11
803 REVIEW ISSN 1462-2416 10.2217/PGS.12.52 © 2012 Future Medicine Ltd Pharmacogenomics (2012) 13(7), 803–813 Pharmacogenomics and public health: implementing ‘populationalized’ medicine Pharmacogenomics, an evolving discipline in translational research aiming to exploit one’s genomic information to predict drug efficacy and/or toxicity, is increasing in both popularity and research, often compared to an explosion of information [1,101] . While most commonly applied in personalized medicine and pharma- ceutical research, population-based analysis and implementation is also possible. The use of pharmacogenomics in personalized medicine has the potential to maximize therapeutic ben- efit and avoid adverse drug reactions (ADRs), whereas in research it enables drug companies to create safer and more efficient drugs. On a population level, particularly in resource- poor settings, it can achieve these same goals while using the funds of the government more efficiently [102] . Multiple drugs are known to be affected by nucleotide changes in the human genome, whereas others are known to be metabolized through highly variable pathways, resulting in either reduced drug efficacy, development of an ADR or possibly both [2–6] . Recent research around tamoxifen has revealed a high level of variability in the CYP2D6 pathway. For example, approximately 10% of Caucasians are unable to metabolize this drug, whereas 29% of Ethiopians metabolize it much too quickly. Both groups are left with suboptimal treatment and potentially dangerous side effects [7,8] . As such, implementation of pharmacogenomics, which could increase effective treatment, is worthy of consideration. Although the public tends to be more inter- ested in predictive genetic testing for disease, the potential benefit of predictive pharmaco- genomic testing may prove more worthwhile [9] . Given the recent global financial crisis, governments of all financial capacities are aim- ing to optimize spending. European countries with emerging economies, such as Greece and Poland, are working towards pharmacogenomic implementation in their national health for- mularies [10] . At the same time, countries with stable economies, such as Germany, are inves- tigating the cost-saving benefits of pharmaco- genomics [10] . In resource-poor settings, where multiple drug choices may not exist, or the burden of infectious diseases is high, a larger potential is expected. For example, Tanzania recently participated in a genotyping cam- paign from the Pharmacogenomics for Every Nation Initiative (PGENI) [103] , which discov- ered that the antimalarial drug provided by the Ministry of Health was less effective and cre- ated a higher risk of adverse drug events in the Tanzanian population [11] . When asked about participating in the PGENI project, Director General of the Commission for Science and Technology Hassan Mshinda was quoted as saying, “If you are poor, actually you need more Pharmacogenomics are frequently considered in personalized medicine to maximize therapeutic benefits and minimize adverse drug reactions. However, there is a movement towards applying this technology to populations, which may produce the same benefits, while saving already scarce health resources. We conducted a narrative literature review to examine how pharmacogenomics and public health can constructively intersect, particularly in resource-poor settings. We identified 27 articles addressing the research question. Real and theoretical connections between public health and pharmacogenomics were presented in the areas of disease, drugs and public policy. Suggested points for consideration, such as educational efforts and cultural acceptability, were also provided. Including pharmacogenomics in public health can result in both health-related and economic benefits. Including pharmacogenomics in public health holds promise but deserves extensive consideration. To fully realize the benefits of this technology, support is needed from private, public and governmental sectors in order to ensure the appropriateness within a society. KEYWORDS: drug use n pharmacogenomics n population n public health Lindsey Mee 1 , Konstannos Mitropoulos 2 , Athanassios Vozikis 3 & George P Patrinos* 1 Charité – Universitätsmedizin, Berlin School of Public Health, Berlin, Germany 2 Golden Helix Instute of Biomedical Research, Athens, Greece 3 University of Piraeus, Economics Department, Piraeus, Greece *Author for correspondence: University of Patras, School of Health Sciences, Department of Pharmacy, University Campus, Rion, GR-26504, Patras, Greece Tel.: +30 2610 969 834 Fax: +30 2610 969 834 [email protected] part of For reprint orders, please contact: [email protected]

Pharmacogenomics and public health: implementing ‘populationalized’ medicine

Embed Size (px)

Citation preview

Page 1: Pharmacogenomics and public health: implementing ‘populationalized’ medicine

803

Review

ISSN 1462-241610.2217/PGS.12.52 © 2012 Future Medicine Ltd Pharmacogenomics (2012) 13(7), 803–813

Pharmacogenomics and public health: implementing ‘populationalized’ medicine

Pharmacogenomics, an evolving discipline in translational research aiming to exploit one’s genomic information to predict drug efficacy and/or toxicity, is increasing in both popularity and research, often compared to an explosion of information [1,101]. While most commonly applied in personalized medicine and pharma­ceutical research, population­based analysis and implementation is also possible. The use of pharmaco genomics in personalized medicine has the potential to maximize therapeutic ben­efit and avoid adverse drug reactions (ADRs), whereas in research it enables drug companies to create safer and more efficient drugs. On a population level, particularly in resource­poor settings, it can achieve these same goals while using the funds of the government more efficiently [102].

Multiple drugs are known to be affected by nucleotide changes in the human genome, whereas others are known to be metabolized through highly variable pathways, resulting in either reduced drug efficacy, development of an ADR or possibly both [2–6]. Recent research around tamoxifen has revealed a high level of variability in the CYP2D6 pathway. For example, approximately 10% of Caucasians are unable to metabolize this drug, whereas 29% of Ethiopians metabolize it much too quickly. Both groups are left with suboptimal treatment and potentially dangerous side effects [7,8]. As such,

implementation of pharmacogenomics, which could increase effective treatment, is worthy of consideration.

Although the public tends to be more inter­ested in predictive genetic testing for disease, the potential benefit of predictive pharmaco­genomic testing may prove more worthwhile [9]. Given the recent global financial crisis, governments of all financial capacities are aim­ing to optimize spending. European countries with emerging economies, such as Greece and Poland, are working towards pharmacogenomic implementation in their national health for­mularies [10]. At the same time, countries with stable economies, such as Germany, are inves­tigating the cost­saving benefits of pharmaco­genomics [10]. In resource­poor settings, where multiple drug choices may not exist, or the burden of infectious diseases is high, a larger potential is expected. For example, Tanzania recently participated in a genotyping cam­paign from the Pharmacogenomics for Every Nation Initiative (PGENI) [103], which discov­ered that the antimalarial drug provided by the Ministry of Health was less effective and cre­ated a higher risk of adverse drug events in the Tanzanian population [11]. When asked about participating in the PGENI project, Director General of the Commission for Science and Technology Hassan Mshinda was quoted as saying, “If you are poor, actually you need more

Pharmacogenomics are frequently considered in personalized medicine to maximize therapeutic benefits and minimize adverse drug reactions. However, there is a movement towards applying this technology to populations, which may produce the same benefits, while saving already scarce health resources. We conducted a narrative literature review to examine how pharmacogenomics and public health can constructively intersect, particularly in resource-poor settings. We identified 27 articles addressing the research question. Real and theoretical connections between public health and pharmacogenomics were presented in the areas of disease, drugs and public policy. Suggested points for consideration, such as educational efforts and cultural acceptability, were also provided. Including pharmacogenomics in public health can result in both health-related and economic benefits. Including pharmacogenomics in public health holds promise but deserves extensive consideration. To fully realize the benefits of this technology, support is needed from private, public and governmental sectors in order to ensure the appropriateness within a society.

KEYWORDS: drug use n pharmacogenomics n population n public health Lindsey Mette1, Konstantinos Mitropoulos2, Athanassios Vozikis3 & George P Patrinos*1Charité – Universitätsmedizin, Berlin School of Public Health, Berlin, Germany 2Golden Helix Institute of Biomedical Research, Athens, Greece 3University of Piraeus, Economics Department, Piraeus, Greece *Author for correspondence: University of Patras, School of Health Sciences, Department of Pharmacy, University Campus, Rion, GR-26504, Patras, Greece Tel.: +30 2610 969 834 Fax: +30 2610 969 834 [email protected]

part of

For reprint orders, please contact: [email protected]

Page 2: Pharmacogenomics and public health: implementing ‘populationalized’ medicine

Pharmacogenomics (2012) 13(7)804 future science group

Review Mette, Mitropoulos, Vozikis & Patrinos

evidence before you invest, rather than if you are rich” [12].

Genetic information is a valuable tool in medi­cal decision­making, both individually and at a population level [5,13]. However, the use of genetic information is not innocuous. While the authors recognize genetic discrimination as a potential threat to both individuals and populations, the topic was excluded from this review. Highly indi­vidualized genetic medicine remains far in the future, and in order for pharmaco genomics to realize its full potential, careful consideration of its implementation is essential. Without a proper reg­ulatory framework, the complete implementation of pharmacogenomics is improbable [13].

In this study, we conducted a narrative litera­ture review surrounding the research question of how pharmacogenomics can be used beneficially in public health. In particular, we reviewed what has been published in the field of both pharmaco­genomics and public health, regarding how the two sciences can constructively intersect, particu­larly in resource­poor settings with the ultimate goal of determining the role pharmacogenomics plays in public health.

MethodsWe conducted a narrative literature review, in which available information was reviewed and analyzed in order to gain knowledge and understanding about the potential connections between pharmacogenomics and public health. Special attention was given to the relationship between pharmacogenomics and infectious dis­eases that pose a major burden in developing countries. Furthermore, recommendations for applying pharmacogenomics to a national health plan were analyzed and synthesized, in order to develop a comprehensive list of considerations which may be applicable to future analyses.

A systematic literature search was carried out multiple times in the PubMed database with Medical Subject Heading (MeSH) search terms. The initial search looked at publications related to the study topic in general, while subsequent searches focused on subpoints. A variety of search terms were used in order to deliver the most rel­evant results. These terms included ‘pharmacoge­netic’, ‘pharmacogenomic’, ‘public health’, ‘policy’, ‘malaria’, and ‘tuberculosis’. Additional terms were used to narrow the results. The search parameters and the number of hits per parameter are listed in Supplementary table 1 (see www.futuremedicine.com/doi/suppl/10.2217/pgs.12.52).

These searches resulted in a total of 1446 studies. Only studies published since 2000 were

considered for review, as the first working draft of the Human Genome Project was completed in 2000. We also employed the limitation of ‘English language’ on account of our language abilities. The previously mentioned limitations narrowed the search results to 1186 publications, of which the Charité – Universitätsmedizin library has access to 945. Thirty one of the retrieved results were duplications. The remaining 914 studies were then evaluated in terms of relevance to the study topic. The exclusion criteria are listed in Supplementary box 1.

Ultimately, a total of 914 publications were obtained including those from the PubMed search. Of those, 887 publications were irrel­evant based upon the abovementioned exclusion criteria. Finally, 27 publications were included in the literature review. The overall search strategy is demonstrated in Figure 1.

n LimitationsThe English language restriction potentially eliminated important and relevant publications, particularly on account of the native languages in developing countries that were a research focus. Also, the literature search was limited to publications indexed within the USA National Library of Medicine PubMed database. While extensive, it certainly does not include all pub­lications. Furthermore, the research was lim­ited to articles which were available through the Charité – Universitätsmedizin library. The limitations imposed by the database and the library may have excluded articles relevant to the study topic. Finally, while the researchers acknowledge the impact of chronic disease on the health of the population globally, the results were restricted to infectious diseases burden­ing low­ and middle­income countries in order to specifically focus on these settings. To this point, specific countries and/or healthcare sys­tems were not analyzed outside of their repre­sentation in the retrieved articles.

Of the 27 publications analyzed, two journals accounted for twelve of the publications (Nature Reviews Genetics: 6; Pharmacogenomics: 6). In addition, four authors appeared in five or more publications (Séguin: 5; Hardy: 5; Singer: 11; Daar: 11). The views of these authors, in com­bination with the aims of the aforementioned journals, therefore shaped a significant portion of the retrieved results.

ResultsThe literature obtained from the PubMed searches was critically read to identify emergent

Page 3: Pharmacogenomics and public health: implementing ‘populationalized’ medicine

www.futuremedicine.com 805future science group

Pharmacogenomics & public health Review

themes reflective of the study objective, including the intersection of pharmacogenomics and public health, and public policy recommendations. table 1 details the analysis parameters. On account of the narrow search parameters, many of the retrieved studies contained overlapping themes such as keys for implementing pharmaco genomics into public policy. All 27 articles analyzed are summarized in Supplementary table 2.

Existing and theoretical connections between pharmacogenomics and public health are well documented [11,14–38], as is the poten­tial for pharmacogenomics to improve public health [14–19,21,23–25,27,31,33–36,38]. We have there­fore decided to summarize our analysis in the themes and subthemes outlined in box 1:

� Interactions between pharmacogenomics and public health;

� Pharmacogenomics and drugs;

� Pharmacogenomics and diseases;

� Benefits of including pharmacogenomics in public health policy;

� Key factors for implementing pharmaco­genomics into public health policy;

� Related points for consideration in policy formation.

Interactions between pharmacogenetics/pharmacogenomics & public healthIncluding pharmacogenomics into national healthcare plans has the potential to make healthcare more cost effective by ensuring bet­ter treatment, proper medication prescription and identification of subpopulations at risk of disease and ADRs [14,17,18,20,21,23,26–29,31,33–39]. Furthermore, it can be a mechanism to save lives through better medicine development and provision [11,15–18,20,21,23,26–28,31,33–39]. Indirect benefits of pharmacogenomics include increased research, which not only holds the evident ben­efit of health translation, but economic benefits as well by creating a knowledge­based economy, developing research capacity and reversing ‘brain drain’ [14,17,18,20,21,23,27,31,34–36,38]. The discus­sion of the appropriateness of this technology in these settings has been divided into two con­cepts: luxuries versus necessities and healthcare inequalities.

n Luxuries versus necessitiesPharmacogenomics has been viewed as a luxury of advanced medical treatment. However, the

idea of luxury versus necessity (e.g. healthcare) varies within and between populations. In the western world, healthcare is a necessity; in low­income countries (LIC) it is a luxury [27]. Similarly, LIC can least afford to waste their already limited resources in healthcare [34]. The use of pharmacogenomics in resource­poor set­tings would introduce cost­effective treatment strategies, better diagnosis and treatment, iden­tify at­risk individuals, guide national health policy decision­making, and perhaps improve the nation’s health, thereby improving their economic capabilities [18,23,27,31,34]. Arguably, pharmacogenomics “should be seen as a public good to benefit all of humanity, especially those most in need” [27].

n Healthcare inequalitiesOne of the hopes of pharmacogenomics is to provide the best treatment, through improved

Potentially relevant articlesidentified and screened

by titlen = 1446

Potentially relevant articlesidentified and screened

by titlen = 1186

Potentially relevant articlesidentified and screened

by titlen = 945

Potentially relevant articlesidentified and screened

by titlen = 914

Articles analyzed for the literature review

n = 27

Articles not available in EnglishArticles published prior to 2000

n = 260

Articles not accessible throughCharité – Universitätsmedizin

n = 241

Duplicate articlesn = 31

Articles not related to study topicn = 887

Figure 1. Screening strategy for the literature review.

Page 4: Pharmacogenomics and public health: implementing ‘populationalized’ medicine

Pharmacogenomics (2012) 13(7)806 future science group

Review Mette, Mitropoulos, Vozikis & Patrinos

diagnosis and prescription [18,20,26,31,32,37,39]. Yet achieving that goal while access to healthcare or medications is still laced with inequality is chal­lenging. Even in settings with universal health­care, inequalities exist [17,19,23,35,39]. As recently stated, “in order to get the benefit of health, of genomics, universal health insurance is probably a pre­requisite” [35]. This is seen in Thailand, which has universal healthcare but access to medications is problematic [35]. Additionally, pharmacogenetic testing is not covered by national health plans. This current pharmaco­genetic trickle­down effect in Thailand has lim­ited access to the wealthy [35]; the same is true in India [23].

The other type of inequality is felt globally. Termed the ‘genomics divide’ it addresses the separation between developed and develop­ing countries in terms of genomic medicine [14,15,19,27,28,38]. Healthcare innovation, tech­nology and research rarely focus on or benefits populations in LIC [14,21,27,38]. Moving forward, the research, development and implementation of pharmacogenomics needs to focus on disad­vantaged populations and ensuring equitable access to and distribution of healthcare resources [17–19,27,28]. Simply put by one scientist, “What can first world science do, not for the west, but for the rest” [28].

Pharmacogenomics & drugsMultiple possibilities exist for improving public health by incorporating pharmacoge­nomics into medicine. One of the hopes for pharmaco genomics is to elucidate the dif­ferences in drug response between patients [15,18,21,23,24,26,29–31,33–35,37,39], which would allow for better drug development [20,21,27,30,37,39] and opportunities to predict patient drug response

prior to dosing [18,23,27,30,36,37,39]. However, due to the varying therapeutic indices across populations, many countries are question­ing the effectiveness within their own popu­lations in comparison to the initial study population [20,26,27,31,35,39].

The cost of ADRs – both social and eco­nomic – is a reality pharmacogenomics hopes to reduce, if not eradicate. By identifying patient responder groups and incorporating pharma­cogenomic technologies in pharmaceutical development, ADRs can largely be prevented [16,18,23,26,27,30,33–35,37,39], thereby improving health and translating to a cost­effective means of healthcare provision [15,16,18,21,27,29,31,33,37]. Especially important in the context of LIC is the access to medicine, both physical and financial. Researchers are moving towards guiding policy­makers in the creation of national drug formu­laries, in order to take population data, variation and cost–effectiveness into account [18,26,31].

Pharmacogenomics & diseasesThe three infectious diseases which pose the larg­est burden globally – malaria, tuberculosis and HIV/AIDS – were the most frequently addressed diseases in the discussion of public health and pharmacogenomics [11,14,16,18,22–24,27,31,32,35–38]. These diseases affect millions of individuals worldwide, primarily in developing countries. While treatments to combat these diseases exist, they are often insufficient, expensive and diffi­cult. Isoniazid, a common drug in tuberculosis treatment, has variable metabolism dependent on the status of multiple genes and is consid­ered one of the first examples of pharmacoge­netics [32]. Accordingly, many have identified tuberculosis as another target for pharmacoge­nomics and genomic technology, with hopes

Table 1. Checklist of relevant topics.

Topic Pharmacogenomics Pharmacogenomic research in global burden diseases

Public policy

Relevance in regards to pharmacogenomics

Clarification of pharmacogenomics and pharmacogenetics

Is pharmacogenomic research being conducted on these diseases? If yes, does it account for variables other than genetic?

Do suggestions exist for incorporating pharmacogenomic technologies into national healthcare public policy?

Relevance in regards to public health

What are the arguments in favor and against pharmacogenomics from the field of public health, including sociological and epidemiological branches?

Are epidemiological studies on these diseases taking genetic factors into account? How does pharmacogenomic research propose to use its results?

Do frameworks exist to guide professionals and policy-makers on whether to use pharmacogenomic data?

Relevance in regards to gender and/or diversity

How are specific minority groups defined? Is pharmacogenomic research gender and ethnically sensitive?

Is the research focused towards a gender or cultural group? Does research have negative implications for a particular group?

What has been published on the protection of individuals or groups participating in genetic research?

Page 5: Pharmacogenomics and public health: implementing ‘populationalized’ medicine

www.futuremedicine.com 807future science group

Pharmacogenomics & public health Review

of increasing treatment efficacy while decreas­ing costs [11,14,15,18,21,38]. India has placed a research priority on drug development, while South Africa is utilizing genomic technology to determine tuberculosis susceptibility and drug metabolism in its population [14,27].

Similarly, the antimalarial drug amodiaquine is not currently recommended for first­line treat­ment, on account of a high frequency of ADRs. However, the drug is metabolized differently across populations, affecting Caucasians much more frequently than Africans, where malaria is much more common and problematic. This varying response to amodiaquine has recently been identif ied by polymorphisms in the CYP2C8 gene [11,22,31]. Appropriately, Zanzibar, Ghana, Malaysia and Bolivia have undertaken efforts in genotyping to determine the preva­lence and significance, of CYP2C8 variants in their populations [11,26,31].

Pharmacogenomics has found success in HIV/AIDS, where the antiretroviral treat­ment, abacavir, has been associated with a hyper sensitivity reaction to patients who har­bor a HLA-B*3701 allele. This reaction varies between populations; however it has been noted to occur in 20% of Thai patients [30,35,37]. An alternative therapy exists, although it is twice as expensive [35]. India and South Africa, two countries with a high HIV prevalence, believe pharmacogenomics has the potential to greatly impact their nation’s health, and have thus named it a research priority [14,38]. It is hoped that by using pharmacogenomics, nations will improve patient treatment and efficacy, which will not only save scarce financial resources, but lives as well [18,27,35,36].

Benefits of including pharmacogenomics in public health policyThe impact of pharmacogenomics on developing countries has remained minimal. Small popula­tions – whether in terms of nationality, genetic profile or disease – have historically been unin­teresting to researchers and pharmaceutical com­panies [15,18,35]. Thus, many describe the necessity of sparsely populated or LIC to invest in pharma­cogenomics within their own borders, to focus on local health needs ignored on the international level [14,18,19,24,25,31,33–38]. By doing so, local disease and drug metabolism patterns will be uncovered, resulting in better and more equitable healthcare and delivery [14,15,18,24,27,33–36,38].

In addition, rationale for the use of pharmaco­genomics in LIC frequently revolves around the

argument for best allocation of resources. In terms of national resources, it is in the poor­est settings where pharmacogenomics can have the largest impact [31]. Using this technology allows for cost­effective medication prescrip­tion [36,37], thereby increasing prevention, diag­nosis and treatment [23,26,33,39], all of which decrease national healthcare costs and allow for a redistribution of scarce resources [14,18,33]. It was suggested that resource­poor settings should replicate existing, successful models in order to implement this strategy to maximize both human and financial resources [38]. Ghana recently implemented pharmacogenomic medi­cine using population­based data as opposed to individual data, which is too expensive for standard use [26].

Furthermore, including pharmacogenomics in public health policy is an investment in both health and research. The lack of communication and collaboration between science and govern­ment was frequently noted [14,15,19,23,31,33,34], as well as that between academic research and the private sector [14,15,17,19,35,38]. The possibility of ‘north–south’ collaborations between developed

Box 1. Themes and subthemes identified in the literature review.

Interactions between pharmacogenomics & public health � Luxuries versus necessities

� Healthcare inequalities

Pharmacogenomics & drugs � Reducing adverse drug reactions

� Developing better drugs

Pharmacogenomics & diseases � Malaria

� Tuberculosis

� HIV/AIDS

� Miscellaneous diseases

Benefits of including pharmacogenomics in public health policy � Local health benefits

� Resource allocation

� Cultivating collaboration

� Using science as a marketable good

Key factors for implementing pharmacogenomics into public health policy � The affordability, accessibility and acceptability (AAA) principle

� Education

� Surveillance

� Political will

� Policy formation

Related points for consideration in policy formation � Genomic sovereignty

� Commercialization

� Ethical, legal and social issues

� Race and ethnicity

Page 6: Pharmacogenomics and public health: implementing ‘populationalized’ medicine

Pharmacogenomics (2012) 13(7)808 future science group

Review Mette, Mitropoulos, Vozikis & Patrinos

and developing countries was similarly pre­sented as an opportunity to build the local sci­ence capacity of LIC [15,19,24,28,31,33–36,38]. These international collaborations increase the likeli­hood of LIC receiving beneficial health­related technologies [34]. However, national regulatory frameworks may prevent international collabora­tion, which could be disadvantageous [23,34,35]. Examples of successful existing international collaborations, such as the Pan­Asian HUGO [34,35,104], PAHO [19,33,105] and PGENI [31,34], were presented as possible models for replication.

Finally, genomic knowledge is a public good for public benefit, not only in countries with large budgets and highly developed science sectors [24,27]. It carries the potential to shape national policy, social understanding and edu­cation [14,18,23]. Through government­supported investment in genomic technologies, countries are afforded an opportunity to enter the global knowledge­based economy [23,24,27,33–35] and leverage their research results to spur local innovation [14,24,33,34]. Furthermore, a commit­ment to this research attracts foreign companies to conduct research in these locations, which further builds scientific capacity and generates capital [23,24,33–35].

Key factors for implementing pharmacogenomics into public healthThe decision to implement pharmacogenomics into public health is multifaceted and requires the combined efforts of multiple sectors of society.

n Affordability, acceptability & appropriateness The affordability, acceptability and appropri­ateness of pharmacogenomics are key elements to its successful implementation. With a par­ticular respect to LIC, the genomic technology and resulting drugs must be affordable, both to individuals in need and the national govern­ments trying to procure them [15,18,19,21,27,34,35]. Governments subsidizing genomic research also must appropriate sufficient funding to promote research without cutting funding in other impor­tant areas [14,15]. Public acceptance of this tech­nology is crucial, and various methods have been suggested to increase social acceptability, includ­ing public education efforts [14,17,25,27], protection against genetic discrimination [23], and spending an appropriate amount of public funding in this field [35]. Finally, the implementation of this technology must be suitable in terms of society,

health and disease, technology and governance. The diseases targeted and the use of this tech­nology must complement the country’s health profile [17,19,24,27,34] while utilizing socially and scientifically acceptable methods [18]. National governments who elect to use this technology must provide researchers appropriate incen­tives while continuing to monitor the research and implementation of pharmacogenomics into public health [14,17,19,24,25,27,34].

n Education & political willTo effectively implement pharmacogenomics into society, politicians must be aware of the capabilities, limitations and implications of pharmacogenomics on a society to ensure appro­priate and effective policy formation [17,24,31,38]. Healthcare professionals must receive increased education in pharmacogenomics in order to comply with the policies and best serve the population [14,17–20,23,24,26,31,35,38]. Finally, edu­cational efforts to the greater public are essential [14,15,17–21,24,25,29,33–35,38]. Thailand and Mexico have government­sponsored educational events and publications distributed in their societ­ies that explain pharmacogenomics [24,33,35]. However, in LIC where education is a privilege and not a guarantee, delivering this information is challenging [15,17,23,24,33].

In addition, implementation of a national public health policy is dependent on the commit­ment of the government. Government funding is essential to not only implement a policy but also to promote research in this area [14,17,23,24,33–35]. Frequent changes in political power threaten existing programs, which creates a continu­ous need for mobilizing political support [14,15,19,28,36,38]. Successful campaigns have dem­onstrated how investing in pharmaco genomics can strengthen a nation’s health and economy [14,15,19,23,24,28,32–35,38]. India, Thailand and Mexico have all succeeded in creating national policies to implement pharmaco genomics [14,23,33–35]. These countries, in addition to South Africa, have all undertaken nationwide research programs in order to maximize the government subsidy and the nation’s benefit.

n Policy formationThe formation of a public policy on pharmaco­genomics is country­specific; however there are a series of suggestions for consideration. Communication between the science sector, stakeholders and the government is of extreme importance to ensure the proper interpretation and beneficial application of research results.

Page 7: Pharmacogenomics and public health: implementing ‘populationalized’ medicine

www.futuremedicine.com 809future science group

Pharmacogenomics & public health Review

Policies must account for healthcare delivery issues [14,17,18,20,21,23,26,38], be feasible [15,36], con­sist of clear goals [14,24] and incorporate proper regulatory frameworks and evaluation methods [17,23,34,35,37]. Policies must also promote further scientific research and translation [23,26], encour­age public–private partnerships [14,17,23,34,38] and define methods to maximize and create invest­ment capital [21,23,24,38]. Socially, the policy must adequately reflect societal needs and values, enhance public education and clearly describe ethical guidelines [14,17,23,31,38,39].

Related points for consideration in policy formationThe benefits and challenges of implementing pharmacogenomics into public health are clearly identified. However, there are additional issues that may arise and require careful consideration, ideally prior to implementing such a policy. One such issue is genomic sovereignty. The human genome is recognized as a global public good, not owned by a single entity or institution and not designed to benefit a select few, but the entire population [33–35]. This is especially true in terms of bio­colonialism and genetic piracy, where research is conducted in populations who are unwilling or unable to research themselves [34,35]. India’s massive population and resulting diver­sity is viewed as a genomic sovereign resource; the same is true of Mexico [23,33,34]. Mexican authorities assume responsibility for discover­ing their own information, in part owing to a concern that Mexico will become dependent on the countries holding this information and sub­sequent technologies it produces [33]. The level of legislation surrounding genomic research varies greatly between countries. Thailand and India lack national legislation, but have guidelines in place regarding the export of genomic informa­tion [23,33,34]. Mexico, conversely, has national laws designed to protect its genomic sovereignty and prohibit the research or transportation of genomic information without government per­mission [33,34]. However, there are concerns that legislation limiting the export of genomic information will impede research collaboration between nations, which can be very valuable to LIC [23,34,35].

Another issue is commercialization. One way to incentivize genomic research within a coun­try is to promote commercialization [14,21,23,33–35,38]. Many articles addressed the need for national support of commercializing research in order to ensure development [14,23,24,33–35,38]. Mexico’s Instituto Nacional de Medicina

Genomica de Mexico (INMEGEN) [106] and Thailand’s Center of Excellence for Life Sciences (TCELS) [107] are two such examples of national support [34,35]. The importance of public–private partnerships in research was emphasized, owing to the limited resources LIC have to dedicate to such research [14,15,17,23,34,35,37,38]. The develop­ment of a commercial research sector is a strat­egy to generate economic and health benefits to developing countries [14,28,34]. Interestingly, only African countries viewed this as an oppor­tunity to foster international collaboration [24,38]. The promotion of commercialized research also raises some concerns. Researchers in Thailand and South Africa felt it was unethical to com­mercialize research results, while researchers in Thailand and India valued peer­reviewed pub­lications more highly than commercial incen­tives [23,34,35]. When a country promotes research commercialization it is vital to balance these commercialization efforts with the principles of public health and human rights [14,37,39].

Ethical issues surrounding the commercializa­tion of research [28,35], biobanking [23], distribu­tion of goods, cost–effectiveness analyses [21,30], informed consent, standard of care [18,27,28,38] and legal issues surrounding intellectual prop­erty must be addressed by leaders in coun­tries implementing pharmacogenomics. Also addressed is the concern of creating a genetic underclass [27,28]. The protection of identity in any form of genomic research is vital to eliminat­ing further opportunity to discriminate against a racial, ethnic or social group within a society [18,21,28,35,39]. There is uncertainty as to whether employing pharmacogenomic research will increase stigmatization [15,25,27,28,38,39] or reduce it [23,27]. What are readily agreed upon are the inequalities and the potential to increase access to healthcare for those currently disadvantaged [15,19,24,28,39]. In order to protect communities and ensure ethical research, capacity building in Ethical, Legal, and Social Issues (ELSI) is essen­tial [14,15,19,24,33,36,38]. To accomplish this, policy­makers must be well­informed regarding genom­ics and associated issues affecting populations [24,33,38]. Equally important is the engagement of the public in supporting genomic research [24,35] and informing the public about genomics and ELSI [17,19,23,24,34]. Finally, the social cost of such technology must be addressed, weighing individual choice against the public good [25,27].

DiscussionAlthough incorporating pharmacogenomics into LIC initially seems illogical, it is gaining

Page 8: Pharmacogenomics and public health: implementing ‘populationalized’ medicine

Pharmacogenomics (2012) 13(7)810 future science group

Review Mette, Mitropoulos, Vozikis & Patrinos

acceptance, since in a resource­poor setting, the financial consequences of ADRs to the national healthcare system, as well as the general popula­tion, will be more severe as compared to devel­oped nations. What was previously considered a tool for personalized medicine is now paving the path towards ‘populationized’ medicine (see Figure 2). However, to realize the benefits of including pharmacogenomics on a national level, both social and political considerations must be taken in order to ensure the desired outcome.

From a policy perspective, laws prohibiting discrimination, particularly genetic discrimi­nation, will be essential to protect the popula­tion. One way to contribute towards a healthier political/medical/social climate is to provide genetics education to the general population, such as TCELS has, through public meetings, media appearances and educational material distribution [35]. Prior to creating educational campaigns, program officials must determine the current social climate and understanding of pharmacogenomics, in order for the campaigns to have the maximum impact. While only a few studies have examined public attitudes regarding genetics – and even fewer regarding pharmaco­genomics – the results have thus far shown a favorable attitude towards genetics and genetic testing services [40].

Increasing social awareness allows the public to be a more informed stakeholder in the dis­cussion surrounding public health policies. The same is true of educating doctors and scientists. Recent surveys in Greece show physicians are more willing to undergo genetic testing than to prescribe it, suggesting a lack of education in how to interpret and apply the results in patient care [40]. Further supporting this suggestion is the gap between patient expectation and physi­cian understanding in clinical genetics [5]. Thus, the scientific community has called for increased

genetic content in medical education programs. European PGENI regional coordinating cen­ter, the Golden Helix Institute of Biomedical Research [41,108], regularly organizes and hosts the ‘Golden Helix Pharmacogenomics Days’ to facilitate knowledge transfer from timely research to clinical practice [42]. Increasing the genetics education will better allow these professionals to effectively implement created policies.

Pharmacogenomics can increase access to medicine by increasing the amount and func­tionality of available medicines, provided that implementing pharmacogenomics results in improved resource allocation and better drug provision; it can also restrict access if the devel­oped drugs are too expensive for regular use. To this end, public health strategies regarding the use of generics and interchangeable drug products can be a solution that has already yielded some positive results in many countries; when combined with pharmacogenomics, not only will the quality of life of the patients be improved but the healthcare cost savings can also be further reduced.

Access to essential medicine is a human right, yet barriers to these medicines remain a major problem. Also important is the appropriateness of this technology in a society. In the event that implementing pharmacogenomics improves treat­ments, improves the health of society, increases education and employment opportunities and discourages discrimination, the appropriateness may still be in question. Medical modernization that does not strive to incorporate traditional healing methods threatens to marginalize tra­ditional healers and their associated practices. Therefore, incorporating this technology has the potential to lead to the abandonment of current traditional and natural medicine treatments, thereby resulting in a loss of culture. Survival International, a human rights organization, warns against the dangers of strictly adhering to western medicine and disregarding traditional treatments and healers, claiming that doing so brings more harm than good [109]. The technology also may be delivered in a culturally unacceptable man­ner. For instance, in a population­based analy­sis, the populations chosen for representation must accurately reflect the structure of society without creating or reinforcing social divisions. Historically, research in pharmacogenomics across populations has used racial criteria, as opposed to ethnic. While well­documented, the clinical relevancy of racial grouping remains in question. The Frequency of Inherited Disorders Pharmacogenomics database (FINDbase) aims

Nat

iona

l G

enet

ic D

atab

aseGenome data

Genome data

EnvironmentPublic health

guidelines

Personalized medicine

‘Populationalized’ medicine

Figure 2. Pharmacogenomics in personalized and ‘populationalized’ medicine.

Page 9: Pharmacogenomics and public health: implementing ‘populationalized’ medicine

www.futuremedicine.com 811future science group

Pharmacogenomics & public health Review

to bring ethnic­specific differences to attention. The database exclusively documents pharmaco­genomic markers in various populations and ethnic groups globally, currently hosting data on over 150 populations [43,110]. Finally, patients must continue to be treated as individuals, not as representatives of a particular subpopulation. Population­based pharmacogenomics should be used as a guide towards national health policy decision­making, and does not serve to under­mine the inherent uniqueness of each individual or the opportunity of individual personalized medicine.

Owing to potential complications arising from genetic information, the task of policy­makers in public health policy formation is difficult. In countries where these policies have already been effectively introduced, the will of the gov­ernment was essential. One successful approach towards gaining favor of the government is being able to prove the economic benefits associated with this type of a policy. Increasing health and gross domestic product, decreasing waste (i.e., through ineffective treatments), and opportu­nities for better resource allocation are attrac­tive outcomes, especially to governments with

extremely limited resources. Furthermore, prom­ises of developing scientific infrastructure and increasing partnerships with global organizations have potential to increase a national economy. By enacting legislation, nations can guard resources, encourage local innovation and foster the abil­ity to participate globally in genomic research. An alternative to self­determined and funded pharmacogenomic research within a country is a partnership model, such as PGENI. This permits Health Ministries to obtain informa­tion and expert consultation for decision­making without the financial commitment. However, the government is still responsible for protecting its residents, its genomic sovereignty, and the deci­sions that are made as a result of this gathered information. Policy­makers should also con­sider sustainability, to ensure continuing drug affordability and continued research translation towards better health.

ConclusionIn areas with limited resources, population­based pharmacogenomics may be a worthy investment, whether self­funded or accomplished through collaboration. However, in order to achieve a

Executive summary

Background � Pharmacogenomics serves personalized medicine to increase treatment benefits and minimize adverse drug reactions. Using pharmacogenomics on a population level can accomplish the same benefits, in addition to using health resources more efficiently.

Methods � Here, we conducted a narrative literature review of what has been researched and published in the field of both pharmacogenomics and public health. The literature review focused on how the two sciences can constructively intersect, particularly in resource-poor settings.

Benefits of including pharmacogenomics in public health policy � The benefits of including pharmacogenomics in public health policy can be realized on multiple levels of society, including local health benefits, resource allocation, increased scientific collaboration and the marketability of scientific knowledge.

Interactions between pharmacogenomics & public health � The implementation of pharmacogenomics into national healthcare plans has the potential to make healthcare more cost effective by ensuring the best treatment of individuals, the proper prescription of medication and identification of subpopulations at risk of disease and adverse drug reactions.

Keys for implementing pharmacogenomics in public health � The recent shift towards incorporating pharmacogenomics into low–medium income countries is gaining acceptance in the science community. However, in order to realize the benefits of including pharmacogenomics in public health policy, both social and political considerations are essential to ensure the desired outcome.

Points for consideration � Stakeholder engagement is necessary to properly assess this issue from areas outside of economic benefit. The feasibility of this technology needs to be assessed in terms of cost–benefit analyses, sustainability, education and scientific infrastructure.

Conclusion � In areas with limited resources, population-based pharmacogenomics may be a worthy investment, whether self-funded or accomplished through collaboration.

Future perspective � Further studies should aim to assess the public’s view on incorporating this technology into national healthcare plans.

Page 10: Pharmacogenomics and public health: implementing ‘populationalized’ medicine

Pharmacogenomics (2012) 13(7)812 future science group

Review Mette, Mitropoulos, Vozikis & Patrinos

public health policy capable of incorporating pharmacogenomics, public and political invest­ment is necessary. Regardless of the country set­ting, the protection of citizens and populations within the country is of the highest importance. One of the acclaims of genomics is the integra­tive approach it takes towards health and disease. A public health genomic policy should aim to be equally as integrative.

Future perspectiveThe use of population­based pharmaco­genomics is in its infancy, but early successes provide a promising outlook. While this lit­erature review focused mainly on infectious diseases and low­income countries, future studies should investigate the impact of phar­macogenomics on chronic disease treatment or how to implement such a strategy in nonuni­versal healthcare systems. Furthermore, addi­tional economic models are needed. To date the majority of cost–effectiveness analyses are

calculated on an individual patient basis, not population­based. In terms of LIC, studies ana­lyzing the increase of a nation’s GDP, the reduc­tion of disability­adjusted life­years in relation to pharmacogenomic healthcare expenditure would be beneficial.

AcknowledgementsThe authors apologize to all of our colleagues whose important work could not be directly cited.

Financial & competing interests disclosureThis work has partly fulfilled the requirements of the Master’s Thesis of L Mette. Part of this work has been finan-cially supported in part by the Golden Helix Institute of Biomedical Research. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

ReferencesPapers of special note have been highlighted as:n of interestnn of considerable interest

1 Xie H, Frueh FW. Pharmacogenomics steps toward personalized medicine. Pers. Med. 2(4), 325–337 (2005).

2 Gil JP, Berglund G. CYP2C8 and antimalarial drug efficacy. Pharmacogenomics 8(2), 187–198 (2007).

3 Gumbo T, Louie A, Liu W et al. Isoniazid bactericidal activity and resistance emergence: integrating pharmacodynamics and pharmacogenomics to predict efficacy in different ethnic populations. Antimicrob. Agents Chemother. 5(7), 2329–2336 (2007).

4 Hasan S, Daugelat S, Rao PS, Schreiber M. Prioritizing genomic drug targets in pathogens: application to Mycobacterium tuberculosis. PLoS Comput. Biol. 2(6), 539–560 (2006).

5 Squassina A, Manchia M, Manolopoulos VG et al. Realities and expectations of pharmacogenomics and personalized medicine: impact of translating genetic knowledge into clinical practice. Pharmacogenomics 11(8), 1149–1167 (2010).

n Comprehensive review of the current applications and future challenges of pharmacogenomics towards its implementation in mainstream medical practice.

6 Lazar A, Tomalik­Scharte T, Fuhr U. Applications of genotyping and phenotyping for clinically­relevant polymorphisms of drug

metabolizing enzymes and drug transporters. In: Handbook of Analytical Separation, Volume 5: Drug Monitoring and Clinical Chemistry. Hempel G (Ed.) Elsevier, Oxford, UK, 321–354 (2004).

7 Bernard S, Neville KA, Nguyen AT, Flockhart DA. Interethnic differences in genetic polymorphisms of CYP2D6 in the US population: clinical implications. Oncologist 11, 126–135 (2006).

8 Dezentjé VO, Guchelaar H, Nortier JWR, van de Velde CJH, Gelderblom H. Clinical implications of CYP2D6 genotyping for tamoxifen treatment for breast cancer. Clin. Cancer Res. 15(1), 15–20 (2009).

9 Sagia A, Cooper DN, Poulas K, Stathakopoulos V, Patrinos GP. Critical appraisal of the private genetic and pharmacogenomics testing environment in Greece. Pers. Med. 8(4), 413–420 (2011).

10 Mitropoulos K, Johnson L, Vozikis A, Patrinos GP. Relevance of pharmacogenomics for developing countries in Europe. Drug Metabol. Drug Interact. 26(4), 143–146 (2011).

11 Cavaco I, Strömberg­Nörklit J, Kaneko A et al. CYP2C8 polymorphism frequencies among malaria patients in Zanzibar. Eur. J. Clin. Pharmacol. 61(1), 15–18 (2005).

12 Oxman AD, Bjørndal A, Becerra­Posada F et al. A framework for mandatory impact evaluation to ensure well informed public policy decisions. Lancet 375, 427–431 (2010).

13 Patrinos GP. General considerations for integrating pharmacogenomics into

mainstream medical practice. Hum. Genomics 4(6), 371–374 (2010).

14 Acharya T, Kumar NK, Muthuswamy V, Daar AS, Singer PA. ‘Harnessing genomics to improve health in India’ – an executive course to support genomics policy. Health Res. Policy Syst. 2(1), 1 (2004).

15 Acharya T, Rab MA, Singer PA, Daar AS. Harnessing genomics to improve health in the Eastern Mediterranean region – an executive course to support genomics policy. Health Res. Policy Syst. 3(1), 1 (2005).

16 Ameen M. Cutaneous leishmanisasis: disease susceptibility and pharmacogenetic implications. Pharmacogenomics 10(3), 451–461 (2009).

17 Burton H, Adams M, Bunton R, Schröder­Bäck P. Developing stakeholder involvement for introducing public health genomics into public policy. Public Health Genomics 12, 11–19 (2009).

18 Daar AS, Singer PA. Pharmacogenetics and geographical ancestry: implications for drug development and global health. Nat. Rev. Genet. 6, 241–246 (2005).

19 Daar AS, Berndtson K, Persad DL, Singer PA. How can developing countries harness biotechnology to improve health? BMC Public Health 7, 346 (2007).

20 Davis RL, Khoury MJ. A public health approach to pharmacogenomics and gene­based diagnostic tests. Pharmacogenomics 7(3), 331–337 (2006).

21 Garrison LP, Carlson RJ, Carlson JJ, Kuszler PC, Meckley LM, Veenstra DL. A review of

Page 11: Pharmacogenomics and public health: implementing ‘populationalized’ medicine

www.futuremedicine.com 813future science group

Pharmacogenomics & public health Review

813www.futuremedicine.com

public policy issues in promoting the development and commercialization of pharmacogenomic applications: challenges and implications. Drug Metab. Rev. 40, 377–401 (2008).

22 Gil JP. Amodiaquine pharmacogenetics. Pharmacogenomics 9(10), 1385–1390 (2008).

23 Hardy B, Séguin B, Singer PA, Mukerji M, Brahmachari SK, Daar AS. From diversity to delivery: the case of the Indian genome variation initiative. Nat. Rev. Genet. 9(Suppl. 1), S9–S14 (2008).

24 Hardy B, Séguin B, Ramesar R, Singer PA, Daar AS. South Africa: from species cradle to genomic applications. Nat. Rev. Genet. 9(Suppl. 1), S19–S23 (2008).

25 Kirkman M. Public health and the challenge of genomics. Aust. N. Z. J. Public Health 29(2), 163–165 (2005).

26 Kudzi W, Adjei O, Ofori­Adjei D, Dodoo AN. Pharmacogenetics in Ghana: reviewing the evidence. Ghana Med. J. 45(2), 73–80 (2011).

27 Oliver C, Williams­Jones B. Pharmacogenomic technologies: a necessary ‘luxury’ for better global public health? Global Health 7(1), 30 (2011).

28 Pang T. The impact of genomics on global health. Am. J. Public Health 92(7), 1077–1079 (2002).

29 Phillips KA, Veenstra D, Van Bebber S, Sakowski J. An introduction to cost–effectiveness and cost–benefit analysis of pharmacogenomics. Pharmacogenomics 4(3), 231–239 (2003).

30 Rai AK. Pharmacogenetic interventions, orphan drugs, and distributive justice: the role of cost–benefit analysis. Soc. Philos. Policy 19(27), 246–270 (2002).

31 Roederer MW, McLeod HL. Applying the genome to national drug formulary policy in the developing world. Pharmacogenomics 11(5), 633–636 (2010).

nn Outline of the Pharmacogenomics for Every Nation Initiative project, aiming to implement pharmacogenomics in resource-poor settings.

32 Roy PD, Majumder M, Roy B. Pharmacogenomics of anti­TB drugs­related

hepatotoxicity. Pharmacogenomics 9(3), 311–321 (2008).

33 Séguin B, Hardy B, Singer PA, Daar AS. Genomic medicine and developing countries: creating a room of their own. Nat. Rev. Genet. 9(6), 481–493 (2008).

34 Séguin B, Hardy B, Singer PA, Daar AS. Genomics, public health and developing countries: the case of the Mexican national institute of genomic medicine (INMEGEN). Nat. Rev. Genet. 9(Suppl. 1), S5–S9 (2008).

35 Séguin B, Hardy B, Singer PA, Daar AS. Universal healthcare, genomic medicine and Thailand: investing in today and tomorrow. Nat. Rev. Genet. 9(Suppl. 1), S14–S19 (2008).

36 Singer PA, Daar AS. Harnessing genomics and biotechnology to improve global health equity. Science 294, 87–89 (2001).

37 Smart A, Martin P. The promise of pharmacogenetics. Assessing the prospects for disease and patient stratification. Stud. Hist. Philos. Biol. Biomed. Sci. 37, 583–601 (2006).

38 Smith AC, Mugabe J, Singer PA, Daar AS. ‘Harnessing genomics to improve health in Africa’ – an executive course to support genomics policy. Health Res. Policy Syst. 3(1), 2 (2005).

39 Lee SS. Racializing drug design: implications of pharmacogenomics for health disparities. Am. J. Public Health 95(12), 2133–2138 (2005).

40 Mai Y, Koromila T, Sagia A et al. A critical view of the general public’s awareness and physicians’ opinions of the trends and potential pitfalls of genetic testing in Greece. Pers. Med. 8(5), 551–561 (2011).

n One of the first studies to assess the general public’s perception of pharmacogenomic testing.

41 Mitropoulos K, Innocenti I, van Schaik RH et al. Golden Helix Institute of Biomedical Research: interdisciplinary research and educational activities in pharmacogenomics and personalized medicine. Pharmacogenomics 13(4), 387–392 (2012).

42 Squassina A, Severino G, Grech G, Fenech A, Borg J, Patrinos GP. Golden Helix Pharmacogenomics Days: educational activities on pharmacogenomics and

personalized medicine. Pharmacogenomics 13(5), 525–528 (2012).

43 Georgitsi M, Viennas E, Gkantouna V et al. Population­specific documentation of pharmacogenomics markers and their allelic frequencies in FINDbase. Pharmacogenomics 12(1), 49–58 (2011).

n Websites101 Public health in an era of genome­based and

personalized medicine. PHG Foundation 2010. www.phgfoundation.org/reports/6617

102 NIH Office of Biotechnology Activities (US). Report of the Secretary’s Advisory Committee of Genetics, Health and Society. Revised edition. Department of Health and Human Services, Bethesda, MD, USA (2008). http://oba.od.nih.gov/oba/SACGHS/reports/SACGHS_PGx_report.pdf

103 PGENI: Pharamcogenetics for Every Nation Initiative. www.pgeni.org

104 HUGO: Pan­Asian Population Genomics Initiative. http://papgi.org/index.php/Main_Page

105 PAHO: Pan American Health Organization. http://new.paho.org

106 INMEGEN: National Institute of Genomic Medicine (Mexico). www.inmegen.gob.mx

107 TCELS: Thailand Center of Excellence for Life Sciences. www.tcels.or.th

108 Golden Helix Institute of Biomedical Research. www.goldenhelix.org

109 Survival International. Progress Can Kill. How Imposed Development Destroys the Health of Tribal Peoples. Woodman J, Grig S (Eds). Survival International, London, UK, 1–43 (2007). http://assets.survivalinternational.org/static/lib/downloads/source/progresscankill/short_report.pdf

110 FINDbase: Frequency of Inherited Disorders database. www.findbase.org