Kocal 2017
Kocal 2017
http://dx.doi.org/10.5772/intechopen.69207
Abstract
© 2017 The Author(s). Licensee InTech. This chapter is distributed under the terms of the Creative Commons
Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use,
distribution, and reproduction in any medium, provided the original work is properly cited.
4 Genetic Polymorphisms
1. Introduction
Genetic and environmental factors are the two main reasons that cause human phenotype
variations. If the genomic DNA sequences of two individuals are compared, substantial
sequence variations can be detected at different points of the whole genome. There are many
forms of these genetic variations [1]. Polymorphism term, arose from the combination of the
Greek words ‘poly’ (meaning as multiple) and ‘morph’ (meaning as form), is used in genet-
ics to describe multiple forms of a single gene that exist in a population. Polymorphisms are
genetic variants and refer to the occurrence of various phenotypes in a certain population.
A polymorphism is a DNA sequence variation and does not classify as mutation. In genetic
polymorphisms, there are two or more equally acceptable sequence of a gene and the com-
mon allele must have a frequency of 1% or more in the population. If the frequency is lower
than 1%, the allele is accepted as a mutation. On the other hand, a mutation is a change in
DNA sequence away from normal allele and forms abnormal variant [2].
1.1.2. Nomenclature
The unique and universal nomenclature to refer specific single nucleotide polymorphisms
(SNPs) is that using the rs number (reference sequence). It stands for Reference SNP clus-
ter ID. The rs number allows the precise identification of a polymorphic variation in the
numerous databases (NCBI, HapMap, SNP500 Cancer, etc.). For instance, a SNP causes a
replacement of an amino acid by another amino acid; this can be defined by the name and
the position of the replaced amino acid, followed by the name of the novel amino acid.
As an example, a common SNP in the DPYD gene is identified as rs 1801160 [V732I or
Val732Ile]. SNPs are also identified by the name and position of nucleotide in the reference
DNA sequence. The same SNP in the DPYD gene, presented with rs 1801160, is identified as
2194G>A. The letters A, T, C, and G can be used for both nucleotides and amino acids, and
this can cause confusion [3].
nucleotide (Figure 1). This type of change generates single nucleotide variants (SNVs). SNPs
can affect gene function due to the change of protein but can also occur in noncoding parts of
the gene so they would not be seen in the protein product [5, 6].
Intrinsic factors and extrinsic factors cause variability in drug response. Extrinsic factors such as
food and concomitant medications may be controllable, but intrinsic factors such as gender, eth-
nicity, age, renal or hepatic function, and genetic differences in the expression of enzymes need
advance knowledge to control [7]. Ethnicity is one of the key factors that can explain the observed
variability in both pharmacokinetics (PK) and pharmacodynamics (PD) of therapeutics, result-
ing in differences in response to drug therapy as well as chemotherapeutics. United States Food
and Drug Administration (FDA) guidelines pay attention to “ethnically sensitive situations”
for some drugs and suggests the types of solutions that may control such ethnic sensitivity.
One of the most important factors that may contribute to this ethnic sensitivity of a drug, include
genetic polymorphisms in metabolic pathways of drugs [7, 8]. To define the variability on the
response or metabolism of specific therapeutics or drug targets, drug companies and presti-
gious research groups are biobanking DNA samples [9]. Although the clinical relevance of some
variants is well characterized, the relevance of some variant alleles is as yet unknown.
There are some challenges on the use of disease-associated polymorphism knowledge. One
of these challenges is the lack of the unique information regarding the frequency of spe-
cific polymorphism in the targeted population. Without a unique presenting style of the
Figure 1. Classes of DNA variation affecting a single nucleotide position. (A) Single nucleotide variant (SNV) in which
two variants differ by having a G nucleotide or a C nucleotide (B) Insertion variation in which variant 1 has exactly
same reference sequence, variant 2 has one more T nucleotide; Deletion variation in which variant 1 has a A nucleotide,
variant 2 does not have; (C) Variable number of tandem repeats (VNTR) in which two variants differ by having repeats
of nucleotides.
6 Genetic Polymorphisms
olymorphism-related data, prevention of the risk for disease and drugs remain unknown.
p
In addition, determining the factors that may affect the association of the allele with disease
or drugs, such as ethnicity, may not be possible without population-based allele frequencies
[10–12]. SNPs can be assigned with an allele frequency—the ratio of chromosomes in the pop-
ulation carrying the less common variant to those with the more common variant. It is impor-
tant to note that there are variations among different populations, so a common SNP allele may
be much rare in one geographical or ethnic group than another [13, 14]. The Hardy-Weinberg
principle can be used to calculate allele frequencies [15]. The Hardy-Weinberg principle (also
known as the Hardy-Weinberg equilibrium, equation, theorem, or law) states that allele fre-
quencies in a population will remain stable from generation to generation in the absence of
other evolutionary factors, such as mutation, polymorphisms, genetic drift, gene flow, meiotic
drive, and mate choice. Hardy-Weinberg principle describes that the ideal condition against
the effects of these factors can be analyzed. The principle is named after Godfrey Harold
Hardy and Wilhem Weinberg, who first demonstrated it mathematically. If Hardy-Weinberg
principle is violated, the key interferences of a genetic polymorphisms-based study may be
compromised. Thus, accumulating evidence suggests that Hardy-Weinberg principle-based
reporting may be optimal in genetic and nongenetic journals, because variability in the ana-
lyzed data can cause errors or peculiarities [15].
Most of the chemotherapeutic agents for cancer treatment affect a minority of cancer patients
and have a narrow therapeutic index that frequently causes life-threatening toxicities and
even death. Even specific molecule-targeted therapies, which are safer than cytotoxic drugs,
are associated with severe adverse events. Thus, novel treatment strategies that can increase
the effectiveness of therapy and decrease the rate of adverse events will be developed. Under
this approach, the aim of personalized medicine is to tailor the therapy options according to
patient’s molecular profile [16, 17]. Establishing the relation between molecular characteris-
tics of patient and drug outcomes is crucial for the identification of predictive biomarkers and
understands the base of personalized therapy. Personalized medicine can also be called as P4
medicine due to the various contents as predictive, personalized, preventive, and participa-
tory medicine (P4); it separates patients into different groups with an individual’s molecular
profile. Thus, personalized medicine covers the determination of the safest and most effec-
tive chemotherapeutic agents [18]. The goal of discipline of pharmacogenetics, first used in
the late 1950s, is to make ‘personalized medicine’ as applicable to various patient groups.
It can be defined as the study of patients’ genotype affecting drug response. In some patient
groups, certain drugs work well but not as well in others. Pharmacogenetics-based studies
(between genotype of patients and the response of therapeutics) allow designing more effec-
tive and population-specific therapeutic treatments (Figure 2). Polymorphism analysis, muta-
tion analysis and genome sequencing are the backbones of discipline of pharmacogenetics.
Polymorphisms in Pharmacogenetics of Personalized Cancer Therapy 7
http://dx.doi.org/10.5772/intechopen.69207
Figure 2. The traditional prescribing approaches in clinical therapies, such as ‘trial and error’, ‘one drug fits all’ and ‘one
dose fits all’, have limits due to the drug safety. Pharmacogenetics combines standard biochemical tests with molecular
genetics based tests to detect DNA variations in the human genome for the application of genomic-based prescribing.
Polymorphisms and their association with diseases should be handled based on gene as bio-
markers, due to the relatively large frequency in the human genome. To learn, how to use and
interpret the polymorphisms analysis and which test(s) should be chosen is essential.
After the deep sequencing of human genome with Human Genome Project, the detection
of population-based DNA polymorphisms, especially that effect the development and the
progress of diseases, is the second phase of human genomics. High-throughput polymor-
phism genotyping process includes fast and cost-effective identification of polymorphisms
in different individuals and lead to the determination of associations between genotype and
phenotype. Generally, genotyping steps start with the isolation of starting material as DNA
from patient; it follows with amplification to increase the sample amount and then finalize
with polymerase chain reaction (PCR), sequencing or array-based technologies. A number of
good polymorphism genotyping technologies are currently in use to meet the needs of clinics
and researches, but only one genotyping method is not ideal for all applications (Table 1).
Slow speed of assays due to the time-consuming protocols, high instrument and consumable
costs, and requirements on the performing multiple assays in parallel are the main challenges
of polymorphism genotyping technologies. Studies on ideal polymorphism genotyping tech-
nologies are on development process [19–21].
8 Genetic Polymorphisms
Mass spectrometry High accuracy (as long as the sample is High instrument and consumable cost;
very pure); Long protocol;
Fast genotyping reactions in seconds; Limited throughput;
Allows thousands of reactions in a day. Requires very pure sample for accuracy.
Invader assay No PCR for amplification; Requires large amount of genomic DNA;
Simple protocol; Requires optimizations for each SNP;
Isothermal reaction Key enzyme is not suitable for general
research use.
Table 1. A summary of popular polymorphism detection methods with their advantages and disadvantages [19–21].
genome, which tumor genome does not present, are the main determinants of toxicity risk
(e.g. polymorphisms on the genes of drug metabolism such as dihydropyrimidine dehydrogenase
gene (DPYD), thiopurine-S-methyltransferase (TPMT), UDP-glucuronosyltransferase (UGT), etc.)
[23]. Genotyping studies have revealed that the gene encoding a specific protein can have a
number of differences in sequence at the nucleotide level. These differences especially called
as polymorphisms, sometimes do not cause significant alterations on the final product, but
may have an effect on the substrate specificity and activity of the product (especially for
enzymes) or other characteristics and functions. For example, polymorphisms in cytochrome
P450 2D6 (CYP2D6) are one of the cytochrome P450 enzymes of the liver that can influ-
ence how humans metabolize cancer drugs, although the enzymes are basically the same
sequence and structure. Polymorphisms in CYP2D6 have been seen in the general popula-
tion about 10% and it has been associated with poor-metabolizer phenotype of enzyme.
This is important for codeine-based pain medications due to the activation of codeine to
morphine and includes CYP2D6-dependent step [24].
Pharmacogenetic tests based on the determination of genetic variants for drug efficacy or
toxicity has begun to use in the 2000s, although genetic-based studies began in the 1950s. FDA
has been working to improve pharmacogenetic technologies in the development, regulation
and use of medications and revised drug labels in terms of pharmacogenetic biomarkers in
oncology area (Table 2) [25, 26].
It is important to describe important gene polymorphisms and their clinical meaning in
oncology field that may determine the optimum pharmacological treatment in terms of
treatment outcomes, tolerability and the occurrence of serious, even life-threatening adverse
reactions.
The UGT super family includes four main UGT families, namely UGT1, UGT2, UGT3, and
UGT8. The UGT1 and UGT2 genes, encode 16 functional proteins, have been extensively
studied and well characterized. A phase II metabolic enzyme, UGT1A1 is the most studied
UGT enzyme due to its main role in glucuronidation of exogenous and endogenous sub-
strates, including bilirubin. UGT1A1 also appears in the metabolism processes of most of the
anti-cancer drugs, such as topoisomerase I inhibitor irinotecan, the topoisomerase II inhibi-
tor etoposide [27]. Alterations on the glucuronidation activity of UGT1A1 caused by genetic
or environmental factors may have significant physiological and pharmacological results
on the metabolism of anticancer agents. Allelic variations have been identified in the pro-
moter region and exon 5 of UGT1A1 region. The wild-type allele of UGT1A1 gene (known
as UGT1A1*1) has six thymine- adenine (TA) repeats in the promoter region of gene (TATA
box). Allelic differences vary from five (UGT1A1*36, proficient allele) to eight (UGT1A1*37,
deficient allele) TA repeats, and these differences affect the UGT1A1-mediated glucuronida-
tion of SN-38 (7-ethyl-10-hydroxycamptothecin), active metabolite of anticancer drug irino-
tecan both in vitro and in vivo. Increasing number of TA repeats has been associated with
decreased transcription of gene and overall UGT1A1 activity (Table 3) [28]. Patients (allele
frequency in Caucasians 8–20% are homozygous, 40–50% are heterozygous) with seven TA
repeat sequence (named UGT1A1*28) have severe toxicity risk after irinotecan treatment
because of decreased gene expression and overall UGT1A1 activity (30% enzyme activity in
*28 relative to *1 allele). These patients [with (TA)7 repeats] have fourfold relative toxicity risk
compared with patients with six repeat sequences [29]. UGT1A1*60 (in linkage disequilib-
rium with TATA box variants) and UGT1A1*93 are the other variants located in the promoter
region. Both of them are found homozygous in around 10% of Caucasians [30]. UGT1A1*6
and UGT1A1*27 are located in coding region exon 1. UGT1A1*6 is the most frequent variant
in Asian populations (not found in Caucasians) and associated with ~30% decreased enzyme
activity in homozygous patients. UGT1A1*27 is almost completely eradicated enzyme activ-
ity. Nearly 3% of Asian people, are homozygous for both *6 and *27 variant [28, 31].
The pyrimidine antimetabolites 5-fluorouracil and its oral prodrug capecitabine are widely
used chemotherapeutic agents in the management of variety of tumor types, including colorec-
tal, breast, and head and neck cancers. They activate metabolically and inhibit thymidylate
synthase enzyme, which takes role in cellular replication. However, 5-FU leads significant
toxicities, such as myelosuppression, mucositis, hand-foot syndrome, and diarrhea. On the
other hand, accumulation of knowledge on 5-FU mechanism has developed new strategies
that increase the treatment efficacy and response [32, 33]. 5-FU is metabolized by dihydropy-
rimidine dehydrogenase (DPD) enzyme, and it converts the fluoropyrimidine to its inactive
metabolite dihydrofluorouracil (Figure 3). DPD enzyme, which is encoded by DPYD gene, is
a rate-limiting enzyme of 5-FU catabolism and is also used for evaluating the variability of
5-FU metabolism among patients [34]. SNPs in the DPYD gene are responsible for insufficient
production of DPD enzyme; therefore, low levels of enzyme increase the half-life of the drug,
Genotype Ref SNP HGVS Drug Enzymatic activity
Region Clinical phenotype
UGT1A1*1 Common allele-Wild type (TA) 6TA Irinotecan Normal enzyme activity
TYMS 2R/2R, rs34743033 28bp VNTR (2R; 3R) 5-fluorouracil Capecitabine Enzyme activity based on the repeats
TYMS 2R/3RG, TYMS Promoter enhancer region 2R/2R: Decreased TYMS expression, increased
3RG/3RG, 5-FU responsiveness, increased risk of toxicity
2R/3RG, 3RG/3RG: Increased TYMS
expression, decreased 5-fluorouracil,
capecitabine responsiveness, poor prognosis.
http://dx.doi.org/10.5772/intechopen.69207
Polymorphisms in Pharmacogenetics of Personalized Cancer Therapy
11
12
TPMT*1 Common allele-Wild type Common allele-Wild type 6-mercaptopurine; Normal enzyme activity
Azathioprine; Thioguanine
CYP2D6*1 Common allele-Wild type Common allele-Wild type Tamoxifen Normal activity
Extensive metabolizer
Note: Guidelines for the description and nomenclature of gene variations are available from the Human Genome Variation Society (HGVS): http://www.hgvs.org/content/
guidelines.
Figure 3. Metabolism of 5-fluorouracil (5-FU). The initial and rate-limiting enzyme of 5-FU is dihydropyrimidine
dehydrogenase (DPD), catalysing the conversion of 5-FU into 5,6-dihydrofluorouracil (DHFU). DHFU is degraded
into fluoro-beta-ureidopropionic acid (FUPA) and fluoro-beta-alanine (FBAL) through degradation cascade. 5-Fluoro-
2′-deoxyuridine-5′-monophosphate (5-FdUMP), is the cytotoxic product resulting from a multi-step activation of
5-FU, inhibits the enzyme thymidylate synthase (TS) enzyme. This inhibition leads to accumulation of deoxy-uridine-
monophospate (dUMP) and depletion of deoxy-thymidine-monophosphate (dTMP), which leads to inhibition of DNA
synthesis. 5-FU can also inhibit RNA synthesis in a pathway that involves 5-fluorouridine monophosphate (5-FUMP)
and subsequent conversion to 5-fluorouridine triphosphate (5-FUTP) via 5-fluorouridine diphosphate (5-FUDP).
thus, resulting in excess drug accumulation and toxicity due to the inefficient catabolism of
drug. Genetic testing of polymorphisms is being used for the classification of patients who
would be at high risk for severe or fatal toxicity when receiving fluoropyrimidine-based che-
motherapy (Table 3) [35]. Complete deficiency of DPD has been seen in approximately 5% of
the overall population and also 3–5% of the population has a partial DPD deficiency due to
sequence variations in DPYD gene [36].
The IVS14+ 1G>A change with the combination of a mutation in intron 14 and a deletion
at 5′-splice consensus sequence of exon 14, the most known and frequent variant (known as
DPYD*2A), is resulting the formation of a truncated enzyme product lacking activity. The
estimated incidence of homozygous genotype of this allelic variant is 0.1% and heterozy-
gous genotype is 0.5–2.0% in Caucasians [37, 38]. Other variants, which are associated with
increased toxicity risk, include 496A>G in exon 6, T1679G (DPYD*13) in exon 13 and 2846A>T
in exon 22 [39–41]. Genetic mutations in DPYD can be analyzed by highly sensitive methods
even for heterozygous variants such as pyrosequencing. But determination of the 5-FU and
dihydrofluorouracil concentration ratio in plasma by high-pressure liquid chromatography
(HPLC) may be more reliable predictor test for toxicity [35]. Besides DPYD, there are some
other pharmacogenetic biomarkers that are being used for the determination of the efficacy
and toxicity of fluoropyrimidine-related therapies, such as thymidylate synthase gene (TYMS).
nucleotides leading to severe toxicity has been shown in people with deficient TPMT alleles,
after receiving a standard dose thiopurine-based treatment. Patients with low TPMT activ-
ity expose more activated thioguanine nucleotides and more treatment toxicity as well as
efficacy. Patient’s genotype predicts thiopurine nucleotide levels and treatment outcomes in
thiopurine drug metabolism that shows tri-modal distribution with 89–94% of patients hav-
ing high enzyme activity, 6–11% of them having intermediate activity (as heterozygous), and
0.3% of them having low or no activity (as homozygous) [53].
Cytochrome P450 2D6 (CYP2D6) deficiency is the first discovered pharmacogenetic deficiency
with the characterization of decreased enzyme expression-related polymorphisms. Decreased
mRNA levels of CY2D6 were determined as the reason of reduced metabolism and adverse
reactions to an anti-hypertensive drug, debrisoquine. These findings based on molecular tech-
nologies display the importance of genotyping on the pharmacological phenotyping involved
in drug metabolism [35]. CYP2D6 is a highly polymorphic enzyme (Table 3). According to
increased or decreased enzyme activity due to SNPs, duplications or deletions, genetic vari-
ants of CYP2D6 were classified into four metabolic phenotypes: ultra-rapid metabolizer,
extensive metabolizer, intermediate metabolizer and poor metabolizer (Table 4). This classifi-
cation system has been used to control treatment recommendations for several drugs, includ-
ing tamoxifen [57].
Tamoxifen is a selective estrogen receptor modulator, which is used in the treatment of estrogen
receptor-positive breast cancer. Tamoxifen is a weak estrogen antagonist by itself, but it is con-
verted into its main active metabolite, antiestrogenic endoxifen, by CYP2D6. CYP2D6 genotype is
predictive of endoxifen exposure that is critical in determining treatment outcome [59]. Increased
endoxifen exposure is associated with increased treatment efficacy and toxicity [60, 61].
18 Genetic Polymorphisms
Phenotype Genotype
Poor metabolizer Two inactive alleles
Notes: Active alleles: *1, *2, *33, *35; decreased activity alleles: *9, *10, *17, *29, *36, *41; inactive alleles: *3, *4, *5, *6, *7,
*8, *11-*16, *19-*21, *38, *40, *42.
Table 4. CYP2D6 phenotypes according to activity status of CYP2D6 alleles (adapted from Swen JJ., et al. 2011) [58].
Pharmacogenetics and its backbone studies in terms of polymorphisms present new devel-
opments and trends in the field of tailored medications and advancements in the modifi-
cation of therapeutic choices utilizing genotypic information from polymorphism analysis.
Pharmacogenetic biomarker studies have multiple processes from discovery to clinical imple-
mentation (Figure 4). The ultimate aim of the biomarker studies is to find a clinically accessible
decision-maker biomarker to improve patient outcomes. However, many of the valid associa-
tions cannot be achieved for clinical implementation due to the lack of sufficient robustness
or clinical importance for the question. Biomarker discovery studies should be performed
as the screening of genotype-phenotype relations in large cohorts with statistical and bioin-
formatics tools. The most significant markers obtained from discovery studies are replicated
for analytical validation in different cohorts with the evaluation of assay reproducibility and
robustness. After successful analytical validity, the biomarker and assay must be evaluated
to confirm its performance in diagnosing the clinical phenotype or predicting outcome of
interest. A clinically valid biomarker assay can undergo translation through prospective con-
firmation of clinical utility to improve patient outcomes. And finally, analytically and clini-
cally validated biomarker assay is ready for implementation phase that includes regulatory
approval and incorporation in clinical practice guidelines, commercialization and coverage
by health insurance [52, 62].
After “Human Genome Project,” science world began to use individual genotypic informa-
tion to predict the risk of diseases, to prognose the disease, to guide therapeutic decision-
making, and to develop targeted medications. Especially, the better prediction ability for
drug response and life-threatening toxic reactions is highly crucial for clinicians due to the
narrow therapeutic index of many cancer chemotherapeutic agents. The aim of personal-
ized medicine is to prescribe a convenient chemotherapeutics to the right patients with right
dose to achieve maximal therapeutic benefit with minimal toxicity. Deep observation of the
human genome is still ongoing, and it will help to discover new targets and select the most
efficacious drug for each patient’s tumor, which is named as genomic prescribing system,
the next evolution of systemic cancer management. Genetic polymorphisms, as significant
biomarkers, have provided significant and crucial information in the management of toxic-
ity and dosing of cancer treatments. So far the achievements have been limited due to the
multifactorial challenges on the polymorphism-based personalized cancer management.
Especially reliable clinical data for the effects of genetic alterations on the disease patho-
genesis, drug metabolism and response are not always available; and performing the large-
scale prospective clinical studies, to understand the associations of polymorphisms and the
application of chemotherapeutics, is often laborious. However, these prospective studies are
required for establishing possible relations for evaluating the utility and cost-effectiveness of
polymorphism-based pharmacogenetic tests and personalized medicine. Ethical, social, and
regulatory issues relevant with pharmacogenetic-based personalized medicine are the other
complex and challenging factors to establish the relations between genetic polymorphisms
and personalized drug responses.
The main application of genetic polymorphism knowledge is improving the futuristic health
care through gene therapy, discovery of new drugs and drug targets and upgrade the discov-
ery processes with advanced technologies. The use of single candidate genes can be useful as
a part of initial treatment, but in near future, it will never be enough to provide fully tailored
treatment decisions on the cancer management. Other omics technologies will complement
the genotype-phenotype association-based pharmacological-pathway approach, such as tran-
scriptomics, epigenomics, and miRNAomics.
• Humans share about 99.9% sequence identity. The other 0.1% are mostly SNPs.
• SNPs are the most common polymorphism type and occur about every 1000 bases.
20 Genetic Polymorphisms
• SNPs can be silent—99% of them are not in coding regions and not in genes and thus cause
harmless, harmful, latent changes.
• Genetic polymorphisms are one of the most important factors that may contribute to ethnic
sensitivity of a drug.
• FDA recommends for following guidelines relating to “ethnically sensitive situations” for
some drugs and suggests solutions that may control such ethnic sensitivity in the context
of therapeutic applications and study designs.
• Personalized medicine separates patients into different groups with an individual’s mo-
lecular profile and covers the determination of the safest and most effective chemothera-
peutic agents.
• Polymorphism in the DPYD gene is responsible for insufficient production of DPD en-
zyme; therefore, low levels of enzyme increase drug accumulation and toxicity due to the
inefficient catabolism of drug.
• Allelic differences of UGT1A1 affect the glucuronidation of SN-38, active metabolite of iri-
notecan both in vitro and in vivo.
• TPMT polymorphisms have different effects on function of TPMT that inactivates thio-
guanine nucleotides. Patients with low TPMT activity expose more activated thioguanine
nucleotides and more treatment toxicity as well as efficacy.
• FDA recommends screening for G6PD deficiency before the beginning of rasburicase
treatment.
Author details
1 Department of Basic Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Turkey
References
[1] Bosch TM, Meijerman I, Beijnen JH, Schellens JH. Genetic polymorphisms of drug-metab-
olising enzymes and drug transporters in the chemotherapeutic treatment of cancer.
Clinical Pharmacokinetics. 2006;45(3):253-285. DOI: 10.2165/00003088-200645030-00003
[2] Lander ES. Initial impact of the sequencing of the human genome. Nature. 2011;470
(7333):187-197. DOI: 10.1038/nature09792
[3] Robert J, Le Morvan V, Giovannetti E, Peters GJ; PAMM Group of EORTC. On the use
of pharmacogenetics in cancer treatment and clinical trials. European Journal of Cancer.
2014;50(15):2532-2543. DOI: 10.1016/j.ejca.2014.07.013
[4] Karki R, Pandya D, Elston RC, Ferlini C. Defining “mutation” and “polymorphism” in the era
of personal genomics. BMC Medical Genomics. 2015;8:37. DOI: 10.1186/s12920-015-0115-z
[7] Huang SM, Temple R, Throckmorton DC, Lesko LJ. Drug interaction studies: Study
design, data analysis, and implications for dosing and labeling. Clinical Pharmacology
& Therapeutics. 2007;81(2):298-304. DOI: 10.1038/sj.clpt.6100054
[8] Andersson T, Flockhart DA, Goldstein DB, Huang SM, Kroetz DL, Milos PM, Ratain MJ,
Thummel K. Drug-metabolizing enzymes: Evidence for clinical utility of pharmacoge-
nomic tests. Clinical Pharmacology & Therapeutics. 2005;78(6):559-581. DOI: 10.1016/j.
clpt.2005.08.013
[9] Bjornsson TD, Callaghan JT, Einolf HJ, Fischer V, Gan L, Grimm S, Kao J, King SP,
Miwa G, Ni L, Kumar G, McLeod J, Obach RS, Roberts S, Roe A, Shah A, Snikeris F,
22 Genetic Polymorphisms
Sullivan JT, Tweedie D, Vega JM, Walsh J, Wrighton SA; Pharmaceutical Research and
Manufacturers of America (PhRMA) Drug Metabolism/Clinical Pharmacology Technical
Working Group.; FDA Center for Drug Evaluation and Research (CDER). The conduct
of in vitro and in vivo drug-drug interaction studies: A Pharmaceutical Research and
Manufacturers of America (PhRMA) perspective. Drug Metabolism and Disposition.
2000;31(7):815-832. DOI: 10.1124/dmd.31.7.815
[10] Galvan A, Ioannidis JP, Dragani TA. Beyond genome-wide association studies: Genetic
heterogeneity and individual predisposition to cancer. Trends in Genetics. 2010;26(3):132-
141. DOI: 10.1016/j.tig.2009.12.008
[11] Dadd T, Dadd T, Weale ME, Lewis CM. A critical evaluation of genomic control methods
for genetic association studies. Genetic Epidemiology. 2009;33(4):290-298. DOI: 10.1002/
gepi.20379
[12] Wang K. Testing for genetic association in the presence of population stratifica-
tion in genome-wide association studies. Genetic Epidemiology. 2009;33(7):637-645.
DOI: 10.1002/gepi.20415
[13] Guan W, Liang L, Boehnke M, Abecasis GR. Genotype-based matching to correct for
population stratification in large-scale case-control genetic association studies. Genetic
Epidemiology. 2009;33(6):508-517. DOI: 10.1002/gepi.20403
[14] Yesupriya A, Evangelou E, Kavvoura FK, Patsopoulos NA, Clyne M, Walsh MC, Lin
BK, Yu W, Gwinn M, Ioannidis JP, Khoury MJ. Reporting of human genome epide-
miology (HuGE) association studies: An empirical assessment. BMC Medical Research
Methodology. 2008;8:31. DOI: 10.1186/1471-2288-8-31
[15] Salanti G, Amountza G, Ntzani EE, Ioannidis JP. Hardy-Weinberg equilibrium in genetic
association studies: An empirical evaluation of reporting, deviations, and power.
European Journal of Human Genetics. 2005;13(7):840-848. DOI: 10.1038/sj.ejhg.5201410
[16] Keefe DM, Bateman EH. Tumor control versus adverse events with targeted anti-
cancer therapies. Nature Reviews Clinical Oncology. 2011;9(2):98-109. DOI: 10.1038/
nrclinonc.2011.192
[18] Tian Q, Price ND, Hood L. Systems cancer medicine: Towards realization of predictive,
preventive, personalized and participatory (P4) medicine. Journal of Internal Medicine.
2012;271(2):111-121. DOI: 10.1111/j.1365-2796.2011.02498.x
[20] Kwok PY. Methods for genotyping single nucleotide polymorphisms. Annual
Review of Genomics and Human Genetics. 2001;2:235-258. DOI: 10.1146/annurev.
genom.2.1.235
[21] Kim S, Misra A. SNP genotyping: Technologies and biomedical applications. Annual
Review of Biomedical Engineering. 2007;9:289-320. DOI: 10.1146/annurev.bioeng.9.060906.
152037
[22] Pirmohamed M, Park BK. Genetic susceptibility to adverse drug reactions. Trends in
Pharmacological Sciences. 2001;22:298-305. DOI: 10.1016/S0165-6147(00)01717-X
[23] Ulrich CM, Robien K, McLeod HL. Cancer pharmacogenetics: Polymorphisms, path-
ways and beyond. Nature Reviews Cancer. 2003;3(12):912-920. DOI: 10.1038/nrc1233
[24] Rogers JF, Nafziger AN, Bertino Jr JS. Pharmacogenetics affects dosing, efficacy, and
toxicity of cytochrome P450-metabolized drugs. American Journal of Medicine.
2002;113(9):746-750. DOI: 10.1016/S0002-9343(02)01363-3
[25] Crews KR, Hicks JK, Pui CH, Relling MV, Evans WE. Pharmacogenomics and individual-
ized medicine: Translating science into practice. Clinical Pharmacology & Therapeutics.
2012;92(4):467-475. DOI: 10.1038/clpt.2012.120
[26] United States Food and Drug Administration. Table of Pharmacogenomic Biomarkers
in Drug Labeling [Internet]. 2017. Available from: https://www.fda.gov/Drugs/Science
Research/ResearchAreas/Pharmacogenetics/ucm083378.htm [Accessed: March 14,
2017]
[27] Bosma PJ, Seppen J, Goldhoorn B, Bakker C, Oude Elferink RP, Chowdhury JR,
Chowdhury NR, Jansen PL. Bilirubin UDP-glucuronosyltransferase 1 is the only rel-
evant bilirubin glucuronidating isoform in man. Journal of Biological Chemistry.
1994;269(27):17960-17964
[28] Etienne-Grimaldi MC, Boyer JC, Thomas F, Quaranta S, Picard N, Loriot MA, Narjoz C,
Poncet D, Gagnieu MC, Ged C, Broly F, Le Morvan V, Bouquié R, Gaub MP, Philibert
L, Ghiringhelli F, Le Guellec C; Collective work by Groupe de Pharmacologie Clinique
Oncologique (GPCO-Unicancer).; French Réseau National de Pharmacogénétique
Hospitalière (RNPGx). UGT1A1 genotype and irinotecan therapy: General review
and implementation in routine practice. Fundamental & Clinical Pharmacology.
2015;29(3):219-237. DOI: 10.1111/fcp.12117
[29] Iyer L, Das S, Janisch L, Wen M, Ramírez J, Karrison T, Fleming GF, Vokes EE, Schilsky
RL, Ratain MJ. UGT1A1*28 polymorphism as a determinant of irinotecan disposition
and toxicity. Pharmacogenomics Journal. 2002;2(1):43-47. DOI: 10.1038/sj/tpj/6500072
[32] Longley DB, Harkin DP, Johnston PG. 5-fluorouracil: Mechanisms of action and clinical
strategies. Nature Reviews Cancer. 2003;3(5):330-338. DOI: 10.1038/nrc.1074
[34] van Staveren MC, Guchelaar HJ, van Kuilenburg AB, et al. Evaluation of predictive tests
for screening for dihydropyrimidine dehydrogenase deficiency. Pharmacogenomics
Journal. 2013;13:389-395. DOI: 10.1038/tpj.2013.25
[35] Aiello M, Vella N, Cannavò C, Scalisi A, Spandidos DA, Toffoli G, Buonadonna A, Libra
M, Stivala F. Role of genetic polymorphisms and mutations in colorectal cancer therapy
(Review). Journal of Molecular Medicine. 2011;4(2):203-208. DOI: 10.3892/mmr.2010.408
[38] Offer SM, Fossum CC, Wegner NJ, Stuflesser AJ, Butterfield GL, Diasio RB. Comparative
functional analysis of DPYD variants of potential clinical relevance to dihydropyrimi-
dine dehydrogenase activity. Cancer Research. 2014;74(9):2545-2554. DOI: 10.1158/0008-
5472.CAN-13-2482
[39] Mattison LK, Ezzeldin H, Carpenter M, Modak A, Johnson MR, Diasio RB. Rapid iden-
tification of dihydropyrimidine dehydrogenase deficiency by using a novel 2-13C-uracil
breath test. Clinical Cancer Research. 2004;10(8):2652-2658. DOI: 10.1158/1078-0432.
CCR-03-0374
[40] Gross E, Ullrich T, Seck K, Mueller V, de Wit M, von Schilling C, Meindl A, Schmitt M,
Kiechle M. Detailed analysis of five mutations in dihydropyrimidine dehydrogenase
detected in cancer patients with 5-fluorouracil-related side effects. Human Mutation.
2003;22(6):498. DOI: 10.1002/humu.9201
[41] van Kuilenburg AB, De Abreu RA, van Gennip AH. Pharmacogenetic and clinical aspects
of dihydropyrimidine dehydrogenase deficiency. Annals of Clinical Biochemistry.
2003;40(Pt 1):41-45. DOI: 10.1258/000456303321016150
Polymorphisms in Pharmacogenetics of Personalized Cancer Therapy 25
http://dx.doi.org/10.5772/intechopen.69207
[42] Marcuello E, Altés A, Menoyo A, Del Rio E, Gómez-Pardo M, Baiget M. UGT1A1 gene
variations and irinotecan treatment in patients with metastatic colorectal cancer. British
Journal of Cancer. 2004;91(4):678-682. DOI: 10.1038/sj.bjc.6602042
[43] Caudle KE, Thorn CF, Klein TE, Swen JJ, McLeod HL, Diasio RB, Schwab M. Clinical
Pharmacogenetics Implementation Consortium guidelines for dihydropyrimidine
dehydrogenase genotype and fluoropyrimidine dosing. Clinical Pharmacology &
Therapeutics. 2013;94(6):640-645. DOI: 10.1038/clpt.2013.172
[44] Hickey SE, Curry CJ, Toriello HV. ACMG practice guideline: Lack of evidence for
MTHFR polymorphism testing. Genetics in Medicine. 2013;15(2):153-156. DOI: 10.1038/
gim.2012.165
[45] Zhou SF, Di YM, Chan E, Du YM, Chow VD, Xue CC, Lai X, Wang JC, Li CG, Tian M,
Duan W. Clinical pharmacogenetics and potential application in personalized medicine.
Current Drug Metabolism. 2008;9(8):738-784. DOI: 10.2174/138920008786049302
[46] Tan BR, Thomas F, Myerson RJ, Zehnbauer B, Trinkaus K, Malyapa RS, Mutch MG,
Abbey EE, Alyasiry A, Fleshman JW, McLeod HL. Thymidylate synthase genotype-
directed neoadjuvant chemoradiation for patients with rectal adenocarcinoma. Journal
of Clinical Oncology. 2011 Mar 1;29(7):875-883. DOI: 10.1200/JCO.2010.32.3212
[47] Lecomte T, Ferraz JM, Zinzindohoué F, Loriot MA, Tregouet DA, Landi B, Berger A,
Cugnenc PH, Jian R, Beaune P, Laurent-Puig P. Thymidylate synthase gene polymorphism
predicts toxicity in colorectal cancer patients receiving 5-fluorouracil-based chemotherapy.
Clinical Cancer Research. 2004;10(17):5880-5888. DOI: 10.1158/1078-0432.CCR-04-0169
[49] Mandola MV, Stoehlmacher J, Zhang W, Groshen S, Yu MC, Iqbal S, Lenz HJ, Ladner
RD. A 6 bp polymorphism in the thymidylate synthase gene causes message instabil-
ity and is associated with decreased intratumoral TS mRNA levels. Pharmacogenetics.
2004;14(5):319-327. DOI: 10.1097/01.fpc.0000114730.08559.df
[50] Ulrich CM, Potter JD. Thymidylate synthase polymorphism and survival of colorectal
cancer patients treated with 5-fluorouracil. British Journal of Cancer. 2002;86(8):1365.
DOI: 10.1038/sj.bjc.6600229
[52] Hertz DL, Rae J. Pharmacogenetics of cancer drugs. Annual Review of Medicine.
2015;66:65-81. DOI: 10.1146/annurev-med-053013-053944
[53] Stanulla M, Schaeffeler E, Flohr T, Cario G, Schrauder A, Zimmermann M, Welte K,
Ludwig WD, Bartram CR, Zanger UM, Eichelbaum M, Schrappe M, Schwab M.
Thiopurine methyltransferase (TPMT) genotype and early treatment response to mer-
captopurine in childhood acute lymphoblastic leukemia. Journal of the American
Medical Association. 2005;293(12):1485-1489. DOI: 10.1001/jama.293.12.1485
[54] Luzzatto L, Seneca E. G6PD deficiency: A classic example of pharmacogenetics with
on-going clinical implications. British Journal of Haematology. 2014;164(4):469-480.
DOI: 10.1111/bjh.12665
[56] Wei CY, Lee MT, Chen YT. Pharmacogenomics of adverse drug reactions: Implementing
personalized medicine. Human Molecular Genetics. 2012;21(R1):R58–R65. DOI: 10.1093/
hmg/dds341
[57] Bell GC, Crews KR, Wilkinson MR, Haidar CE, Hicks JK, Baker DK, Kornegay NM, Yang
W, Cross SJ, Howard SC, Freimuth RR, Evans WE, Broeckel U, Relling MV, Hoffman JM.
Development and use of active clinical decision support for preemptive pharmacoge-
nomics. Journal of the American Medical Informatics Association. 2014;21(e1):e93–e99.
DOI: 10.1136/amiajnl-2013-001993
[58] 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.
Clinical Pharmacology & Therapeutics. 2011;89(5):662-673. DOI: 10.1038/clpt.2011.34
[59] Borges S, Desta Z, Jin Y, Faouzi A, Robarge JD, Philips S, Nguyen A, Stearns V,
Hayes D, Rae JM, Skaar TC, Flockhart DA, Li L. Composite functional genetic and
comedication CYP2D6 activity score in predicting tamoxifen drug exposure among
breast cancer patients. The Journal of Clinical Pharmacology. 2010;50(4):450-458.
DOI: 10.1177/0091270009359182
[60] Madlensky L, Natarajan L, Tchu S, Pu M, Mortimer J, Flatt SW, Nikoloff DM, Hillman
G, Fontecha MR, Lawrence HJ, Parker BA, Wu AH, Pierce JP. Tamoxifen metabolite con-
centrations, CYP2D6 genotype, and breast cancer outcomes. Clinical Pharmacology &
Therapeutics. 2011;89(5):718-725. DOI: 10.1038/clpt.2011.32
[61] Lorizio W, Wu AH, Beattie MS, Rugo H, Tchu S, Kerlikowske K, Ziv E. Clinical and bio-
marker predictors of side effects from tamoxifen. Breast Cancer Research and Treatment.
2012;132(3):1107-1118. DOI: 10.1007/s10549-011-1893-4
[62] Goossens N, Nakagawa S, Sun X, Hoshida Y. Cancer biomarker discovery and validation.
Translational Cancer Research. 2015;4(3):256-269. DOI: 10.3978/j.issn.2218-676X.2015.06.04