ArticlePDF Available

Perception of the Usefulness of Drug/Gene Pairs and Barriers for Pharmacogenomics in Latin America

Authors:

Abstract and Figures

Pharmacogenetics and Pharmacogenomics areas are currently emerging fields focused to manage pharmacotherapy that may prevent undertreatment while avoiding associated drug toxicity in patients. Large international differences in the awareness and in the use of pharmacogenomic testing are presumed, but not well assessed to date. In the present study we review the awareness of Latin American scientific community about pharmacogenomic testing and the perceived barriers for their clinical application. In order to that, we have compiled information from 9 countries of the region using a structured survey which is compared with surveys previously performed in USA and Spain. The most relevant group of barriers was related to the need for clear guidelines for the use of pharmacogenomics in clinical practice, followed by insufficient awareness about pharmacogenomics among clinicians and the absence of regulatory institutions that facilitate the use of pharmacogenetic tests. The higher ranked pairs were TPMT/thioguanine, TPMT/azathioprine, CYP2C9/warfarin, UGT1A1/irinotecan, CYP2D6/amitriptiline, CYP2C19/citalopram and CYP2D6/clozapine. The lower ranked pairs were SLCO1B1/simvastatin, CYP2D6/metoprolol and GP6D/chloroquine. Compared with USA and Spanish surveys, 25 pairs were of lower importance for Latin American respondents. Only CYP2C19/esomeprazole, CYP2C19/omeprazole, CYP2C19/celecoxib and G6PD/dapsone were ranked higher or similarly to the USA and Spanish surveys. Integration of pharmacogenomics in clinical practice needs training of healthcare professionals and citizens, but in addition legal and regulatory guidelines and safeguards will be needed. We propose that the approach offered by pharmacogenomics should be incorporated into the decision-making plans in Latin America.
Content may be subject to copyright.
Perception of the usefulness of drug/gene pairs and barriers for
pharmacogenomics in Latin America
Quiñones LA1, Lavanderos MA1, Cayún JP1, García-Martin E2, Agúndez JA3, Cáceres DD1,4, Roco AM1,5,
Morales JE6, Herrera L7, Encina G8, Isaza C9, Redal MA10, Laróvere LE11, Soria NW12, Eslava-Schmalbach
J13, Castañeda-Hernández G14, López-Cortés A15, Magno LA16, López M17, Chiurillo M18, Rodeiro I19, Castro
de Guerra D20, Terán E21, Estevez-Carrizo, F22, Lares-Assef I23.
1. Laboratory of Chemical Carcinogenesis and Pharmacogenetics, IFT, Molecular and Clinical
Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile.
2. Department of Biochemistry, University of Extremadura, Cáceres, Spain.
3. Department of Pharmacology, University of Extremadura, Cáceres, Spain.
4. School of Public Health, Faculty of Medicine, University of Chile, Santiago, Chile.
5. Servicio Metropolitano de Salud Occidente, Santiago, Chile.
6. Dr. Luis Calvo Mackenna Hospital, Santiago, Chile.
7. Human Genetic Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile.
8. Laboratorio de Oncología y Genética Molecular, Clínica Las Condes, Santiago, Chile.
9. Facultades de medicina de las universidades Tecnológica de Pereira y Autónoma de las Américas, Pereira,
Colombia.
10. Unidad de Medicina Molecular y Genómica-Hospital Italiano de Buenos Aires-CP1199, Argentina
11. CEMECO, Hospital de Niño, FCM, Universidad Nacional de Córdoba, Argentina.
12. Facultad de Ciencias Químicas - Universidad Católica de Córdoba - Campus Universitario, Córdoba -
Argentina.
13. Universidad Nacional de Colombia, Ciudad Universitaria, Facultad de Medicina, Bogotá, Colombia.
14. Centro de Investigación y de Estudios Avanzados del IPN, México, D.F., Mexico.
15. Instituto de Investigaciones Biomédicas. Universidad de las Américas, Quito, Ecuador.
16. INCT de Medicina Molecular, Faculdade de Medicina, Universidade Federal de Minas Gerais, Brazil.
17. Departamento de Sistemas Biológicos, División Ciencias Biológicas y de la Salud, Universidad Aunoma
Metropolitana-Xochimilco, México.
18. Universidad Centroccidental Lisandro Alvarado, Decanato de Ciencias de la Salud, Barquisimeto,
Venezuela.
19. Centro de Bioproductos Marinos (CEBIMAR), La Habana, Cuba.
20. Instituto Venezolano de Investigaciones Científicas, Centro de Medicina Experimental, Laboratorio de
genética Humana, Caracas, Venezuela.
21. Colegio de Ciencias de la Salud, Universidad San Francisco de Quito. Quito, Ecuador.
22. Center for Clinical Pharmacology Research, Italian Hospital. pc 11600 Montevideo. Uruguay
23. Instituto Politécnico Nacional, CIIDIR. Academia de Farmacogenómica y Biomedicina Molecular.
Durango, México.
Running Title: Drug/gene pairs and barriers for pharmacogenomics in Latin America
Key Words: biomarkers, adverse drug reactions, pharmacogenomics, clinical recommendations,
clinical relevance.
Corresponding author: Dr. Luis Quiñones S., Laboratory of Chemical Carcinogenesis and
Pharmacogenetics, IFT, Molecular and Clinical Pharmacology Program, ICBM, Faculty of Medicine,
University of Chile. E-mail: lquinone@med.uchile.cl, Phone number: 56-2-6817756, Fax number: 56-2-
6822406, P.O. Box 70111. Carlos Schachtebeck 299, Quinta Normal, Santiago, Chile.
ABSTRACT
Pharmacogenetics and Pharmacogenomics areas are currently emerging fields focused to manage
pharmacotherapy that may prevent undertreatment wh ile avoiding associated drug toxicity in patients. Large
international differences in the awareness and in the use of pharmacogenomic testing are presumed, but not
well assessed to date. In the pr esent study we review the awareness of Latin American scientific community
about pharmacogenomic testing and the perceived barriers for their clinical application. In order to that, we
have compiled information from 9 countries of the region using a structured survey which is compared with
surveys previously performed in USA and Spain.
The most relevant group of barriers was related to the need for clear guidelines for the use of
pharmacogenomics in clinical practice, followed by insufficient awareness about pharmacogenomics among
clinicians and the absence of regulatory institutions that facilitate the use of pharmacogenetic tests.
The higher ranked pairs were TPMT/thioguanine, TPMT/azathioprine, CYP2C9/warfarin,
UGT1A1/irinotecan, CYP2D6/amitriptiline, CYP2C19/citalopram and CYP2D6/clozapine. The lower ranked
pairs were SLCO1B1/simvastatin, CYP2D6/metoprolol and GP6D/chloroquine. Compared with USA and
Spanish surveys, 25 pairs were of lower importance for Latin American respondents. Only
CYP2C19/esomeprazole, CYP2C19/omeprazole, CYP2C19/celecoxib and G6PD/dapsone were ranked higher
or similarly to the USA and Spanish surveys.
Integration of pharmacogenomics in clinical practice needs training of healthcare professionals and
citizens, but in addition legal and regulatory guidelines and safeguards will be needed. We propose that the
approach offered by pharmacogenomics should be incorporated into the decision-making plans in Latin
America.
RATIONALE FOR THIS STUDY
It is well known that the efficacy and safety of drug therapy show substantial inter-individual
variability which is based on genetic variations affecting pharmacokinetic and/or pharmacodynamic factors
[1]. But it is also known that there are non-genetic factors affecting drug response, for example age, sex,
organ function, concomitant therapies, drug interactions, evolution of disease, nutritional factors, smoking,
alcohol, the presence of virus, among others. We see that the ineffectiveness or toxicity of drug therapy is due
to the interaction of genes with environmental factors. A drug that is well tolerated and causes a strong
response in some patients may be ineffective, toxic or may cause adverse drug reactions in other patients. In
fact, it has been reported that 1 in 15 hospital admissions in the United Kingdom are due to adverse drug
reactions [2] and that adverse drug effects in hospitalized patients are the fifth leading cause of death in the
United States [3]. It has been reported that approximately 2 million adverse drug reactions lead to a spending
of U$100 billion annually [4].
Variability in drug metabolism and response is not a new idea. In 1892 Sir William Osler pointed out
“If it were not for the great variability among individuals, medicine might as well be a science, not an art" and
in 1895 Dr. Claude Bernard said “the excessive complexity of the physiological processes and organisms
wher e are observed commands respect by saying that no two patients are alike”. Both ancient sentences
reinforce the idea that it is necessary not to treat illnesses but ill people.
In this sense, it is known that pharmacokinetic factors affecting absorption, distribution,
biotransformation and excretion influence the plasma and tissue concentration reached by drugs. Therefore,
polymorphism of genes encoding drug transporters, biotransformation enzymes and drug targets will
influence drug efficacy and safety. Accordingly, the current practices for the dosing of therapeutic agents
should be improved through the understanding of gene variation associated with “drug life” inside the body.
Therefore, in order to be able to predict patient’s predispositions to treatment complications and poor outcome
it is essential to examine all candidate loci influencing response to drugs. We should also investigate
metabolic pathways for activation or inactivation of drugs, the interaction between drugs, age and gender
sensitivities, the impact of ethnicity and environmental factors to understand the individual and population
variability in drug response.
Pharmacogenetics and Pharmacogenomics areas are currently emerging fields focused to manage
pharmacotherapy that may prevent undertreatment while avoiding associated drug toxicity in patients. Large
international differences in the awareness and in the use of pharmacogenomic testing are presumed, but not
well assessed to date. In the present study we review the awareness of Latin American scientific community
about pharmacogenomic testing and the perceived barriers for their clinical application. A major focus of
current research entails clinical evaluation of polymorphisms on their impact on gene function.
There has been an increase in the number of research articles and clinical trials of
pharmacogenomics/pharmacogenetics studies since 1961, just after the German pharmacologist Friedrich
Vogel (1959) [5] coined the term pharmacogenetics. As it is observed in Figure 1 from Vogel’s definition the
number of publications has constantly increased, especially in the last 15 years, concomitantly the
development of pharmacogenomics has evolved.
While the most conservative use of pharmacogenomics aims to stratify patient populations into poor,
extensive, intermediate and rapid/ultrarapid metabolizers, which leads to a selection criteria for those who
should or should not receive a given drug [6], other researchers promote guidelines intended to adjust drug
dosage based on pharmacogenomics tests [7]. Of course, in both cases, pharmacogenomic testing could be
more useful in outlier patients.
In the present work we review and analyze the knowledge of Latin American scientific and clinical
community about pharmacogenomic testing and the barriers for their clinical application. In order to that, we
have compiled the information of a number of countries of the region (Figure 2) using a structured survey
which is compared to the USA and Spanish surveys previously performed [8, 9].
The survey was structurated into two items, first, a list of 15 potential barriers to the clinical
application of biomarkers were evaluated in terms of their importance on the scale of 1 to 10 (10 being the
highest) (Figure 3). Second, a list of 51 gene/drug pairs were evaluated on the scale of 1-5 (5 being the
highest) to ascribe association between biomarkers and their response to genomic medicine (Figure 4).
Using the Scopus database and several key words related to pharmacogenics/pharmacogenetics we
searched for potential respondents of the survey in Latin America. This search was based on papers published
between 1990 to 2013. The used keywords were: polymorphisms, pharmacogenetics, pharmacogenomics,
biomarkers, adverse drug reactions, clinical pharmacology. We searched for clinicians and biomedical
researchers from all Latin American countries. Our search yielded 44 potential respondents from 13 countries.
We did not find any potential respondent from Antigua and Barbuda, Belize, Bolivia, Dominican Republic, El
Salvador, Guiana, Guatemala, Nicaragua, Panama, Paraguay, Peru, Suriname and Trinidad and Tobago, and
even though we contact researchers from Bolivia, Costa Rica, Haiti and Honduras they did not respondr the
survey. Therefore, the results were obtained from 20 respondents from 9 countries, Argentina, Brazil, Chile,
Colombia, Cuba, Ecuador, Mexico, Uruguay and Venezuela. The response rate was 45.45%.
The profile of the participants was as follows: 60% medical doctors, 20% biochemists, and a
miscellaneous group comprising one pharmacist, one veterinary doctor, one biologist and one anthropologist,
all of them working in the field of pharmacology. 20% of the participants were affiliated to clinical centers,
45% were affiliated to universities, 30% were affiliated to clinical centers associated with academic activities
in universities and 15% were affiliated to research centers. We were not able to find any participants affiliated
to the pharmaceutical industry.
Figure 3 summarizes the results about the perceived importance of barriers for implementing the use
of pharmacogenomics testing in clinical practice. Our results showed three major groups of barriers. The most
commom group was related to the need for clear guidelines for the use of pharmacogenomics in clinical
practice (8.76 points on a scale of 1 to 10). The second was the insufficient awareness about
pharmacogenomics among clinicians (8.52) and the third group was the absence of a regulatory institution
that facilitates the use of pharmacogenetic tests (8.47). As expected, all three barriers are closely related.
(8.33 and 8.14, respectively). We believe that these are the barriers specific to Latin America, taking account
that in the Spanish survey these are lower ranked barriers. Moreover, both, Spanish and Latin America
surveys consider that ethical, legal and social implication are not important barriers as they were evaluated
with the lowest importance. We believe that the insufficient awareness of the clinicians leads to the lack of
regulatory institutions and guidelines.
Because most of the main barriers were related to lack of clinical guidelines and protocols, we
included in our survey the same 29 gene/drug pairings a criteria listed in two previous studies [8,9] which
were obtained from members of the Spanish Societies of Pharmacology and Clinical Pharmacology, the
Clinical Pharmacogenetics Implementation Consortium (CPIC; see http://www.pharmgkb.org/page/cpic) and
members of the American Society for Clinical Pharmacology and Therapeutics. This was in order to obtain
comparative results. However we also include 22 additional gene/drug pairs selected from the recently
published list of pharmacogenomic biomarkers by FDA (2013) [10] (Figure 4).
Data related to the percentages of respondents (frequencies) shown in figure 4 were defined as
respondents who ranked the gene/drug pairs as 3, 4 or 5 in relation to the total responses (on a scale of 1–5)
are plotted along the y-axis. We used scale of 1 to 5 for gene/drug pair evaluation to make results comparable
to those from the Spanish and US survey [8,9].
Based on the survey results, the perceived importance of the data linking the drug to the gene
variation the higher ranked pairs were TPMT/thioguanine, TPMT/azathioprine and CYP2C9/warfarin, with a
very close ranking for UGT1A1/irinotecan (Figure 4A and 4B). From the additional 22 gene/drug pairs
CYP2D6/amitriptiline, CYP2C19/citalopram and CYP2D6/clozapine wer e the higher ranked pairs (Figure
4C), wh ile the lower ranked pairs were SLCO1B1/simvastatin, CYP2D6/metoprolol and GP6D/chloroquine.
In comparison with USA and Spanish surveys from the 29 pairs, 25 were of lower importance to the Latin
American and the Caribbean respondents. Only CYP2C19/esomeprazole, CYP2C19/omeprazole,
CYP2C19/celecoxib and G6PD/dapsone were ranked higher or similar to the USA and Spanish surveys. We
believe this is mainly due to insufficient scientific information available in our countries giving rise to an
underestimation of the importance of gene/drug pairs. The higher ranked pairs in the US study were
CYP2C9/warfarin, UGT1A1/irinotecan, VKORC1/warfarin, and for the Spanish study were HLA-B/abacavir,
UGT1A1/irinotecan and CYP2D6/tamoxifen. Therefore, UGT1A1/irinotecan seems to be very important in
all analyzed countries and the importance of CYP2C9/warfarin seems to be ratified. On the other hand
CYP2D6/amitriptiline, CYP2C19/citalopram and CYP2D6/clozapine, the higher ranked additional pairs are
related to psychiatric drugs giving rise to the idea that in the Latin American countries the variability in the
response to these drugs (e.g. antidepressants) is fairly important. Dramatically lower ranked pairs in this study
(LAC) in comparison with the US and Spain studies (less than half the importance in the evaluation) were
SLCO1B1/simvastatin and DPYD/5-fluorouracil (gene/drug pairs). We have no an explanation for this result
and neither for the low(er) ranking of SLCO1B1/simvastatin, CYP2D6/metoprolol and GP6D/chloroquine in
this study.
Some limitations of this survey must be pointed. First, the poor developed pharmacogenomic research in this
region does not allow us to have a large number of participants, thus not all Latin American countries
participated in the survey. This can question the representativeness of the survey. However, we are confident
enough that the study participants from each country are the most knowledgeable people in the field of
pharmacogenomics. Second, the importance of the gene/drug pairings in different countries could be
evaluated differently due to the absence of some drugs in the market according to drug acquisition policies of
each Ministry of Health.
DICUSSION AND FUTURE PERSPECTIVES
Since the completion of the human genome project and its potential ability to change the practice of
medicine, great expectations and enthusiasm regarding possible applications were positioned in the scientific
community. Together, millions of SNPs have been identified [11] and the effects of each specific SNPs are
still under study [12]. However, human genome sequence defined how similar people are (99.9%) and not
how different they are. Thus, actually many researchers believe that pharmacogenomics can be one of the first
successes in the study of individual differences, at least in relation to drug response.
The ultimate goal of pharmacogenetic research is to predict individual’s responses to drug therapy
and subsequently to adapt the therapeutic strategy. In this regard, it is estimated that gene polymorphisms
account for 20 to 95% of the variability in therapeutic response and toxicity [13]. Of all the known drugs
involved in adverse reactions about 80% are metabolized by polymorphic enzymes [14]. Since 2004, several
drugs refer to this study in the labeling information, some of them considered sufficient to guide decision-
treatment decisions [15]. In 2005 the FDA issued a guidance document for the industry about the referral data
for genotyping drug metabolizing enzymes. Some authors estimate that in the next 5-10 years, 10 to 20% of
new drugs approved will include genetic study. In 2005 the FDA approved the marketing of the first
laboratory test system based on cytochrome P450 genotypes [16], which allows the use of genetic information
to select appropriate doses of drugs and drugs for a wide variety of common conditions. However, in Latin
America the test seems to have suboptimal results, which could be due to ethnic differences between Latin
Americans and other human populations.
Currently, the FDA recommends more than 100 drugs for pharmacogenomic monitoring to improve
prescription dosage including antivirals, antibiotics, psychiatry drugs, analgesic and anticancer agents (FDA,
2013)[10]. In some cases this information has been incorporated into the dataset long after the drug was
approved by regulatory agencies. In other cases, pharmacogenetic data has been obtained during the process
of drug development and has been taken into account for approval.
Therefore, the current challenge for personalized therapy is to define genetic profiles to predict the
response to drugs and the progression of the diseases [9, 17, 18, 19, 20, 21]. Information to address this
challenge can only be obtained from case-control and prospective studies with a pharmacogenomics basis.
Despite the enormous amount of known information about the genetic basis of variable response to
drugs, it has little influence on its application to the current clinical practice. Thus, acceptance of
pharmacogenomic studies in medical practice is gradual. Several issues have prevented its rapid
implementation, such as, a) lack of readily available clinical laboratories which can perform these tests
quickly and cost-effectively, b) shortage of health care professionals who can interpret the test data and
associated clinical pharmacology and c) doubts whether insurance companies will pay for thie study. In
addition, many ethical questions pose continuing challenges. However, the number of drugs approved with a
reference to the genetic study in labeling information is increasing.
Of course pharmacogenomics has several limitations to its application in clinical practice which
should be addressed, some of them have been analyzed previously by Agúndez et al [20]. These limitations
include the lack of sufficient evidence for cost–efficiency, the need for the identification of new biomarkers
for drug toxicity and response, technical limitations and ethnicity questions. Together, we know that inter-
individual variability to drug response exists, even in individuals with identical pharmacogenomic profile,
giving rise to the idea that pharmacogenomics is only one of the several factors to be considered in dose
adjustment. Therefore algorithms including anthropometric, lifestyle and environmental factors appear to be
the best approach.
Another restriction for the use of pharmacogenomics is the poor information about pharmagenes in
Latin America populations, which prevents direct extrapolation of the dosage of drugs with clinical studies
performed in other ethnic groups. Since profound variation in the effect of drugs have been described to be
associated to the genetic polymorphisms in diverse populations, ethnicity appears to be an important issue in
Latin America. In this sense, in order to have a first approach, particularly in American Hispanic populations,
we have previously discussed the implications of interethnic and intraethnic genetic variability [22, 23, 24, 25,
26]. In this respect, it is clear that there is a need for developing more and well designed studies in Latin
Americanpopulations to better address the issue that the introduction of pharmacogenomics in clinical
practice. These studies should include ethnic comparison of pharmacogenomic profiles, the impact of
polymorphism on phenotype, gene expression and regulation, metabolic profiles of patients with a given drug
and relevant environmental factors that influence drug response.
Clinical practice guidelines and protocols may help to overtake the major groups of barriers shown in
Figure 3 and, in consequence, this will hopefully lessen the impact of the first ranked and the most
determinant barrier. Similarly, we believe that governmental support and promotion for the use of
pharmacogenomics biomarkers in the countries of this region will greatly influence the relevance of the other
barriers.
The healthcare professionals (prescribers, insurers and regulators) will want to know if there is a
substantial impact of pharmacogenomics on the safety and efficacy of the drug on an individual. Of course,
before use in the clinical routine selected pharmagenes must demonstrate, in retrospective and prospective
studies, a value sufficient to have good cost-effectiveness.
Pharmacology of the future intends to conduct individualized pharmacotherapeutic treatment for the
manifestation of a disease and the appropriate dose for the therapeutic effect in a given patient, minimizing
the risk of adverse reactions. Therefore the main idea is the accomplishment of the five “R” for drug therapy
“the Right dose of the Right drug for the Right indication in the Right patient at the Right time”. For instance,
nowadays the individualized treatments are a pressing need. The current formula of standard
pharmacotherapy is not ideal according to the great variability between patients.
The rapidly evolving field of pharmacogenetics holds great promise for assisting the selection of
patient-individualized treatment regimens and dosages. A vast number of single nucleotide polymorphisms
have been discovered in genes thought to be involved in the regulation of drug metabolism; however,
relatively few studies have been conducted that establish a link between genotype, efficacy and safety of
drugs.
In short, integration of pharmacogenomics in clinical practice needs training of healthcare
professionals and citizens, moreover legal and regulatory guidelines and safeguards will be needed. The
answers to the question of which patient should receive which drug and dose will be not easy, but we believe
that the approach offered by pharmacogenomics should be incorporated into the decision-making process. A
more rational use of expensive treatment drugs together with actions to minimize patient toxic events and its
consequences, would dramatically reduce medical costs, as an added benefit.
Financial Support:
The work in the author’s laboratory have been financed by Grants FONDECYT 3020043, DI, U. de Chile nº
1102-002, Chile; PS09/00943, PS09/00469, PI12/00241, PI12/00324 and RETICS RD12/0013/0002 from
Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Spain, and GR10068 from Junta de
Extremadura, Spain. Financed in part with FEDER funds from the European Union.
Acknowledgements: The authors wish to thank to Dr. Marcia Llacuachaqui, from the Women’s College
Research Institute, Women's College Research Hospital , Canada, for her assistance in language editing.
REFERENCES
[1] Evans WE. Pharmacogenomics: marshalling the human genome to individualise drug therapy.
Gut 2003, 52 Suppl 2, 10-8.
[2] Pirmohamed M, James S, Meakin S, Green C, Scott AK, Walley TJ, Farrar K, Park BK,
Breckenridge AM. Adverse drug reactions as cause of admission to hospital: prospective analysis of
18 820 patients. BMJ 2004, 329(7456), 15-9.
[3] Mancinelli L, Cronin M, Sadee W. Pharmacogenomics: the promise of personalized medicine.
AAPS Pharm. Sci. 2000, 2: E4.
[4] Ross CJ, Visscher H, Rassekh SR, Castro-Pastrana LI, Shereck E, Carleton B, Hayden MR.
“Pharmacogenomics of serious adverse drug reactions in pediatric oncology”, J. Popul. Ther. Clin.
Pharmacol. 2011, 18: 134-151.
[5] Vogel F. Moderne Problem der humangenetik. Ergebn. Inn. Med. Kinderheilkd 1959, 12, 52
125
[6] Green ED, Guyer MS. Charting a course for genomic medicine from base pairs to bedside.
Nature 2011, 10, 470 (7333):204-213.
[7] Swen JJ, Nijenhuis M, de Boer A, Grandia L, Maitland-van der Zee AH, Mulder H, Rongen GA,
van Schaik RH, Schalekamp T, Touw DJ, van der Weide J, Wilffert B, Deneer VH, Guchelaar HJ.
Pharmacogenetics: from bench to byte--an update of guidelines. Clin. Pharmacol. Ther. 2011, 89(5),
662-73.
[8] Relling MV and Klein TE. CPIC: Clinical Pharmacogenetics Implementation Consortium of the
Pharmacogenomics Research Network. Clin. Pharmacol. Ther. 2011, 89, 464–467.
[9] Agúndez JA, Abad-Santos F, Aldea A, Alonso-Navarro H, Bernal ML, Borobia AM, Borrás E,
Carballo M, Carvajal A, García-Muñiz JD, Gervasini G, Jiménez-Jiménez FJ, Lucena MI, Martínez
C, Sacristán JA, Salado I, Sinués B, Vicente J, García-Martín E. Toward a clinical practice guide in
pharmacogenomics testing for functional polymorphisms of drug-metabolizing enzymes. Gene/drug
pairs and barriers perceived in Spain. Front. Genet. 2012a, 3:273.
[10] Food and Drug Administration-USA (FDA). Table of Pharmacogenomic Biomarkers in Drug
Labels. Available on
http://www.fda.gov/drugs/scienceresearch/researchareas/pharmacogenetics/ucm083378.htm
(accessed: July 30, 2013).
[11] SNP Single Nucleotide Polymorphism consortium, http://snp.cshl.org/ (accessed: July 23,
2013).
[12] GWAS. Genome Wide Association Studies. 2008. http://gwas.nih.gov/ (accessed: July 23,
2013)
[13] Evans WE, Mcleod HL. Pharmacogenomics-drug disposition, drug targets, and side effects. N.
Engl. J. Med. 2003, 348, 538-549.
[14] Phillips KA, Veenstra DL, Oren E, Lee JK, Sadee W. Potential role of pharmacogenomics in
reducing adverse drug reactions: a systematic review. JAMA 2001, 286(18), 2270-9.
[15] Zineh I, Gerhard T, Aquilante CL, et al. Availability of pharmacogenomics-based prescribing
information in drug package inserts for currently approved drugs. Pharmacogenomics J. 2004, 4(6),
354-8.
[16] AmpliChip CYP450 Test. The Medical Letter Inc. Med. Lett. Drugs. Ther. 2005; 47: 71–2.
[17] Gardiner SJ, Begg EJ. Pharmacogenetics, drug-metabolizing enzymes, and clinical practice.
Pharmacol. Rev. 2006, 58, 521-90.
[18] Huang SM, Goodsaid F, Rahman A, Frueh F. Application of pharmacogenetics in clinical
pharmacology. Toxicol. Mechanisms and Methods 2006, 16, 89-99.
[19] Gurwitz D, Lunshof JE. Personalized pharmacotherapy: genotypes, biomarkers, and beyond.
Clin. Pharmacol. Ther. 2009, 85(2), 142.
[20] Agúndez JA, Del Barrio J, Padró T, Stephens C, Farré M, Andrade RJ, Badimon L, García-
Martín E, Vilahur G, Lucena MI. Trends in qualifying biomarkers in drug safety. Consensus of the
2011 meeting of the Spanish Society of Clinical Pharmacology, Front. Pharmacol. 2012b, 3:2.
[21] Gladding PA. Clinical applications of pharmacogenetics: Present and near future. Cleve Clin. J.
Med. 2013, 80(8):477-82.
[22] Garcia-Martin E, Martinez C, Ladero JM and Agúndez JA. Interethnic and intraethnic
variability of CYP2C8 and CYP2C9 polymorphisms in healthy individuals. Mol. Diagn. Ther. 2006,
10, 29–40.
[23] Garcia-Martin E, 2008
[24] Borobia AM, López-Parra AM, Tabarés B, Ramirez E, Baeza C, Arroyo-Pardo E, Carcas AJ.
CYP2C9 polymorphism in five autochthonous population of the same geographic area (Spanish
Pyrenees). Pharmacol. Res. 2009, 59, 107–111.
[25] Restrepo JG, Martínez C, García-Agúndez A, Gaviria E, Laguna JJ, García-Martín E, Agúndez
JA. Cytochrome P450 CYP2B6 genotypes and haplotypes in a Colombian population: identification
of novel variant CYP2B6 alleles. Pharmacogenet. Genomics 2011, 21, 773–778.
[26] Roco A, Quiñones L, Agúndez JA, García-Martín E, Squicciarini V, Miranda C, Garay J, Farfán
N, Saavedra I, Cáceres D, Ibarra C, Varela N. Allele frequencies of 23 functionally significant
variant alleles related with metabolism of antineoplastic drugs in the Chilean population: comparison
with Caucasian and Asian populations. Front. Genetics 2012, 3(art 229): 1 – 9.
[27] ClinicalTrials.gov. Bethesda MD: National Library of Medicine. Available at:
http://clinicaltrials.gov. Accessed: July 3, 2013.
Conflict of Interest Statement: The authors declare that do not have any potential conflict of interest.
FIGURE 1: Variation in number of publications [Scopus] and clinical trials [27] including pharmacogenomics/pharmacogenetics studies from
1961.
12410 8 8 30 51 30 39 43 31 57 75 58 45 48 82 64 51 38 62 58 54 59 93 70 43 69 55 56 60 61 55 76 71 96 148
262
445
637
846
1045
1154
1427
1521
1608
1891
1768
1928
1890
1985
1961
1962
1965
1968
1969
1970
1971
1973
1974
1975
1977
1978
1979
1982
1983
1984
1985
1988
1991
1992
1993
1994
1996
1997
1998
2000
2001
2002
2005
2006
2007
2008
2010
2011
Publications
22487
70 61 64
95 95
123
92 83
Clinical trials
Figure 2: Latin American countries participating in the survey (in gray).
Figure 3. Highest-ranking barriers for implementing the use of pharmacogenomics testing, based on a survey in Latin American Scientific and
Clinical Researchers. Data related to average importance on a scale of 1 (low) to 10 (high) + standard deviation are plotted along the X-axis.
Figure 4. Highest- ranking gene/drug pairs, based on the survey of Latin American scientific and
clinical community, compared to the published survey of Spanish Societies for Pharmacology
and Clinical Pharmacology members in 2012 and the American Society for Clinical Pharmacology
and Therapeutics (ASCPT) members conducted by CPIC. Data related to the percentages of
respondents who ranked the gene/drug pairs as 3, 4, and 5 in relation to the total evaluations
(on a scale of 1(low)–5(high)) are plotted along the Y-axis.
A
B
Percentage of Respondents
C
36
27
13
93
46 46 50
20
36
70 72
80
92
59
41 46
35
82
69
57 59
26
39
69
83
65
90 89
40
19 25
39 33 39 43
25
15
47 43 50 46 43
40
50
27
36
54
67
54
42 42 38
25
67
82
63 63
25
79
21 28 25
90
65
42
55
43
27
42
87
57
48
23
39
29 32 27
47 42
29 31 35
17
30
42
25
35
Span ish surv ey ASCPT survey LAC survey
42
10 13
21 25
31
42
29
25
29
15 19
41
10
32 28
23 23 23
38
27
20
... Advances in high-throughput technologies, particularly in large-scale DNA sequencing, have improved our understanding of the molecular mechanisms underlying gastric cancer. This progress has been instrumental in identifying cancer driver genes Lawrence et al., 2014), germline mutations (Lu et al., 2015), cancer driver variants in both coding and non-coding regions (Sjöblom et al., 2006;Tamborero et al., 2013;Porta-Pardo et al., 2017;Rheinbay et al., 2020), druggable proteins (Rubio-Perez et al., 2015), drug resistance mechanisms (Vasan et al., 2019), pharmacogenomics (PGx) clinical guidelines (Quinones et al., 2014;López-Cortés et al., 2020c;Varela et al., 2021), and the development of artificial intelligence algorithms (López-Cortés et al., 2020a;Jumper et al., 2021). ...
... In recent years, it has become evident that patients with the same cancer type do not uniformly respond to standard treatments (Raguz and Yagüe, 2008;Mansoori et al., 2017). Precision oncology addresses this variability by providing personalized treatment options, including appropriate medications and dosages, considering individual patient needs, their ethnicity, and treatment timing (Garraway et al., 2013;Quinones et al., 2014;Pérez-Villa et al., 2023). Therefore, identifying actionable genomic alterations is a primary goal of cancer research, particularly in the driver gene landscape of gastric cancer, to devise effective therapeutic strategies and PGx clinical guidelines (ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium, 2020). ...
... These challenges encompass the formulation of extensive PGx clinical guidelines to guide treatment decisions across a culturally and genetically varied patient base. Moreover, there is a pressing need for rigorous cost-effectiveness analysis, suitable regulatory frameworks, and increased gene/drug trials (Quinones et al., 2014;López-Cortés et al., 2017;Salas-Hernández et al., 2023). ...
Article
Full-text available
Introduction: Gastric cancer is one of the most prevalent types of cancer worldwide. The World Health Organization (WHO), the International Agency for Research on Cancer (IARC), and the Global Cancer Statistics (GLOBOCAN) reported an age standardized global incidence rate of 9.2 per 100,000 individuals for gastric cancer in 2022, with a mortality rate of 6.1. Despite considerable progress in precision oncology through the efforts of international consortia, understanding the genomic features and their influence on the effectiveness of anti-cancer treatments across diverse ethnic groups remains essential. Methods: Our study aimed to address this need by conducting integrated in silico analyses to identify actionable genomic alterations in gastric cancer driver genes, assess their impact using deleteriousness scores, and determine allele frequencies across nine global populations: European Finnish, European non-Finnish, Latino, East Asian, South Asian, African, Middle Eastern, Ashkenazi Jewish, and Amish. Furthermore, our goal was to prioritize targeted therapeutic strategies based on pharmacogenomics clinical guidelines, in silico drug prescriptions, and clinical trial data. Results: Our comprehensive analysis examined 275,634 variants within 60 gastric cancer driver genes from 730,947 exome sequences and 76,215 whole-genome sequences from unrelated individuals, identifying 13,542 annotated and predicted oncogenic variants. We prioritized the most prevalent and deleterious oncogenic variants for subsequent pharmacogenomics testing. Additionally, we discovered actionable genomic alterations in the ARID1A, ATM, BCOR, ERBB2, ERBB3, CDKN2A, KIT, PIK3CA, PTEN, NTRK3, TP53, and CDKN2A genes that could enhance the efficacy of anti-cancer therapies, as suggested by in silico drug prescription analyses, reviews of current pharmacogenomics clinical guidelines, and evaluations of phase III and IV clinical trials targeting gastric cancer driver proteins. Discussion: These findings underline the urgency of consolidating efforts to devise effective prevention measures, invest in genomic profiling for underrepresented populations, and ensure the inclusion of ethnic minorities in future clinical trials and cancer research in developed countries.
... Finally, there is lethargy in obtaining laboratory accreditation for genetic testing in countries that offer direct-to-customer testing. This stems from the absence of national guidelines for genetic testing in most SSA countries and other less developed regions [17]. ...
... The successful implementation of PGx testing will require acceptance and adequate knowledge of PGx by health-care workers, especially physicians. Nevertheless, competency on PGx testing amongst clinicians in African populations is lacking, which has also been reported as one of the major barriers for implementing PGx in other under-resourced clinical settings such as in Latin America [17,74]. The lack of competency stems from the absence of or limited PGx training programs in health-care training institutions and universities in Africa. ...
... Most countries, particularly in SSA and other less developed regions [17], lack specific and clear regulatory policies for the implementation of genetic testing, and in particular PGx testing. For instance, only South Africa, Nigeria, and Malawi amongst SSA countries provide clear and specific guidelines for genetic testing and research [78]. ...
Article
Full-text available
Clinical research in high-income countries is increasingly demonstrating the cost- effectiveness of clinical pharmacogenetic (PGx) testing in reducing the incidence of adverse drug reactions and improving overall patient care. Medications are prescribed based on an individual’s genotype (pharmacogenes), which underlies a specific phenotypic drug response. The advent of cost-effective high-throughput genotyping techniques coupled with the existence of Clinical Pharmacogenetics Implementation Consortium (CPIC) dosing guidelines for pharmacogenetic “actionable variants” have increased the clinical applicability of PGx testing. The implementation of clinical PGx testing in sub-Saharan African (SSA) countries can significantly improve health care delivery, considering the high incidence of communicable diseases, the increasing incidence of non-communicable diseases, and the high degree of genetic diversity in these populations. However, the implementation of PGx testing has been sluggish in SSA, prompting this review, the aim of which is to document the existing barriers. These include under-resourced clinical care logistics, a paucity of pharmacogenetics clinical trials, scientific and technical barriers to genotyping pharmacogene variants, and socio-cultural as well as ethical issues regarding health-care stakeholders, among other barriers. Investing in large-scale SSA PGx research and governance, establishing biobanks/bio-databases coupled with clinical electronic health systems, and encouraging the uptake of PGx knowledge by health-care stakeholders, will ensure the successful implementation of pharmacogenetically guided treatment in SSA.
... Over the past years it has become clear that oncological patients, diagnosed with the same cancer type, may have different responses to generic treatments such as radiation or chemotherapy. To overcome these variable responses, cancer precision medicine aims to provide the right dose of the right drug for the right patient at the right time (Quinones et al., 2014). Thus, precision medicine has become an important tool in cancer treatment; it allows the identification of specific mutations in driver genes responsible for tumor progression (ICGC/ TCGA Pan-Cancer Analysis of Whole Genomes Consortium, 2020). ...
... On the one hand, the most relevant cancer genome projects worldwide, such as TCGA(The Cancer Genome Atlas Research Network, 2013), TARGET or PCAWGC (ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium, 2020), are overrepresented by Caucasian individuals (91.1%), and do not include enough individuals from minority populations (Guerrero et al., 2018). On the other hand, developing regions lack of investment in cancer genomics tests, have fragmented healthcare systems, and have insufficient characterization of pharmacogenetics variability in their populations (Quinones et al., 2014). Therefore, in this study we proposed a new insight for identification of the most frequent oncogenic variants in the Latino, African, Ashkenazi Jewish, East Asian, South Asian, European Finnish, and European non-Finnish populations in order to focus economic resources on analyzing the most frequent and relevant molecular targets. ...
Book
Full-text available
Since the pharmacogenetics and pharmacogenomics (PGx) field started to rise, the information about the relationship between actionable genes, genotypes, and response to drugs has increased exponentially (Nicholson et al., 2021). There is evidence of the utility and impact of genetics in the choice of therapeutic regimens improving their effectiveness and safety (Arbitrio et al., 2021). Even some international efforts have created clinical guidelines that allow to implementation of pharmacogenomics in daily clinical practice. In addition to clinical outcomes, Economic benefits have been associated with the translation from “the bench to the bedside”. Moreover, several major PGx expert organizations such as the Clinical Pharmacogenetics Implementation Consortium (CPIC, 2021) and the Dutch Pharmacogenetics Working Group (DPWG), provide gene-drug guidelines for actionable variants. In addition, Ubiquitous Pharmacogenomics (U-PGx, 2021), the Latin American Network for the Implementation and Validation of Pharmacogenomics Guidelines (RELIVAF-CYTED, 2021), and the Southeast Asian Pharmacogenomics Research Network (SEAPharm, Chumnumwat et al., 2019) have investigated pharmacotherapeutic recommendations guided by pharmacogenetics. In this respect, based on scientific evidence the Food and Drug Administration (FDA) has published a list of PGx biomarkers for drug labelling (FDA, 2021).
... Unfortunately, the clinical application of this knowledge has progressed very slowly. A variety of reasons have been identified to contribute to this latency in clinical implementation [15,[19][20][21][22]. Arguably among the most important factors is the substantial amount of underpowered preliminary studies that report interactions between specific variants and pharmacological phenotypes without replication in independent cohorts or corroborative mechanistic data [23]. ...
... All over the world, several issues have prevented rapid implementation of pharmacogenomics, such as (a) lack of readily available clinical laboratories which can perform these tests quickly and cost-effectively, (b) shortage of healthcare professionals who can interpret the test data and associated clinical pharmacology, and (c) doubts whether insurance companies will pay for the study. In addition, many ethical questions pose continuing challenges [19,35]. However, the number of drugs approved with a reference to the genetic study in labeling information is increasing. ...
... Over the past years it has become clear that oncological patients, diagnosed with the same cancer type, may have different responses to generic treatments such as radiation or chemotherapy. To overcome these variable responses, cancer precision medicine aims to provide the right dose of the right drug for the right patient at the right time (Quinones et al., 2014). Thus, precision medicine has become an important tool in cancer treatment; it allows the identification of specific mutations in driver genes responsible for tumor progression (ICGC/ TCGA Pan-Cancer Analysis of Whole Genomes Consortium, 2020). ...
... On the one hand, the most relevant cancer genome projects worldwide, such as TCGA(The Cancer Genome Atlas Research Network, 2013), TARGET or PCAWGC (ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium, 2020), are overrepresented by Caucasian individuals (91.1%), and do not include enough individuals from minority populations (Guerrero et al., 2018). On the other hand, developing regions lack of investment in cancer genomics tests, have fragmented healthcare systems, and have insufficient characterization of pharmacogenetics variability in their populations (Quinones et al., 2014). Therefore, in this study we proposed a new insight for identification of the most frequent oncogenic variants in the Latino, African, Ashkenazi Jewish, East Asian, South Asian, European Finnish, and European non-Finnish populations in order to focus economic resources on analyzing the most frequent and relevant molecular targets. ...
Article
Full-text available
Background: Breast cancer (BRCA) and prostate cancer (PRCA) are the most commonly diagnosed cancer types in Latin American women and men, respectively. Although in recent years large-scale efforts from international consortia have focused on improving precision oncology, a better understanding of genomic features of BRCA and PRCA in developing regions and racial/ethnic minority populations is still required. Methods: To fill in this gap, we performed integrated in silico analyses to elucidate oncogenic variants from BRCA and PRCA driver genes; to calculate their deleteriousness scores and allele frequencies from seven human populations worldwide, including Latinos; and to propose the most effective therapeutic strategies based on precision oncology. Results: We analyzed 339,100 variants belonging to 99 BRCA and 82 PRCA driver genes and identified 18,512 and 15,648 known/predicted oncogenic variants, respectively. Regarding known oncogenic variants, we prioritized the most frequent and deleterious variants of BRCA (n = 230) and PRCA (n = 167) from Latino, African, Ashkenazi Jewish, East Asian, South Asian, European Finnish, and European non-Finnish populations, to incorporate them into pharmacogenomics testing. Lastly, we identified which oncogenic variants may shape the response to anti-cancer therapies, detailing the current status of pharmacogenomics guidelines and clinical trials involved in BRCA and PRCA cancer driver proteins. Conclusion: It is imperative to unify efforts where developing countries might invest in obtaining databases of genomic profiles of their populations, and developed countries might incorporate racial/ethnic minority populations in future clinical trials and cancer researches with the overall objective of fomenting pharmacogenomics in clinical practice and public health policies.
... Additionally, they analyzed a paired list of 54 genes/drugs associations to determine relationships between biomarkers and responses to genomic medicine. The regional structured survey was compared to a previous survey conducted in 2014 to assess progress in the region (Quinones et al., 2014). The results indicated that LAC countries have contributed 3.44% of total publications and 2.45% of the PGxrelated clinical trials worldwide thus far. ...
... In addition, in Latin America, arguably the largest recently mixed population among European, African and Amerindians is underrepresented in world databases (Salzano and Sans, 2014;STATISTA, 2022). This issue and other barriers for personalized medicine could explain that pharmacogenomics are only scarcely available in this region (Quinones et al., 2014). Consequently, it is of paramount importance that we determine and establish a specific map for population-specific pharmacogenomic biomarkers that have the potential to directly impact and promote the clinical implementation of pharmacogenomics in very specific and unrepresented populations. ...
... Specifically for low-and-middle income countries, there is a gap in the access to health, including a timely quality diagnosis and treatment. Additionally, several studies have highlighted the need to diversify oncological studies to populations representing several ethnic groups along with the development of novel strategies to enhance race/ethnicity data recording and reporting [86][87][88][89][90] . In this sense, multi-omics technologies and public-private investment related to identifying therapeutic targets improving metastatic disease treatments are crucial to reduce inequalities in health and strengthen mechanisms that can improve survival rates of different types of cancer. ...
Article
Full-text available
Many primary-tumor subregions exhibit low levels of molecular oxygen and restricted access to nutrients due to poor vascularization in the tissue, phenomenon known as hypoxia. Hypoxic tumors are able to regulate the expression of certain genes and signaling molecules in the microenvironment that shift it towards a more aggressive phenotype. The transcriptional landscape of the tumor favors malignant transformation of neighboring cells and their migration to distant sites. Herein, we focused on identifying key proteins that participate in the signaling crossroads between hypoxic environment and metastasis progression that remain poorly defined. To shed light on these mechanisms, we performed an integrated multi-omics analysis encompassing genomic/transcriptomic alterations of hypoxia-related genes and Buffa hypoxia scores across 17 pancarcinomas taken from the PanCancer Atlas project from The Cancer Genome Atlas consortium, protein–protein interactome network, shortest paths from hypoxia-related proteins to metastatic and angiogenic phenotypes, and drugs involved in current clinical trials to treat the metastatic disease. As results, we identified 30 hypoxia-related proteins highly involved in metastasis and angiogenesis. This set of proteins, validated with the MSK-MET Project, could represent key targets for developing therapies. The upregulation of mRNA was the most prevalent alteration in all cancer types. The highest frequencies of genomic/transcriptomic alterations and hypoxia score belonged to tumor stage 4 and positive metastatic status in all pancarcinomas. The most significantly associated signaling pathways were HIF-1, PI3K-Akt, thyroid hormone, ErbB, FoxO, mTOR, insulin, MAPK, Ras, AMPK, and VEGF. The interactome network revealed high-confidence interactions among hypoxic and metastatic proteins. The analysis of shortest paths revealed several ways to spread metastasis and angiogenesis from hypoxic proteins. Lastly, we identified 23 drugs enrolled in clinical trials focused on metastatic disease treatment. Six of them were involved in advanced-stage clinical trials: aflibercept, bevacizumab, cetuximab, erlotinib, ipatasertib, and panitumumab.
... Some recently regional formed scientific societies (RELAGH, 2014; SOLFAGEM, 2021) and international efforts (RELIVAF-CYTED) are looking to shorten the region's gap of evidence and information. In this respect, Latin America is a vast region with some characteristics that do not allow easy implementation of research made in other settings (Quiñones et al., 2014). It is one of the most genetically diverse areas having frequencies or polymorphisms not found in other regions. ...
Article
Full-text available
Since the pharmacogenetics and pharmacogenomics (PGx) field started to rise, the information about the relationship between actionable genes, genotypes, and response to drugs has increased exponentially (Nicholson et al., 2021). There is evidence of the utility and impact of genetics in the choice of therapeutic regimens improving their effectiveness and safety (Arbitrio et al., 2021). Even some international efforts have created clinical guidelines that allow to implementation of pharmacogenomics in daily clinical practice. In addition to clinical outcomes, Economic benefits have been associated with the translation from “the bench to the bedside-
Article
Full-text available
Pharmacogenomics (PGx) is considered an emergent field in developing countries. Research on PGx in the Latin American and the Caribbean (LAC) region remains scarce, with limited information in some populations. Thus, extrapolations are complicated, especially in mixed populations. In this paper, we reviewed and analyzed pharmacogenomic knowledge among the LAC scientific and clinical community and examined barriers to clinical application. We performed a search for publications and clinical trials in the field worldwide and evaluated the contribution of LAC. Next, we conducted a regional structured survey that evaluated a list of 14 potential barriers to the clinical implementation of biomarkers based on their importance. In addition, a paired list of 54 genes/drugs was analyzed to determine an association between biomarkers and response to genomic medicine. This survey was compared to a previous survey performed in 2014 to assess progress in the region. The search results indicated that Latin American and Caribbean countries have contributed 3.44% of the total publications and 2.45% of the PGx-related clinical trials worldwide thus far. A total of 106 professionals from 17 countries answered the survey. Six major groups of barriers were identified. Despite the region’s continuous efforts in the last decade, the primary barrier to PGx implementation in LAC remains the same, the “need for guidelines, processes, and protocols for the clinical application of pharmacogenetics/pharmacogenomics”. Cost-effectiveness issues are considered critical factors in the region. Items related to the reluctance of clinicians are currently less relevant. Based on the survey results, the highest ranked (96%–99%) gene/drug pairs perceived as important were CYP2D6 /tamoxifen, CYP3A5 /tacrolimus, CYP2D6 /opioids, DPYD /fluoropyrimidines, TMPT /thiopurines, CYP2D6 /tricyclic antidepressants, CYP2C19 /tricyclic antidepressants, NUDT15 /thiopurines, CYP2B6 /efavirenz, and CYP2C19 /clopidogrel. In conclusion, although the global contribution of LAC countries remains low in the PGx field, a relevant improvement has been observed in the region. The perception of the usefulness of PGx tests in biomedical community has drastically changed, raising awareness among physicians, which suggests a promising future in the clinical applications of PGx in LAC.
Article
Full-text available
The development of clinical practice recommendations or guidelines for the clinical use of biomarkers is an issue of great importance with regard to adverse drug reactions. The potential of pharmacogenomic biomarkers has been extensively investigated in recent years. However, several barriers to implementing the use of pharmacogenomics testing exist. We conducted a survey among members of the Spanish Societies of Pharmacology and Clinical Pharmacology to obtain information about the perception of such barriers and to compare the perceptions of participants about the relative importance of major gene/drug pairs. Of 11 potential barriers, the highest importance was attributed to lack of institutional support for pharmacogenomics testing, and to the issues related to the lack of guidelines. Of the proposed gene/drug pairs the highest importance was assigned to HLA-B/abacavir, UGT1A1/irinotecan, and CYP2D6/tamoxifen. In this perspective article, we compare the relative importance of 29 gene/drug pairs in the Spanish study with that of the same pairs in the American Society for Clinical Pharmacology and Therapeutics study, and we provide suggestions and areas of focus to develop a guide for clinical practice in pharmacogenomics testing.
Article
Full-text available
Cancer is a leading cause of death worldwide. The cancer incidence rate in Chile is 133.7/100,000 inhabitants and it is the second cause of death, after cardiovascular diseases. Most of the antineoplastic drugs are metabolized to be detoxified, and some of them to be activated. Genetic polymorphisms of drug-metabolizing enzymes can induce deep changes in enzyme activity, leading to individual variability in drug efficacy and/or toxicity. The present research describes the presence of genetic polymorphisms in the Chilean population, which might be useful in public health programs for personalized treatment of cancer, and compares these frequencies with those reported for Asian and Caucasian populations, as a contribution to the evaluation of ethnic differences in the response to chemotherapy. We analyzed 23 polymorphisms in a group of 253 unrelated Chilean volunteers from the general population. The results showed that CYP2A6*2, CYP2A6*3, CYP2D6*3, CYP2C19*3, and CYP3A4*17 variant alleles are virtually absent in Chileans. CYP1A1*2A allele frequency (0.37) is similar to that of Caucasians and higher than that reported for Japanese people. Allele frequencies for CYP3A5*3(0.76) and CYP2C9*3(0.04) are similar to those observed in Japanese people. CYP1A1*2C(0.32), CYP1A2*1F(0.77), CYP3A4*1B(0.06), CYP2D6*2(0.41), and MTHFR T(0.52) allele frequencies are higher than the observed either in Caucasian or in Japanese populations. Conversely, CYP2C19*2 allelic frequency (0.12), and genotype frequencies for GSTT1 null (0.11) and GSTM1 null (0.36) are lower than those observed in both populations. Finally, allele frequencies for CYP2A6*4(0.04), CYP2C8*3(0.06), CYP2C9*2(0.06), CYP2D6*4(0.12), CYP2E1*5B(0.14), CYP2E1*6(0.19), and UGT2B7*2(0.40) are intermediate in relation to those described in Caucasian and in Japanese populations, as expected according to the ethnic origin of the Chilean population. In conclusion, our findings support the idea that ethnic variability must be considered in the pharmacogenomic assessment of cancer pharmacotherapy, especially in mixed populations and for drugs with a narrow safety range.
Article
Full-text available
In this paper we discuss the consensus view on the use of qualifying biomarkers in drug safety, raised within the frame of the XXIV meeting of the Spanish Society of Clinical Pharmacology held in Málaga (Spain) in October, 2011. The widespread use of biomarkers as surrogate endpoints is a goal that scientists have long been pursuing. Thirty years ago, when molecular pharmacogenomics evolved, we anticipated that these genetic biomarkers would soon obviate the routine use of drug therapies in a way that patients should adapt to the therapy rather than the opposite. This expected revolution in routine clinical practice never took place as quickly nor with the intensity as initially expected. The concerted action of operating multicenter networks holds great promise for future studies to identify biomarkers related to drug toxicity and to provide better insight into the underlying pathogenesis. Today some pharmacogenomic advances are already widely accepted, but pharmacogenomics still needs further development to elaborate more precise algorithms and many barriers to implementing individualized medicine exist. We briefly discuss our view about these barriers and we provide suggestions and areas of focus to advance in the field.
Article
Full-text available
Adverse drug reactions (ADRs) rank as one of the top ten leading causes of death and illness in the developed world. In cancer therapy, more patients are surviving cancer than ever before, but 40% of cancer survivors suffer life-threatening or permanently disabling severe ADRs and are left with long-term sequelae. ADRs are often more frequent and more severe in children, and the consequences for children who experience a severe ADR can be catastrophic. Pharmacogenomics has the potential to improve the safety of these drugs. This review highlights severe ADRs that can occur in cancer therapy that are more frequent and more severe in children, and the pharmacogenomics research that aims to understand, predict, and ultimately prevent these severe reactions.
Article
Full-text available
Currently, there are very few guidelines linking the results of pharmacogenetic tests to specific therapeutic recommendations. Therefore, the Royal Dutch Association for the Advancement of Pharmacy established the Pharmacogenetics Working Group with the objective of developing pharmacogenetics-based therapeutic (dose) recommendations. After systematic review of the literature, recommendations were developed for 53 drugs associated with genes coding for CYP2D6, CYP2C19, CYP2C9, thiopurine-S-methyltransferase (TPMT), dihydropyrimidine dehydrogenase (DPD), vitamin K epoxide reductase (VKORC1), uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1), HLA-B44, HLA-B*5701, CYP3A5, and factor V Leiden (FVL).
Article
Information on CYP2B6 allele frequencies and detrimental genotypes in mixed human populations is scarce. The aim of this study was to analyze the frequencies and haplotypes of nonsynonymous CYP2B6 single nucleotide polymorphisms (SNPs) in a Colombian population. One hundred and fifty-two healthy individuals were analyzed for five nonsynonymous CYP2B6 SNPs, namely rs8192709, rs3745274, rs2279343 rs28399499, and rs3211371. Besides eight known variant alleles, we identified two as yet unknown variant alleles combining, respectively, the SNPs rs3745274 and rs3211371 and rs8192709 and rs3745274. Comparison of Colombian mestizo individuals with other mestizo population indicates statistically significant differences (P<0.001) for the gain-of-function CYP2B6*4 allele and for combined detrimental CYP2B6 alleles. In addition, we observed a low linkage between the SNPs rs3745274 and rs2279343, which are often assumed as linked. In conclusion, large interethnic and intraethnic variability exists for CYP2B6 polymorphisms, thus reinforcing the need for tailored genotyping protocols for CYP2B6 testing as a biomarker of drug response.
Article
There has been much progress in genomics in the ten years since a draft sequence of the human genome was published. Opportunities for understanding health and disease are now unprecedented, as advances in genomics are harnessed to obtain robust foundational knowledge about the structure and function of the human genome and about the genetic contributions to human health and disease. Here we articulate a 2011 vision for the future of genomics research and describe the path towards an era of genomic medicine.
Article
The slow rate at which pharmacogenetic tests are being adopted in clinical practice is partly due to the lack of specific guidelines on how to adjust medications on the basis of the genetic test results. One of the goals of the Clinical Pharmacogenetics Implementation Consortium (CPIC) of the National Institutes of Health's Pharmacogenomics Research Network (http://www.pgrn.org) and the Pharmacogenomics Knowledge Base (PharmGKB, http://www.pharmgkb.org) is to provide peer-reviewed, updated, evidence-based, freely accessible guidelines for gene/drug pairs. These guidelines will facilitate the translation of pharmacogenomic knowledge from bench to bedside.Clinical Pharmacology & Therapeutics (2011) 89 3, 464-467. doi:10.1038/clpt.2010.279