE, taking into account the (fixed) incremental threshold of λ.
If C(S)py 5 λ, then the graphical representation of C(T )py and Et is a
straight line. If C(S)py . λ (C(S)py , λ), then the graphical representation
is a downward (upward) curve with an asymptotic plateau of λ. Figure 8.4
depicts, for illustrative purposes, the case where C(S)py , λ. Figure 8.4
depicts that the threshold approach leads to decisions that result in
increased expenditures and thus raises concerns about the funding sustain-
ability. Figure 8.5 shows the opposite case. Given a fixed budget constraint
that has been exhausted, λ has a decreasing trend, is not constant across
Cpy
λ
C(T)py
Equilibrium given the
budget constraint
Budget constraint
map
E
Figure 8.4 Average cost and expanded budget constraint given a fixed λ. Cpy: aver-
age cost per patient per year; λ: willingness to pay; C(T)py: average cost per patient
per year for the new intervention; budget constraint refers to the available budget
per patient for his/her life span; E: effectiveness.
138 Economic Evaluation in Genomic Medicine
Decreasing λ in a
given budget constraint
Es Et E
Figure 8.5 Willingness to pay as a function of a fixed budget constraint and effec-
tiveness. λ: willingness to pay; E: effectiveness; Es: effectiveness of the standard/con-
ventional intervention; Et: effectiveness of the new intervention.
ΔE, and becomes asymptotically zero. Thus, λ tends to be zero in the case
of—potential—new interventions with high ΔE, because the society is
unable to afford the incremental cost of innovation. If the ΔE tends to be
small, then λ could be high; nonetheless, society probably is unwilling to
pay a high incremental cost for a small ΔE.
RELAXATION OF FIXED λ
In this paragraph, we go a step further concerning λ. The presented
approach is sensitive to the actual incremental effectiveness of a new
intervention. Let us assume that the budget B2t is fixed. The model
assumes there is a maximum expected incremental effectiveness (ΔEmax)
that could be additionally reimbursed. Hence, we could introduce a non-
linear S-shape function that determines λ as a function of actual ΔE,
where ΔE $ 0. As shown in Figure 8.6, the willingness to pay threshold
belongs to the S-shape family, ensuring that λ is different across ΔE in
every single point of the cost-effectiveness plane. For a very small ΔE
(i.e., a “me too” drug), and without any substantial differentiation point
from existing interventions, λ is expected to be low. If ΔE is fair or small,
then a higher λ with an increased marginal utility for the additional
QALYs can be expected. This is in accordance with economic theory
A New Methodological Approach for Cost-Effectiveness Analysis in Genomic Medicine 139
ΔC
Very innovative drugs
almost zero marginal utility
of QALYs, high λ
Innovative drugs
decreased marginal utility
=4
5, b
of QALYs, high λ h
ac
a= pro
ap
ne ant λ
4
=
l i
b
t t
igh cons
2,
tra
a=
S
=4
Fair drugs
,b
1.3
increased marginal utility
of QALYs, small λ
a=
“Me too” drugs
small λ
ΔEmax ΔE
Figure 8.6 A nonlinear S-shape function that determines the willingness to pay as a
function of actual ΔE. ΔE: difference in the effectiveness between the standard/con-
ventional and the new intervention; ΔC: difference in the total cost between the
standard/conventional and new intervention per patient; ΔCmax: available budget
(per patient) is willing to afford the budget holder to capture the maximum
expected effectiveness (ΔEmax); a and b are parameters of the equation:
b
1 2 eð2αΔE Þ
λ5 ΔCmax
ΔE
CðSÞpy Es 1 λðΔEÞ
CðT Þpy 5 ð8:5Þ
Es 1 ΔE
Thus, the path of average cost, the budget constraint map, and the
path of λ changes depend on the form of function of the willingness to
140 Economic Evaluation in Genomic Medicine
pay threshold. Let us assume that λ has a flexible form derived from the
Weibull family (Yin et al., 2003):
1 2 eð2αΔE Þ
b
λ5 ΔCmax ð8:6Þ
ΔE
where:
ΔCmax 5 The maximum available budget (per patient) the budget
holder is willing to afford to capture the maximum expected differ-
ence in effectiveness (ΔEmax).
ΔEmax has been assumed and a and b are constants. In such a case, the
straight line converts to a line segment (bounded by 0 to the left and
ΔEmax to the right). The new model partly incorporates the classical
model because we could find appropriate values for the parameters a and
b, which produce an approximately straight line segment or, to postulate
it, a line in the cost-effectiveness plane. It is paramount to consider that
in this model the budget upper limit is predetermined, and Figure 8.6
reveals the path of willingness to pay as we get closer to the budget
constraint.
λ5 50;000
ΔE
assuming that a and b are parameters that have been estimated from real
data. If a new intervention has a ΔE that is higher than expected (i.e.,
ΔE 5 1.5), then λ is estimated as λ 5 ΔCmax/ΔE or λ 5 50,000/1.5 5
h33,000 per LY. In that case, the manufacturer must accept this λ due to
budget constraints of the health care system and the inability of budget
holders to absorb the cost of such an innovative (in term of ΔE) inter-
vention. In the extreme scenario where the actual ΔE was even higher, λ
tends to be zero, as depicted in Figure 8.5, but the benefit of new
A New Methodological Approach for Cost-Effectiveness Analysis in Genomic Medicine 141
λ5 310
ΔE
where ΔCmax 5 h310.0, different acceptability curves are determined.
The results of the probabilistic analysis are shown in Figure 8.7B. It must
be noted that the scenarios with ΔEmax 5 0.027 are too “ambitious” and
“demanding” because they require that PGx must have a difference of
0.027 QALYs against N-PGx to invest all the additional h310, and thus
A New Methodological Approach for Cost-Effectiveness Analysis in Genomic Medicine 143
(A)
100.0%
90.0%
80.0%
70.0%
60.0%
50.0%
40.0%
30.0%
20.0%
10.0%
0.0%
5. 0
9. 0
13 0
17 0
21 00
25 00
29 00
33 00
37 00
41 0
45 00
49 00
53 00
57 00
61 00
65 00
69 0
73 00
77 00
81 00
85 00
89 00
93 00
97 0
00
00
00
00
0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
1.
(B)
100%
90%
80%
70%
60%
50%
40%
30%
20%
10%
0%
5. 0
9. 0
13 0
17 00
21 00
25 00
29 00
33 00
37 00
41 00
45 00
49 00
53 00
57 00
61 00
65 00
69 00
73 00
77 00
81 00
85 00
89 00
93 00
97 00
00
00
00
00
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
.0
1.
Figure 8.7 Cost-effectiveness acceptability curve for PGx group versus the N-PGx
control group using the classical model (A) and the new model (B). ΔEmax: maximum
expected difference in effectiveness between the N-PGx and PGx groups, which
could be additionally reimbursed given that the budget is 0.01 and 0.027 QALYs in
the two scenarios, respectively. λ: ðð1 2 eð25ΔE Þ Þ=ΔEÞ310 for the new models.
3
IMPLICATIONS
Today, the cost of innovation in health care is expected to be covered by
the third-party payers and usually leads to uncontrolled growth in health
care expenditure. In view of the scarcity of resources, economic evalua-
tion provides a criterion for the final decision concerning the adoption of
certain new interventions but has certain inconsistencies and drawbacks.
In this analysis, a new methodological approach was proposed taking into
account the budget constraint, the effectiveness of a new intervention,
and the willingness to pay in a flexible way.
In this new cost-effectiveness analysis model, it was assumed that the
budget is exogenous and has been set by the budget holders. It must be
noted that the health care budgets frequently depend on historical, politi-
cal, and social criteria without taking into account the economic models
and their assumptions (Schwappach, 2002). Furthermore, uncertainty in
the determination of costs and outcomes (Ramsey et al., 2005), lack of
data or knowledge for preferences (Weyler and Gandjour, 2011), lack of
training of policymakers (Veney et al., 1997), established status quo, and
other policy relevant issues set restrictions in a rational decision-making
process for the health maximization problem. Thus, the quantitative
determination of constraints and the application of prominent instruments
such as mathematical programming (Flessa, 2000) or other methodologi-
cal approaches (Sendi and Briggs, 2001) seem difficult to be implemented
practically for the optimum allocation of the health care budgets.
In this model, a far less ambiguous approach was adopted to estimate
the equilibrium that is driven by the de facto budget availability. The
model allows the determination of λ (via the budget constraint and the
S-curve for λ) to vary across ΔE. This argument of varying determina-
tion of λ is in accordance with related literature (Bridges et al., 2010;
Zhao et al., 2011). There are some notable differences between the pro-
posed new and classical models. In the proposed new model, the
A New Methodological Approach for Cost-Effectiveness Analysis in Genomic Medicine 145
CONCLUSIONS
The model presented in this chapter provides a new conceptual
methodology for the betterment of our understanding in decision-
making process, as well as a practical tool for educational purposes. The
simplification of the model’s assumptions and the calibration of the
146 Economic Evaluation in Genomic Medicine
REFERENCES
Barton, G.R., Briggs, A.H., Fenwick, E.A., 2008. Optimal cost-effectiveness decisions:
the role of the cost-effectiveness acceptability curve (CEAC), the cost-effectiveness
acceptability frontier (CEAF), and the expected value of perfection information
(EVPI). Value Health 11 (5), 886897.
Birch, S., Gafni, A., 2006. The biggest bang for the buck or bigger bucks for the bang:
the fallacy of the cost-effectiveness threshold. J. Health Serv. Res. Policy 11 (1),
4651.
Bridges, J.F., Onukwugha, E., Mullins, C.D., 2010. Healthcare rationing by proxy: cost-
effectiveness analysis and the misuse of the $50,000 threshold in the US.
Pharmacoeconomics 28 (3), 175184.
Cartwright, W.S., 1999. Methods for the economic evaluation of health care programmes,
second edition. By Michael F. Drummond, Bernie O’Brien, Greg L. Stoddart,
George W. Torrance. Oxford: Oxford University Press, 1997. J. Ment. Health Policy
Econ. 2 (1), 43.
Donaldson, C., Birch, S., Gafni, A., 2002b. The distribution problem in economic evalu-
ation: income and the valuation of costs and consequences of health care programmes.
Health Econ. 11 (1), 5570.
Donaldson, C., Currie, G., Mitton, C., 2002a. Cost effectiveness analysis in health care:
contraindications. BMJ 325 (7369), 891894.
Eckermann, S., Briggs, A., Willan, A.R., 2008. Health technology assessment in the cost-
disutility plane. Med. Decis. Making 28 (2), 172181.
Flessa, S., 2000. Where efficiency saves lives: a linear programme for the optimal alloca-
tion of health care resources in developing countries. Health Care Manage. Sci. 3 (3),
249267.
Gafni, A., 1998. Willingness to pay. What’s in a name? Pharmacoeconomics 14 (5),
465470.
Gafni, A., Birch, S., 2006. Incremental cost-effectiveness ratios (ICERs): the silence of the
lambda. Soc. Sci. Med. 62 (9), 20912100.
Georgitsi, M., Viennas, E., Gkantouna, V., Christodoulopoulou, E., Zagoriti, Z., Tafrali,
C., et al., 2011. Population-specific documentation of pharmacogenomic markers and
their allelic frequencies in FINDbase. Pharmacogenomics 12 (1), 4958.
Gyrd-Hansen, D., 2005. Willingness to pay for a QALY: theoretical and methodological
issues. Pharmacoeconomics 23 (5), 423432.
Kvamme, M.K., Gyrd-Hansen, D., Olsen, J.A., Kristiansen, I.S., 2010. Increasing mar-
ginal utility of small increases in life-expectancy? Results from a population survey. J.
Health Econ. 29 (4), 541548.
McCabe, C., Claxton, K., Culyer, A.J., 2008. The NICE cost-effectiveness threshold:
what it is and what that means. Pharmacoeconomics 26 (9), 733744.
A New Methodological Approach for Cost-Effectiveness Analysis in Genomic Medicine 147
Nelson, A.L., Cohen, J.T., Greenberg, D., Kent, D.M., 2009. Much cheaper, almost as
good: decrementally cost-effective medical innovation. Ann. Intern. Med. 151 (9),
662667.
O’Brien, B.J., Briggs, A.H., 2002. Analysis of uncertainty in health care cost-effectiveness
studies: an introduction to statistical issues and methods. Stat. Methods Med. Res. 11
(6), 455468.
O’Brien, B.J., Gertsen, K., Willan, A.R., Faulkner, L.A., 2002. Is there a kink in consu-
mers’ threshold value for cost-effectiveness in health care? Health Econ. 11 (2),
175180.
Ramsey, S., Willke, R., Briggs, A., et al., 2005. Good research practices for cost-
effectiveness analysis alongside clinical trials: the ISPOR RCT-CEA Task Force
report. Value Health 8 (5), 521533.
Schwappach, D.L., 2002. Resource allocation, social values and the QALY: a review of
the debate and empirical evidence. Health Expect. 5 (3), 210222.
Sendi, P., Gafni, A., Birch, S., 2002. Opportunity costs and uncertainty in the economic
evaluation of health care interventions. Health Econ. 11 (1), 2331.
Sendi, P.P., Briggs, A.H., 2001. Affordability and cost-effectiveness: decision-making on
the cost-effectiveness plane. Health Econ. 10 (7), 675680.
Stinnett, A.A., Paltiel, A.D., 1996. Mathematical programming for the efficient allocation
of health care resources. J. Health Econ. 15 (5), 641653.
Towse, A., 2009. Should NICE’s threshold range for cost per QALY be raised? Yes. BMJ
338, b181.
Veney, J.E., Williams, P., Hatzell, T., 1997. In search of failure: guidelines for ministries of
health. J. Health Popul. Dev. Ctries. 1 (1), 115.
Wailoo, A., Tsuchiya, A., McCabe, C., 2009. Weighting must wait: incorporating equity
concerns into cost-effectiveness analysis may take longer than expected.
Pharmacoeconomics 27 (12), 983989.
Weinstein, M., Zeckhauser, R., 1973. Critical ratios and efficient allocation. J. Public
Econ. 2, 21472157.
Weyler, E.J., Gandjour, A., 2011. Empirical validation of patient versus population prefer-
ences in calculating QALYs. Health Serv. Res.
Whitehead, S.J., Ali, S., 2010. Health outcomes in economic evaluation: the QALY and
utilities. Br. Med. Bull. 96, 521.
Yin, X., Goudriaan, J., Lantinga, E.A., Vos, J., Spiertz, H.J., 2003. A flexible sigmoid
function of determinate growth. Ann. Bot. 91 (3), 361371.
Zhao, F.L., Yue, M., Yang, H., Wang, T., Wu, J.H., Li, S.C., 2011. Willingness to pay per
quality-adjusted life year: is one threshold enough for decision-making? results from a
study in patients with chronic prostatitis. Med. Care 49 (3), 267272.
CHAPTER 9
INTRODUCTION
The previous chapters outlined the various technical aspects and selected
applications of economic evaluation in genomic medicine. If applied
correctly, Cost-effectiveness analysis (CEA) and Cost utility analysis
(CUA) are promising technical analyses, but they are also characterized by
certain drawbacks. Some of these can be resolved if handled appropri-
ately; others involve political issues and conflicts of interest characteristic
of the health care sector in general. There are also challenges associated
with the nature of the subject itself.
Lack of Education
In many cases, health care decision-makers cannot understand the meth-
odological aspects of economic evaluation if they are not familiar with
them, which may lead to a lack of trust in the results. For example, they
are unable to understand basic concepts, such as the quality-adjusted life-
year (QALY), or the fact that a cost-effective treatment does not neces-
sarily mean savings in resources and that it could be just the “socially
acceptable option”—much less the concept of the “acceptability curve.”
They may also conclude that the results are not relevant because the anal-
yses were not done from the perspective of “their system.”
This problem is soluble in that policymakers can familiarize themselves
with the principles of economic analysis and can utilize experts conver-
sant with this scientific field and “translate” research findings into a lan-
guage that can be understood by the general public.
It Is not “Ethical”
Economic evaluation seeks to maximize social welfare through statistical
approaches, whereas clinical scientists seek to maximize the welfare of
their patient (personalized medicine). In this sense, the clinical scientist’s
approach can never fully coincide with the economist’s because it has
been said that it is “unethical” to prescribe a treatment that we know not
to be the most effective one based on pharmacoeconomic criteria. To this
end, there are several ethical issues that are complex and therefore beyond
the scope of this book.
It Increases Costs
It has also been said that economic evaluation is mostly used by compa-
nies to convince insurance funds to reimburse their expensive treatments
without specifically indicating which competing treatment should be
rejected from the reimbursement system to meet the relevant budget
goals. Studies proposing immediate discontinuation of reimbursement of
specific treatments that are not cost-effective are rare or nonexistent. The
analysis usually proceeds in only one direction: the introduction of even
more treatments. This puts tremendous pressure on insurance funds,
which collapse under the burden of the economic obligations created by
the expectations of patient, providers, and policymakers.
Despite the negative statements regarding the disadvantages of eco-
nomic evaluation, it should be clear from this book that there is potential
for its utilization in the current political climate. Preparing an economic
evaluation is now a prerequisite for a medication to be reimbursed in
most European countries, which is a significant sign of the acceptance of
this approach. In several countries, the first steps have already been taken
and we are quickly proceeding in the direction of full implementation.
This development creates a basis that will help all shareholders in the
health care sector take into account the cost of the treatments they offer,
along with their benefit.
We believe that for new treatments it is very important to prepare
economic evaluations, both to estimate the cost-effectiveness ratio and to
determine their burden on the health system. This is not as important for
already existing health technologies, because these are established in clini-
cal practice and any studies performed are not expected to lead to signifi-
cant changes in practice.
154 Economic Evaluation in Genomic Medicine
CONCLUSIONS
Economic evaluations are of utmost importance for genomic medicine to
demonstrate not only the cost-effectiveness of a certain intervention that
directly impacts on the national health care expenditure but also, most
importantly, its potential to improve the quality of life of the patients.
This is an emerging and highly promising field (Snyder et al., 2014) con-
sidering the very few economic evaluation studies in pharmacogenomics
and genomic medicine.
Furthermore, there is an urgent need at the moment to perform eco-
nomic evaluation studies in various developing countries to demonstrate
the cost-effectiveness of the individualization of certain therapeutic inter-
ventions that may not necessarily be cost-effective in developed countries.
For example, individualization of warfarin treatment has been shown to be
cost-effective for elderly Croatian patients (Mitropoulou et al., 2015), sug-
gesting that, contrary to other developed countries, pharmacogenomics-
guided warfarin treatment not only represents a cost-effective therapy
option for the management of elderly patients with atrial fibrillation in
Croatia but also may be the case for the same and other anticoagulation
treatment modalities in neighboring countries.
This fact dictates such economic evaluation studies to be replicated in
every health care system. To this end, a generic economic evaluation
model would be extremely useful to assess cost-effectiveness of individual-
ized treatment modalities. Such a tool could be used to perform a cost-
effectiveness threshold analysis across a variety of input conditions, such as
drugs, costs, and biomarkers, and would enable entities with no specific
expertise in economic analysis but with access to the necessary parameters
and cost items (see also Chapter 7) to reach a rapid decision regarding
Conclusions and Future Perspectives 155
REFERENCES
Berger, A.C., Olson, S., 2013. The Economics of Genomic Medicine: Workshop
Summary. Institute of Medicine of the National Academies, National Academies
Press, Washington, DC.
Graham, R., Mancher, M., Miller-Wolman, D., Greenfield, S., Steinberg, E., 2011.
Clinical Practice Guidelines We Can Trust. Institute of Medicine of the National
Academies, National Academies Press, Washington, DC.
Knight, J., 2003. Negative results: null and void. Nature 422, 554 555.
Mitropoulou, C., Fragoulakis, V., Bozina, N., Vozikis, A., Supe, S., Bozina, T., et al.,
2015. Economic evaluation for pharmacogenomic-guided warfarin treatment for
elderly Croatian patients with atrial fibrillation. Pharmacogenomics.
Snyder, S.R., Mitropoulou, C., Patrinos, G.P., Williams, M.S., 2014. Economic evalua-
tion of pharmacogenomics: a value-based approach to pragmatic decision-making in
the face of complexity. Public Health Genomics 17, 256 264.
INDEX
Note: Page numbers followed by “f ” and “t” refer to figures and tables, respectively.
157
158 Index
E G
Economic evaluation of health services, Genetic discrimination/stigmatization,
scope of, 1 4 18 19
Economic stakeholders, 2 3, 5 Genetic Information Non-discrimination
Education and training costs, 119 120 Act (GINA), 19
Education, lack of, 149 Genetic testing, 11, 13 14, 16, 18 19
Effectiveness, estimates of, 72 74 insurance and privacy issues, 18 19
clinical databases, 74 National Institutes of
clinical trials, 72 Health Department of Energy
controlled experiments, 72 Task Force on, 16 17
databases, 73 Genomic era, economic evaluation in, 97
expert panels, 74 balancing adverse events and efficacy,
medical records, 73 74 100 104
meta-analysis, 72 73 cost-effectiveness analysis in genomic
Epidermal growth factor receptor (EGFR), medicine and developing world,
11 108 110
EQ-5D questionnaire, 56 diseases, 104 105
Ethics in genomics, 153 performance characteristics, defining,
European Alliance for Personalized 107 108
Medicine (EAPM), 20 21 perspectives on perspective, 105 107
European Medicines Agency (EMA), pharmacogenomics and adverse events
10 11, 115 116 prevention, 97 100
European Network on Methodology and Genomic medicine, 9
Application of Economic central dogma of, 9
Evaluation Techniques, 60 61 clinical utility, demonstration of, 16 17
EuroQol EQ-5D, 53 54 current challenges of, 14 15
Evaluation, defined, 3 economic evaluation in, 15f
Evaluation of Genomic Applications in education of health professionals, 15 16
Practice and Prevention (EGAPP), future perspectives, 19 21
11, 13 14, 16 17 health care costs, 17 18
160 Index
U W
UGT1A1 28, 11 Warfarin, 12 13, 30 32
Unhealthy habits, avoidance of, 79 80 for atrial fibrillation, 141 142
Unilateral consumption, 2 concept of, 1 6
United States Preventive Services Task dosing algorithms, 120 121
Force (USPSTF), 5 genome-guided warfarin treatment,
118 119
V Whole-exome sequencing, 115
Value framework, 5f, 42f Whole-genome sequencing, 19 20,
Value of information and Bayesian analysis, 32 34, 115 116, 118 119, 125
91 94 Willingness to accept (WTA), 46 47,
Value-based health care, 4 131 132
V-erb-b2 erythroblastic leukemia viral Willingness to pay (WTP), 46 47, 138f, 139f
oncogene homolog 2 nonlinear S-shape function, 139f
(HER2/ERBB2), 11 relaxation of fixed λ, 138 140
Vitamin K epoxide reductase (VKORC1), WTA. See Willingness to accept (WTA)
13, 30 31 WTP. See Willingness to pay (WTP)