SlideShare a Scribd company logo
1 of 61
Download to read offline
Pharmacogenetics and Drug Response 
Author & Course Teacher 
MAIZBHA UDDIN AHMED 
Assistant Professor 
Department of Clinical Pharmacy and Pharmacology 
Faculty of Pharmacy 
University of Dhaka
Contents 
1 Pharmacogenetics 5 
1.1 Pharmacogenetics and Drug Response . . . . . . . . . . . . . . . . . . . . . . 6 
1.2 Pharmacogenetics and Drug safety: Pharmacogenetics of Irinotecan . . . . . . 6 
1.3 Genotype Phenotype correlation: CYP3A4 . . . . . . . . . . . . . . . . . . . . 8 
1.3.1 Genotype . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9 
1.3.2 Phenotype . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9 
1.3.3 Phenotypic diversity due to heterogeneity of mutations . . . . . . . . . 9 
1.4 Drug Metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10 
1.5 Classification of Metabolizer groups . . . . . . . . . . . . . . . . . . . . . . . 11 
1.6 CYP3A4 genotype and Phenotype . . . . . . . . . . . . . . . . . . . . . . . . 12 
1.6.1 CYP3A4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12 
1.6.2 CYP3A4 genotyping . . . . . . . . . . . . . . . . . . . . . . . . . . . 12 
1.6.3 CYP3A4 phenotyping . . . . . . . . . . . . . . . . . . . . . . . . . . 13 
1.7 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15 
2 Pharmacogenetics of Warfarin 17 
2.1 Warfarin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17 
2.1.1 Warfarin Pharmacgology . . . . . . . . . . . . . . . . . . . . . . . . . 17 
2.1.2 Mechanism of Action . . . . . . . . . . . . . . . . . . . . . . . . . . . 18 
2.2 Metabolism of Warfarin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19 
2.3 Pharmacogenetics of Warfarin . . . . . . . . . . . . . . . . . . . . . . . . . . 19 
2.3.1 CYP2C9 and Warfarin dosing . . . . . . . . . . . . . . . . . . . . . . 21 
2.3.2 VKORC1 and Warfarin . . . . . . . . . . . . . . . . . . . . . . . . . . 21 
2.3.3 CYP4F2 and Warfarin . . . . . . . . . . . . . . . . . . . . . . . . . . 21 
2.4 CPIC dosing guideline for Warfarin . . . . . . . . . . . . . . . . . . . . . . . 21 
2.5 References: . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21 
3 Pharmacogenetics of Clopidogrel 23 
3.1 Clopidogrel . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23 
3.1.1 Pharmacology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23 
3.1.2 Why it is important to study CYP2C19 polymorphism . . . . . . . . . 24 
3.2 Bioactivation of Clopidogrel and Prasugrel . . . . . . . . . . . . . . . . . . . . 24 
3.3 Clopidogrel and CYP2C19 . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24 
3.4 Genetic variation in CYP2C19 influences individual response to clopidogrel . . 25 
3.5 Choosing the best option: Clopidogrel? Prasugrel? or Ticagrelor? . . . . . . . 26 
3.6 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28 
1
CONTENTS 2 
4 Pharmacogenetics of NAT2 30 
4.1 NAT localization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30 
4.2 Human NAT2 Alleles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30 
4.3 NAT and Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31 
4.4 NAT2 and Drug Response . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31 
4.4.1 NAT2 and Isoniazid . . . . . . . . . . . . . . . . . . . . . . . . . . . 31 
4.4.2 NAT2 and Environmental carcinogen . . . . . . . . . . . . . . . . . . 32 
4.4.3 NAT2 and Procainamide . . . . . . . . . . . . . . . . . . . . . . . . . 33 
4.5 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33 
5 Pharmacogenetics of CYP2D6 35 
5.1 CYP2D6 allelic variants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35 
5.2 CYP2D6 and Antipsychotics Pharmacology . . . . . . . . . . . . . . . . . . . 37 
5.2.1 Metabolism of Amytriptyline . . . . . . . . . . . . . . . . . . . . . . 38 
5.2.2 Dosing recommendations for amitriptyline/nortriptyline based on CYP2D6 
phenotype . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38 
5.2.3 Recommended dosing of codeine by CYP2D6 Phenotype . . . . . . . . 40 
5.3 CYP2D6 and Tamoxifen: Why endoxifen is a better option than Tamoxifen . . 41 
5.4 Why Endoxifen? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42 
5.4.1 Why Endoxifen Treatment may have better therapeutic outcome . . . . 42 
5.5 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43 
6 Pharmacogenetics of cancer 46 
6.1 Interindividual variation in Chemotherapeutic drug response . . . . . . . . . . 46 
6.2 Genetic variations affecting drug response and toxicity with cancer chemotherapy 47 
6.3 Polymorphism in Drug-Metabolizing Enzymes . . . . . . . . . . . . . . . . . 48 
6.3.1 Thiopurine methyltransferase and 6-Mercaptopurine . . . . . . . . . . 48 
6.3.2 UDP-glucuronosyltransferase 1A1 and Irinotecan . . . . . . . . . . . . 49 
6.3.3 Dihydropyrimidine dehydrogenase and 5-FU . . . . . . . . . . . . . . 50 
6.3.4 MTHFR and Methotrexate . . . . . . . . . . . . . . . . . . . . . . . . 50 
6.4 Polymorphisms in Drug Transporters . . . . . . . . . . . . . . . . . . . . . . . 53 
6.4.1 Clinical Annotation for ABCB1 (rs1045642) and 5-fluorouracil . . . . 54 
6.4.2 Clinical Annotation for rs1045642 and paclitaxel . . . . . . . . . . . . 55 
6.5 Polymorphisms in Drug Targets . . . . . . . . . . . . . . . . . . . . . . . . . 55 
6.6 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
List of Figures 
1.1 Principle of Pharmacogenetics . . . . . . . . . . . . . . . . . . . . . . . . . . 6 
1.2 Pharmacogenetics based Dosing/Diagnostics . . . . . . . . . . . . . . . . . . . 7 
1.3 Metabolism of Irinotecan. Abbreviations: APC, 7-ethyl-10[4-N-(5-aminopentanoic 
acid)-1-piperidino]-carbonyloxycamptothecin; CYP3A4, cytochrome P450 3A4 
enzyme; CYP3A5, cytochrome P450, family 3, subfamily A, polypeptide 5; 
NPC, 7-ethyl-10[4-(1-piperidino)-1-amino]-carbonyloxycamptothecin; SN-38, 
7-ethyl-10-hydroxy-camptothecin; SN-38G, glucuronidated SN-38; Topo 1, DNA 
topoisomerase I. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8 
1.4 Irinotecan Metabolizer and Toxicities . . . . . . . . . . . . . . . . . . . . . . 8 
1.5 Genetics of Sickle cell anaemia . . . . . . . . . . . . . . . . . . . . . . . . . . 9 
1.6 Overview of Drug Metabolism and Excretion . . . . . . . . . . . . . . . . . . 10 
1.7 Genotype-Phenotype Association of Drug Metabolism and Pharmacology . . . 11 
1.8 Genotyping protocol of CYP3A4 . . . . . . . . . . . . . . . . . . . . . . . . . 13 
2.1 Warfarin Metabolism and Mechanism of Action . . . . . . . . . . . . . . . . . 18 
2.2 Warfarin Metabolic Pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . 19 
3.1 Comparative metabolic pattern of Clopidogrel and Prasugrel . . . . . . . . . . 25 
3.2 Comparative Pharmacokinetic pathway of Clopidogrel, Prasugrel and Ticagrelor 27 
4.1 Metabolism of Isoniazid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32 
5.1 Metabolism of Amytriptyline . . . . . . . . . . . . . . . . . . . . . . . . . . . 39 
5.2 Tamoxifen metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43 
5.3 Tamoxifen: Interaction with other drugs . . . . . . . . . . . . . . . . . . . . . 44 
6.1 Metabolism of 6-Mercaptopurine . . . . . . . . . . . . . . . . . . . . . . . . . 48 
6.2 6-Mercaptopurine dosing according to TPMT genotype . . . . . . . . . . . . . 49 
6.3 5-FU metabolism and Rate limiting role of DPYD enzyme . . . . . . . . . . . 50 
6.4 Illustrates some of the key enzymes involved in metabolism of MTX: RFC, 
reduced folate carrier; MTX-PGs, polyglutamated MTX; FPGS, folylpolyglu-tamate 
synthase; GGH, gamma-glutamyl hydrolase; DHF, dihydrofolate; THF, 
tetrahydrofolate; TYMS, thymidylate synthase; DHFR, dihydrofolate reduc-tase; 
MTHFR, methylenetetrahydrofolate reductase; ATIC, 5-aminoimidazole- 
4-carboxamide ribonucleotide transformylase. . . . . . . . . . . . . . . . . . . 52 
3
List of Tables 
1.1 Metabolizer’s Genotype and Phenotype in different CYP450 isoenzymes . . . . 12 
1.2 PCR mixture . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13 
1.3 PCR condition and PCR size . . . . . . . . . . . . . . . . . . . . . . . . . . . 14 
1.4 Primers for PCR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14 
2.1 Enzymatic activity of different CYP2C9 allelic variants . . . . . . . . . . . . . 20 
2.2 Genotype based Dosing guideline by Clinical Pharmacogenetics Implementa-tion 
Consortium (CPIC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21 
3.1 Common polymorphisms in the CYP2C19 gene that influence anti-platelet re-sponse 
to clopidogrel . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26 
3.2 Allelic Frequency of common CYP2C19 variants by ethnicity . . . . . . . . . 26 
4.1 Common NAT2 alleles and their phenotype . . . . . . . . . . . . . . . . . . . 34 
5.1 CYP2D6 Substrate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35 
5.2 CYP2D6 Allelic variants with enzyme function in different ethnic groups . . . 36 
5.3 Genotype and Phenotype of CYP2D6 Alleles . . . . . . . . . . . . . . . . . . 37 
5.4 Metabolizer classification of CYP2D6 . . . . . . . . . . . . . . . . . . . . . . 38 
6.1 Examples of genetic variations in major Phase-I and Phase-II enzymes as-sociated 
with changes in patient responses to chemotherapy agents. Ab-breviations: 
CYP = cytochrome P450; DPD = dihydropyrimidine dehydroge-nase; 
GST = glutathione S-transferase; MDR1 = multidrug resistance 1; MRP = 
multidrug-resistancerelated protein; MTHFR = 5,10-methylenetetrahydrofolate 
reductase; NAT = N-acetyl transferase; TPMT = thiopurine methyltransferase; 
TS = thymidylate synthase; UGT1A1 = UDP-glucuronosyltransferase 1A1; 5- 
FU = 5-fluorouracil; 6-MP = 6-mercaptopurine. . . . . . . . . . . . . . . . . . 47 
6.2 Frequency of MTHFR allelic variation among different ethnic groups . . . . . 51 
6.3 Clinical Annotation for Methotrexate in different MTHFR alleles . . . . . . . . 53 
6.4 Clinical Annotation of ABCB1 rs1045642 and 5-FU . . . . . . . . . . . . . . 54 
6.5 Clinical Annotation of ABCB1 rs1045642 and Paclitaxel . . . . . . . . . . . . 55 
4
Chapter 1 
Pharmacogenetics 
Pharmacogenetics is the study of inherited genetic differences in drug metabolic pathways 
which can affect individual responses to drugs, both in terms of therapeutic effect as well as 
adverse effects.The term pharmacogenetics is often used interchangeably with the term phar-macogenomics 
which also investigates the role of acquired and inherited genetic differences in 
relation to drug response and drug behavior through a systematic examination of genes, gene 
products, and inter- and intra-individual variation in gene expression and function. 
In oncology, pharmacogenetics historically is the study of germline mutations (e.g., single-nucleotide 
polymorphisms affecting genes coding for liver enzymes responsible for drug de-position 
and pharmacokinetics), whereas pharmacogenomics refers to somatic mutations in tu-moral 
DNA leading to alteration in drug response (e.g., KRAS mutations in patients treated 
with anti-Her1 biologics). 
Pharmacogenetics is a rising concern in clinical oncology, because the therapeutic win-dow 
of most anticancer drugs is narrow and patients with impaired ability to detoxify drugs 
will undergo life-threatenting toxicities. In particular, genetic deregulations affecting genes 
coding for DPD, UGT1A1, TPMT, CDA and Cyp2D6 are now considered as critical issues 
for patients treated with 5-FU/capecitabine, irinotecan, mercaptopurine/azathioprine, gemc-itabine/ 
capecitabine/AraC and tamoxifen, respectively. The decision to use pharmacogenetic 
techniques is influenced by the relative costs of genotyping technologies and the cost of provid-ing 
a treatment to a patient with an incompatible genotype. When available, phenotype-based 
approaches proved their usefulness while being cost-effective. 
Pharmacogenetics has several advantages: 
 The genotype of an individual is essentially invariable and remains unaffected by the 
treatment itself. 
 Molecular biology techniques provide an accurate assessment of the genotype of an indi-vidual. 
 There has been a dramatic increase in the amount of genomic information that is available. 
This information provides the necessary data for comprehensive studies of individual 
genes and broad investigation of genome-wide variation. 
 The ease of accessibility to genotype information through peripheral blood or saliva sam-pling 
and advances in molecular techniques has increased the feasibility of DNA collec-tion 
and genotyping in large-scale clinical trials. 
5
CHAPTER 1. PHARMACOGENETICS 6 
1.1 Pharmacogenetics and Drug Response 
1. Dose adaptation:Variability in drug pharmacokinetics resulting from polymorphisms af-fecting 
transport and metabolism might be alleviated, in part, by appropriate dose adjust-ments. 
Dose adjustment according to Pharmacogenetics based Diagnostics (PGDx) from 
fields of drug therapy where compensation for differences in individual drug exposure 
can be expected to have a beneficial effect on the therapeutic outcome. [Figure 1.1] 
Figure 1.1: Principle of Pharmacogenetics 
2. Drug selection using PGDx: Polymorphisms in receptors or other targets of a drug can 
affect the probability of obtaining efficacy for that drug. It is unlikely that any physician 
would restrict a patient from treatment with a drug because the chance of response might 
be only 40% in carriers of a certain PGx variant, when the average chance is 70% in 
carriers of the respective wild-type genotype. However, such PGx information might be 
helpful if alternative medications are available or if a better response might be achieved 
by a change in dose or by a specific additive medication. [Figure 1.2] 
1.2 Pharmacogenetics and Drug safety: Pharmacogenetics of 
Irinotecan 
Irinotecan is another example of a drug that is metabolized differently in some patients; these 
differences can predispose a subset of patients to increased drug toxicity. The addition of 
irinotecan to the combination of fluorouracil and leucovorin in 1996 represented the first major 
advance in the treatment of colorectal cancer since the late 1950s, when fluorouracil originally 
became the standard of care. Regimens that contained irinotecan resulted in a near-doubling 
of response rates, from around 20% to 40%, and prolongation of overall survival by up to 3 
months. These advances, however, came with the risk of significant and unpredictable toxicity. 
The most common toxicities of irinotecan are acute and delayed diarrhea and neutropenia.
CHAPTER 1. PHARMACOGENETICS 7 
Figure 1.2: Pharmacogenetics based Dosing/Diagnostics 
The drug-associated mortality from bolus fluorouracil and leucovorin regimens that con-tained 
irinotecan was threefold higher than that of regimens that did not contain the drug. Death 
often resulted from drug-related gastrointestinal and vascular syndromes. Differences between 
individuals with respect to the drug’s metabolism were a rational route of exploration because 
of the noted correlation between active drug concentration and toxicity. 
Irinotecan is primarily metabolized by carboxylesterase to the active metabolite, SN-38, 
which is responsible for inhibition of DNA topoisomerase I. SN-38 is glucuronidated by UGT1A1 
to the inactive metabolite SN-38G, which is subsequently excreted. Other irinotecan inactiva-tion 
pathways include oxidation by CYP3A4 and CYP3A5 into APC and NPC, which can also 
be metabolized by carboxylesterase to SN-38. [Figure 1.3] 
Patients with the UGT1A1*28 allele have lower glucuronidating ability than patients with 
wild-type alleles. In the Caucasian population, 917% of individuals are homozygous (7/7) 
and 2836% are heterozygous (6/7) for the UGT1A1*28 allele. In African-Americans, 1733% 
and 3850% are homozygous and heterozygous, respectively. This variant allele is seen to a 
lesser extent in the Asian population; only 14% of individuals are homozygous and 1531% are 
heterozygous for the UGT1A1*28 allele. [Figure 1.4]
CHAPTER 1. PHARMACOGENETICS 8 
Figure 1.3: Metabolism of Irinotecan. Abbreviations: APC, 7-ethyl-10[4-N-(5-aminopentanoic 
acid)-1-piperidino]-carbonyloxycamptothecin; CYP3A4, cytochrome P450 3A4 enzyme; 
CYP3A5, cytochrome P450, family 3, subfamily A, polypeptide 5; NPC, 7-ethyl-10[4- 
(1-piperidino)-1-amino]-carbonyloxycamptothecin; SN-38, 7-ethyl-10-hydroxy-camptothecin; 
SN-38G, glucuronidated SN-38; Topo 1, DNA topoisomerase I. 
Figure 1.4: Irinotecan Metabolizer and Toxicities 
1.3 Genotype Phenotype correlation: CYP3A4 
An organisms genotype is the set of genes that it carries. An organisms phenotype is all of its 
observable characteristicswhich are influenced both by its genotype and by the environment. 
So in defining evolution, we are really concerned with changes in the genotypes that make up 
a population from generation to generation. However, since an organisms genotype generally 
affects its phenotype, the phenotypes that make up the population are also likely to change.
CHAPTER 1. PHARMACOGENETICS 9 
1.3.1 Genotype 
This is the ”internally coded, inheritable information” carried by all living organisms. This 
stored information is used as a ”blueprint” or set of instructions for building and maintaining a 
living creature. These instructions are found within almost all cells (the ”internal” part), they 
are written in a coded language (the genetic code), they are copied at the time of cell division 
or reproduction and are passed from one generation to the next (”inheritable”). These instruc-tions 
are intimately involved with all aspects of the life of a cell or an organism. They control 
everything from the formation of protein macromolecules, to the regulation of metabolism and 
synthesis. 
1.3.2 Phenotype 
This is the ”outward, physical manifestation” of the organism. These are the physical parts, the 
sum of the atoms, molecules, macromolecules, cells, structures, metabolism, energy utilization, 
tissues, organs, reflexes and behaviors; anything that is part of the observable structure, function 
or behavior of a living organism. 
1.3.3 Phenotypic diversity due to heterogeneity of mutations 
Mutations that alter the structure of gene products, missense mutations, may give rise to a 
remarkable diversity of phenotypes associated with mutations at the same gene locus. One of 
the best examples comes from the inherited disorders of haemoglobin. 
Figure 1.5: Genetics of Sickle cell anaemia 
The structure of human haemoglobin (Hb) changes during embryonic, fetal and adult life. 
All normal haemoglobins are tetramers of two pairs of dissimilar globin chains. Adult and fetal 
haemoglobins have a chains combined with b (HbA a2b2) or g chains (HbF a2g2). Over 400 
structural haemoglobin variants have been identified, many of which cause no clinical disabil-ity. 
However, some, because the amino acid substitution alters the stability or function of the 
haemoglobin molecule, result in a disease phenotype. For example, the substitution of glutamic
CHAPTER 1. PHARMACOGENETICS 10 
acid for valine in the sixth position in the b chain causes the haemoglobin molecules to form 
linear stacks in the deoxy configuration which, in turn, causes the red cells to assume a sickled 
configuration. The resulting disease in homozygotes, sickle cell anaemia, is characterized by 
chronic anaemia and tissue damage due to blockage of the micro-circulation with aggregates 
of sickled red cells. Other amino acid substitutions result in instability of the haemoglobin 
molecule, which precipitates in the red cells during their life in the circulation. This results in 
damage to their membranes and increased rigidity, and hence in a variable degree of anaemia 
due to their premature destruction. 
1.4 Drug Metabolism 
Drug metabolism also known as xenobiotic metabolism is the biochemical modification of phar-maceutical 
substances or xenobiotics respectively by living organisms, usually through special-ized 
enzymatic systems. Drug metabolism often converts lipophilic chemical compounds into 
more readily excreted hydrophilic products. 
Figure 1.6: Overview of Drug Metabolism and Excretion 
In the process of metabolism, drugs will be more water soluble, therefore more accessible 
for renal excretion. During drug metabolism, sometimes toxic compounds are formed. In many 
cases, metabolism is responsible for the activation of the prodrug. This process can be divided 
into two different types of reaction. Phase-I reactions are: oxydation, reduction and hydrolysis. 
Phase-II (conjugation) reactions include sulfation, methylation, glucuronidation, acetylation. 
Both reactions-whose names do not indicate the succession of the reactions - normally make the 
originally lipophilic compound more hydrophilic. The genes coding the proteins responsible for 
these processes are usually highly polymorphic. This means that in some cases, the individual 
response after the administration of a specific drug can be attributed to the genetic background 
of the patient. It also means that in some cases, the knowledge of the patients genetic status
CHAPTER 1. PHARMACOGENETICS 11 
makes individualized therapy possible. Individualized therapy has two major goals: it might 
help not only in quickly establishing the correct dose of certain drug, but also in avoiding the 
dangerous, sometimes life-threatening side effects. 
1.5 Classification of Metabolizer groups 
Drug or other xenobiotics metabolizers can be classified according to their metabolic capacities 
and their genetic make up. 
1. Ultra Rapid Metabolizer/Ultra Metabolizer: Ultra-rapid metabolizers have one or 
more alleles which result in increased enzyme activity compared to extensive metabo-lizers. 
2. Rapid/Extensive Metabolizer: Extensive metabolizers have 2 normally functioning al-leles 
and therefore have normal enzyme activity. 
3. Intermediate Metabolizer: Intermediate metabolizers have one non-functional allele 
and one normally functioning allele, and therefore have decreased enzyme activity. 
4. Poor Metabolizer: Poor metabolizers have two non-functional alleles and therefore have 
little to no enzyme activity. 
Figure 1.7: Genotype-Phenotype Association of Drug Metabolism and Pharmacology
CHAPTER 1. PHARMACOGENETICS 12 
Table 1.1: Metabolizer’s Genotype and Phenotype in different CYP450 isoenzymes 
1.6 CYP3A4 genotype and Phenotype 
1.6.1 CYP3A4 
Cytochrome P450 3A4 (abbreviated CYP3A4) (EC 1.14.13.97), is an important enzyme in the 
body, mainly found in the liver and in the intestine. Its purpose is to oxidize small foreign 
organic molecules (xenobiotics), such as toxins or drugs, so that they can be removed from 
the body. While many drugs are deactivated by CYP3A4, there are also some drugs which are 
activated by the enzyme. Some substances, such as grapefruit juice and some drugs, interfere 
with the action of CYP3A4. These substances will therefore either amplify or weaken the action 
of those drugs that are modified by CYP3A4. 
CYP3A4 is a member of the cytochrome P450 family of oxidizing enzymes. Several other 
members of this family are also involved in drug metabolism, but CYP3A4 is the most common 
and the most versatile one. Like all members of this family, it is a hemoprotein, i.e. a protein 
containing a heme group with an iron atom. In humans, the CYP3A4 protein is encoded by 
the CYP3A4 gene. This gene is part of a cluster of cytochrome P450 genes on chromosome 
7q21.1. 
Cytochrome P450 (CYP) enzymes metabolize more than 70% of drugs for clinical use. 
Among them, CYP3A4 is quantitatively the most important P450 enzyme in adults. It is ex-pressed 
to a major extent not only in the human liver (95%) but also in the small intestine, thus 
contributing to the presystemic and systemic metabolism of 30% of all drugs. 
1.6.2 CYP3A4 genotyping 
Genotyping is normally done by two step gel electrophoresis: a) one after PCR and b) Another 
after restriction enzyme digestion. A typical PCR mixture contains: forward primer, reverse 
primer, dNTP, Taq DNA polymerase, Buffer and a template DNA. PCR condition will be initial 
denaturation of 2min at 94o, 35 cycles of 30 sec at 94o, anneling of 30sec at 60o, chain elon-
CHAPTER 1. PHARMACOGENETICS 13 
gation of 30 sec at 72o and final chain elongation for 5min and cooling forever until machine 
stops. A flowchart of CYP3A4 genotyping is given below: 
Figure 1.8: Genotyping protocol of CYP3A4 
A typical PCR mixture contains the following reagents: 
Table 1.2: PCR mixture 
1.6.3 CYP3A4 phenotyping 
CYP3A is involved in the metabolism of endogenous cortisol to 6b-hydroxy-cortisol. The 
urinary excretion of 6b-hydroxycortisol and its ratio to free cortisol reflected its activities. It
CHAPTER 1. PHARMACOGENETICS 14 
Table 1.3: PCR condition and PCR size 
Table 1.4: Primers for PCR 
has been reported that measuring the urinary morning spot of 6b-hydroxy-cortisol/cortisol ra-tio 
is a good indication of CYP3A activity. Hsieh et al. reported that, from the 6b-hydroxy-cortisol/ 
cortisol ratio, those withCYP3A4*4, *5and*6 alleles have shown below average CYP3A4 
activity implying reduced catalytic activity for the corresponding protein variants compared to 
those with no mutations shown. 
Phenotyping is normally done by measuring the ratio of 6b-hydroxycortisol (6b-OH-CS) 
and cortisol (CS) in urine by HPLC method. Extensive class group of individual will be repre-sented 
by higher cortisol metabolite and poor metabolizer will be phenotyped by lower cortisol 
metabolite concentration.
CHAPTER 1. PHARMACOGENETICS 15 
1.7 References 
1. Klotz, U. (2007). ”The role of pharmacogenetics in the metabolism of antiepileptic 
drugs: pharmacokinetic and therapeutic implications.”. Clin Pharmacokinet 46 (4): 2719. 
doi:10.2165/00003088-200746040-00001. PMID 17375979 
2. ”Center for Pharmacogenomics and Individualized Therapy”. Retrieved 2014-10-28. 
3. Roses AD (June 2000). ”Pharmacogenetics and the practice of medicine”. Nature 405 
(6788): 85765. doi:10.1038/35015728. PMID 10866212. 
4. Lazarou J, Pomeranz BH, Corey PN (April 1998). ”Incidence of adverse drug reactions 
in hospitalized patients: a meta-analysis of prospective studies”. JAMA 279 (15): 12005. 
doi:10.1001/jama.279.15.1200. PMID 9555760. 
5. Ingelman-Sundberg M, Rodriguez-Antona C (August 2005). ”Pharmacogenetics of drug-metabolizing 
enzymes: implications for a safer and more effective drug therapy”. Philos. 
Trans. R. Soc. Lond., B, Biol. Sci. 360 (1460): 156370. doi:10.1098/rstb.2005.1685. 
PMC 1569528. PMID 16096104. 
6. Kirchheiner J, Seeringer A, Viviani R (2010). ”Pharmacogenetics in psychiatry–a useful 
clinical tool or wishful thinking for the future?”. Curr. Pharm. Des. 16 (2): 13644. 
doi:10.2174/138161210790112728. PMID 20205659. 
7. AlazrakiM(2011). ”The 10 Biggest-Selling Drugs That Are About to Lose Their Patent”. 
DailyFinance. Retrieved 2012-05-06. 
8. Shuldiner AR, O’Connell JR, Bliden KP, Gandhi A, Ryan K, Horenstein RB, Damcott 
CM, Pakyz R, Tantry US, Gibson Q, Pollin TI, PostW, Parsa A, Mitchell BD, Faraday N, 
Herzog W, Gurbel PA (August 2009). ”Association of cytochrome P450 2C19 genotype 
with the antiplatelet effect and clinical efficacy of clopidogrel therapy”. JAMA 302 (8): 
84957. doi:10.1001/jama.2009.1232. PMID 19706858. 
9. The Creative Destruction of Medicine: How the Digital Revolution Will Create Better 
Health Care. New York: Basic Books. 2012. ISBN 0-465-02550-1. 
10. Farbstein D, Blum S, Pollak M, Asaf R, Viener HL, Lache O, Asleh R, Miller-Lotan 
R, Barkay I, Star M, Schwartz A, Kalet-Littman S, Ozeri D, Vaya J, Tavori H, Vardi 
M, Laor A, Bucher SE, Anbinder Y, Moskovich D, Abbas N, Perry N, Levy Y, Levy 
AP (November 2011). ”Vitamin E therapy results in a reduction in HDL function in 
individuals with diabetes and the haptoglobin 2-1 genotype”. Atherosclerosis 219 (1): 
2404. doi:10.1016/j.atherosclerosis.2011.06.005. PMC 3200506. PMID 21722898. 
11. Yang CG, Ciccolini J, Blesius A, Dahan L, Bagarry-Liegey D, Brunet C, Varoquaux A, 
Frances N, Marouani H, Giovanni A, Ferri-Dessens RM, Chefrour M, Favre R, Duffaud F, 
Seitz JF, Zanaret M, Lacarelle B, Mercier C (January 2011). ”DPD-based adaptive dosing 
of 5-FU in patients with head and neck cancer: impact on treatment efficacy and toxicity”. 
Cancer Chemother. Pharmacol. 67 (1): 4956. doi:10.1007/s00280-010-1282-4. PMID 
20204365. 
12. Malhotra AK (2010). ”The state of pharmacogenetics”. Psychiatry Times 27 (4): 3841, 
62.
CHAPTER 1. PHARMACOGENETICS 16 
13. Gardiner SJ, Begg EJ (September 2006). ”Pharmacogenetics, drug-metabolizing en-zymes, 
and clinical practice”. Pharmacol. Rev. 58 (3): 52190. doi:10.1124/pr.58.3.6. 
PMID 16968950. 
14. Genetic Science Learning Center. ”Your Doctor’s New Genetic Tools.”. Lern.Genetics. 
Retrieved 15 April 2012. 
15. ”Pharmacogenomics: Personalizing Medicine”. Discovery’s Edge. Mayo Clinic. Re-trieved 
April 15, 2012. 
16. Ge D, Fellay J, Thompson AJ, Simon JS, Shianna KV, Urban TJ, Heinzen EL, Qiu P, Ber-telsen 
AH, Muir AJ, Sulkowski M, McHutchison JG, Goldstein DB (September 2009). 
”Genetic variation in IL28B predicts hepatitis C treatment-induced viral clearance”. Na-ture 
461 (7262): 399401. doi:10.1038/nature08309. PMID 19684573. 
17. Thomas DL, Thio CL, Martin MP, Qi Y, Ge D, O’Huigin C, Kidd J, Kidd K, Khakoo 
SI, Alexander G, Goedert JJ, Kirk GD, Donfield SM, Rosen HR, Tobler LH, Busch MP, 
McHutchison JG, Goldstein DB, Carrington M (October 2009). ”Genetic variation in 
IL28B and spontaneous clearance of hepatitis C virus”. Nature 461 (7265): 798801. 
doi:10.1038/nature08463. PMC 3172006. PMID 19759533. 
18. Frueh FW, Amur S, Mummaneni P, Epstein RS, Aubert RE, DeLuca TM, Verbrugge RR, 
Burckart GJ, Lesko LJ (August 2008). ”Pharmacogenomic biomarker information in drug 
labels approved by the United States food and drug administration: prevalence of related 
drug use”. Pharmacotherapy 28 (8): 9928. doi:10.1592/phco.28.8.992. PMID 18657016. 
19. Corrigan OP (2011). ”Personalized Medicine in a Consumer Age”. Current Pharmacoge-nomics 
and Personalized Medicine 9: 168176. doi:10.2174/187569211796957566. 
20. Paul NW, Fangerau H (December 2006). ”Why should we bother? Ethical and social is-sues 
in individualized medicine”. Curr Drug Targets 7 (12): 17217. doi:10.2174/138945006779025428. 
PMID 17168846. 
21. Payne K, Shabaruddin FH (May 2010). ”Cost-effectiveness analysis in pharmacoge-nomics”. 
Pharmacogenomics 11 (5): 6436. doi:10.2217/pgs.10.45. PMID 20415553. 
22. Oxford Nanopore Technologies. 2012. Press releases - News - Oxford Nanopore Tech-nologies. 
Available from: http://www.nanoporetech.com/news/press-releases/view/39 
23. Breckenridge A, Lindpaintner K, Lipton P, McLeod H, Rothstein M,Wallace H (Septem-ber 
2004). ”Pharmacogenetics: ethical problems and solutions”. Nat. Rev. Genet. 5 (9): 
67680. doi:10.1038/nrg1431. PMID 15372090. 
24. Corrigan OP (March 2005). ”Pharmacogenetics, ethical issues: review of the Nuffield 
Council on Bioethics Report”. J. Med. Ethics 31 (3): 1448. doi:10.1136/jme.2004.007229. 
PMC 1734105. PMID 15738433.
Chapter 2 
Pharmacogenetics ofWarfarin 
2.1 Warfarin 
Warfarin (also known by the brand names Coumadin, Jantoven, Marevan, Uniwarfin) is an 
anticoagulant normally used in the prevention of thrombosis and thromboembolism, the forma-tion 
of blood clots in the blood vessels and their migration elsewhere in the body, respectively. 
Warfarin is commonly but incorrectly referred to as a blood thinner. 
Despite its effectiveness, treatment with warfarin has several shortcomings. Many com-monly 
used medications interact with warfarin, as do some foods (particularly leaf vegetable 
foods or ”greens,” since these typically contain large amounts of vitamin K1) and its activity 
has to be monitored by blood testing for the international normalized ratio (INR) to ensure an 
adequate yet safe dose is taken. A high INR predisposes to a high risk of bleeding, while an 
INR below the therapeutic target indicates the dose of warfarin is insufficient to protect against 
thromboembolic events. 
2.1.1 Warfarin Pharmacgology 
Warfarin and related 4-hydroxycoumarin-containing molecules decrease blood coagulation by 
inhibiting vitamin K epoxide reductase, an enzyme that recycles oxidized vitamin K1 to its 
reduced form after it has participated in the carboxylation of several blood coagulation proteins, 
mainly prothrombin and factor VII. Despite being labeled a vitamin K antagonist, warfarin does 
not antagonize the action of vitamin K1, but rather antagonizes vitamin K1 recycling, depleting 
active vitamin K1. 
Warfarin is used to decrease the tendency for thrombosis or as secondary prophylaxis (pre-vention 
of further episodes) in those individuals who have already formed a blood clot (throm-bus). 
Warfarin treatment can help prevent formation of future blood clots and help reduce the 
risk of embolism (migration of a thrombus to a spot where it blocks blood supply to a vital 
organ). 
Warfarin is best suited for anticoagulation (clot formation inhibition) in areas of slowly run-ning 
blood (such as in veins and the pooled blood behind artificial and natural valves) and in 
blood pooled in dysfunctional cardiac atria. Thus, common clinical indications for warfarin 
use are atrial fibrillation, the presence of artificial heart valves, deep venous thrombosis, and 
pulmonary embolism (where the embolized clots first form in veins). Warfarin is also used 
in antiphospholipid syndrome. It has been used occasionally after heart attacks (myocardial 
infarctions), but is far less effective at preventing new thromboses in coronary arteries. Preven-tion 
of clotting in arteries is usually undertaken with antiplatelet drugs, which act by a different 
17
CHAPTER 2. PHARMACOGENETICS OF WARFARIN 18 
mechanism from warfarin (which normally has no effect on platelet function). 
2.1.2 Mechanism of Action 
Warfarin inhibits the vitamin K-dependent synthesis of biologically active forms of the calcium-dependent 
clotting factors II, VII, IX and X, as well as the regulatory factors protein C, protein 
S, and protein Z. Other proteins not involved in blood clotting, such as osteocalcin, or matrix 
Gla protein, may also be affected. The precursors of these factors require carboxylation of their 
Figure 2.1: Warfarin Metabolism and Mechanism of Action 
glutamic acid residues to allow the coagulation factors to bind to phospholipid surfaces inside 
blood vessels, on the vascular endothelium. The enzyme that carries out the carboxylation of 
glutamic acid is gamma-glutamyl carboxylase. The carboxylation reaction will proceed only 
if the carboxylase enzyme is able to convert a reduced form of vitamin K (vitamin K hydro-quinone) 
to vitamin K epoxide at the same time. The vitamin K epoxide is in turn recycled back 
to vitamin K and vitamin K hydroquinone by another enzyme, the vitamin K epoxide reduc-tase 
(VKOR). Warfarin inhibits epoxide reductase (specifically the VKORC1 subunit), thereby 
diminishing available vitamin K and vitamin K hydroquinone in the tissues, which inhibits the 
carboxylation activity of the glutamyl carboxylase. When this occurs, the coagulation factors 
are no longer carboxylated at certain glutamic acid residues, and are incapable of binding to 
the endothelial surface of blood vessels, and are thus biologically inactive. As the body’s stores
CHAPTER 2. PHARMACOGENETICS OF WARFARIN 19 
of previously produced active factors degrade (over several days) and are replaced by inactive 
factors, the anticoagulation effect becomes apparent. The coagulation factors are produced, 
but have decreased functionality due to undercarboxylation; they are collectively referred to as 
PIVKAs (proteins induced [by] vitamin K absence/antagonism), and individual coagulation fac-tors 
as PIVKA-number (e.g.PIVKA-II). The end result of warfarin use, therefore, is to diminish 
blood clotting in the patient. 
2.2 Metabolism of Warfarin 
Warfarin is a natural product and given as racemic mixture of the R and S stereoisomers of 
the drug. S-warfarin is 3-5 times more potent an inhibitor of the vitamin K epoxide reductase 
complex, the target of action, than R-warfarin. The stereoisomers are metabolized by differ-ent 
phase 1 enzymes; the predominant metabolism of the S isomer is via CYP2C9 whereas 
metabolism of R-warfarin is mainly via CYP3A4 with involvement of CYP1A1, CYP1A2, 
CYP2C8, CYP2C9, CYP2C18 and CYP2C19 as depicted in the Warfarin Pharmacokinetics 
Pathway [Figure 2.2]. Phase 2 metabolism of warfarin has not been well studied and is not 
depicted in this pathway representation, although it is known that sulfated and glucuronyl con-jugates 
can be formed. Elimination is predominantly renal however warfarin has been shown to 
interact with the ABCB1 transporter in liver. 
Figure 2.2: Warfarin Metabolic Pathway 
2.3 Pharmacogenetics ofWarfarin 
The association between warfarin dose and the CYP2C9 and VKORC1genes is now unquestion-able, 
as evidenced by numerous studies. Single nucleotide polymorphisms in the cytochrome 
P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKOR) genes have been shown to have
CHAPTER 2. PHARMACOGENETICS OF WARFARIN 20 
a significant effect on warfarin dose requirement. Other genes mediating the action of warfarin 
make either little or no contribution to dose requirement. Although the polymorphisms in- 
CYP2C9andVKORC1explain a significant proportion of the interindividual variability in war-farin 
dose requirement, currently available evidence based on a few small studies relating to 
the use of pharmacogenetics-guided dosing in the initiation of warfarin therapy has not shown 
improved outcomes in either safety or efficacy of therapy. Better clinical evidence of beneficial 
effects on patient outcome, particularly at the extremes of the dose requirements in geographi-cally 
and ethnically diverse patient populations, is needed before the role of a pharmacogenomic 
approach to oral anticoagulation therapy in clinical practice can be established. 
Patients carrying CYP2C9 variant alleles had a higher rate of above-range INR values, took 
longer to reach stable dosing, and had a higher risk of serious and life-threatening bleeding 
events than patients with the wild-type allele. Similar results were noted in a study of warfarin-treated 
patients whereCYP2C92 or 3 compound heterozygotes and homozygotes had low war-farin 
requirements and increased rates of excessive (INR6.0) anticoagulation and bleeding 
com-pared with wild-type patients. 
Table 2.1: Enzymatic activity of different CYP2C9 allelic variants
CHAPTER 2. PHARMACOGENETICS OF WARFARIN 21 
2.3.1 CYP2C9 andWarfarin dosing 
CYP2C9*1 metabolizes warfarin normally, CYP2C9*2 reduces warfarin metabolism by 30%, 
and CYP2C9*3 reduces warfarin metabolism by 90%. Because warfarin given to patients with 
*2 or *3 variants will be metabolized less efficiently, the drug will remain in circulation longer, 
so lower warfarin doses will be needed to achieve anticoagulation. 
2.3.2 VKORC1 andWarfarin 
In the VKORC1 1639 (or 3673) SNP, the common G allele is replaced by the A allele. Because 
people with an A allele (or the ”A haplotype”) produce less VKORC1 than do those with the G 
allele (or the ”non-A haplotype”), lower warfarin doses are needed to inhibit VKORC1 and to 
produce an anticoagulant effect in carriers of the A allele. The prevalence of these variants also 
varies by race, with 37% of Caucasians and 14% of Africans carrying the A allele. 
2.3.3 CYP4F2 andWarfarin 
Recent genome wide association studies have not only confirmed these observations but also 
identified a novel association between rs2108622 in CYP4F2 and reduced hepatic CYP4F2, 
higher levels of hepatic vitamin K, and higher warfarin dose requirements. 
2.4 CPIC dosing guideline for Warfarin 
Approach to pharmacogenetic-based warfarin dosing without access to dosing algorithms: In 
2007, the FDA modified the warfarin label, stating that CYP2C9 and VKORC1 genotypes may 
be useful in determining the optimal initial dose of warfarin. The label was further updated in 
2010 to include a table describing recommendations for initial dosing ranges for patients with 
different combinations of CYP2C9 and VKORC1 genotypes. Genetics-based algorithms also 
better predict warfarin dose than the FDA-approved warfarin label table. Therefore, the use of 
pharmacogenetic algorithm-based dosing is recommended when possible, although if electronic 
means for such dosing are not available, the table-based dosing approaches are suggested. 
Table 2.2: Genotype based Dosing guideline by Clinical Pharmacogenetics Implementation 
Consortium (CPIC) 
2.5 References: 
1. Johnson JA, Gong L, Whirl-Carrillo M, Gage BF, Scott SA, Stein CM, Anderson JL, 
Kimmel SE, Lee MT, Pirmohamed M,Wadelius M, Klein TE, Altman RB; Clinical Phar-
CHAPTER 2. PHARMACOGENETICS OF WARFARIN 22 
macogenetics Implementation Consortium. Clinical Pharmacogenetics Implementation 
Consortium Guidelines for CYP2C9 and VKORC1 genotypes and warfarin dosing. Clin 
Pharmacol Ther. 2011 Oct;90(4):625-9. doi: 10.1038/clpt.2011.185. Epub 2011 Sep 7. 
2. Holford, NH (December 1986). ”Clinical Pharmacokinetics and Pharmacodynamics of 
Warfarin Understanding the Dose-Effect Relationship”. Clinical Pharamacokinetics (Springer 
International Publishing) 11 (6): 483504. doi:10.2165/00003088-198611060-00005. PMID 
3542339 
3. Holbrook AM, Pereira JA, Labiris R, McDonald H, Douketis JD, Crowther M, Wells PS 
(May 2005). ”Systematic overview of warfarin and its drug and food interactions”. Arch. 
Intern. Med. 165 (10): 1095106. doi:10.1001/archinte.165.10.1095. PMID 15911722 
4. Ansell J, Hirsh J, Hylek E, et al. (2008). ”Pharmacology and management of the vitamin 
K antagonists: American College of Chest Physicians evidence-based clinical practice 
guidelines (8th Edition)”. Chest 133 (6 Suppl): 160S198S. doi:10.1378/chest.08-0670. 
PMID 18574265 
5. Hirsh J, Fuster V, Ansell J, Halperin JL (2003). ”American Heart Association/American 
College of Cardiology Foundation guide to warfarin therapy”. J. Am. Coll. Cardiol. 41 
(9): 163352. doi:10.1016/S0735-1097(03)00416-9. PMID 12742309 
6. Ansell J, Jacobson A, Levy J, Vller H, Hasenkam JM (March 2005). ”Guidelines for im-plementation 
of patient self-testing and patient self-management of oral anticoagulation. 
International consensus guidelines prepared by International Self-Monitoring Association 
for Oral Anticoagulation”. Int. J. Cardiol. 99 (1): 3745. doi:10.1016/j.ijcard.2003.11.008. 
PMID 15721497 
7. Aithal GP, Day CP, Kesteven PJ, Daly AK. 1999. Association of polymorphism in the 
cytochrome P450 CYP2C9 with warfarin dose requirements and risk of bleeding compli-cations. 
Lancet353:71719 
8. Daly AK, King BP. 2003. Pharmacogenetics of oral anticoagulants.Pharmacogenetics13:24752 
9. DAndrea G, DAmbrosio RL, Di Perna P, et al. 2005. A polymorphism in the VKORC1 
gene is associated with an interindividual variability in the dose-anticoagulant effect of 
warfarin.Blood105:64549 
10. Scordo MG, Aklillu E, Yasar U, et al. 2001. Genetic polymorphism of cytochrome P450 
2C9 in a Caucasian and a black African population.Br. J. Clin. Pharmacol.52:44750
Chapter 3 
Pharmacogenetics of Clopidogrel 
3.1 Clopidogrel 
Clopidogrel (INN) is an oral, thienopyridine-class antiplatelet agent used to inhibit blood clots 
in coronary artery disease, peripheral vascular disease, cerebrovascular disease, and to prevent 
myocardial infarction (heart attack). It is marketed by Bristol-Myers Squibb and Sanofi under 
the trade name Plavix. The drug works by irreversibly inhibiting a receptor called P2Y12, 
an adenosine diphosphate (ADP) chemoreceptor on platelet cell membranes. Adverse effects 
include hemorrhage, severe neutropenia, and thrombotic thrombocytopenic purpura. 
Clopidogrel is used to prevent myocardial infarction (heart attack) and stroke in people who 
are at high risk of these events, including those with a history of myocardial infarction and other 
forms of acute coronary syndrome, stroke and those with peripheral artery disease. Treatment 
with clopidogrel or a related drug is recommended by the American Heart Association and the 
American College of Cardiology for people who: 
1. Present for treatment with a myocardial infarction with ST-elevation, including 
(a) A loading dose given in advance of percutaneous coronary intervention (PCI), fol-lowed 
by a full year of treatment for those receiving a vascular stent 
(b) A loading dose given in advance of fibrinolytic therapy, continued for at least 14 
days 
2. Present for treatment of a non-ST elevation myocardial infarction (NSTEMI) or unstable 
angina. 
(a) Including a loading dose and maintenance therapy in those receiving PCI and unable 
to tolerate aspirin therapy 
(b) Maintenance therapy for up to 12 months in those at medium to high risk for which 
a non-invasive treatment strategy is chosen 
3. In those with stable ischemic heart disease,[4] treatment with clopidogrel is described as a 
”reasonable” option for monotherapy in those who cannot tolerate aspirin, as is treatment 
with clopidogrel in combination with aspirin in certain high risk patients. 
3.1.1 Pharmacology 
Clopidogrel is a prodrug, which requires CYP2C19 for its activation. It acts on the ADP recep-tor 
on platelet cell membranes. The drug specifically and irreversibly inhibits the P2Y12 subtype 
23
CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 24 
of ADP receptor, which is important in activation of platelets and eventual cross-linking by the 
protein fibrin. Platelet inhibition can be demonstrated two hours after a single dose of oral clopi-dogrel, 
but the onset of action is slow, so a loading dose of either 600 or 300 mg is administered 
when a rapid effect is needed. 
3.1.2 Why it is important to study CYP2C19 polymorphism 
1. Antiplatelet drugs reduce the risk of heart attack, unstable angina, stroke, and cardiovas-cular 
death in patients with coronary heart disease by making platelets less likely to form 
blood clots. 
2. Antiplatelet drug does not have its antiplatelet effects until it is metabolized into its active 
form by CYP2C19. 
3. Study by De Morais et al, 1994 detects the *17 allele as an ultra fast metabolizer of 
antiplatelet drugs. A recent report indicates the increased risk for bleeding in individuals 
treated with clopidogrel who carry the *17 allele. 
4. People who have reduced functioning of their CYP2C19 liver enzyme due to inactivating 
polymorphisms in their CYP2C19 gene cannot effectively convert Plavix to its active 
form. As a result, Plavix may be less effective in altering platelet activity in those people. 
These poor metabolizers may not receive the full benefit of Plavix treatment and may 
remain at risk for heart attack, stroke, and cardiovascular death. 
5. It is estimated that 1.5% to 19.2% of the Asian population are ultra fast metabolizers. 
3.2 Bioactivation of Clopidogrel and Prasugrel 
Clopidogrel is a prodrug, and its activation is complex (Figure 3.1). Some 85% of the dose 
administered is hydrolyzed in the liver, where it is converted into an inactive metabolite (SR 
26334); the remainder is metabolized by CYP2C19 and to a lesser degree by CYP1A2 and 
CYP2B6 to 2-oxo-clopidogrel. Around half of this metabolite, which is also inactive, is hy-drolyzed 
and converted into an inactive thiolactone, while the other half is metabolized by 
the 3A4, 3A5, 2B6, 2C9 and 2C19 isoenzymes, finally producing the active metabolite (R- 
130964). The fact that a significant proportion of the absorbed drug is wasted (converted to 
inactive metabolites), the complex activation of the prodrug requiring two oxidation steps, and 
its heavy dependence on the function of the CYP2C19 isoenzyme (which is involved in the me-tabolization 
of numerous drugs and whose reduced-function genetic variants have a prevalence 
of around 25%), are the main pharmacokinetic factors affecting the variability of response to 
clopidogrel. 
3.3 Clopidogrel and CYP2C19 
One of the main causes of this clopidogrel nonresponsiveness is interindividual variability in the 
metabolic activation of clopidogrel. Polymorphisms in the CYP2C19 gene affect clopidogrel 
pharmacokinetics, causing the CYP2C19 enzyme that converts Plavix to its active form to be 
more or less effective. The most common CYP2C19 polymorphisms,*2, *3, and *17, result 
in highly variable enzyme activity, and are used to classify individuals as to metabolizer type:
CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 25 
Figure 3.1: Comparative metabolic pattern of Clopidogrel and Prasugrel 
poor, intermediate, extensive (normal), or ultrarapid. These polymorphisms may be useful to 
identify clopidogrel nonresponders (carrying the *2 or *3 alleles) who may benefit by taking an 
alternative antiplatelet agent, such as prasugrel and ticagrelor, or individuals who may be at risk 
of bleeding due to clopidogrel overactivity (*17 allele carriers). 
A loss-of-function polymorphism in the CYP2C19 gene, known as the CYP2C19*2 allelic 
variant, has been associated with higher levels of ADP-induced platelet aggregation values in 
clopidogrel-treated patients and consequently a higher risk of major adverse cardio vascular 
events, including the occurrence of stent thrombosis (ST). 
A novel allelic variant, CYP2C19*17, results in an increased enzyme function of CYP2C19 
because of a mutation (806CT) in the 5-flanking region of the gene that causes an increased 
transcription of CYP2C19. Such increased transcriptional activity of CYP2C19 may confer 
a rapid metabolization of CYP2C19 substrates, which may lead to an enhanced response to 
antiplatelet treatment. Although this may improve the prevention of thrombotic events, it also 
may increase the risk of bleeding. 
3.4 Genetic variation in CYP2C19 influences individual re-sponse 
to clopidogrel 
The CYP2C19 gene is highly polymorphic with over 25 known variants. Functional polymor-phisms 
in the CYP2C19 gene, the most common of which are *2, *3, and *17, result in highly 
variable enzyme activity, influencing an individuals ability to activate clopidogrel. These vari-ants 
cause the CYP2C19 enzyme that converts Plavix to its active form to be more or less 
effective and are used to classify individuals as to metabolizer type: extensive metabolizers
CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 26 
(normal conversion to the active metabolite), intermediate/poor metabolizers (incomplete con-version 
to the active metabolite), and ultrarapid metabolizers (increased conversion to the active 
metabolite). 
Table 3.1: Common polymorphisms in the CYP2C19 gene that influence anti-platelet response 
to clopidogrel 
Approximately 2-4% of Caucasian, 4% of African or African/American, and 14-20% of 
Asian populations are poor metabolizers (PM) because these individuals have two loss-of-function 
alleles and very low CYP2C19 activity. Intermediate metabolizers have one CYP2C19 
loss-of-function allele. Approximately 30-50% of the population are intermediate metabolizers 
( 30% of Caucasians, 40% of African-Americans, and more than 55% of Asians). 
Table 3.2: Allelic Frequency of common CYP2C19 variants by ethnicity 
3.5 Choosing the best option: Clopidogrel? Prasugrel? or 
Ticagrelor? 
Prasugrel and ticagrelor are newer antiplatelet drugs that could be an alternative to clopidogrel. 
Their effect is less variable and fewer ischemic events are associated with their use; however,
CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 27 
more bleeding is related to the use of these drugs. Pharmacogenetic studies are currently con-sidering 
these drugs. So far, no association has been found. 
Prasugrel are found to be a risk factor for intracranial bleeding resulting in strokes in around 
2.5% population. CYP2C19 variants shows differences in pharmacodynamic and Pharmacoki-netic 
properties variation of Clopidogrel. 
Figure 3.2: Comparative Pharmacokinetic pathway of Clopidogrel, Prasugrel and Ticagrelor 
Clopidogrel is activated in a 2-step process mediated by oxidative biotransformation in the 
liver, in which CYP2C19 and CYP3A have particularly important roles. The parent com-pound 
clopidogrel, and to a lesser extent 2-oxo-clopidogrel, are both substrates and inhibitors 
of CYP1A2, CYP2B6, and CYP2C19. Clopidogrel and 2-oxo-clopidogrel are extensively hy-drolyzed 
to inactive metabolites, potentially magnifying the effects of CYP2C19 inhibitors and
CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 28 
polymorphisms. 
Prasugrel is also a pro-drug that requires biotransformation to active metabolites by cy-tochrome 
P-450 enzymes, including CYP3A isoforms, CYP2B6, CYP2C9, and CYP2C19. 
Prasugrel is hydrolyzed to a thiolactone derivative in the intestine and then oxidized to its ac-tive 
metabolite in both the intestine and the liver. Reduced-function CYP2C19 alleles are not 
believed to have a clinically meaningful effect in prasugrel-treated patients. 
Ticagrelor (AZD6140) is an orally active cyclopentyltriazolopyrimidine adenosine triphos-phate 
analog that reversibly inhibits P2Y12 platelet receptors. Ticagrelor, which is not yet 
approved in the United States, is an active compound and is metabolized by CYP3A4 to an 
active metabolite. Ticagrelor and its active metabolite are both metabolized and glucuronidated 
in the liver before elimination in the urine. Genetic variations in CYP isoenzymes do not appear 
to affect metabolism of ticagrelor. 
3.6 References 
1. Desta Z, Zhao X, Shin JG, Flockhart DA. Clinical significance of the cytochrome P450 
2C19 genetic polymorphism. Clin Pharmacokinet. 2002;41(12):913-958. 
2. Campo G, Fileti L, Valgimigli M, et al. Poor response to clopidogrel: current and future 
options for its management. J Thromb Thrombolysis. 2010;30(3):319-31. 
3. Gurbel PA, Tantry US. Selecting optimal antiplatelet therapy based on platelet function 
monitoring in patients with coronary artery disease. Curr Treat Options Cardiovasc Med. 
2009;11(1):22-32. 
4. Kuliczkowski W, Witkowski A, Polonski L, et al. Interindividual variability in the re-sponse 
to oral antiplatelet drugs: a position paper of the Working Group on antiplatelet 
drugs resistance appointed by the Section of Cardiovascular Interventions of the Polish 
Cardiac Society, endorsed by theWorking Group on Thrombosis of the European Society 
of Cardiology. Eur Heart J. 2009;30(4):426-435. 
5. Terpening C. Clopidogrel: A pharmacogenomic perspective on its use in coronary artery 
disease. Clin Med Insights Cardiol. 2010;4:117-128. 
6. Sharma RK, Reddy HK, Singh VN, et al. Aspirin and clopidogrel hyporesponsiveness 
and nonresponsiveness in patients with coronary artery stenting. Vasc Health Risk Manag. 
2009;5:965-972. 
7. Simon T, Verstuyft C, Mary-Krause M, et al. Genetic determinants of response to clopi-dogrel 
and cardiovascular events. New Engl J Med. 2009;360(4):363-375. 
8. Sofi F, Marcucci R, Gori AM, Abbate R, Gensini GF. Clopidogrel non-responsiveness 
and risk of cardiovascular morbidity. An updated meta-analysis. Thromb Haemost. 
2010;103(4):841-848. 
9. Hulot J, Bura A, Villard E, Azizi M, et al. P450 2C19 loss-of-function polymorphism. 
Cytochrome is a major determinant of clopidogrel responsiveness in healthy subjects. 
Blood. 2006;108(7):2244-2247.
CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 29 
10. Geisler T, Schaeffeler E, Dipporr J, et al. CYP2C19 and nongenetic factors predict poor 
responsiveness to clopidogrel loading dose after coronary stent implantation. Pharma-cogenomics. 
2008;9(9):1251-1259. 
11. Mega JL, Close SL, Wiviott SD, et al. Cytochrome P-450 polymorphisms and response 
to clopidogrel. N Engl J Med. 2009;360(4):354-362. 
12. http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationfor Patientsand- 
Providers/ucm203888.htm. (Accessed Jun 8, 2011). 
13. Product information for Plavix (Sanofi/Aventis US). Label information, approved Feb 
2011. (htt p : ==www:accessdata: f da:gov=drugsat f dadocs=label=2011=020839s052lbl:) 
14. Kazui M, Nishiya Y, Ishizuka T, et al. Identification of the human cytochrome P450 
enzymes involved in the two oxidative steps in the bioactivation of clopidogrel to its 
pharmacologically active metabolite. Drug Metab Dispos. 2010;38(1):92-99. 
15. Dahl M, Gunes A. Implications of inter-individual differences in clopidogrel metabolism, 
with Focus on Pharmacogenetics. Pharmaceuticals. 2010;3(4):782-794. 
16. Pare G, Mehta SR, Yusuf S, et al. Effects of CYP2C19 genotype on outcomes of clopi-dogrel 
treatment. N Engl J Med. 2010;363(18):1704-1714. 
17. Sim SC, Risinger C, Dahl ML, et al. A common novel CYP2C19 gene variant causes 
ultrarapid drug metabolism relevant for the drug response to proton pump inhibitors and 
antidepressants. Clin Pharmacol Ther. 2006;79(1):103-113. 
18. Li-wan-po A, Girard T, Farndon P, Cooley C, Lithgow J. Pharmacogenetics of CYP2C19: 
functional and clinical implications of a new variant CYP2C19*17. Br J Clin Pharmacol. 
2010;69(3):222-230. 
19. Shuldiner AR, OConnell JR, Bliden KP, Ghandhi A, et al. Association of cytochrome 
P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel ther-apy. 
JAMA. 2009;302(8):849-858. 
20. Human cytochrome P450 allele nomenclature (htt p : ==www:cypalleles:ki:se=cyp2c19:htm) 
21. Mega, J.L. et al. Reduced-function CYP2C19 genotype and risk of adverse clinical out-comes 
among patients treated with clopidogrel predominantly for PCI: a meta-analysis. 
JAMA. 2010;304(16):1821-1830. 
22. Mega JL, Hochholzer W, Frelinger AL, Kluk MJ, et al. Dosing clopidogrel based on 
CYP2C19 genotype and the effect on platelet reactivity in patients with stable cardiovas-cular 
disease. JAMA. 2011;306(20):2221-2228. 
23. Trenk D, Hochholzer W, Fromm MF, Chialda L, et al. Cytochrome P450 2C19 681G¿A 
polymorphism and high on-clopidogrel platelet reactivity associated with adverse 1-Year 
clinical outcome of elective percutaneous coronary intervention with drug-eluting or bare-metal 
stents. J Am Coll Cardiol. 2008;51(20):1925-1934. 
24. Giusti B, Goria AM, Marcuccia R, et al. Cytochrome P450 2C19 loss-of-function poly-morphism, 
but not CYP3A4 IVS10 + 12G/A and P2Y12 T744C polymorphisms, is as-sociated 
with response variability to dual antiplatelet treatment in high-risk vascular pa-tients. 
Pharmacogenet Genomics. 2007;17:1057-1064.
Chapter 4 
Pharmacogenetics of NAT2 
The arylamine N-acetyltransferases (NATs) are found in nearly all species from bacteria to 
humans. They catalyse the acetyltransfer from acetylcoenzyme A to an aromatic amine, hete-rocyclic 
amine or hydrazine compound. In humans, acetylation is a major route of biotransfor-mation 
for many arylamine and hydrazine drugs, as well as for a number of known carcinogens 
present in the diet, cigarette smoke and the environment. The reaction pathway is catalysed by 
two cytoplasmic acetyltransferases (NAT; EC 2.3.1.5), N-acetyltransferase Type I (NAT1) and 
N-acetyltransferase Type II (NAT2). 
The arylamine N-acetyltransferases (NATs) are involved in the metabolism of a variety of 
different compounds that we are exposed to on a daily basis. Many drugs and chemicals found 
in the environment, such as those in cigarette smoke, car exhaust fumes and in foodstuffs, can 
be either detoxified by NATs and eliminated from the body or bioactivated to metabolites that 
have the potential to cause toxicity and/or cancer. NATs have been implicated in some adverse 
drug reactions and as risk factors for several different types of cancers. As a result, the levels of 
NATs in the body have important consequences with regard to an individual’s susceptibility to 
certain drug-induced toxicities and cancers. 
4.1 NAT localization 
Two NAT isoenzymes have been identified in humans, namely NAT1 and NAT2, which are the 
products of distinct genetic loci, designated NAT1 and NAT2, respectively. A related pseu-dogene, 
NATP1, has also been identified, which contains multiple frameshift and nonsense 
mutations. The two functional NAT genes share an 87% nucleotide identity, which translates 
to an 81% homology at the amino acid level. While the entire transcript of NAT1 is derived 
from a single exon, that of NAT2 is derived from the protein encoding exon together with a sec-ond 
noncoding exon of 100 bp located about 8kb upstream of the translation start site. Human 
NAT1 and NAT2, as well as NATP1, have been localised to the short arm of chromosome 8, 
more specifically in region 8p22. The NAT loci are separated by only 170-360kb and are in the 
orientation NAT1!NATP1!NAT2. 
4.2 Human NAT2 Alleles 
Since the human NAT2 locus was established as the site of the classical acetylation polymor-phism, 
the study of NAT2 allelic variation has been an area of intense investigation. To date, 29 
different NAT2 alleles have been detected in human populations. Each of the variant alleles is 
30
CHAPTER 4. PHARMACOGENETICS OF NAT2 31 
comprised of between one and four nucleotide substitutions, of which 13 have been identified, 
located in the protein encoding region of the gene. 
Nine of these lead to a change in the encoded amino acid (C190T, G191A, T341C, A434C, 
G499A, G590A, A803G, A845C, and G857A), while the remaining four are silent (T111C, 
C282T, C481T, and C759T). 
Several studies have been performed that show clear correlations between NAT2 genotype 
and phenotype. Early genotyping studies screened for the presence of the C481T (M1), the 
G590A (M2), the G857A (M3) and sometimes the G191A (M4) nucleotide changes, all of 
which were shown to cause a slow acetylation phenotype. Moreover, there was a gene-dosage 
effect. Individuals who were homozygous for NAT2 polymorphisms had a slow acetylator 
phenotype, individuals heterozygous for NAT2 polymorphisms had an intermediate acetylator 
phenotype, and individuals who lacked NAT2 polymorphisms had a rapid acetylator phenotype. 
It should be noted that the method of detection of the above polymorphisms only identifies a 
subset of the variant alleles found in human populations, and there is potential for the misclas-sification 
of genotype and deduced phenotypes. 
4.3 NAT and Disease 
The association between acetylator status and the risk of various diseases has been extensively 
reported, and reviewed in detail. Altered risk with either the slow or rapid phenotype has been 
observed for bladder, colon and breast cancer, systemic lupus erythematosis, diabetes, Gilbert’s 
disease, Parkinson’s disease and Alzheimer’s disease. These associations imply a role for envi-ronmental 
factors that are metabolised by the NATs, in particular NAT2, in each disorder. How-ever, 
identifying those factors has remained elusive. Humans are exposed to many toxic NAT 
substrates including the food-derived heterocyclics present in the diet as well as arylamines such 
as 4-aminobiphenyl and -naphthylamine present in tobacco smoke. Moreover, occupational ex-posure 
to arylamine carcinogens such as benzidine has also been reported. 
Because of the role of acetylation in the metabolic activation and detoxification of ary-lamine 
and heterocyclic carcinogens, acetylator status and cancer risk has been widely investi-gated. 
Unlike the relatively rare but highly penetrant genes involved in familial cancers, those 
genes responsible for metabolic polymorphisms have low penetrance and cause only a moder-ate 
increase in cancer risk. Nevertheless, their widespread occurrence in the general population 
suggests they are a significant contributor to individual risk. 
When acetylator phenotype has been linked to carcinogen exposure, more consistent results 
have been reported. For example, the rapid phenotype has emerged as a strong risk factor for 
colorectal cancer in those individuals who have a higher exposure to the food-derived hete-rocyclic 
amines. Recently, the NAT2 acetylator phenotype has been linked to increased risk 
associated with neurodegenerative diseases such as Parkinson’s disease and Alzheimer’s dis-ease. 
4.4 NAT2 and Drug Response 
4.4.1 NAT2 and Isoniazid 
The genetic polymorphism in N-acetyltransferase activity was first discovered in patients treated 
with isoniazid for tuberculosis. This drug is primarily excreted following acetylation catalysed 
by NAT2.
CHAPTER 4. PHARMACOGENETICS OF NAT2 32 
Isoniazid (INH), a key drug of antituberculosis therapy, is metabolized by arylamine N-acetyltransferase2 
(NAT2), cytochrome P450 2E1 (CYP2E1) and glutathione S-transferase (GST). 
The metabolism of INH into acetyl isoniazid (AcINH) by N-acetyltransferase 2 (NAT2), which 
is estimated to account for 50-90% of INH metabolism. A portion of AcINH is further con-verted 
to acetyl hydrazine (AcHz) by amidase-catalyzed hydrolysis. There is another amidase-catalyzed 
route where INH is directly hydrolyzed to yield hydrazine (Hz). NAT2 then catalyzes 
the acetylation of Hz to yield AcHz and subsequently to diacetyl hydrazine before excretion. 
AcHz can also enter the cytochrome P450 2E1 (CYP2E1)-mediated metabolic pathway in the 
liver, which is linked to the glutathione-S-transferase (GST)-mediated metabolic pathway, to 
become an excretable metabolite. 
Figure 4.1: Metabolism of Isoniazid 
NAT2 genetic polymorphisms and the resulting variation in acetylation capacity of NAT2 
are implicated in the risk of INH-induced hepatotoxicity. Slow acetylator genotype of NAT2 
will activate the CYP2E1 pathway resulting in reactive metabolite, hence will cause hepatotox-icity 
(Hepatic Necrosis). But there are some contradictory result also, like RA (rapid or fast 
acetylator) genotype also have a significant role in INH induced hepatotoxicity. Hydrazine and 
N-actyl Hydrazine are supposed to be hepatotoxic so are RA groups. So it is necessary to con-sider 
the role of CYP2E1 and GST in INH metabolism to draw a conclusion here without any 
conflict. 
4.4.2 NAT2 and Environmental carcinogen 
Biological Amines are not reactive and converted into a highly electrophilic reactive metabolite, 
acts as a carcinogen and later detoxified by the action of NAT2 enzymes into an excretable non-
CHAPTER 4. PHARMACOGENETICS OF NAT2 33 
reactive form. Slow-acetylator phenotype should have a higher risk of developing cancer by 
this biological amine than in fast-acetylator phenotype. 
4.4.3 NAT2 and Procainamide 
Procainamide is a class Ia antiarrhythmic used for the treatment of several different arrhyth-mias. 
It is metabolized by n-acetyltransferases (NAT) to n-acetylprocainamide (NAPA), an ac-tive 
metabolite, which also possesses antiarrhythmic properties. NAT1 is consistently expressed 
in post patients; however, NAT2 is variably expressed and plays a major role in the production 
of NAPA. The majority of patients reveiving procainamide therapy will develop autoantibodies 
over time and possibly drug induced lupus. An early study showed that subjects who had a 
slow-acteylator phenotype developed antinuclear antibodies earlier than rapid acetylators. This 
was later confirmed in a study of subjects who received long term procainamide therapy. The-oretically 
patients with NAT2 genotype associated with rapid-acetylation may have increased 
NAPA concentrations and thus increased anti-arrhythmic effects and possible excessive QT 
prolongation. 
4.5 References 
1. DWHein. N-acetyltransferase 2 genetic polymorphism: effects of carcinogen and haplo-type 
on urinary bladder cancer risk. Oncogene (2006) 25, 16491658. doi:10.1038/sj.onc.1209374 
2. Alexandra King. NAT2 and GSTM1 polymorphisms affect the risk of bladder cancer. 
Nature Reviews Clinical Oncology 2, 540 (November 2005)—doi:10.1038/ncponc0307 
3. T SOTSUKA, Y SASAKI, S HIRAI, F YAMAGISHI and K UENO. Association of 
Isoniazid-metabolizing Enzyme Genotypes and Isoniazid-induced Hepatotoxicity in Tu-berculosis 
Patients. In Vivo. 2011. 25(5): 803-812. 
4. Evans DA (1989). ”N-acetyltransferase”. Pharmacology  Therapeutics 42 (2): 157234. 
doi:10.1016/0163-7258(89)90036-3. PMID 2664821.
CHAPTER 4. PHARMACOGENETICS OF NAT2 34 
Table 4.1: Common NAT2 alleles and their phenotype
Chapter 5 
Pharmacogenetics of CYP2D6 
5.1 CYP2D6 allelic variants 
More than 50 human CYP isozymes have been identified to date. Of these, more than 20 are 
encoded by genes that are functionally polymorphic, including CYP2A6, CYP2C9, CYP2C19, 
and CYP2D6. Consequently, approximately 40% of CYP-dependent drug metabolism is carried 
out by polymorphic enzymes. 
Table 5.1: CYP2D6 Substrate 
CYP2D6 is the best characterized of these polymorphic CYP isozymes, with more than 75 
allelic variants currently identified. These variants result from point mutations, deletions or 
additions, gene rearrangements, and deletion or duplication of the entire gene, and result in an 
increase, reduction, or complete loss of activity. While it accounts for only 2%5% of all hepatic 
CYP isozymes, CYP2D6 metabolizes approximately 25% of all clinically used medications, 
including some cytotoxics, tamoxifen, and many agents used to treat associated complications 
such as antiarrythmics, antiemetics, antidepressants, antipsychotics, and analgesics. Since the 
identification of the CYP2D6 polymorphism in the 1970s, several studies have shown that the 
frequencies of the alternative CYP2D6 phenotypes vary significantly among different ethnic 
35
CHAPTER 5. PHARMACOGENETICS OF CYP2D6 36 
groups and that such polymorphisms may play a role in the induction of adverse effects from 
administration of some therapeutic agents. Variation in CYP2D6 expression is also thought 
to increase the potential for drugdrug interactions, an important implication because of the 
increasing number of drugs being prescribed. For example, in the oncology setting, patients 
with advanced cancer receiving palliative therapy receive an average of five or more medications 
for symptom relief. 
Table 5.2: CYP2D6 Allelic variants with enzyme function in different ethnic groups 
A number of specific polymorphisms have been found in the CYP2D6 gene that results in 
enzymatic deficiencies. The frequency of these polymorphisms varies within the major ethnic 
groups. CYP2D6 polymorphisms that produce poor metabolizers are found with frequencies of 
7% to 10% in Caucasians, 2% in Africans and African Americans, and 1% in Asians. Identifica-tion 
of patient CYP2D6 genotypes can help clinicians individualize drug treatment by selection 
of appropriate therapies. These measures may improve patient outcome by ensuring maximum 
drug efficacy with minimal adverse drug reactions (ADR).
CHAPTER 5. PHARMACOGENETICS OF CYP2D6 37 
Table 5.3: Genotype and Phenotype of CYP2D6 Alleles 
CYP2D6 polymorphisms can be classified according to one of four levels of activity: poor 
metabolizers (PMs), intermediate metabolizers (IMs), extensive metabolizers (EMs), and ul-trarapid 
metabolizers (UMs) [Figure 5.4]. The EM phenotype is expressed by the majority of 
the population and is therefore considered the norm. PMs inherit two deficient CYP2D6 alleles 
and, as a result, metabolize drugs at a notably slower rate. This leads to an accumulation of high 
levels of unmetabolized drugs that are CYP2D6 substrates, a concomitant greater potential for 
adverse events and drugdrug interactions, and lower efficacy for drugs requiring CYP2D6 acti-vation. 
The UM phenotype is caused by the duplication, multiduplication, or amplification of 
active CYP2D6 genes, including primarily the CYP2D6*2 allele, but also involving CYP2D6*1 
and others. Individuals with the UM phenotype metabolize drugs at an ultrarapid rate, which 
may lead to a loss of therapeutic efficacy at standard doses. Individuals who are heterozygous 
for a defective CYP2D6 allele often demonstrate an IM phenotype. This phenotype has a wide 
spectrum of metabolic activity that can range from marginally better than the PM phenotype to 
activity that is close to that of the EM phenotype. 
5.2 CYP2D6 and Antipsychotics Pharmacology 
CYP2D6 is described as the most relevant enzyme in the metabolism of many antipsychotic 
drugs. Its contribution to the interindividual differences in drug response is reviewed here high-lighting 
its role in the kinetics of antipsychotic drugs and the occurrence of drug interactions. 
The activity of CYP2D6 is inherited as a monogenetic trait and the CYP2D6 gene appears 
highly polymorphic in humans. The polymorphic alleles may lead to altered activity of the
CHAPTER 5. PHARMACOGENETICS OF CYP2D6 38 
Table 5.4: Metabolizer classification of CYP2D6 
CYP enzymes causing absent, decreased (poor), or increased (ultrarapid) metabolism that in 
turn influence the disposition of the antipsychotic drugs. Antipsychotic drug biotransformation 
is mainly determined by genetic factors mediating CYP2D6 gene polymorphism, however the 
importance of environmental factors (dietary, smoking, diseases, etc.) is also recognized. Ad-ditionally, 
the potential interaction between CYP2D6 and the endogenous metabolism must be 
taken into consideration. 
5.2.1 Metabolism of Amytriptyline 
Amytriptyline (AT) is metabolized mainly by demethylation forming nortriptyline (NT), and by 
hydroxylation, leading to the formation of E-10-hydroxy (EHAT) and Z-10-hydroxyamitriptyline 
(ZHAT). NT is further demethylated to desmethylnortriptyline (NNT) and hydroxylated to E- 
10-hydroxy (EHNT) and Z-10-hydroxynortriptyline (ZHNT). The demethylation of AT and NT 
is mainly catalyzed by CYP2C19, with the participation of other CYP enzyme forms in higher 
drug concentrations. The formation of the E-10-hydroxy metabolites is dependent on the activ-ity 
of CYP2D6, with stereospecificity to the (-)-EHAT and (-)-EHNT metabolites. 
5.2.2 Dosing recommendations for amitriptyline/nortriptyline based on 
CYP2D6 phenotype 
1. Likely phenotype: Ultrarapid metabolizer ( 1-2% of patients)
CHAPTER 5. PHARMACOGENETICS OF CYP2D6 39 
Figure 5.1: Metabolism of Amytriptyline 
(a) Genotypes: An individual carrying more than two copies of functional alleles. 
(b) Examples of diplotypes: *1/*1xN, *1/*2xN 
(c) Implications for TCA metabolism: Increased metabolism of tricyclics to less ac-tive 
compounds when compared to extensive metabolizers. Lower plasma concen-trations 
will increase probability of pharmacotherapy failure. 
(d) Therapeutic Recommendations: Avoid tricyclic use due to potential lack of effi-cacy. 
Consider alternative drug not metabolized by CYP2D6. If a tricyclic is war-ranted, 
consider increasing the starting dose.a Utilize therapeutic drug monitoring 
to guide dose adjustments. 
(e) Classification of recommendation for amitriptyline/nortriptyline therapy: Strong 
2. Likely phenotype: Extensive metabolizer ( 77-92% of patients) 
(a) Genotypes: An individual carrying two alleles encoding full or reduced function 
or one full function allele together with either one nonfunctional or one reduced-function 
allele. 
(b) Examples of diplotypes: *1/*1, *1/*2, *2/*2, *1/*41, *1/*4, *2/*5, *10/*10 
(c) Implications for TCA metabolism: Normal metabolism of tricyclics.
CHAPTER 5. PHARMACOGENETICS OF CYP2D6 40 
(d) Therapeutic Recommendations: Initiate therapy with recommended starting dose. 
(e) Classification of recommendation for amitriptyline/nortriptyline therapy: Strong 
3. Likely phenotype: Intermediate metabolizer ( 2-11% of patients) 
(a) Genotypes: An individual carrying one reduced and one nonfunctional allele, 
(b) Examples of diplotypes: *4/*10, *5/*41 
(c) Implications for TCA metabolism: Reduced metabolism of tricyclics to less ac-tive 
compounds when compared to extensive metabolizers. Higher plasma concen-trations 
will increase the probability of side effects. 
(d) Therapeutic Recommendations: Consider 25% reduction of recommended start-ing 
dose. Utilize therapeutic drug monitoring to guide dose adjustments. 
(e) Classification of recommendation for amitriptyline/nortriptyline therapy: Mod-erate 
4. Likely phenotype: Poor metabolizer ( 5-10% of patients) 
(a) Genotypes: An individual carrying no functional alleles. 
(b) Examples of diplotypes: *4/*4, *4/*5, *5/*5, *4/*6 
(c) Implications for TCA metabolism: Greatly reduced metabolism of tricyclics to 
less active compounds when compared to extensive metabolizers. Higher plasma 
concentrations will increase the probability of side effects. 
(d) Therapeutic Recommendations: Avoid tricyclic use due to potential for side ef-fects. 
Consider alternative drug not metabolized by CYP2D6. If a tricyclic is war-ranted, 
consider 50% reduction of recommended starting dose. Utilize therapeutic 
drug monitoring to guide dose adjustments. 
(e) Classification of recommendation for amitriptyline/nortriptyline therapy: Strong 
5.2.3 Recommended dosing of codeine by CYP2D6 Phenotype 
1. Likely phenotype: Ultrarapid metabolizer ( 1-2% of patients) 
(a) Genotypes: An individual carrying more than two copies of functional alleles. 
(b) Examples of diplotypes: *1/*1xN, *1/*2xN 
(c) Implications for codeine metabolism: Increased formation of morphine following 
codeine administration, leading to higher risk of toxicity. 
(d) Therapeutic Recommendations: Avoid codeine use due to potential for toxicity. 
(e) Classification of recommendation for codeine therapy: Strong 
(f) Considerations for alternative opioids: Alternatives that are not affected by this 
CYP2D6 phenotype include morphine and non-opioid analgesics. Tramadol, and to 
a lesser extent hydrocodone and oxycodone, are not good alternatives because their 
metabolism is affected by CYP2D6 activity. 
2. Likely phenotype: Extensive metabolizer ( 77-92% of patients)
CHAPTER 5. PHARMACOGENETICS OF CYP2D6 41 
(a) Genotypes: An individual carrying two alleles encoding full or reduced function 
or one full function allele together with either one nonfunctional or one reduced-function 
allele. 
(b) Examples of diplotypes: *1/*1, *1/*2, *2/*2, *1/*41, *1/*4, *2/*5, *10/*10 
(c) Implications for codeine metabolism: Normal morphine formation 
(d) Therapeutic Recommendations: Use label recommended age- or weight-specific 
dosing. 
(e) Classification of recommendation for codeine therapy: Strong 
(f) Considerations for alternative opioids: Not required. 
3. Likely phenotype: Intermediate metabolizer ( 2-11% of patients) 
(a) Genotypes: An individual carrying one reduced and one nonfunctional allele. 
(b) Examples of diplotypes: *4/*10, *5/*41 
(c) Implications for codeine metabolism: Reduced morphine formation. 
(d) Therapeutic Recommendations: Use label recommended age- or weight-specific 
dosing. If no response, consider alternative analgesics such as morphine or a non-opioid. 
(e) Classification of recommendation for codeine therapy: Moderate 
(f) Considerations for alternative opioids: Monitor tramadol use for response. 
4. Likely phenotype: Poor metabolizer ( 5-10% of patients) 
(a) Genotypes: An individual carrying no functional alleles. 
(b) Examples of diplotypes: *4/*4, *4/*5, *5/*5, *4/*6 
(c) Implications for codeine metabolism: Greatly reduced morphine formation fol-lowing 
codeine administration, leading to insufficient pain relief. 
(d) Therapeutic Recommendations: Avoid codeine use due to lack of efficacy. 
(e) Classification of recommendation for codeine therapy: Strong. 
(f) Considerations for alternative opioids: Alternatives that are not affected by this 
CYP2D6 phenotype include morphine and non-opioid analgesics. Tramadol, and to 
a lesser extent hydrocodone and oxycodone, are not good alternatives because their 
metabolism is affected by CYP2D6 activity; these agents should be avoided. 
5.3 CYP2D6 and Tamoxifen: Why endoxifen is a better op-tion 
than Tamoxifen 
The selective estrogen receptor modulator (SERM) tamoxifen has been used for many years as 
endocrine treatment for hormone receptorpositive breast cancer, with indications in the metastatic, 
adjuvant, and preventative settings. The minimal acute toxicity of endocrine treatments such as 
tamoxifen compared with chemotherapy helps maintain patients quality of life, and increasing 
emphasis is being placed on these treatments to delay the use of cytotoxic therapies for as long 
as possible.
CHAPTER 5. PHARMACOGENETICS OF CYP2D6 42 
Tamoxifens adverse-event profile includes venous thrombosis, endometrial cancer, and, 
most commonly, hot flashes, which are often treated with an SSRI antidepressant. Tamoxifen 
is extensively hepatically metabolized to several primary and secondary metabolites by multi-ple 
CYP enzymes, including CYP2D6. With some SSRIs (e.g., paroxetine) known to inhibit 
CYP2D6, a study was recently conducted to investigate the effects of SSRI coadministration on 
plasma concentrations of tamoxifen and its metabolites in addition to the effects of individual 
patient genotypes. A strong association was shown between CYP2D6 activity and plasma levels 
of the active tamoxifen metabolite, endoxifen. Concomitant use of paroxetine was associated 
with lower endoxifen plasma concentrations, with the magnitude of this difference being de-pendent 
on CYP2D6 genotype. It is possible, therefore, that patients taking some SSRIs may 
be less responsive to tamoxifen because of a lower rate of formation of its active metabolite, 
which may be exacerbated in individuals with slowed CYP2D6 metabolism. The possibility 
that tamoxifens antitumoral activity could be affected in this way needs further testing in clini-cal 
trials designed to record clinical outcome in conjunction with genotype and coadministered 
medication with the potential to affect CYP2D6. 
5.4 Why Endoxifen? 
Endoxifen is an active metabolite of Tamoxifen, a widely used breast cancer drug. Treat-ment 
of breast cancer directly with Endoxifen may eliminate Tamoxifen metabolism associ-ated 
variability and avoid a potential serious drug-drug interaction. Upon oral administration, 
tamoxifen (TAM) is extensively metabolized by cytochrome P450(CYP) enzymes into active 
metabolites including 4-hydroxy tamoxifen and endoxifen (4-hydroxy N-desmethyl Tamox-ifen) 
Endoxifen is one hundred times more potent than TAM and is generated via CYP3A4/5- 
mediated N-demethylation and CYP2D6-mediated hydroxylation. Due to genetic polymor-phism 
of CYP2D6, there are large variations among patients in both the therapeutic efficacy 
and side effects. Furthermore, CYP2D6 interacts with specific selective serotonin reuptake in-hibitors 
(SSRIs), frequently used to prevent the hot flushes experienced by up to 45% of patients 
taking TAM. As a consequence, significant numbers of women might not receive optimal bene-fit 
from TAM treatment. Our pre-clinical studies have validated the concept of using Endoxifen 
for the treatment of breast cancer (Ahmad et al. 2010). Treatment of breast cancer directly with 
Endoxifen may eliminate TAM metabolism associated variability and avoid a potential serious 
drug-drug interaction. 
5.4.1 Why Endoxifen Treatment may have better therapeutic outcome 
 Endoxifen may overcome the unpredictable therapeutic outcome due to pharmacoge-nomic 
variation of Tamoxifen and Aromatase inhibitors 
 Endoxifen would help women taking anti-depressants who have problems metabolizing 
Tamoxifen 
 Endoxifen acts directly at the target 
 Tamoxifen have been shown to be more effective than Aromatase inhibitors in patients 
with wild type CYP2D6 
 Showed enhanced therapeutic efficacy against established human tumor model in mice
CHAPTER 5. PHARMACOGENETICS OF CYP2D6 43 
Figure 5.2: Tamoxifen metabolism 
 Only potential drug for ER positive premenopausal women with CYP2D6 deficiency 
5.5 References 
1. Wang B, Yang LP, Zhang XZ, Huang SQ, Bartlam M, Zhou SF (2009). ”New insights into 
the structural characteristics and functional relevance of the human cytochrome P450 2D6 
enzyme”. Drug Metab. Rev. 41 (4): 573643. doi:10.1080/03602530903118729. PMID 
19645588. 
2. Teh LK, Bertilsson L (2012). ”Pharmacogenomics of CYP2D6: molecular genetics, in-terethnic 
differences and clinical importance”. Drug Metab. Pharmacokinet. 27 (1):
CHAPTER 5. PHARMACOGENETICS OF CYP2D6 44 
Figure 5.3: Tamoxifen: Interaction with other drugs 
5567. doi:10.2133/dmpk.DMPK-11-RV-121. PMID 22185816. 
3. Walko CM, McLeod H (April 2012). ”Use of CYP2D6 genotyping in practice: tamox-ifen 
dose adjustment”. Pharmacogenomics 13 (6): 6917. doi:10.2217/pgs.12.27. PMID 
22515611. 
4. ”Entrez Gene: CYP2D6 cytochrome P450, family 2, subfamily D, polypeptide 6”. 
5. Ostille DO., Warren AM., Kulkarni, J. The role of pharmacogenomic testing in psychia-try. 
Aust New Zealand J Psychiatry 2014 Jan 10. PMID 24413808 
6. Bertilsson L, Dahl ML, Daln P, Al-Shurbaji A (February 2002). ”Molecular genetics of 
CYP2D6: clinical relevance with focus on psychotropic drugs”. Br J Clin Pharmacol 53 
(2): 11122. doi:10.1046/j.0306-5251.2001.01548.x. PMC 1874287. PMID 11851634.
CHAPTER 5. PHARMACOGENETICS OF CYP2D6 45 
7. Llerena A, Dorado P, Peas-Lled EM (January 2009). ”Pharmacogenetics of debrisoquine 
and its use as a marker for CYP2D6 hydroxylation capacity”. Pharmacogenomics 10 (1): 
1728. doi:10.2217/14622416.10.1.17. PMID 19102711. 
8. Lynch T, Price A (August 2007). ”The effect of cytochrome P450 metabolism on drug 
response, interactions, and adverse effects”. Am Fam Physician 76 (3): 3916. PMID 
17708140. 
9. Droll K, Bruce-Mensah K, Otton SV, Gaedigk A, Sellers EM, Tyndale RF (1998). ”Com-parison 
of three CYP2D6 probe substrates and genotype in Ghanaians, Chinese and Cau-casians”. 
Pharmacogenetics 8 (4): 32533. doi:10.1097/00008571-199808000-00006. 
PMID 9731719. 
10. Gaedigk A, Bradford LD, Marcucci KA, Leeder JS (2002). ”Unique CYP2D6 activity 
distribution and genotype-phenotype discordance in black Americans”. Clin. Pharmacol. 
Ther. 72 (1): 7689. doi:10.1067/mcp.2002.125783. PMID 12152006. 
11. McLellan RA, Oscarson M, Seidegrd J, Evans DA, Ingelman-Sundberg M (June 1997). 
”Frequent occurrence of CYP2D6 gene duplication in Saudi Arabians”. Pharmacogenet-ics 
7 (3): 18791. doi:10.1097/00008571-199706000-00003. PMID 9241658. 
12. PHARMACOGENETICS AND PHARMACOGENOMICS. J. Steven Leeder PharmD, 
PhD Pediatric Clinics of North America - Volume 48, Issue 3 (June 2001). doi:10.1016/S0031- 
3955%2805%2970338-2. 
13. Hoskins JM, Carey LA, McLeod HL (August 2009). ”CYP2D6 and tamoxifen: DNA 
matters in breast cancer”. Nat. Rev. Cancer 9 (8): 57686. doi:10.1038/nrc2683. PMID 
19629072. 
14. Foster BC, Sockovie ER, Vandenhoek S, Bellefeuille N, Drouin CE, Krantis A, Budzinski 
JW, Livesey J, and Arnason JT (2004). ”In Vitro Activity of St. John’s Wort Against Cy-tochrome 
P450 Isozymes and P-Glycoprotein”. Pharmaceutical Biology 42 (2): 159169. 
doi:10.1080/13880200490512034.
Chapter 6 
Pharmacogenetics of cancer 
Advances in molecular biology and genetics over the past 60 years have facilitated develop-ment 
of multiple chemotherapeutic agents that are active against most common malignancies. 
However, significant heterogeneity in the efficacy and toxicity of these agents is consistently ob-served 
across human populations. Ad-ministration of the same dose of a given anticancer drug 
to a population of patients can result in any-where from minimal toxicity to severe, potentially 
life-threatening events. Many clinical factors are associated with drug toxicity, but genetic dif-ferences, 
or polymorphisms, in the proteins affecting drug disposition also have a great impact. 
In addition, the enzymes that metabolize the majority of chemotherapy agents contain genetic 
polymorphisms, but prospective identification of patients likely to be harmed by chemotherapy 
currently is not possible for most therapeutic regimens. 
6.1 Interindividual variation in Chemotherapeutic drug re-sponse 
Interindividual differences in tumor response and normal tissue toxicities are consistently ob-served 
with most chemotherapeutic agents or regimens. While many clinical variables have 
been associated with drug responses (e.g., age, gender, diet, drug-drug interactions), inherited 
variations in drug disposition (metabolism and transport) genes and drug target genes also likely 
con-tribute to the observed variability in cancer treatment outcome. 
Pharmacogenomic studies aim to elucidate the genetic bases for interindividual differences 
and to use such genetic information to predict the safety, toxicity, and/or efficacy of drugs. There 
exist several clinically relevant examples of the utility of pharmacogenomics that associate spe-cific 
genetic polymorphisms in drug metabolizing enzymes (e.g., TPMT, UGT1A1, DPD), drug 
transporters (MDR1), and drug target enzymes (TS) with clinical outcomes in patients treated 
with commonly prescribed chemotherapy drugs, such as 5-fluorouracil and irinotecan. Tech-niques 
to discover and evaluate the functional significance of these polymorphisms have evolved 
in recent years and may soon be applied to clinical practice and clinical trials of currently pre-scribed 
anticancer drugs as well as new therapeutic agents. 
Clearly, a better understanding of the genetic determinants of chemotherapeutic response 
will enable prospective identification of patients at risk for severe toxicity or those most likely 
to benefit from a particular treatment regimen. Such studies can be translated to clinical practice 
via molecular diagnostics (genotyping) in order to guide selection of the optimal drug combina-tion 
and dosage for the individual patient. A number of detailed reviews on cancer pharmacoge-nomics 
have been published recently. This article focuses on the current and future applications 
46
CHAPTER 6. PHARMACOGENETICS OF CANCER 47 
of pharmacogenomics in clinical cancer therapy and cancer drug development. 
6.2 Genetic variations affecting drug response and toxicity 
with cancer chemotherapy 
Table 6.1: Examples of genetic variations in major Phase-I and Phase-II enzymes asso-ciated 
with changes in patient responses to chemotherapy agents. Abbreviations: CYP = 
cytochrome P450; DPD = dihydropyrimidine dehydrogenase; GST = glutathione S-transferase; 
MDR1 = multidrug resistance 1; MRP = multidrug-resistancerelated protein; MTHFR = 5,10- 
methylenetetrahydrofolate reductase; NAT = N-acetyl transferase; TPMT = thiopurine methyl-transferase; 
TS = thymidylate synthase; UGT1A1 = UDP-glucuronosyltransferase 1A1; 5-FU 
= 5-fluorouracil; 6-MP = 6-mercaptopurine. 
Pharmacogenomic approaches have been applied to many existing chemotherapy agents in 
an effort to identify relevant inherited variations that may better predict patient response to 
chemotherapy. Genetic variations include nucleotide repeats, insertions, deletions, and single 
nucleotide polymorphisms (SNPs), which can alter the amino acid sequence of the encoded pro-teins, 
RNA splicing, and gene transcription. Such genetic polymorphisms in drug-metabolizing
CHAPTER 6. PHARMACOGENETICS OF CANCER 48 
enzymes, transporters, and molecular targets have been actively explored with regard to func-tional 
changes in phenotype (altered expression levels and/or activity of the encoded proteins) 
and their contribution to variable drug response. Recent studies also indicate that genetic varia-tions 
vary substantially among different ethnic groups and that the evaluation of the haplotypes 
(combination of polymorphisms that are inherited together) can often result in better correla-tion 
with phenotypes than with individual polymorphisms. The following sections describe 
some clinically relevant exam-ples of genetic polymorphisms to illustrate the relevance of can-cer 
pharmacogenomics in optimizing chemotherapy as a way to enhance efficacy and safety 
[Figure 6.1]. 
6.3 Polymorphism in Drug-Metabolizing Enzymes 
6.3.1 Thiopurine methyltransferase and 6-Mercaptopurine 
6-Mercaptopurine (6-MP) is a purine antimetabolite used in the treatment of leukemia. The 
antitumor activity of 6-MP is via the inhibition of the formation of nucleotides necessary for 
DNA and RNA synthesis. Thiopurine methyltransferase (TPMT) catalyzes the S-methylation 
of 6-MP to form inactive metabolites. Genetic variations in the TPMT gene have profound 
effects on the bioavailability and toxicity of 6-MP. It has been demonstrated that about 1 in 300 
individuals inherit TPMT deficiency as an autosomal recessive trait. Patients who carry TPMT 
polymorphisms are at risk for severe hematologic toxicities when treated with 6-MP because 
these polymorphisms lead to a decrease in the rate of 6-MP metabolism. 
Figure 6.1: Metabolism of 6-Mercaptopurine 
The molecular basis for polymorphic TPMT activity has been well defined. Three particular 
TPMTalleles, designated as TPMT*2, TMPT*3A, and TPMT*3C, have been shown to account 
for nearly 95% of the observed cases of TPMT defi-ciency. Each of these mutant alleles encodes 
TPMT pro-teins that undergo rapid degradation, leading to enzyme deficiency. The types and 
frequencies of TPMTalleles have been reported to differ among ethnic groups. A recent trial 
estimated that 71% of patients with bone marrow intolerance to 6-MP were phenotypically 
TPMT deficient and that these patients were more likely to be hospitalized, receive platelet 
transfusions, and miss scheduled doses of chemotherapy.
CHAPTER 6. PHARMACOGENETICS OF CANCER 49 
Figure 6.2: 6-Mercaptopurine dosing according to TPMT genotype 
Appropriate 6-MP dose reductions for TPMT-deficient patients have allowed for similar tox-icity 
and survival outcomes as patients with normal TPMT levels. Genotyping methods have 
been established for the molecular diagnosis of TPMT deficiency and can assist with determin-ing 
a safe starting dose for 6-MP therapy. 
6.3.2 UDP-glucuronosyltransferase 1A1 and Irinotecan 
[Irinotecan metabolism and Genotype based toxicities are discussed in Chapter 1: Figure 
1.3 and Figure 1.4] 
Variations in UGT1A1 activity most commonly arise from polymorphisms in the UGT1A1 
promoter region that contains several repeating TA elements. The presence of seven TA re-peats 
(referred to as UGT1A1*28), instead of the wild-type number of six, results in reduced 
UGT1A1 expression and activity. Accordingly, UGT1A1*28has been shown to be associated 
with reduced glucuronidation of SN-38, increased exposure to SN-38, and increased clini-cal 
toxicity for patients treated with irinotecan. The frequencies of UGT1A1*28alleles vary 
significantly among different ethnic groups: UGT1A1*28alleles are present in approximately 
35% of Caucasians and African Americans while their frequency is much lower in Asians.. 
A recent prospective study demonstrated, with sufficient statistical power, that patients with a 
UGT1A1*28 allele are at higher risk of grade 4 neutropenia. While that study clearly illustrates 
the importance of UGT1A1 pharmacogenetics as a molecular predictor of irinotecan toxicity, 
clinical guidelines have yet to be developed with regard to dosage adjustment or selection of
CHAPTER 6. PHARMACOGENETICS OF CANCER 50 
alternative non irinotecan-containing regimens. 
6.3.3 Dihydropyrimidine dehydrogenase and 5-FU 
Based on its activity against a variety of tumors and synergistic interactions with other chemother-apy 
agents, 5-fluorouracil (5-FU) and its derivatives remain some of the most commonly pre-scribed 
chemotherapy agents. Approximately 5% of administered 5-FU undergoes anabolism 
into cytotoxic nucleotides responsible for its antitumor activity, whereas the other 80%-95% 
under-goes catabolism into biologically inactive metabolites that are excreted in the urine and 
bile. Dihydropyrimidine dehydrogenase (DPD) catalyzes the rate-limiting step in 5-FU catabolism; 
therefore, variability in this enzyme activity is one of the major factors that influences systemic 
exposure to fluorodeoxyuridine monophosphate (FdUMP) and the incidence of adverse effects 
to 5-FU. DPD activity is completely or partially deficient in 0.1% and 3%5% of individuals 
in the general population, respectively, and DPD deficiency has been associated with severe 
toxicity and fatal outcomes after 5-FU treatment. 
Figure 6.3: 5-FU metabolism and Rate limiting role of DPYD enzyme 
DPD deficiency appears to be a genetic disorder arising from multiple polymorphisms in the 
DPYDgene resulting in decreased enzyme activity. Analyses of the preva-lence of the various 
mutations in the DPYD allele have shown that a guanidine to adenine point mutation in the 
invariant splice donor site (DPYD*2A) is by far the most common . 
6.3.4 MTHFR and Methotrexate 
Methotrexate inhibits dihydrofolate reductase (DHFR), which in turn causes accumulation of 
dihydrofolate and depletion of cellular folates needed for DNA synthesis. Methotrexate is asso-ciated 
with significant side effects and carries multiple black box warnings regarding potential 
toxicities, including acute renal failure and bone marrow suppression. Side effects can range
CHAPTER 6. PHARMACOGENETICS OF CANCER 51 
from mild and self limiting to more severe and sometimes fatal. There is significant interindi-vidual 
variation in response to methotrexate therapy. As many as one third of patients fail to 
respond to treatment either due to lack of efficacy or adverse events. 
The cytotoxic effect of methotrexate is dependent on inhibition of folate metabolism, so the 
folate pathway has been examined for enzyme variants that could be associated with efficacy 
and/or toxicity. One of these enzymes, 5,10-methylenetetrahydrofolate reductase (MTHFR), 
plays a key role, modulating DNA synthesis and intracellular folate pools. Two tightly linked, 
common variants in the MTHFR gene, C677T and A1298C, have been shown to cause reduced 
levels of MTHFR enzyme activity and corresponding reductions in available folate. Therefore, 
these variants may act as a useful marker for predicting individual differences in efficacy or 
toxicity of methotrexate. 
Table 6.2: Frequency of MTHFR allelic variation among different ethnic groups 
To date, numerous studies have examined the effect of the C677T and A1298C variants on 
toxicity and efficacy of methotrexate across disease states, methods of dosing, and ethnicities-and 
have shown conflicting results. 
1. Two recent meta-analyses of rheumatoid arthritis patients (RA) concluded that there is 
no significant relationship between MTHFR variant alleles and toxicity or efficacy of 
methotrexate. The authors acknowledge that the differences in their analyses compared 
to the previous meta-analysis cited could be due to the data sets used, heterogeneity of 
patients, and differences in methods of analysis. 
2. In recent studies of psoriatic arthritis and juvenile ALL, homozygosity of the C677 variant 
has been associated with toxicity. 
3. A small study of juvenile ALL patients showed that patients with normal MTHFR alleles 
tolerated a higher dosage of methotrexate with lower risk of toxicity than patient with 
variants treated with a standard dosage, suggesting that the MTHFR genotype can help 
guide dosage in this population.
Pharmacogenetics and drug response
Pharmacogenetics and drug response
Pharmacogenetics and drug response
Pharmacogenetics and drug response
Pharmacogenetics and drug response
Pharmacogenetics and drug response
Pharmacogenetics and drug response
Pharmacogenetics and drug response
Pharmacogenetics and drug response

More Related Content

What's hot

What's hot (20)

Cytochrome p450
Cytochrome p450Cytochrome p450
Cytochrome p450
 
Metabolism I
Metabolism IMetabolism I
Metabolism I
 
Biosimilars
BiosimilarsBiosimilars
Biosimilars
 
Anticancer drugs 3 antimetabolites
Anticancer drugs 3 antimetabolitesAnticancer drugs 3 antimetabolites
Anticancer drugs 3 antimetabolites
 
Drug Discovery Process by Kashikant Yadav
Drug Discovery Process by Kashikant YadavDrug Discovery Process by Kashikant Yadav
Drug Discovery Process by Kashikant Yadav
 
cyp450 system
cyp450 systemcyp450 system
cyp450 system
 
Cytochrome p450
Cytochrome p450Cytochrome p450
Cytochrome p450
 
Drug transporters
Drug transportersDrug transporters
Drug transporters
 
Drug discovery
Drug discoveryDrug discovery
Drug discovery
 
Anti-cancers and their mechanism of action
Anti-cancers and their mechanism of actionAnti-cancers and their mechanism of action
Anti-cancers and their mechanism of action
 
Pharmacophore modeling and docking techniques
Pharmacophore modeling and docking techniquesPharmacophore modeling and docking techniques
Pharmacophore modeling and docking techniques
 
Basic principles of chemotherapy
Basic principles of chemotherapyBasic principles of chemotherapy
Basic principles of chemotherapy
 
Target identification in drug discovery
Target identification in drug discoveryTarget identification in drug discovery
Target identification in drug discovery
 
Cytochrome p450 by Faizan Deshmukh
Cytochrome p450 by Faizan DeshmukhCytochrome p450 by Faizan Deshmukh
Cytochrome p450 by Faizan Deshmukh
 
Lead identification
Lead identification Lead identification
Lead identification
 
Pharmacogenetics
PharmacogeneticsPharmacogenetics
Pharmacogenetics
 
Prodrugs - concept & Applications
Prodrugs - concept & ApplicationsProdrugs - concept & Applications
Prodrugs - concept & Applications
 
various approaches to drug discovery
various approaches to drug discoveryvarious approaches to drug discovery
various approaches to drug discovery
 
Concept of clearance & factors affecting renal excretion
Concept of clearance & factors affecting renal excretionConcept of clearance & factors affecting renal excretion
Concept of clearance & factors affecting renal excretion
 
Vinca alkaloids
Vinca alkaloidsVinca alkaloids
Vinca alkaloids
 

Viewers also liked

Pharmacogenetics and Warfarin
Pharmacogenetics and WarfarinPharmacogenetics and Warfarin
Pharmacogenetics and Warfarin
Andrew Guvetis
 
Metabolism final
Metabolism finalMetabolism final
Metabolism final
pankaj
 
Drug Target Identification
Drug Target IdentificationDrug Target Identification
Drug Target Identification
Arvind306
 

Viewers also liked (20)

Pharmacogenetics Ppt
Pharmacogenetics PptPharmacogenetics Ppt
Pharmacogenetics Ppt
 
Pharmacogenetics and Warfarin
Pharmacogenetics and WarfarinPharmacogenetics and Warfarin
Pharmacogenetics and Warfarin
 
Pharmacogenetics
PharmacogeneticsPharmacogenetics
Pharmacogenetics
 
Pharmacogenetics and Pharmacogenomics
Pharmacogenetics and PharmacogenomicsPharmacogenetics and Pharmacogenomics
Pharmacogenetics and Pharmacogenomics
 
Pharmacogenetics and individual variation of drug response
Pharmacogenetics and individual variation of drug responsePharmacogenetics and individual variation of drug response
Pharmacogenetics and individual variation of drug response
 
Pharmacogenetics and Pharmacogenomics
Pharmacogenetics and PharmacogenomicsPharmacogenetics and Pharmacogenomics
Pharmacogenetics and Pharmacogenomics
 
Pharmacogenomics
PharmacogenomicsPharmacogenomics
Pharmacogenomics
 
Pharmacogenetics
PharmacogeneticsPharmacogenetics
Pharmacogenetics
 
Pharmacogenomics
PharmacogenomicsPharmacogenomics
Pharmacogenomics
 
Anti platelets- facts and controversies
Anti platelets- facts and controversies Anti platelets- facts and controversies
Anti platelets- facts and controversies
 
Unit 6 pharmacogenomics
Unit 6   pharmacogenomicsUnit 6   pharmacogenomics
Unit 6 pharmacogenomics
 
Demystifying Pharmacogenetics
Demystifying PharmacogeneticsDemystifying Pharmacogenetics
Demystifying Pharmacogenetics
 
Clopidogrelandpolymorphisms
ClopidogrelandpolymorphismsClopidogrelandpolymorphisms
Clopidogrelandpolymorphisms
 
Drug Discovery: Target Identification and Validation
Drug Discovery: Target Identification and Validation Drug Discovery: Target Identification and Validation
Drug Discovery: Target Identification and Validation
 
Clinical Pharmacokinetic of thenophylline
Clinical Pharmacokinetic of thenophyllineClinical Pharmacokinetic of thenophylline
Clinical Pharmacokinetic of thenophylline
 
NSARG Meeting Feb 18 2016 - Pharmacogenetics in Benefit Plans - Personalized ...
NSARG Meeting Feb 18 2016 - Pharmacogenetics in Benefit Plans - Personalized ...NSARG Meeting Feb 18 2016 - Pharmacogenetics in Benefit Plans - Personalized ...
NSARG Meeting Feb 18 2016 - Pharmacogenetics in Benefit Plans - Personalized ...
 
Metabolism final
Metabolism finalMetabolism final
Metabolism final
 
Drug Target Identification
Drug Target IdentificationDrug Target Identification
Drug Target Identification
 
Pharmacogenomics in Cardiovascular Diseases
Pharmacogenomics in Cardiovascular DiseasesPharmacogenomics in Cardiovascular Diseases
Pharmacogenomics in Cardiovascular Diseases
 
Antiplatelet resistence - significance and how to deal ?
Antiplatelet resistence - significance and how to deal ?Antiplatelet resistence - significance and how to deal ?
Antiplatelet resistence - significance and how to deal ?
 

Similar to Pharmacogenetics and drug response

[] Molecular cancer_therapeutics_strategies_for_d(book_zz.org)
[] Molecular cancer_therapeutics_strategies_for_d(book_zz.org)[] Molecular cancer_therapeutics_strategies_for_d(book_zz.org)
[] Molecular cancer_therapeutics_strategies_for_d(book_zz.org)
Hoàng Lê
 
Integrin Signalling And Cancer Front Pgs
Integrin Signalling And Cancer   Front PgsIntegrin Signalling And Cancer   Front Pgs
Integrin Signalling And Cancer Front Pgs
efreiter
 
patterson-ghrelinthedefenderoffatinthefaceofstressimplications
patterson-ghrelinthedefenderoffatinthefaceofstressimplicationspatterson-ghrelinthedefenderoffatinthefaceofstressimplications
patterson-ghrelinthedefenderoffatinthefaceofstressimplications
Zach Patterson, PhD
 
利用分子動力學電腦模擬研究聚穀氨醯胺及胰
利用分子動力學電腦模擬研究聚穀氨醯胺及胰利用分子動力學電腦模擬研究聚穀氨醯胺及胰
利用分子動力學電腦模擬研究聚穀氨醯胺及胰
Hsin-Lin Chiang
 
Antipsychotic induced hyperprolactinaemia mechanisms, clinical features and m...
Antipsychotic induced hyperprolactinaemia mechanisms, clinical features and m...Antipsychotic induced hyperprolactinaemia mechanisms, clinical features and m...
Antipsychotic induced hyperprolactinaemia mechanisms, clinical features and m...
guestd3f209b
 
msb_cannabis_report
msb_cannabis_reportmsb_cannabis_report
msb_cannabis_report
Sergio Scro
 
STEP 1 KAPLAN Pharmacology_booksmedicos.org.pdf
STEP 1 KAPLAN Pharmacology_booksmedicos.org.pdfSTEP 1 KAPLAN Pharmacology_booksmedicos.org.pdf
STEP 1 KAPLAN Pharmacology_booksmedicos.org.pdf
MarisolRivasCcente
 
TECO_Katalog_Diabetes_GB_0113_F
TECO_Katalog_Diabetes_GB_0113_FTECO_Katalog_Diabetes_GB_0113_F
TECO_Katalog_Diabetes_GB_0113_F
Peter Haima, Ph.D.
 

Similar to Pharmacogenetics and drug response (20)

[] Molecular cancer_therapeutics_strategies_for_d(book_zz.org)
[] Molecular cancer_therapeutics_strategies_for_d(book_zz.org)[] Molecular cancer_therapeutics_strategies_for_d(book_zz.org)
[] Molecular cancer_therapeutics_strategies_for_d(book_zz.org)
 
Integrin Signalling And Cancer Front Pgs
Integrin Signalling And Cancer   Front PgsIntegrin Signalling And Cancer   Front Pgs
Integrin Signalling And Cancer Front Pgs
 
patterson-ghrelinthedefenderoffatinthefaceofstressimplications
patterson-ghrelinthedefenderoffatinthefaceofstressimplicationspatterson-ghrelinthedefenderoffatinthefaceofstressimplications
patterson-ghrelinthedefenderoffatinthefaceofstressimplications
 
Isl1408681688437
Isl1408681688437Isl1408681688437
Isl1408681688437
 
Epigen princip
Epigen principEpigen princip
Epigen princip
 
Waters protein therapeutics application proctocols
Waters protein therapeutics application proctocolsWaters protein therapeutics application proctocols
Waters protein therapeutics application proctocols
 
利用分子動力學電腦模擬研究聚穀氨醯胺及胰
利用分子動力學電腦模擬研究聚穀氨醯胺及胰利用分子動力學電腦模擬研究聚穀氨醯胺及胰
利用分子動力學電腦模擬研究聚穀氨醯胺及胰
 
The Clinical Drug Therapy Rationales for Nursing Practice Field
The Clinical Drug Therapy Rationales for Nursing Practice FieldThe Clinical Drug Therapy Rationales for Nursing Practice Field
The Clinical Drug Therapy Rationales for Nursing Practice Field
 
At
AtAt
At
 
PhD Thesis Book
PhD Thesis BookPhD Thesis Book
PhD Thesis Book
 
Antipsychotic induced hyperprolactinaemia mechanisms, clinical features and m...
Antipsychotic induced hyperprolactinaemia mechanisms, clinical features and m...Antipsychotic induced hyperprolactinaemia mechanisms, clinical features and m...
Antipsychotic induced hyperprolactinaemia mechanisms, clinical features and m...
 
St nar-40-ebook
St nar-40-ebookSt nar-40-ebook
St nar-40-ebook
 
msb_cannabis_report
msb_cannabis_reportmsb_cannabis_report
msb_cannabis_report
 
STEP 1 KAPLAN Pharmacology_booksmedicos.org.pdf
STEP 1 KAPLAN Pharmacology_booksmedicos.org.pdfSTEP 1 KAPLAN Pharmacology_booksmedicos.org.pdf
STEP 1 KAPLAN Pharmacology_booksmedicos.org.pdf
 
Glicolisis y el cancer
Glicolisis y el cancerGlicolisis y el cancer
Glicolisis y el cancer
 
Passarge toc
Passarge tocPassarge toc
Passarge toc
 
TECO_Katalog_Diabetes_GB_0113_F
TECO_Katalog_Diabetes_GB_0113_FTECO_Katalog_Diabetes_GB_0113_F
TECO_Katalog_Diabetes_GB_0113_F
 
Metal protein attenuating compounds for the treatment of alzheimer's dementia
Metal protein attenuating compounds for the treatment of alzheimer's dementiaMetal protein attenuating compounds for the treatment of alzheimer's dementia
Metal protein attenuating compounds for the treatment of alzheimer's dementia
 
Sma12 4668: Clinical Drug Testing in Primary Care
Sma12 4668: Clinical Drug Testing in Primary CareSma12 4668: Clinical Drug Testing in Primary Care
Sma12 4668: Clinical Drug Testing in Primary Care
 
M.tech Thesis
M.tech ThesisM.tech Thesis
M.tech Thesis
 

Recently uploaded

Call Girls in Gagan Vihar (delhi) call me [🔝 9953056974 🔝] escort service 24X7
Call Girls in Gagan Vihar (delhi) call me [🔝  9953056974 🔝] escort service 24X7Call Girls in Gagan Vihar (delhi) call me [🔝  9953056974 🔝] escort service 24X7
Call Girls in Gagan Vihar (delhi) call me [🔝 9953056974 🔝] escort service 24X7
9953056974 Low Rate Call Girls In Saket, Delhi NCR
 
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Dipal Arora
 

Recently uploaded (20)

(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
(👑VVIP ISHAAN ) Russian Call Girls Service Navi Mumbai🖕9920874524🖕Independent...
 
Call Girls in Gagan Vihar (delhi) call me [🔝 9953056974 🔝] escort service 24X7
Call Girls in Gagan Vihar (delhi) call me [🔝  9953056974 🔝] escort service 24X7Call Girls in Gagan Vihar (delhi) call me [🔝  9953056974 🔝] escort service 24X7
Call Girls in Gagan Vihar (delhi) call me [🔝 9953056974 🔝] escort service 24X7
 
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any TimeTop Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
Top Quality Call Girl Service Kalyanpur 6378878445 Available Call Girls Any Time
 
Best Rate (Patna ) Call Girls Patna ⟟ 8617370543 ⟟ High Class Call Girl In 5 ...
Best Rate (Patna ) Call Girls Patna ⟟ 8617370543 ⟟ High Class Call Girl In 5 ...Best Rate (Patna ) Call Girls Patna ⟟ 8617370543 ⟟ High Class Call Girl In 5 ...
Best Rate (Patna ) Call Girls Patna ⟟ 8617370543 ⟟ High Class Call Girl In 5 ...
 
Call Girls Bangalore Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Bangalore Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Bangalore Just Call 8250077686 Top Class Call Girl Service Available
 
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Jabalpur Just Call 8250077686 Top Class Call Girl Service Available
 
Call Girls Haridwar Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Haridwar Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Haridwar Just Call 8250077686 Top Class Call Girl Service Available
 
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
Night 7k to 12k Navi Mumbai Call Girl Photo 👉 BOOK NOW 9833363713 👈 ♀️ night ...
 
Premium Call Girls In Jaipur {8445551418} ❤️VVIP SEEMA Call Girl in Jaipur Ra...
Premium Call Girls In Jaipur {8445551418} ❤️VVIP SEEMA Call Girl in Jaipur Ra...Premium Call Girls In Jaipur {8445551418} ❤️VVIP SEEMA Call Girl in Jaipur Ra...
Premium Call Girls In Jaipur {8445551418} ❤️VVIP SEEMA Call Girl in Jaipur Ra...
 
Call Girls Ooty Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Ooty Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Ooty Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Ooty Just Call 8250077686 Top Class Call Girl Service Available
 
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
 
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...
 
Premium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort Service
Premium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort ServicePremium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort Service
Premium Call Girls Cottonpet Whatsapp 7001035870 Independent Escort Service
 
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 9332606886 𖠋 Will You Mis...
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 9332606886 𖠋 Will You Mis...The Most Attractive Hyderabad Call Girls Kothapet 𖠋 9332606886 𖠋 Will You Mis...
The Most Attractive Hyderabad Call Girls Kothapet 𖠋 9332606886 𖠋 Will You Mis...
 
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
Call Girls Bhubaneswar Just Call 9907093804 Top Class Call Girl Service Avail...
 
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...
 
Call Girls Service Jaipur {9521753030} ❤️VVIP RIDDHI Call Girl in Jaipur Raja...
Call Girls Service Jaipur {9521753030} ❤️VVIP RIDDHI Call Girl in Jaipur Raja...Call Girls Service Jaipur {9521753030} ❤️VVIP RIDDHI Call Girl in Jaipur Raja...
Call Girls Service Jaipur {9521753030} ❤️VVIP RIDDHI Call Girl in Jaipur Raja...
 
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
 
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟ 9332606886 ⟟ Call Me For G...
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟  9332606886 ⟟ Call Me For G...Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟  9332606886 ⟟ Call Me For G...
Top Rated Bangalore Call Girls Ramamurthy Nagar ⟟ 9332606886 ⟟ Call Me For G...
 

Pharmacogenetics and drug response

  • 1. Pharmacogenetics and Drug Response Author & Course Teacher MAIZBHA UDDIN AHMED Assistant Professor Department of Clinical Pharmacy and Pharmacology Faculty of Pharmacy University of Dhaka
  • 2. Contents 1 Pharmacogenetics 5 1.1 Pharmacogenetics and Drug Response . . . . . . . . . . . . . . . . . . . . . . 6 1.2 Pharmacogenetics and Drug safety: Pharmacogenetics of Irinotecan . . . . . . 6 1.3 Genotype Phenotype correlation: CYP3A4 . . . . . . . . . . . . . . . . . . . . 8 1.3.1 Genotype . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9 1.3.2 Phenotype . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9 1.3.3 Phenotypic diversity due to heterogeneity of mutations . . . . . . . . . 9 1.4 Drug Metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10 1.5 Classification of Metabolizer groups . . . . . . . . . . . . . . . . . . . . . . . 11 1.6 CYP3A4 genotype and Phenotype . . . . . . . . . . . . . . . . . . . . . . . . 12 1.6.1 CYP3A4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12 1.6.2 CYP3A4 genotyping . . . . . . . . . . . . . . . . . . . . . . . . . . . 12 1.6.3 CYP3A4 phenotyping . . . . . . . . . . . . . . . . . . . . . . . . . . 13 1.7 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15 2 Pharmacogenetics of Warfarin 17 2.1 Warfarin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17 2.1.1 Warfarin Pharmacgology . . . . . . . . . . . . . . . . . . . . . . . . . 17 2.1.2 Mechanism of Action . . . . . . . . . . . . . . . . . . . . . . . . . . . 18 2.2 Metabolism of Warfarin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19 2.3 Pharmacogenetics of Warfarin . . . . . . . . . . . . . . . . . . . . . . . . . . 19 2.3.1 CYP2C9 and Warfarin dosing . . . . . . . . . . . . . . . . . . . . . . 21 2.3.2 VKORC1 and Warfarin . . . . . . . . . . . . . . . . . . . . . . . . . . 21 2.3.3 CYP4F2 and Warfarin . . . . . . . . . . . . . . . . . . . . . . . . . . 21 2.4 CPIC dosing guideline for Warfarin . . . . . . . . . . . . . . . . . . . . . . . 21 2.5 References: . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21 3 Pharmacogenetics of Clopidogrel 23 3.1 Clopidogrel . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23 3.1.1 Pharmacology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23 3.1.2 Why it is important to study CYP2C19 polymorphism . . . . . . . . . 24 3.2 Bioactivation of Clopidogrel and Prasugrel . . . . . . . . . . . . . . . . . . . . 24 3.3 Clopidogrel and CYP2C19 . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24 3.4 Genetic variation in CYP2C19 influences individual response to clopidogrel . . 25 3.5 Choosing the best option: Clopidogrel? Prasugrel? or Ticagrelor? . . . . . . . 26 3.6 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28 1
  • 3. CONTENTS 2 4 Pharmacogenetics of NAT2 30 4.1 NAT localization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30 4.2 Human NAT2 Alleles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30 4.3 NAT and Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31 4.4 NAT2 and Drug Response . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31 4.4.1 NAT2 and Isoniazid . . . . . . . . . . . . . . . . . . . . . . . . . . . 31 4.4.2 NAT2 and Environmental carcinogen . . . . . . . . . . . . . . . . . . 32 4.4.3 NAT2 and Procainamide . . . . . . . . . . . . . . . . . . . . . . . . . 33 4.5 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33 5 Pharmacogenetics of CYP2D6 35 5.1 CYP2D6 allelic variants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35 5.2 CYP2D6 and Antipsychotics Pharmacology . . . . . . . . . . . . . . . . . . . 37 5.2.1 Metabolism of Amytriptyline . . . . . . . . . . . . . . . . . . . . . . 38 5.2.2 Dosing recommendations for amitriptyline/nortriptyline based on CYP2D6 phenotype . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38 5.2.3 Recommended dosing of codeine by CYP2D6 Phenotype . . . . . . . . 40 5.3 CYP2D6 and Tamoxifen: Why endoxifen is a better option than Tamoxifen . . 41 5.4 Why Endoxifen? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42 5.4.1 Why Endoxifen Treatment may have better therapeutic outcome . . . . 42 5.5 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43 6 Pharmacogenetics of cancer 46 6.1 Interindividual variation in Chemotherapeutic drug response . . . . . . . . . . 46 6.2 Genetic variations affecting drug response and toxicity with cancer chemotherapy 47 6.3 Polymorphism in Drug-Metabolizing Enzymes . . . . . . . . . . . . . . . . . 48 6.3.1 Thiopurine methyltransferase and 6-Mercaptopurine . . . . . . . . . . 48 6.3.2 UDP-glucuronosyltransferase 1A1 and Irinotecan . . . . . . . . . . . . 49 6.3.3 Dihydropyrimidine dehydrogenase and 5-FU . . . . . . . . . . . . . . 50 6.3.4 MTHFR and Methotrexate . . . . . . . . . . . . . . . . . . . . . . . . 50 6.4 Polymorphisms in Drug Transporters . . . . . . . . . . . . . . . . . . . . . . . 53 6.4.1 Clinical Annotation for ABCB1 (rs1045642) and 5-fluorouracil . . . . 54 6.4.2 Clinical Annotation for rs1045642 and paclitaxel . . . . . . . . . . . . 55 6.5 Polymorphisms in Drug Targets . . . . . . . . . . . . . . . . . . . . . . . . . 55 6.6 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
  • 4. List of Figures 1.1 Principle of Pharmacogenetics . . . . . . . . . . . . . . . . . . . . . . . . . . 6 1.2 Pharmacogenetics based Dosing/Diagnostics . . . . . . . . . . . . . . . . . . . 7 1.3 Metabolism of Irinotecan. Abbreviations: APC, 7-ethyl-10[4-N-(5-aminopentanoic acid)-1-piperidino]-carbonyloxycamptothecin; CYP3A4, cytochrome P450 3A4 enzyme; CYP3A5, cytochrome P450, family 3, subfamily A, polypeptide 5; NPC, 7-ethyl-10[4-(1-piperidino)-1-amino]-carbonyloxycamptothecin; SN-38, 7-ethyl-10-hydroxy-camptothecin; SN-38G, glucuronidated SN-38; Topo 1, DNA topoisomerase I. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8 1.4 Irinotecan Metabolizer and Toxicities . . . . . . . . . . . . . . . . . . . . . . 8 1.5 Genetics of Sickle cell anaemia . . . . . . . . . . . . . . . . . . . . . . . . . . 9 1.6 Overview of Drug Metabolism and Excretion . . . . . . . . . . . . . . . . . . 10 1.7 Genotype-Phenotype Association of Drug Metabolism and Pharmacology . . . 11 1.8 Genotyping protocol of CYP3A4 . . . . . . . . . . . . . . . . . . . . . . . . . 13 2.1 Warfarin Metabolism and Mechanism of Action . . . . . . . . . . . . . . . . . 18 2.2 Warfarin Metabolic Pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . 19 3.1 Comparative metabolic pattern of Clopidogrel and Prasugrel . . . . . . . . . . 25 3.2 Comparative Pharmacokinetic pathway of Clopidogrel, Prasugrel and Ticagrelor 27 4.1 Metabolism of Isoniazid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32 5.1 Metabolism of Amytriptyline . . . . . . . . . . . . . . . . . . . . . . . . . . . 39 5.2 Tamoxifen metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43 5.3 Tamoxifen: Interaction with other drugs . . . . . . . . . . . . . . . . . . . . . 44 6.1 Metabolism of 6-Mercaptopurine . . . . . . . . . . . . . . . . . . . . . . . . . 48 6.2 6-Mercaptopurine dosing according to TPMT genotype . . . . . . . . . . . . . 49 6.3 5-FU metabolism and Rate limiting role of DPYD enzyme . . . . . . . . . . . 50 6.4 Illustrates some of the key enzymes involved in metabolism of MTX: RFC, reduced folate carrier; MTX-PGs, polyglutamated MTX; FPGS, folylpolyglu-tamate synthase; GGH, gamma-glutamyl hydrolase; DHF, dihydrofolate; THF, tetrahydrofolate; TYMS, thymidylate synthase; DHFR, dihydrofolate reduc-tase; MTHFR, methylenetetrahydrofolate reductase; ATIC, 5-aminoimidazole- 4-carboxamide ribonucleotide transformylase. . . . . . . . . . . . . . . . . . . 52 3
  • 5. List of Tables 1.1 Metabolizer’s Genotype and Phenotype in different CYP450 isoenzymes . . . . 12 1.2 PCR mixture . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13 1.3 PCR condition and PCR size . . . . . . . . . . . . . . . . . . . . . . . . . . . 14 1.4 Primers for PCR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14 2.1 Enzymatic activity of different CYP2C9 allelic variants . . . . . . . . . . . . . 20 2.2 Genotype based Dosing guideline by Clinical Pharmacogenetics Implementa-tion Consortium (CPIC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21 3.1 Common polymorphisms in the CYP2C19 gene that influence anti-platelet re-sponse to clopidogrel . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26 3.2 Allelic Frequency of common CYP2C19 variants by ethnicity . . . . . . . . . 26 4.1 Common NAT2 alleles and their phenotype . . . . . . . . . . . . . . . . . . . 34 5.1 CYP2D6 Substrate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35 5.2 CYP2D6 Allelic variants with enzyme function in different ethnic groups . . . 36 5.3 Genotype and Phenotype of CYP2D6 Alleles . . . . . . . . . . . . . . . . . . 37 5.4 Metabolizer classification of CYP2D6 . . . . . . . . . . . . . . . . . . . . . . 38 6.1 Examples of genetic variations in major Phase-I and Phase-II enzymes as-sociated with changes in patient responses to chemotherapy agents. Ab-breviations: CYP = cytochrome P450; DPD = dihydropyrimidine dehydroge-nase; GST = glutathione S-transferase; MDR1 = multidrug resistance 1; MRP = multidrug-resistancerelated protein; MTHFR = 5,10-methylenetetrahydrofolate reductase; NAT = N-acetyl transferase; TPMT = thiopurine methyltransferase; TS = thymidylate synthase; UGT1A1 = UDP-glucuronosyltransferase 1A1; 5- FU = 5-fluorouracil; 6-MP = 6-mercaptopurine. . . . . . . . . . . . . . . . . . 47 6.2 Frequency of MTHFR allelic variation among different ethnic groups . . . . . 51 6.3 Clinical Annotation for Methotrexate in different MTHFR alleles . . . . . . . . 53 6.4 Clinical Annotation of ABCB1 rs1045642 and 5-FU . . . . . . . . . . . . . . 54 6.5 Clinical Annotation of ABCB1 rs1045642 and Paclitaxel . . . . . . . . . . . . 55 4
  • 6. Chapter 1 Pharmacogenetics Pharmacogenetics is the study of inherited genetic differences in drug metabolic pathways which can affect individual responses to drugs, both in terms of therapeutic effect as well as adverse effects.The term pharmacogenetics is often used interchangeably with the term phar-macogenomics which also investigates the role of acquired and inherited genetic differences in relation to drug response and drug behavior through a systematic examination of genes, gene products, and inter- and intra-individual variation in gene expression and function. In oncology, pharmacogenetics historically is the study of germline mutations (e.g., single-nucleotide polymorphisms affecting genes coding for liver enzymes responsible for drug de-position and pharmacokinetics), whereas pharmacogenomics refers to somatic mutations in tu-moral DNA leading to alteration in drug response (e.g., KRAS mutations in patients treated with anti-Her1 biologics). Pharmacogenetics is a rising concern in clinical oncology, because the therapeutic win-dow of most anticancer drugs is narrow and patients with impaired ability to detoxify drugs will undergo life-threatenting toxicities. In particular, genetic deregulations affecting genes coding for DPD, UGT1A1, TPMT, CDA and Cyp2D6 are now considered as critical issues for patients treated with 5-FU/capecitabine, irinotecan, mercaptopurine/azathioprine, gemc-itabine/ capecitabine/AraC and tamoxifen, respectively. The decision to use pharmacogenetic techniques is influenced by the relative costs of genotyping technologies and the cost of provid-ing a treatment to a patient with an incompatible genotype. When available, phenotype-based approaches proved their usefulness while being cost-effective. Pharmacogenetics has several advantages: The genotype of an individual is essentially invariable and remains unaffected by the treatment itself. Molecular biology techniques provide an accurate assessment of the genotype of an indi-vidual. There has been a dramatic increase in the amount of genomic information that is available. This information provides the necessary data for comprehensive studies of individual genes and broad investigation of genome-wide variation. The ease of accessibility to genotype information through peripheral blood or saliva sam-pling and advances in molecular techniques has increased the feasibility of DNA collec-tion and genotyping in large-scale clinical trials. 5
  • 7. CHAPTER 1. PHARMACOGENETICS 6 1.1 Pharmacogenetics and Drug Response 1. Dose adaptation:Variability in drug pharmacokinetics resulting from polymorphisms af-fecting transport and metabolism might be alleviated, in part, by appropriate dose adjust-ments. Dose adjustment according to Pharmacogenetics based Diagnostics (PGDx) from fields of drug therapy where compensation for differences in individual drug exposure can be expected to have a beneficial effect on the therapeutic outcome. [Figure 1.1] Figure 1.1: Principle of Pharmacogenetics 2. Drug selection using PGDx: Polymorphisms in receptors or other targets of a drug can affect the probability of obtaining efficacy for that drug. It is unlikely that any physician would restrict a patient from treatment with a drug because the chance of response might be only 40% in carriers of a certain PGx variant, when the average chance is 70% in carriers of the respective wild-type genotype. However, such PGx information might be helpful if alternative medications are available or if a better response might be achieved by a change in dose or by a specific additive medication. [Figure 1.2] 1.2 Pharmacogenetics and Drug safety: Pharmacogenetics of Irinotecan Irinotecan is another example of a drug that is metabolized differently in some patients; these differences can predispose a subset of patients to increased drug toxicity. The addition of irinotecan to the combination of fluorouracil and leucovorin in 1996 represented the first major advance in the treatment of colorectal cancer since the late 1950s, when fluorouracil originally became the standard of care. Regimens that contained irinotecan resulted in a near-doubling of response rates, from around 20% to 40%, and prolongation of overall survival by up to 3 months. These advances, however, came with the risk of significant and unpredictable toxicity. The most common toxicities of irinotecan are acute and delayed diarrhea and neutropenia.
  • 8. CHAPTER 1. PHARMACOGENETICS 7 Figure 1.2: Pharmacogenetics based Dosing/Diagnostics The drug-associated mortality from bolus fluorouracil and leucovorin regimens that con-tained irinotecan was threefold higher than that of regimens that did not contain the drug. Death often resulted from drug-related gastrointestinal and vascular syndromes. Differences between individuals with respect to the drug’s metabolism were a rational route of exploration because of the noted correlation between active drug concentration and toxicity. Irinotecan is primarily metabolized by carboxylesterase to the active metabolite, SN-38, which is responsible for inhibition of DNA topoisomerase I. SN-38 is glucuronidated by UGT1A1 to the inactive metabolite SN-38G, which is subsequently excreted. Other irinotecan inactiva-tion pathways include oxidation by CYP3A4 and CYP3A5 into APC and NPC, which can also be metabolized by carboxylesterase to SN-38. [Figure 1.3] Patients with the UGT1A1*28 allele have lower glucuronidating ability than patients with wild-type alleles. In the Caucasian population, 917% of individuals are homozygous (7/7) and 2836% are heterozygous (6/7) for the UGT1A1*28 allele. In African-Americans, 1733% and 3850% are homozygous and heterozygous, respectively. This variant allele is seen to a lesser extent in the Asian population; only 14% of individuals are homozygous and 1531% are heterozygous for the UGT1A1*28 allele. [Figure 1.4]
  • 9. CHAPTER 1. PHARMACOGENETICS 8 Figure 1.3: Metabolism of Irinotecan. Abbreviations: APC, 7-ethyl-10[4-N-(5-aminopentanoic acid)-1-piperidino]-carbonyloxycamptothecin; CYP3A4, cytochrome P450 3A4 enzyme; CYP3A5, cytochrome P450, family 3, subfamily A, polypeptide 5; NPC, 7-ethyl-10[4- (1-piperidino)-1-amino]-carbonyloxycamptothecin; SN-38, 7-ethyl-10-hydroxy-camptothecin; SN-38G, glucuronidated SN-38; Topo 1, DNA topoisomerase I. Figure 1.4: Irinotecan Metabolizer and Toxicities 1.3 Genotype Phenotype correlation: CYP3A4 An organisms genotype is the set of genes that it carries. An organisms phenotype is all of its observable characteristicswhich are influenced both by its genotype and by the environment. So in defining evolution, we are really concerned with changes in the genotypes that make up a population from generation to generation. However, since an organisms genotype generally affects its phenotype, the phenotypes that make up the population are also likely to change.
  • 10. CHAPTER 1. PHARMACOGENETICS 9 1.3.1 Genotype This is the ”internally coded, inheritable information” carried by all living organisms. This stored information is used as a ”blueprint” or set of instructions for building and maintaining a living creature. These instructions are found within almost all cells (the ”internal” part), they are written in a coded language (the genetic code), they are copied at the time of cell division or reproduction and are passed from one generation to the next (”inheritable”). These instruc-tions are intimately involved with all aspects of the life of a cell or an organism. They control everything from the formation of protein macromolecules, to the regulation of metabolism and synthesis. 1.3.2 Phenotype This is the ”outward, physical manifestation” of the organism. These are the physical parts, the sum of the atoms, molecules, macromolecules, cells, structures, metabolism, energy utilization, tissues, organs, reflexes and behaviors; anything that is part of the observable structure, function or behavior of a living organism. 1.3.3 Phenotypic diversity due to heterogeneity of mutations Mutations that alter the structure of gene products, missense mutations, may give rise to a remarkable diversity of phenotypes associated with mutations at the same gene locus. One of the best examples comes from the inherited disorders of haemoglobin. Figure 1.5: Genetics of Sickle cell anaemia The structure of human haemoglobin (Hb) changes during embryonic, fetal and adult life. All normal haemoglobins are tetramers of two pairs of dissimilar globin chains. Adult and fetal haemoglobins have a chains combined with b (HbA a2b2) or g chains (HbF a2g2). Over 400 structural haemoglobin variants have been identified, many of which cause no clinical disabil-ity. However, some, because the amino acid substitution alters the stability or function of the haemoglobin molecule, result in a disease phenotype. For example, the substitution of glutamic
  • 11. CHAPTER 1. PHARMACOGENETICS 10 acid for valine in the sixth position in the b chain causes the haemoglobin molecules to form linear stacks in the deoxy configuration which, in turn, causes the red cells to assume a sickled configuration. The resulting disease in homozygotes, sickle cell anaemia, is characterized by chronic anaemia and tissue damage due to blockage of the micro-circulation with aggregates of sickled red cells. Other amino acid substitutions result in instability of the haemoglobin molecule, which precipitates in the red cells during their life in the circulation. This results in damage to their membranes and increased rigidity, and hence in a variable degree of anaemia due to their premature destruction. 1.4 Drug Metabolism Drug metabolism also known as xenobiotic metabolism is the biochemical modification of phar-maceutical substances or xenobiotics respectively by living organisms, usually through special-ized enzymatic systems. Drug metabolism often converts lipophilic chemical compounds into more readily excreted hydrophilic products. Figure 1.6: Overview of Drug Metabolism and Excretion In the process of metabolism, drugs will be more water soluble, therefore more accessible for renal excretion. During drug metabolism, sometimes toxic compounds are formed. In many cases, metabolism is responsible for the activation of the prodrug. This process can be divided into two different types of reaction. Phase-I reactions are: oxydation, reduction and hydrolysis. Phase-II (conjugation) reactions include sulfation, methylation, glucuronidation, acetylation. Both reactions-whose names do not indicate the succession of the reactions - normally make the originally lipophilic compound more hydrophilic. The genes coding the proteins responsible for these processes are usually highly polymorphic. This means that in some cases, the individual response after the administration of a specific drug can be attributed to the genetic background of the patient. It also means that in some cases, the knowledge of the patients genetic status
  • 12. CHAPTER 1. PHARMACOGENETICS 11 makes individualized therapy possible. Individualized therapy has two major goals: it might help not only in quickly establishing the correct dose of certain drug, but also in avoiding the dangerous, sometimes life-threatening side effects. 1.5 Classification of Metabolizer groups Drug or other xenobiotics metabolizers can be classified according to their metabolic capacities and their genetic make up. 1. Ultra Rapid Metabolizer/Ultra Metabolizer: Ultra-rapid metabolizers have one or more alleles which result in increased enzyme activity compared to extensive metabo-lizers. 2. Rapid/Extensive Metabolizer: Extensive metabolizers have 2 normally functioning al-leles and therefore have normal enzyme activity. 3. Intermediate Metabolizer: Intermediate metabolizers have one non-functional allele and one normally functioning allele, and therefore have decreased enzyme activity. 4. Poor Metabolizer: Poor metabolizers have two non-functional alleles and therefore have little to no enzyme activity. Figure 1.7: Genotype-Phenotype Association of Drug Metabolism and Pharmacology
  • 13. CHAPTER 1. PHARMACOGENETICS 12 Table 1.1: Metabolizer’s Genotype and Phenotype in different CYP450 isoenzymes 1.6 CYP3A4 genotype and Phenotype 1.6.1 CYP3A4 Cytochrome P450 3A4 (abbreviated CYP3A4) (EC 1.14.13.97), is an important enzyme in the body, mainly found in the liver and in the intestine. Its purpose is to oxidize small foreign organic molecules (xenobiotics), such as toxins or drugs, so that they can be removed from the body. While many drugs are deactivated by CYP3A4, there are also some drugs which are activated by the enzyme. Some substances, such as grapefruit juice and some drugs, interfere with the action of CYP3A4. These substances will therefore either amplify or weaken the action of those drugs that are modified by CYP3A4. CYP3A4 is a member of the cytochrome P450 family of oxidizing enzymes. Several other members of this family are also involved in drug metabolism, but CYP3A4 is the most common and the most versatile one. Like all members of this family, it is a hemoprotein, i.e. a protein containing a heme group with an iron atom. In humans, the CYP3A4 protein is encoded by the CYP3A4 gene. This gene is part of a cluster of cytochrome P450 genes on chromosome 7q21.1. Cytochrome P450 (CYP) enzymes metabolize more than 70% of drugs for clinical use. Among them, CYP3A4 is quantitatively the most important P450 enzyme in adults. It is ex-pressed to a major extent not only in the human liver (95%) but also in the small intestine, thus contributing to the presystemic and systemic metabolism of 30% of all drugs. 1.6.2 CYP3A4 genotyping Genotyping is normally done by two step gel electrophoresis: a) one after PCR and b) Another after restriction enzyme digestion. A typical PCR mixture contains: forward primer, reverse primer, dNTP, Taq DNA polymerase, Buffer and a template DNA. PCR condition will be initial denaturation of 2min at 94o, 35 cycles of 30 sec at 94o, anneling of 30sec at 60o, chain elon-
  • 14. CHAPTER 1. PHARMACOGENETICS 13 gation of 30 sec at 72o and final chain elongation for 5min and cooling forever until machine stops. A flowchart of CYP3A4 genotyping is given below: Figure 1.8: Genotyping protocol of CYP3A4 A typical PCR mixture contains the following reagents: Table 1.2: PCR mixture 1.6.3 CYP3A4 phenotyping CYP3A is involved in the metabolism of endogenous cortisol to 6b-hydroxy-cortisol. The urinary excretion of 6b-hydroxycortisol and its ratio to free cortisol reflected its activities. It
  • 15. CHAPTER 1. PHARMACOGENETICS 14 Table 1.3: PCR condition and PCR size Table 1.4: Primers for PCR has been reported that measuring the urinary morning spot of 6b-hydroxy-cortisol/cortisol ra-tio is a good indication of CYP3A activity. Hsieh et al. reported that, from the 6b-hydroxy-cortisol/ cortisol ratio, those withCYP3A4*4, *5and*6 alleles have shown below average CYP3A4 activity implying reduced catalytic activity for the corresponding protein variants compared to those with no mutations shown. Phenotyping is normally done by measuring the ratio of 6b-hydroxycortisol (6b-OH-CS) and cortisol (CS) in urine by HPLC method. Extensive class group of individual will be repre-sented by higher cortisol metabolite and poor metabolizer will be phenotyped by lower cortisol metabolite concentration.
  • 16. CHAPTER 1. PHARMACOGENETICS 15 1.7 References 1. Klotz, U. (2007). ”The role of pharmacogenetics in the metabolism of antiepileptic drugs: pharmacokinetic and therapeutic implications.”. Clin Pharmacokinet 46 (4): 2719. doi:10.2165/00003088-200746040-00001. PMID 17375979 2. ”Center for Pharmacogenomics and Individualized Therapy”. Retrieved 2014-10-28. 3. Roses AD (June 2000). ”Pharmacogenetics and the practice of medicine”. Nature 405 (6788): 85765. doi:10.1038/35015728. PMID 10866212. 4. Lazarou J, Pomeranz BH, Corey PN (April 1998). ”Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies”. JAMA 279 (15): 12005. doi:10.1001/jama.279.15.1200. PMID 9555760. 5. Ingelman-Sundberg M, Rodriguez-Antona C (August 2005). ”Pharmacogenetics of drug-metabolizing enzymes: implications for a safer and more effective drug therapy”. Philos. Trans. R. Soc. Lond., B, Biol. Sci. 360 (1460): 156370. doi:10.1098/rstb.2005.1685. PMC 1569528. PMID 16096104. 6. Kirchheiner J, Seeringer A, Viviani R (2010). ”Pharmacogenetics in psychiatry–a useful clinical tool or wishful thinking for the future?”. Curr. Pharm. Des. 16 (2): 13644. doi:10.2174/138161210790112728. PMID 20205659. 7. AlazrakiM(2011). ”The 10 Biggest-Selling Drugs That Are About to Lose Their Patent”. DailyFinance. Retrieved 2012-05-06. 8. Shuldiner AR, O’Connell JR, Bliden KP, Gandhi A, Ryan K, Horenstein RB, Damcott CM, Pakyz R, Tantry US, Gibson Q, Pollin TI, PostW, Parsa A, Mitchell BD, Faraday N, Herzog W, Gurbel PA (August 2009). ”Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy”. JAMA 302 (8): 84957. doi:10.1001/jama.2009.1232. PMID 19706858. 9. The Creative Destruction of Medicine: How the Digital Revolution Will Create Better Health Care. New York: Basic Books. 2012. ISBN 0-465-02550-1. 10. Farbstein D, Blum S, Pollak M, Asaf R, Viener HL, Lache O, Asleh R, Miller-Lotan R, Barkay I, Star M, Schwartz A, Kalet-Littman S, Ozeri D, Vaya J, Tavori H, Vardi M, Laor A, Bucher SE, Anbinder Y, Moskovich D, Abbas N, Perry N, Levy Y, Levy AP (November 2011). ”Vitamin E therapy results in a reduction in HDL function in individuals with diabetes and the haptoglobin 2-1 genotype”. Atherosclerosis 219 (1): 2404. doi:10.1016/j.atherosclerosis.2011.06.005. PMC 3200506. PMID 21722898. 11. Yang CG, Ciccolini J, Blesius A, Dahan L, Bagarry-Liegey D, Brunet C, Varoquaux A, Frances N, Marouani H, Giovanni A, Ferri-Dessens RM, Chefrour M, Favre R, Duffaud F, Seitz JF, Zanaret M, Lacarelle B, Mercier C (January 2011). ”DPD-based adaptive dosing of 5-FU in patients with head and neck cancer: impact on treatment efficacy and toxicity”. Cancer Chemother. Pharmacol. 67 (1): 4956. doi:10.1007/s00280-010-1282-4. PMID 20204365. 12. Malhotra AK (2010). ”The state of pharmacogenetics”. Psychiatry Times 27 (4): 3841, 62.
  • 17. CHAPTER 1. PHARMACOGENETICS 16 13. Gardiner SJ, Begg EJ (September 2006). ”Pharmacogenetics, drug-metabolizing en-zymes, and clinical practice”. Pharmacol. Rev. 58 (3): 52190. doi:10.1124/pr.58.3.6. PMID 16968950. 14. Genetic Science Learning Center. ”Your Doctor’s New Genetic Tools.”. Lern.Genetics. Retrieved 15 April 2012. 15. ”Pharmacogenomics: Personalizing Medicine”. Discovery’s Edge. Mayo Clinic. Re-trieved April 15, 2012. 16. Ge D, Fellay J, Thompson AJ, Simon JS, Shianna KV, Urban TJ, Heinzen EL, Qiu P, Ber-telsen AH, Muir AJ, Sulkowski M, McHutchison JG, Goldstein DB (September 2009). ”Genetic variation in IL28B predicts hepatitis C treatment-induced viral clearance”. Na-ture 461 (7262): 399401. doi:10.1038/nature08309. PMID 19684573. 17. Thomas DL, Thio CL, Martin MP, Qi Y, Ge D, O’Huigin C, Kidd J, Kidd K, Khakoo SI, Alexander G, Goedert JJ, Kirk GD, Donfield SM, Rosen HR, Tobler LH, Busch MP, McHutchison JG, Goldstein DB, Carrington M (October 2009). ”Genetic variation in IL28B and spontaneous clearance of hepatitis C virus”. Nature 461 (7265): 798801. doi:10.1038/nature08463. PMC 3172006. PMID 19759533. 18. Frueh FW, Amur S, Mummaneni P, Epstein RS, Aubert RE, DeLuca TM, Verbrugge RR, Burckart GJ, Lesko LJ (August 2008). ”Pharmacogenomic biomarker information in drug labels approved by the United States food and drug administration: prevalence of related drug use”. Pharmacotherapy 28 (8): 9928. doi:10.1592/phco.28.8.992. PMID 18657016. 19. Corrigan OP (2011). ”Personalized Medicine in a Consumer Age”. Current Pharmacoge-nomics and Personalized Medicine 9: 168176. doi:10.2174/187569211796957566. 20. Paul NW, Fangerau H (December 2006). ”Why should we bother? Ethical and social is-sues in individualized medicine”. Curr Drug Targets 7 (12): 17217. doi:10.2174/138945006779025428. PMID 17168846. 21. Payne K, Shabaruddin FH (May 2010). ”Cost-effectiveness analysis in pharmacoge-nomics”. Pharmacogenomics 11 (5): 6436. doi:10.2217/pgs.10.45. PMID 20415553. 22. Oxford Nanopore Technologies. 2012. Press releases - News - Oxford Nanopore Tech-nologies. Available from: http://www.nanoporetech.com/news/press-releases/view/39 23. Breckenridge A, Lindpaintner K, Lipton P, McLeod H, Rothstein M,Wallace H (Septem-ber 2004). ”Pharmacogenetics: ethical problems and solutions”. Nat. Rev. Genet. 5 (9): 67680. doi:10.1038/nrg1431. PMID 15372090. 24. Corrigan OP (March 2005). ”Pharmacogenetics, ethical issues: review of the Nuffield Council on Bioethics Report”. J. Med. Ethics 31 (3): 1448. doi:10.1136/jme.2004.007229. PMC 1734105. PMID 15738433.
  • 18. Chapter 2 Pharmacogenetics ofWarfarin 2.1 Warfarin Warfarin (also known by the brand names Coumadin, Jantoven, Marevan, Uniwarfin) is an anticoagulant normally used in the prevention of thrombosis and thromboembolism, the forma-tion of blood clots in the blood vessels and their migration elsewhere in the body, respectively. Warfarin is commonly but incorrectly referred to as a blood thinner. Despite its effectiveness, treatment with warfarin has several shortcomings. Many com-monly used medications interact with warfarin, as do some foods (particularly leaf vegetable foods or ”greens,” since these typically contain large amounts of vitamin K1) and its activity has to be monitored by blood testing for the international normalized ratio (INR) to ensure an adequate yet safe dose is taken. A high INR predisposes to a high risk of bleeding, while an INR below the therapeutic target indicates the dose of warfarin is insufficient to protect against thromboembolic events. 2.1.1 Warfarin Pharmacgology Warfarin and related 4-hydroxycoumarin-containing molecules decrease blood coagulation by inhibiting vitamin K epoxide reductase, an enzyme that recycles oxidized vitamin K1 to its reduced form after it has participated in the carboxylation of several blood coagulation proteins, mainly prothrombin and factor VII. Despite being labeled a vitamin K antagonist, warfarin does not antagonize the action of vitamin K1, but rather antagonizes vitamin K1 recycling, depleting active vitamin K1. Warfarin is used to decrease the tendency for thrombosis or as secondary prophylaxis (pre-vention of further episodes) in those individuals who have already formed a blood clot (throm-bus). Warfarin treatment can help prevent formation of future blood clots and help reduce the risk of embolism (migration of a thrombus to a spot where it blocks blood supply to a vital organ). Warfarin is best suited for anticoagulation (clot formation inhibition) in areas of slowly run-ning blood (such as in veins and the pooled blood behind artificial and natural valves) and in blood pooled in dysfunctional cardiac atria. Thus, common clinical indications for warfarin use are atrial fibrillation, the presence of artificial heart valves, deep venous thrombosis, and pulmonary embolism (where the embolized clots first form in veins). Warfarin is also used in antiphospholipid syndrome. It has been used occasionally after heart attacks (myocardial infarctions), but is far less effective at preventing new thromboses in coronary arteries. Preven-tion of clotting in arteries is usually undertaken with antiplatelet drugs, which act by a different 17
  • 19. CHAPTER 2. PHARMACOGENETICS OF WARFARIN 18 mechanism from warfarin (which normally has no effect on platelet function). 2.1.2 Mechanism of Action Warfarin inhibits the vitamin K-dependent synthesis of biologically active forms of the calcium-dependent clotting factors II, VII, IX and X, as well as the regulatory factors protein C, protein S, and protein Z. Other proteins not involved in blood clotting, such as osteocalcin, or matrix Gla protein, may also be affected. The precursors of these factors require carboxylation of their Figure 2.1: Warfarin Metabolism and Mechanism of Action glutamic acid residues to allow the coagulation factors to bind to phospholipid surfaces inside blood vessels, on the vascular endothelium. The enzyme that carries out the carboxylation of glutamic acid is gamma-glutamyl carboxylase. The carboxylation reaction will proceed only if the carboxylase enzyme is able to convert a reduced form of vitamin K (vitamin K hydro-quinone) to vitamin K epoxide at the same time. The vitamin K epoxide is in turn recycled back to vitamin K and vitamin K hydroquinone by another enzyme, the vitamin K epoxide reduc-tase (VKOR). Warfarin inhibits epoxide reductase (specifically the VKORC1 subunit), thereby diminishing available vitamin K and vitamin K hydroquinone in the tissues, which inhibits the carboxylation activity of the glutamyl carboxylase. When this occurs, the coagulation factors are no longer carboxylated at certain glutamic acid residues, and are incapable of binding to the endothelial surface of blood vessels, and are thus biologically inactive. As the body’s stores
  • 20. CHAPTER 2. PHARMACOGENETICS OF WARFARIN 19 of previously produced active factors degrade (over several days) and are replaced by inactive factors, the anticoagulation effect becomes apparent. The coagulation factors are produced, but have decreased functionality due to undercarboxylation; they are collectively referred to as PIVKAs (proteins induced [by] vitamin K absence/antagonism), and individual coagulation fac-tors as PIVKA-number (e.g.PIVKA-II). The end result of warfarin use, therefore, is to diminish blood clotting in the patient. 2.2 Metabolism of Warfarin Warfarin is a natural product and given as racemic mixture of the R and S stereoisomers of the drug. S-warfarin is 3-5 times more potent an inhibitor of the vitamin K epoxide reductase complex, the target of action, than R-warfarin. The stereoisomers are metabolized by differ-ent phase 1 enzymes; the predominant metabolism of the S isomer is via CYP2C9 whereas metabolism of R-warfarin is mainly via CYP3A4 with involvement of CYP1A1, CYP1A2, CYP2C8, CYP2C9, CYP2C18 and CYP2C19 as depicted in the Warfarin Pharmacokinetics Pathway [Figure 2.2]. Phase 2 metabolism of warfarin has not been well studied and is not depicted in this pathway representation, although it is known that sulfated and glucuronyl con-jugates can be formed. Elimination is predominantly renal however warfarin has been shown to interact with the ABCB1 transporter in liver. Figure 2.2: Warfarin Metabolic Pathway 2.3 Pharmacogenetics ofWarfarin The association between warfarin dose and the CYP2C9 and VKORC1genes is now unquestion-able, as evidenced by numerous studies. Single nucleotide polymorphisms in the cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKOR) genes have been shown to have
  • 21. CHAPTER 2. PHARMACOGENETICS OF WARFARIN 20 a significant effect on warfarin dose requirement. Other genes mediating the action of warfarin make either little or no contribution to dose requirement. Although the polymorphisms in- CYP2C9andVKORC1explain a significant proportion of the interindividual variability in war-farin dose requirement, currently available evidence based on a few small studies relating to the use of pharmacogenetics-guided dosing in the initiation of warfarin therapy has not shown improved outcomes in either safety or efficacy of therapy. Better clinical evidence of beneficial effects on patient outcome, particularly at the extremes of the dose requirements in geographi-cally and ethnically diverse patient populations, is needed before the role of a pharmacogenomic approach to oral anticoagulation therapy in clinical practice can be established. Patients carrying CYP2C9 variant alleles had a higher rate of above-range INR values, took longer to reach stable dosing, and had a higher risk of serious and life-threatening bleeding events than patients with the wild-type allele. Similar results were noted in a study of warfarin-treated patients whereCYP2C92 or 3 compound heterozygotes and homozygotes had low war-farin requirements and increased rates of excessive (INR6.0) anticoagulation and bleeding com-pared with wild-type patients. Table 2.1: Enzymatic activity of different CYP2C9 allelic variants
  • 22. CHAPTER 2. PHARMACOGENETICS OF WARFARIN 21 2.3.1 CYP2C9 andWarfarin dosing CYP2C9*1 metabolizes warfarin normally, CYP2C9*2 reduces warfarin metabolism by 30%, and CYP2C9*3 reduces warfarin metabolism by 90%. Because warfarin given to patients with *2 or *3 variants will be metabolized less efficiently, the drug will remain in circulation longer, so lower warfarin doses will be needed to achieve anticoagulation. 2.3.2 VKORC1 andWarfarin In the VKORC1 1639 (or 3673) SNP, the common G allele is replaced by the A allele. Because people with an A allele (or the ”A haplotype”) produce less VKORC1 than do those with the G allele (or the ”non-A haplotype”), lower warfarin doses are needed to inhibit VKORC1 and to produce an anticoagulant effect in carriers of the A allele. The prevalence of these variants also varies by race, with 37% of Caucasians and 14% of Africans carrying the A allele. 2.3.3 CYP4F2 andWarfarin Recent genome wide association studies have not only confirmed these observations but also identified a novel association between rs2108622 in CYP4F2 and reduced hepatic CYP4F2, higher levels of hepatic vitamin K, and higher warfarin dose requirements. 2.4 CPIC dosing guideline for Warfarin Approach to pharmacogenetic-based warfarin dosing without access to dosing algorithms: In 2007, the FDA modified the warfarin label, stating that CYP2C9 and VKORC1 genotypes may be useful in determining the optimal initial dose of warfarin. The label was further updated in 2010 to include a table describing recommendations for initial dosing ranges for patients with different combinations of CYP2C9 and VKORC1 genotypes. Genetics-based algorithms also better predict warfarin dose than the FDA-approved warfarin label table. Therefore, the use of pharmacogenetic algorithm-based dosing is recommended when possible, although if electronic means for such dosing are not available, the table-based dosing approaches are suggested. Table 2.2: Genotype based Dosing guideline by Clinical Pharmacogenetics Implementation Consortium (CPIC) 2.5 References: 1. Johnson JA, Gong L, Whirl-Carrillo M, Gage BF, Scott SA, Stein CM, Anderson JL, Kimmel SE, Lee MT, Pirmohamed M,Wadelius M, Klein TE, Altman RB; Clinical Phar-
  • 23. CHAPTER 2. PHARMACOGENETICS OF WARFARIN 22 macogenetics Implementation Consortium. Clinical Pharmacogenetics Implementation Consortium Guidelines for CYP2C9 and VKORC1 genotypes and warfarin dosing. Clin Pharmacol Ther. 2011 Oct;90(4):625-9. doi: 10.1038/clpt.2011.185. Epub 2011 Sep 7. 2. Holford, NH (December 1986). ”Clinical Pharmacokinetics and Pharmacodynamics of Warfarin Understanding the Dose-Effect Relationship”. Clinical Pharamacokinetics (Springer International Publishing) 11 (6): 483504. doi:10.2165/00003088-198611060-00005. PMID 3542339 3. Holbrook AM, Pereira JA, Labiris R, McDonald H, Douketis JD, Crowther M, Wells PS (May 2005). ”Systematic overview of warfarin and its drug and food interactions”. Arch. Intern. Med. 165 (10): 1095106. doi:10.1001/archinte.165.10.1095. PMID 15911722 4. Ansell J, Hirsh J, Hylek E, et al. (2008). ”Pharmacology and management of the vitamin K antagonists: American College of Chest Physicians evidence-based clinical practice guidelines (8th Edition)”. Chest 133 (6 Suppl): 160S198S. doi:10.1378/chest.08-0670. PMID 18574265 5. Hirsh J, Fuster V, Ansell J, Halperin JL (2003). ”American Heart Association/American College of Cardiology Foundation guide to warfarin therapy”. J. Am. Coll. Cardiol. 41 (9): 163352. doi:10.1016/S0735-1097(03)00416-9. PMID 12742309 6. Ansell J, Jacobson A, Levy J, Vller H, Hasenkam JM (March 2005). ”Guidelines for im-plementation of patient self-testing and patient self-management of oral anticoagulation. International consensus guidelines prepared by International Self-Monitoring Association for Oral Anticoagulation”. Int. J. Cardiol. 99 (1): 3745. doi:10.1016/j.ijcard.2003.11.008. PMID 15721497 7. Aithal GP, Day CP, Kesteven PJ, Daly AK. 1999. Association of polymorphism in the cytochrome P450 CYP2C9 with warfarin dose requirements and risk of bleeding compli-cations. Lancet353:71719 8. Daly AK, King BP. 2003. Pharmacogenetics of oral anticoagulants.Pharmacogenetics13:24752 9. DAndrea G, DAmbrosio RL, Di Perna P, et al. 2005. A polymorphism in the VKORC1 gene is associated with an interindividual variability in the dose-anticoagulant effect of warfarin.Blood105:64549 10. Scordo MG, Aklillu E, Yasar U, et al. 2001. Genetic polymorphism of cytochrome P450 2C9 in a Caucasian and a black African population.Br. J. Clin. Pharmacol.52:44750
  • 24. Chapter 3 Pharmacogenetics of Clopidogrel 3.1 Clopidogrel Clopidogrel (INN) is an oral, thienopyridine-class antiplatelet agent used to inhibit blood clots in coronary artery disease, peripheral vascular disease, cerebrovascular disease, and to prevent myocardial infarction (heart attack). It is marketed by Bristol-Myers Squibb and Sanofi under the trade name Plavix. The drug works by irreversibly inhibiting a receptor called P2Y12, an adenosine diphosphate (ADP) chemoreceptor on platelet cell membranes. Adverse effects include hemorrhage, severe neutropenia, and thrombotic thrombocytopenic purpura. Clopidogrel is used to prevent myocardial infarction (heart attack) and stroke in people who are at high risk of these events, including those with a history of myocardial infarction and other forms of acute coronary syndrome, stroke and those with peripheral artery disease. Treatment with clopidogrel or a related drug is recommended by the American Heart Association and the American College of Cardiology for people who: 1. Present for treatment with a myocardial infarction with ST-elevation, including (a) A loading dose given in advance of percutaneous coronary intervention (PCI), fol-lowed by a full year of treatment for those receiving a vascular stent (b) A loading dose given in advance of fibrinolytic therapy, continued for at least 14 days 2. Present for treatment of a non-ST elevation myocardial infarction (NSTEMI) or unstable angina. (a) Including a loading dose and maintenance therapy in those receiving PCI and unable to tolerate aspirin therapy (b) Maintenance therapy for up to 12 months in those at medium to high risk for which a non-invasive treatment strategy is chosen 3. In those with stable ischemic heart disease,[4] treatment with clopidogrel is described as a ”reasonable” option for monotherapy in those who cannot tolerate aspirin, as is treatment with clopidogrel in combination with aspirin in certain high risk patients. 3.1.1 Pharmacology Clopidogrel is a prodrug, which requires CYP2C19 for its activation. It acts on the ADP recep-tor on platelet cell membranes. The drug specifically and irreversibly inhibits the P2Y12 subtype 23
  • 25. CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 24 of ADP receptor, which is important in activation of platelets and eventual cross-linking by the protein fibrin. Platelet inhibition can be demonstrated two hours after a single dose of oral clopi-dogrel, but the onset of action is slow, so a loading dose of either 600 or 300 mg is administered when a rapid effect is needed. 3.1.2 Why it is important to study CYP2C19 polymorphism 1. Antiplatelet drugs reduce the risk of heart attack, unstable angina, stroke, and cardiovas-cular death in patients with coronary heart disease by making platelets less likely to form blood clots. 2. Antiplatelet drug does not have its antiplatelet effects until it is metabolized into its active form by CYP2C19. 3. Study by De Morais et al, 1994 detects the *17 allele as an ultra fast metabolizer of antiplatelet drugs. A recent report indicates the increased risk for bleeding in individuals treated with clopidogrel who carry the *17 allele. 4. People who have reduced functioning of their CYP2C19 liver enzyme due to inactivating polymorphisms in their CYP2C19 gene cannot effectively convert Plavix to its active form. As a result, Plavix may be less effective in altering platelet activity in those people. These poor metabolizers may not receive the full benefit of Plavix treatment and may remain at risk for heart attack, stroke, and cardiovascular death. 5. It is estimated that 1.5% to 19.2% of the Asian population are ultra fast metabolizers. 3.2 Bioactivation of Clopidogrel and Prasugrel Clopidogrel is a prodrug, and its activation is complex (Figure 3.1). Some 85% of the dose administered is hydrolyzed in the liver, where it is converted into an inactive metabolite (SR 26334); the remainder is metabolized by CYP2C19 and to a lesser degree by CYP1A2 and CYP2B6 to 2-oxo-clopidogrel. Around half of this metabolite, which is also inactive, is hy-drolyzed and converted into an inactive thiolactone, while the other half is metabolized by the 3A4, 3A5, 2B6, 2C9 and 2C19 isoenzymes, finally producing the active metabolite (R- 130964). The fact that a significant proportion of the absorbed drug is wasted (converted to inactive metabolites), the complex activation of the prodrug requiring two oxidation steps, and its heavy dependence on the function of the CYP2C19 isoenzyme (which is involved in the me-tabolization of numerous drugs and whose reduced-function genetic variants have a prevalence of around 25%), are the main pharmacokinetic factors affecting the variability of response to clopidogrel. 3.3 Clopidogrel and CYP2C19 One of the main causes of this clopidogrel nonresponsiveness is interindividual variability in the metabolic activation of clopidogrel. Polymorphisms in the CYP2C19 gene affect clopidogrel pharmacokinetics, causing the CYP2C19 enzyme that converts Plavix to its active form to be more or less effective. The most common CYP2C19 polymorphisms,*2, *3, and *17, result in highly variable enzyme activity, and are used to classify individuals as to metabolizer type:
  • 26. CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 25 Figure 3.1: Comparative metabolic pattern of Clopidogrel and Prasugrel poor, intermediate, extensive (normal), or ultrarapid. These polymorphisms may be useful to identify clopidogrel nonresponders (carrying the *2 or *3 alleles) who may benefit by taking an alternative antiplatelet agent, such as prasugrel and ticagrelor, or individuals who may be at risk of bleeding due to clopidogrel overactivity (*17 allele carriers). A loss-of-function polymorphism in the CYP2C19 gene, known as the CYP2C19*2 allelic variant, has been associated with higher levels of ADP-induced platelet aggregation values in clopidogrel-treated patients and consequently a higher risk of major adverse cardio vascular events, including the occurrence of stent thrombosis (ST). A novel allelic variant, CYP2C19*17, results in an increased enzyme function of CYP2C19 because of a mutation (806CT) in the 5-flanking region of the gene that causes an increased transcription of CYP2C19. Such increased transcriptional activity of CYP2C19 may confer a rapid metabolization of CYP2C19 substrates, which may lead to an enhanced response to antiplatelet treatment. Although this may improve the prevention of thrombotic events, it also may increase the risk of bleeding. 3.4 Genetic variation in CYP2C19 influences individual re-sponse to clopidogrel The CYP2C19 gene is highly polymorphic with over 25 known variants. Functional polymor-phisms in the CYP2C19 gene, the most common of which are *2, *3, and *17, result in highly variable enzyme activity, influencing an individuals ability to activate clopidogrel. These vari-ants cause the CYP2C19 enzyme that converts Plavix to its active form to be more or less effective and are used to classify individuals as to metabolizer type: extensive metabolizers
  • 27. CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 26 (normal conversion to the active metabolite), intermediate/poor metabolizers (incomplete con-version to the active metabolite), and ultrarapid metabolizers (increased conversion to the active metabolite). Table 3.1: Common polymorphisms in the CYP2C19 gene that influence anti-platelet response to clopidogrel Approximately 2-4% of Caucasian, 4% of African or African/American, and 14-20% of Asian populations are poor metabolizers (PM) because these individuals have two loss-of-function alleles and very low CYP2C19 activity. Intermediate metabolizers have one CYP2C19 loss-of-function allele. Approximately 30-50% of the population are intermediate metabolizers ( 30% of Caucasians, 40% of African-Americans, and more than 55% of Asians). Table 3.2: Allelic Frequency of common CYP2C19 variants by ethnicity 3.5 Choosing the best option: Clopidogrel? Prasugrel? or Ticagrelor? Prasugrel and ticagrelor are newer antiplatelet drugs that could be an alternative to clopidogrel. Their effect is less variable and fewer ischemic events are associated with their use; however,
  • 28. CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 27 more bleeding is related to the use of these drugs. Pharmacogenetic studies are currently con-sidering these drugs. So far, no association has been found. Prasugrel are found to be a risk factor for intracranial bleeding resulting in strokes in around 2.5% population. CYP2C19 variants shows differences in pharmacodynamic and Pharmacoki-netic properties variation of Clopidogrel. Figure 3.2: Comparative Pharmacokinetic pathway of Clopidogrel, Prasugrel and Ticagrelor Clopidogrel is activated in a 2-step process mediated by oxidative biotransformation in the liver, in which CYP2C19 and CYP3A have particularly important roles. The parent com-pound clopidogrel, and to a lesser extent 2-oxo-clopidogrel, are both substrates and inhibitors of CYP1A2, CYP2B6, and CYP2C19. Clopidogrel and 2-oxo-clopidogrel are extensively hy-drolyzed to inactive metabolites, potentially magnifying the effects of CYP2C19 inhibitors and
  • 29. CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 28 polymorphisms. Prasugrel is also a pro-drug that requires biotransformation to active metabolites by cy-tochrome P-450 enzymes, including CYP3A isoforms, CYP2B6, CYP2C9, and CYP2C19. Prasugrel is hydrolyzed to a thiolactone derivative in the intestine and then oxidized to its ac-tive metabolite in both the intestine and the liver. Reduced-function CYP2C19 alleles are not believed to have a clinically meaningful effect in prasugrel-treated patients. Ticagrelor (AZD6140) is an orally active cyclopentyltriazolopyrimidine adenosine triphos-phate analog that reversibly inhibits P2Y12 platelet receptors. Ticagrelor, which is not yet approved in the United States, is an active compound and is metabolized by CYP3A4 to an active metabolite. Ticagrelor and its active metabolite are both metabolized and glucuronidated in the liver before elimination in the urine. Genetic variations in CYP isoenzymes do not appear to affect metabolism of ticagrelor. 3.6 References 1. Desta Z, Zhao X, Shin JG, Flockhart DA. Clinical significance of the cytochrome P450 2C19 genetic polymorphism. Clin Pharmacokinet. 2002;41(12):913-958. 2. Campo G, Fileti L, Valgimigli M, et al. Poor response to clopidogrel: current and future options for its management. J Thromb Thrombolysis. 2010;30(3):319-31. 3. Gurbel PA, Tantry US. Selecting optimal antiplatelet therapy based on platelet function monitoring in patients with coronary artery disease. Curr Treat Options Cardiovasc Med. 2009;11(1):22-32. 4. Kuliczkowski W, Witkowski A, Polonski L, et al. Interindividual variability in the re-sponse to oral antiplatelet drugs: a position paper of the Working Group on antiplatelet drugs resistance appointed by the Section of Cardiovascular Interventions of the Polish Cardiac Society, endorsed by theWorking Group on Thrombosis of the European Society of Cardiology. Eur Heart J. 2009;30(4):426-435. 5. Terpening C. Clopidogrel: A pharmacogenomic perspective on its use in coronary artery disease. Clin Med Insights Cardiol. 2010;4:117-128. 6. Sharma RK, Reddy HK, Singh VN, et al. Aspirin and clopidogrel hyporesponsiveness and nonresponsiveness in patients with coronary artery stenting. Vasc Health Risk Manag. 2009;5:965-972. 7. Simon T, Verstuyft C, Mary-Krause M, et al. Genetic determinants of response to clopi-dogrel and cardiovascular events. New Engl J Med. 2009;360(4):363-375. 8. Sofi F, Marcucci R, Gori AM, Abbate R, Gensini GF. Clopidogrel non-responsiveness and risk of cardiovascular morbidity. An updated meta-analysis. Thromb Haemost. 2010;103(4):841-848. 9. Hulot J, Bura A, Villard E, Azizi M, et al. P450 2C19 loss-of-function polymorphism. Cytochrome is a major determinant of clopidogrel responsiveness in healthy subjects. Blood. 2006;108(7):2244-2247.
  • 30. CHAPTER 3. PHARMACOGENETICS OF CLOPIDOGREL 29 10. Geisler T, Schaeffeler E, Dipporr J, et al. CYP2C19 and nongenetic factors predict poor responsiveness to clopidogrel loading dose after coronary stent implantation. Pharma-cogenomics. 2008;9(9):1251-1259. 11. Mega JL, Close SL, Wiviott SD, et al. Cytochrome P-450 polymorphisms and response to clopidogrel. N Engl J Med. 2009;360(4):354-362. 12. http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationfor Patientsand- Providers/ucm203888.htm. (Accessed Jun 8, 2011). 13. Product information for Plavix (Sanofi/Aventis US). Label information, approved Feb 2011. (htt p : ==www:accessdata: f da:gov=drugsat f dadocs=label=2011=020839s052lbl:) 14. Kazui M, Nishiya Y, Ishizuka T, et al. Identification of the human cytochrome P450 enzymes involved in the two oxidative steps in the bioactivation of clopidogrel to its pharmacologically active metabolite. Drug Metab Dispos. 2010;38(1):92-99. 15. Dahl M, Gunes A. Implications of inter-individual differences in clopidogrel metabolism, with Focus on Pharmacogenetics. Pharmaceuticals. 2010;3(4):782-794. 16. Pare G, Mehta SR, Yusuf S, et al. Effects of CYP2C19 genotype on outcomes of clopi-dogrel treatment. N Engl J Med. 2010;363(18):1704-1714. 17. Sim SC, Risinger C, Dahl ML, et al. A common novel CYP2C19 gene variant causes ultrarapid drug metabolism relevant for the drug response to proton pump inhibitors and antidepressants. Clin Pharmacol Ther. 2006;79(1):103-113. 18. Li-wan-po A, Girard T, Farndon P, Cooley C, Lithgow J. Pharmacogenetics of CYP2C19: functional and clinical implications of a new variant CYP2C19*17. Br J Clin Pharmacol. 2010;69(3):222-230. 19. Shuldiner AR, OConnell JR, Bliden KP, Ghandhi A, et al. Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel ther-apy. JAMA. 2009;302(8):849-858. 20. Human cytochrome P450 allele nomenclature (htt p : ==www:cypalleles:ki:se=cyp2c19:htm) 21. Mega, J.L. et al. Reduced-function CYP2C19 genotype and risk of adverse clinical out-comes among patients treated with clopidogrel predominantly for PCI: a meta-analysis. JAMA. 2010;304(16):1821-1830. 22. Mega JL, Hochholzer W, Frelinger AL, Kluk MJ, et al. Dosing clopidogrel based on CYP2C19 genotype and the effect on platelet reactivity in patients with stable cardiovas-cular disease. JAMA. 2011;306(20):2221-2228. 23. Trenk D, Hochholzer W, Fromm MF, Chialda L, et al. Cytochrome P450 2C19 681G¿A polymorphism and high on-clopidogrel platelet reactivity associated with adverse 1-Year clinical outcome of elective percutaneous coronary intervention with drug-eluting or bare-metal stents. J Am Coll Cardiol. 2008;51(20):1925-1934. 24. Giusti B, Goria AM, Marcuccia R, et al. Cytochrome P450 2C19 loss-of-function poly-morphism, but not CYP3A4 IVS10 + 12G/A and P2Y12 T744C polymorphisms, is as-sociated with response variability to dual antiplatelet treatment in high-risk vascular pa-tients. Pharmacogenet Genomics. 2007;17:1057-1064.
  • 31. Chapter 4 Pharmacogenetics of NAT2 The arylamine N-acetyltransferases (NATs) are found in nearly all species from bacteria to humans. They catalyse the acetyltransfer from acetylcoenzyme A to an aromatic amine, hete-rocyclic amine or hydrazine compound. In humans, acetylation is a major route of biotransfor-mation for many arylamine and hydrazine drugs, as well as for a number of known carcinogens present in the diet, cigarette smoke and the environment. The reaction pathway is catalysed by two cytoplasmic acetyltransferases (NAT; EC 2.3.1.5), N-acetyltransferase Type I (NAT1) and N-acetyltransferase Type II (NAT2). The arylamine N-acetyltransferases (NATs) are involved in the metabolism of a variety of different compounds that we are exposed to on a daily basis. Many drugs and chemicals found in the environment, such as those in cigarette smoke, car exhaust fumes and in foodstuffs, can be either detoxified by NATs and eliminated from the body or bioactivated to metabolites that have the potential to cause toxicity and/or cancer. NATs have been implicated in some adverse drug reactions and as risk factors for several different types of cancers. As a result, the levels of NATs in the body have important consequences with regard to an individual’s susceptibility to certain drug-induced toxicities and cancers. 4.1 NAT localization Two NAT isoenzymes have been identified in humans, namely NAT1 and NAT2, which are the products of distinct genetic loci, designated NAT1 and NAT2, respectively. A related pseu-dogene, NATP1, has also been identified, which contains multiple frameshift and nonsense mutations. The two functional NAT genes share an 87% nucleotide identity, which translates to an 81% homology at the amino acid level. While the entire transcript of NAT1 is derived from a single exon, that of NAT2 is derived from the protein encoding exon together with a sec-ond noncoding exon of 100 bp located about 8kb upstream of the translation start site. Human NAT1 and NAT2, as well as NATP1, have been localised to the short arm of chromosome 8, more specifically in region 8p22. The NAT loci are separated by only 170-360kb and are in the orientation NAT1!NATP1!NAT2. 4.2 Human NAT2 Alleles Since the human NAT2 locus was established as the site of the classical acetylation polymor-phism, the study of NAT2 allelic variation has been an area of intense investigation. To date, 29 different NAT2 alleles have been detected in human populations. Each of the variant alleles is 30
  • 32. CHAPTER 4. PHARMACOGENETICS OF NAT2 31 comprised of between one and four nucleotide substitutions, of which 13 have been identified, located in the protein encoding region of the gene. Nine of these lead to a change in the encoded amino acid (C190T, G191A, T341C, A434C, G499A, G590A, A803G, A845C, and G857A), while the remaining four are silent (T111C, C282T, C481T, and C759T). Several studies have been performed that show clear correlations between NAT2 genotype and phenotype. Early genotyping studies screened for the presence of the C481T (M1), the G590A (M2), the G857A (M3) and sometimes the G191A (M4) nucleotide changes, all of which were shown to cause a slow acetylation phenotype. Moreover, there was a gene-dosage effect. Individuals who were homozygous for NAT2 polymorphisms had a slow acetylator phenotype, individuals heterozygous for NAT2 polymorphisms had an intermediate acetylator phenotype, and individuals who lacked NAT2 polymorphisms had a rapid acetylator phenotype. It should be noted that the method of detection of the above polymorphisms only identifies a subset of the variant alleles found in human populations, and there is potential for the misclas-sification of genotype and deduced phenotypes. 4.3 NAT and Disease The association between acetylator status and the risk of various diseases has been extensively reported, and reviewed in detail. Altered risk with either the slow or rapid phenotype has been observed for bladder, colon and breast cancer, systemic lupus erythematosis, diabetes, Gilbert’s disease, Parkinson’s disease and Alzheimer’s disease. These associations imply a role for envi-ronmental factors that are metabolised by the NATs, in particular NAT2, in each disorder. How-ever, identifying those factors has remained elusive. Humans are exposed to many toxic NAT substrates including the food-derived heterocyclics present in the diet as well as arylamines such as 4-aminobiphenyl and -naphthylamine present in tobacco smoke. Moreover, occupational ex-posure to arylamine carcinogens such as benzidine has also been reported. Because of the role of acetylation in the metabolic activation and detoxification of ary-lamine and heterocyclic carcinogens, acetylator status and cancer risk has been widely investi-gated. Unlike the relatively rare but highly penetrant genes involved in familial cancers, those genes responsible for metabolic polymorphisms have low penetrance and cause only a moder-ate increase in cancer risk. Nevertheless, their widespread occurrence in the general population suggests they are a significant contributor to individual risk. When acetylator phenotype has been linked to carcinogen exposure, more consistent results have been reported. For example, the rapid phenotype has emerged as a strong risk factor for colorectal cancer in those individuals who have a higher exposure to the food-derived hete-rocyclic amines. Recently, the NAT2 acetylator phenotype has been linked to increased risk associated with neurodegenerative diseases such as Parkinson’s disease and Alzheimer’s dis-ease. 4.4 NAT2 and Drug Response 4.4.1 NAT2 and Isoniazid The genetic polymorphism in N-acetyltransferase activity was first discovered in patients treated with isoniazid for tuberculosis. This drug is primarily excreted following acetylation catalysed by NAT2.
  • 33. CHAPTER 4. PHARMACOGENETICS OF NAT2 32 Isoniazid (INH), a key drug of antituberculosis therapy, is metabolized by arylamine N-acetyltransferase2 (NAT2), cytochrome P450 2E1 (CYP2E1) and glutathione S-transferase (GST). The metabolism of INH into acetyl isoniazid (AcINH) by N-acetyltransferase 2 (NAT2), which is estimated to account for 50-90% of INH metabolism. A portion of AcINH is further con-verted to acetyl hydrazine (AcHz) by amidase-catalyzed hydrolysis. There is another amidase-catalyzed route where INH is directly hydrolyzed to yield hydrazine (Hz). NAT2 then catalyzes the acetylation of Hz to yield AcHz and subsequently to diacetyl hydrazine before excretion. AcHz can also enter the cytochrome P450 2E1 (CYP2E1)-mediated metabolic pathway in the liver, which is linked to the glutathione-S-transferase (GST)-mediated metabolic pathway, to become an excretable metabolite. Figure 4.1: Metabolism of Isoniazid NAT2 genetic polymorphisms and the resulting variation in acetylation capacity of NAT2 are implicated in the risk of INH-induced hepatotoxicity. Slow acetylator genotype of NAT2 will activate the CYP2E1 pathway resulting in reactive metabolite, hence will cause hepatotox-icity (Hepatic Necrosis). But there are some contradictory result also, like RA (rapid or fast acetylator) genotype also have a significant role in INH induced hepatotoxicity. Hydrazine and N-actyl Hydrazine are supposed to be hepatotoxic so are RA groups. So it is necessary to con-sider the role of CYP2E1 and GST in INH metabolism to draw a conclusion here without any conflict. 4.4.2 NAT2 and Environmental carcinogen Biological Amines are not reactive and converted into a highly electrophilic reactive metabolite, acts as a carcinogen and later detoxified by the action of NAT2 enzymes into an excretable non-
  • 34. CHAPTER 4. PHARMACOGENETICS OF NAT2 33 reactive form. Slow-acetylator phenotype should have a higher risk of developing cancer by this biological amine than in fast-acetylator phenotype. 4.4.3 NAT2 and Procainamide Procainamide is a class Ia antiarrhythmic used for the treatment of several different arrhyth-mias. It is metabolized by n-acetyltransferases (NAT) to n-acetylprocainamide (NAPA), an ac-tive metabolite, which also possesses antiarrhythmic properties. NAT1 is consistently expressed in post patients; however, NAT2 is variably expressed and plays a major role in the production of NAPA. The majority of patients reveiving procainamide therapy will develop autoantibodies over time and possibly drug induced lupus. An early study showed that subjects who had a slow-acteylator phenotype developed antinuclear antibodies earlier than rapid acetylators. This was later confirmed in a study of subjects who received long term procainamide therapy. The-oretically patients with NAT2 genotype associated with rapid-acetylation may have increased NAPA concentrations and thus increased anti-arrhythmic effects and possible excessive QT prolongation. 4.5 References 1. DWHein. N-acetyltransferase 2 genetic polymorphism: effects of carcinogen and haplo-type on urinary bladder cancer risk. Oncogene (2006) 25, 16491658. doi:10.1038/sj.onc.1209374 2. Alexandra King. NAT2 and GSTM1 polymorphisms affect the risk of bladder cancer. Nature Reviews Clinical Oncology 2, 540 (November 2005)—doi:10.1038/ncponc0307 3. T SOTSUKA, Y SASAKI, S HIRAI, F YAMAGISHI and K UENO. Association of Isoniazid-metabolizing Enzyme Genotypes and Isoniazid-induced Hepatotoxicity in Tu-berculosis Patients. In Vivo. 2011. 25(5): 803-812. 4. Evans DA (1989). ”N-acetyltransferase”. Pharmacology Therapeutics 42 (2): 157234. doi:10.1016/0163-7258(89)90036-3. PMID 2664821.
  • 35. CHAPTER 4. PHARMACOGENETICS OF NAT2 34 Table 4.1: Common NAT2 alleles and their phenotype
  • 36. Chapter 5 Pharmacogenetics of CYP2D6 5.1 CYP2D6 allelic variants More than 50 human CYP isozymes have been identified to date. Of these, more than 20 are encoded by genes that are functionally polymorphic, including CYP2A6, CYP2C9, CYP2C19, and CYP2D6. Consequently, approximately 40% of CYP-dependent drug metabolism is carried out by polymorphic enzymes. Table 5.1: CYP2D6 Substrate CYP2D6 is the best characterized of these polymorphic CYP isozymes, with more than 75 allelic variants currently identified. These variants result from point mutations, deletions or additions, gene rearrangements, and deletion or duplication of the entire gene, and result in an increase, reduction, or complete loss of activity. While it accounts for only 2%5% of all hepatic CYP isozymes, CYP2D6 metabolizes approximately 25% of all clinically used medications, including some cytotoxics, tamoxifen, and many agents used to treat associated complications such as antiarrythmics, antiemetics, antidepressants, antipsychotics, and analgesics. Since the identification of the CYP2D6 polymorphism in the 1970s, several studies have shown that the frequencies of the alternative CYP2D6 phenotypes vary significantly among different ethnic 35
  • 37. CHAPTER 5. PHARMACOGENETICS OF CYP2D6 36 groups and that such polymorphisms may play a role in the induction of adverse effects from administration of some therapeutic agents. Variation in CYP2D6 expression is also thought to increase the potential for drugdrug interactions, an important implication because of the increasing number of drugs being prescribed. For example, in the oncology setting, patients with advanced cancer receiving palliative therapy receive an average of five or more medications for symptom relief. Table 5.2: CYP2D6 Allelic variants with enzyme function in different ethnic groups A number of specific polymorphisms have been found in the CYP2D6 gene that results in enzymatic deficiencies. The frequency of these polymorphisms varies within the major ethnic groups. CYP2D6 polymorphisms that produce poor metabolizers are found with frequencies of 7% to 10% in Caucasians, 2% in Africans and African Americans, and 1% in Asians. Identifica-tion of patient CYP2D6 genotypes can help clinicians individualize drug treatment by selection of appropriate therapies. These measures may improve patient outcome by ensuring maximum drug efficacy with minimal adverse drug reactions (ADR).
  • 38. CHAPTER 5. PHARMACOGENETICS OF CYP2D6 37 Table 5.3: Genotype and Phenotype of CYP2D6 Alleles CYP2D6 polymorphisms can be classified according to one of four levels of activity: poor metabolizers (PMs), intermediate metabolizers (IMs), extensive metabolizers (EMs), and ul-trarapid metabolizers (UMs) [Figure 5.4]. The EM phenotype is expressed by the majority of the population and is therefore considered the norm. PMs inherit two deficient CYP2D6 alleles and, as a result, metabolize drugs at a notably slower rate. This leads to an accumulation of high levels of unmetabolized drugs that are CYP2D6 substrates, a concomitant greater potential for adverse events and drugdrug interactions, and lower efficacy for drugs requiring CYP2D6 acti-vation. The UM phenotype is caused by the duplication, multiduplication, or amplification of active CYP2D6 genes, including primarily the CYP2D6*2 allele, but also involving CYP2D6*1 and others. Individuals with the UM phenotype metabolize drugs at an ultrarapid rate, which may lead to a loss of therapeutic efficacy at standard doses. Individuals who are heterozygous for a defective CYP2D6 allele often demonstrate an IM phenotype. This phenotype has a wide spectrum of metabolic activity that can range from marginally better than the PM phenotype to activity that is close to that of the EM phenotype. 5.2 CYP2D6 and Antipsychotics Pharmacology CYP2D6 is described as the most relevant enzyme in the metabolism of many antipsychotic drugs. Its contribution to the interindividual differences in drug response is reviewed here high-lighting its role in the kinetics of antipsychotic drugs and the occurrence of drug interactions. The activity of CYP2D6 is inherited as a monogenetic trait and the CYP2D6 gene appears highly polymorphic in humans. The polymorphic alleles may lead to altered activity of the
  • 39. CHAPTER 5. PHARMACOGENETICS OF CYP2D6 38 Table 5.4: Metabolizer classification of CYP2D6 CYP enzymes causing absent, decreased (poor), or increased (ultrarapid) metabolism that in turn influence the disposition of the antipsychotic drugs. Antipsychotic drug biotransformation is mainly determined by genetic factors mediating CYP2D6 gene polymorphism, however the importance of environmental factors (dietary, smoking, diseases, etc.) is also recognized. Ad-ditionally, the potential interaction between CYP2D6 and the endogenous metabolism must be taken into consideration. 5.2.1 Metabolism of Amytriptyline Amytriptyline (AT) is metabolized mainly by demethylation forming nortriptyline (NT), and by hydroxylation, leading to the formation of E-10-hydroxy (EHAT) and Z-10-hydroxyamitriptyline (ZHAT). NT is further demethylated to desmethylnortriptyline (NNT) and hydroxylated to E- 10-hydroxy (EHNT) and Z-10-hydroxynortriptyline (ZHNT). The demethylation of AT and NT is mainly catalyzed by CYP2C19, with the participation of other CYP enzyme forms in higher drug concentrations. The formation of the E-10-hydroxy metabolites is dependent on the activ-ity of CYP2D6, with stereospecificity to the (-)-EHAT and (-)-EHNT metabolites. 5.2.2 Dosing recommendations for amitriptyline/nortriptyline based on CYP2D6 phenotype 1. Likely phenotype: Ultrarapid metabolizer ( 1-2% of patients)
  • 40. CHAPTER 5. PHARMACOGENETICS OF CYP2D6 39 Figure 5.1: Metabolism of Amytriptyline (a) Genotypes: An individual carrying more than two copies of functional alleles. (b) Examples of diplotypes: *1/*1xN, *1/*2xN (c) Implications for TCA metabolism: Increased metabolism of tricyclics to less ac-tive compounds when compared to extensive metabolizers. Lower plasma concen-trations will increase probability of pharmacotherapy failure. (d) Therapeutic Recommendations: Avoid tricyclic use due to potential lack of effi-cacy. Consider alternative drug not metabolized by CYP2D6. If a tricyclic is war-ranted, consider increasing the starting dose.a Utilize therapeutic drug monitoring to guide dose adjustments. (e) Classification of recommendation for amitriptyline/nortriptyline therapy: Strong 2. Likely phenotype: Extensive metabolizer ( 77-92% of patients) (a) Genotypes: An individual carrying two alleles encoding full or reduced function or one full function allele together with either one nonfunctional or one reduced-function allele. (b) Examples of diplotypes: *1/*1, *1/*2, *2/*2, *1/*41, *1/*4, *2/*5, *10/*10 (c) Implications for TCA metabolism: Normal metabolism of tricyclics.
  • 41. CHAPTER 5. PHARMACOGENETICS OF CYP2D6 40 (d) Therapeutic Recommendations: Initiate therapy with recommended starting dose. (e) Classification of recommendation for amitriptyline/nortriptyline therapy: Strong 3. Likely phenotype: Intermediate metabolizer ( 2-11% of patients) (a) Genotypes: An individual carrying one reduced and one nonfunctional allele, (b) Examples of diplotypes: *4/*10, *5/*41 (c) Implications for TCA metabolism: Reduced metabolism of tricyclics to less ac-tive compounds when compared to extensive metabolizers. Higher plasma concen-trations will increase the probability of side effects. (d) Therapeutic Recommendations: Consider 25% reduction of recommended start-ing dose. Utilize therapeutic drug monitoring to guide dose adjustments. (e) Classification of recommendation for amitriptyline/nortriptyline therapy: Mod-erate 4. Likely phenotype: Poor metabolizer ( 5-10% of patients) (a) Genotypes: An individual carrying no functional alleles. (b) Examples of diplotypes: *4/*4, *4/*5, *5/*5, *4/*6 (c) Implications for TCA metabolism: Greatly reduced metabolism of tricyclics to less active compounds when compared to extensive metabolizers. Higher plasma concentrations will increase the probability of side effects. (d) Therapeutic Recommendations: Avoid tricyclic use due to potential for side ef-fects. Consider alternative drug not metabolized by CYP2D6. If a tricyclic is war-ranted, consider 50% reduction of recommended starting dose. Utilize therapeutic drug monitoring to guide dose adjustments. (e) Classification of recommendation for amitriptyline/nortriptyline therapy: Strong 5.2.3 Recommended dosing of codeine by CYP2D6 Phenotype 1. Likely phenotype: Ultrarapid metabolizer ( 1-2% of patients) (a) Genotypes: An individual carrying more than two copies of functional alleles. (b) Examples of diplotypes: *1/*1xN, *1/*2xN (c) Implications for codeine metabolism: Increased formation of morphine following codeine administration, leading to higher risk of toxicity. (d) Therapeutic Recommendations: Avoid codeine use due to potential for toxicity. (e) Classification of recommendation for codeine therapy: Strong (f) Considerations for alternative opioids: Alternatives that are not affected by this CYP2D6 phenotype include morphine and non-opioid analgesics. Tramadol, and to a lesser extent hydrocodone and oxycodone, are not good alternatives because their metabolism is affected by CYP2D6 activity. 2. Likely phenotype: Extensive metabolizer ( 77-92% of patients)
  • 42. CHAPTER 5. PHARMACOGENETICS OF CYP2D6 41 (a) Genotypes: An individual carrying two alleles encoding full or reduced function or one full function allele together with either one nonfunctional or one reduced-function allele. (b) Examples of diplotypes: *1/*1, *1/*2, *2/*2, *1/*41, *1/*4, *2/*5, *10/*10 (c) Implications for codeine metabolism: Normal morphine formation (d) Therapeutic Recommendations: Use label recommended age- or weight-specific dosing. (e) Classification of recommendation for codeine therapy: Strong (f) Considerations for alternative opioids: Not required. 3. Likely phenotype: Intermediate metabolizer ( 2-11% of patients) (a) Genotypes: An individual carrying one reduced and one nonfunctional allele. (b) Examples of diplotypes: *4/*10, *5/*41 (c) Implications for codeine metabolism: Reduced morphine formation. (d) Therapeutic Recommendations: Use label recommended age- or weight-specific dosing. If no response, consider alternative analgesics such as morphine or a non-opioid. (e) Classification of recommendation for codeine therapy: Moderate (f) Considerations for alternative opioids: Monitor tramadol use for response. 4. Likely phenotype: Poor metabolizer ( 5-10% of patients) (a) Genotypes: An individual carrying no functional alleles. (b) Examples of diplotypes: *4/*4, *4/*5, *5/*5, *4/*6 (c) Implications for codeine metabolism: Greatly reduced morphine formation fol-lowing codeine administration, leading to insufficient pain relief. (d) Therapeutic Recommendations: Avoid codeine use due to lack of efficacy. (e) Classification of recommendation for codeine therapy: Strong. (f) Considerations for alternative opioids: Alternatives that are not affected by this CYP2D6 phenotype include morphine and non-opioid analgesics. Tramadol, and to a lesser extent hydrocodone and oxycodone, are not good alternatives because their metabolism is affected by CYP2D6 activity; these agents should be avoided. 5.3 CYP2D6 and Tamoxifen: Why endoxifen is a better op-tion than Tamoxifen The selective estrogen receptor modulator (SERM) tamoxifen has been used for many years as endocrine treatment for hormone receptorpositive breast cancer, with indications in the metastatic, adjuvant, and preventative settings. The minimal acute toxicity of endocrine treatments such as tamoxifen compared with chemotherapy helps maintain patients quality of life, and increasing emphasis is being placed on these treatments to delay the use of cytotoxic therapies for as long as possible.
  • 43. CHAPTER 5. PHARMACOGENETICS OF CYP2D6 42 Tamoxifens adverse-event profile includes venous thrombosis, endometrial cancer, and, most commonly, hot flashes, which are often treated with an SSRI antidepressant. Tamoxifen is extensively hepatically metabolized to several primary and secondary metabolites by multi-ple CYP enzymes, including CYP2D6. With some SSRIs (e.g., paroxetine) known to inhibit CYP2D6, a study was recently conducted to investigate the effects of SSRI coadministration on plasma concentrations of tamoxifen and its metabolites in addition to the effects of individual patient genotypes. A strong association was shown between CYP2D6 activity and plasma levels of the active tamoxifen metabolite, endoxifen. Concomitant use of paroxetine was associated with lower endoxifen plasma concentrations, with the magnitude of this difference being de-pendent on CYP2D6 genotype. It is possible, therefore, that patients taking some SSRIs may be less responsive to tamoxifen because of a lower rate of formation of its active metabolite, which may be exacerbated in individuals with slowed CYP2D6 metabolism. The possibility that tamoxifens antitumoral activity could be affected in this way needs further testing in clini-cal trials designed to record clinical outcome in conjunction with genotype and coadministered medication with the potential to affect CYP2D6. 5.4 Why Endoxifen? Endoxifen is an active metabolite of Tamoxifen, a widely used breast cancer drug. Treat-ment of breast cancer directly with Endoxifen may eliminate Tamoxifen metabolism associ-ated variability and avoid a potential serious drug-drug interaction. Upon oral administration, tamoxifen (TAM) is extensively metabolized by cytochrome P450(CYP) enzymes into active metabolites including 4-hydroxy tamoxifen and endoxifen (4-hydroxy N-desmethyl Tamox-ifen) Endoxifen is one hundred times more potent than TAM and is generated via CYP3A4/5- mediated N-demethylation and CYP2D6-mediated hydroxylation. Due to genetic polymor-phism of CYP2D6, there are large variations among patients in both the therapeutic efficacy and side effects. Furthermore, CYP2D6 interacts with specific selective serotonin reuptake in-hibitors (SSRIs), frequently used to prevent the hot flushes experienced by up to 45% of patients taking TAM. As a consequence, significant numbers of women might not receive optimal bene-fit from TAM treatment. Our pre-clinical studies have validated the concept of using Endoxifen for the treatment of breast cancer (Ahmad et al. 2010). Treatment of breast cancer directly with Endoxifen may eliminate TAM metabolism associated variability and avoid a potential serious drug-drug interaction. 5.4.1 Why Endoxifen Treatment may have better therapeutic outcome Endoxifen may overcome the unpredictable therapeutic outcome due to pharmacoge-nomic variation of Tamoxifen and Aromatase inhibitors Endoxifen would help women taking anti-depressants who have problems metabolizing Tamoxifen Endoxifen acts directly at the target Tamoxifen have been shown to be more effective than Aromatase inhibitors in patients with wild type CYP2D6 Showed enhanced therapeutic efficacy against established human tumor model in mice
  • 44. CHAPTER 5. PHARMACOGENETICS OF CYP2D6 43 Figure 5.2: Tamoxifen metabolism Only potential drug for ER positive premenopausal women with CYP2D6 deficiency 5.5 References 1. Wang B, Yang LP, Zhang XZ, Huang SQ, Bartlam M, Zhou SF (2009). ”New insights into the structural characteristics and functional relevance of the human cytochrome P450 2D6 enzyme”. Drug Metab. Rev. 41 (4): 573643. doi:10.1080/03602530903118729. PMID 19645588. 2. Teh LK, Bertilsson L (2012). ”Pharmacogenomics of CYP2D6: molecular genetics, in-terethnic differences and clinical importance”. Drug Metab. Pharmacokinet. 27 (1):
  • 45. CHAPTER 5. PHARMACOGENETICS OF CYP2D6 44 Figure 5.3: Tamoxifen: Interaction with other drugs 5567. doi:10.2133/dmpk.DMPK-11-RV-121. PMID 22185816. 3. Walko CM, McLeod H (April 2012). ”Use of CYP2D6 genotyping in practice: tamox-ifen dose adjustment”. Pharmacogenomics 13 (6): 6917. doi:10.2217/pgs.12.27. PMID 22515611. 4. ”Entrez Gene: CYP2D6 cytochrome P450, family 2, subfamily D, polypeptide 6”. 5. Ostille DO., Warren AM., Kulkarni, J. The role of pharmacogenomic testing in psychia-try. Aust New Zealand J Psychiatry 2014 Jan 10. PMID 24413808 6. Bertilsson L, Dahl ML, Daln P, Al-Shurbaji A (February 2002). ”Molecular genetics of CYP2D6: clinical relevance with focus on psychotropic drugs”. Br J Clin Pharmacol 53 (2): 11122. doi:10.1046/j.0306-5251.2001.01548.x. PMC 1874287. PMID 11851634.
  • 46. CHAPTER 5. PHARMACOGENETICS OF CYP2D6 45 7. Llerena A, Dorado P, Peas-Lled EM (January 2009). ”Pharmacogenetics of debrisoquine and its use as a marker for CYP2D6 hydroxylation capacity”. Pharmacogenomics 10 (1): 1728. doi:10.2217/14622416.10.1.17. PMID 19102711. 8. Lynch T, Price A (August 2007). ”The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects”. Am Fam Physician 76 (3): 3916. PMID 17708140. 9. Droll K, Bruce-Mensah K, Otton SV, Gaedigk A, Sellers EM, Tyndale RF (1998). ”Com-parison of three CYP2D6 probe substrates and genotype in Ghanaians, Chinese and Cau-casians”. Pharmacogenetics 8 (4): 32533. doi:10.1097/00008571-199808000-00006. PMID 9731719. 10. Gaedigk A, Bradford LD, Marcucci KA, Leeder JS (2002). ”Unique CYP2D6 activity distribution and genotype-phenotype discordance in black Americans”. Clin. Pharmacol. Ther. 72 (1): 7689. doi:10.1067/mcp.2002.125783. PMID 12152006. 11. McLellan RA, Oscarson M, Seidegrd J, Evans DA, Ingelman-Sundberg M (June 1997). ”Frequent occurrence of CYP2D6 gene duplication in Saudi Arabians”. Pharmacogenet-ics 7 (3): 18791. doi:10.1097/00008571-199706000-00003. PMID 9241658. 12. PHARMACOGENETICS AND PHARMACOGENOMICS. J. Steven Leeder PharmD, PhD Pediatric Clinics of North America - Volume 48, Issue 3 (June 2001). doi:10.1016/S0031- 3955%2805%2970338-2. 13. Hoskins JM, Carey LA, McLeod HL (August 2009). ”CYP2D6 and tamoxifen: DNA matters in breast cancer”. Nat. Rev. Cancer 9 (8): 57686. doi:10.1038/nrc2683. PMID 19629072. 14. Foster BC, Sockovie ER, Vandenhoek S, Bellefeuille N, Drouin CE, Krantis A, Budzinski JW, Livesey J, and Arnason JT (2004). ”In Vitro Activity of St. John’s Wort Against Cy-tochrome P450 Isozymes and P-Glycoprotein”. Pharmaceutical Biology 42 (2): 159169. doi:10.1080/13880200490512034.
  • 47. Chapter 6 Pharmacogenetics of cancer Advances in molecular biology and genetics over the past 60 years have facilitated develop-ment of multiple chemotherapeutic agents that are active against most common malignancies. However, significant heterogeneity in the efficacy and toxicity of these agents is consistently ob-served across human populations. Ad-ministration of the same dose of a given anticancer drug to a population of patients can result in any-where from minimal toxicity to severe, potentially life-threatening events. Many clinical factors are associated with drug toxicity, but genetic dif-ferences, or polymorphisms, in the proteins affecting drug disposition also have a great impact. In addition, the enzymes that metabolize the majority of chemotherapy agents contain genetic polymorphisms, but prospective identification of patients likely to be harmed by chemotherapy currently is not possible for most therapeutic regimens. 6.1 Interindividual variation in Chemotherapeutic drug re-sponse Interindividual differences in tumor response and normal tissue toxicities are consistently ob-served with most chemotherapeutic agents or regimens. While many clinical variables have been associated with drug responses (e.g., age, gender, diet, drug-drug interactions), inherited variations in drug disposition (metabolism and transport) genes and drug target genes also likely con-tribute to the observed variability in cancer treatment outcome. Pharmacogenomic studies aim to elucidate the genetic bases for interindividual differences and to use such genetic information to predict the safety, toxicity, and/or efficacy of drugs. There exist several clinically relevant examples of the utility of pharmacogenomics that associate spe-cific genetic polymorphisms in drug metabolizing enzymes (e.g., TPMT, UGT1A1, DPD), drug transporters (MDR1), and drug target enzymes (TS) with clinical outcomes in patients treated with commonly prescribed chemotherapy drugs, such as 5-fluorouracil and irinotecan. Tech-niques to discover and evaluate the functional significance of these polymorphisms have evolved in recent years and may soon be applied to clinical practice and clinical trials of currently pre-scribed anticancer drugs as well as new therapeutic agents. Clearly, a better understanding of the genetic determinants of chemotherapeutic response will enable prospective identification of patients at risk for severe toxicity or those most likely to benefit from a particular treatment regimen. Such studies can be translated to clinical practice via molecular diagnostics (genotyping) in order to guide selection of the optimal drug combina-tion and dosage for the individual patient. A number of detailed reviews on cancer pharmacoge-nomics have been published recently. This article focuses on the current and future applications 46
  • 48. CHAPTER 6. PHARMACOGENETICS OF CANCER 47 of pharmacogenomics in clinical cancer therapy and cancer drug development. 6.2 Genetic variations affecting drug response and toxicity with cancer chemotherapy Table 6.1: Examples of genetic variations in major Phase-I and Phase-II enzymes asso-ciated with changes in patient responses to chemotherapy agents. Abbreviations: CYP = cytochrome P450; DPD = dihydropyrimidine dehydrogenase; GST = glutathione S-transferase; MDR1 = multidrug resistance 1; MRP = multidrug-resistancerelated protein; MTHFR = 5,10- methylenetetrahydrofolate reductase; NAT = N-acetyl transferase; TPMT = thiopurine methyl-transferase; TS = thymidylate synthase; UGT1A1 = UDP-glucuronosyltransferase 1A1; 5-FU = 5-fluorouracil; 6-MP = 6-mercaptopurine. Pharmacogenomic approaches have been applied to many existing chemotherapy agents in an effort to identify relevant inherited variations that may better predict patient response to chemotherapy. Genetic variations include nucleotide repeats, insertions, deletions, and single nucleotide polymorphisms (SNPs), which can alter the amino acid sequence of the encoded pro-teins, RNA splicing, and gene transcription. Such genetic polymorphisms in drug-metabolizing
  • 49. CHAPTER 6. PHARMACOGENETICS OF CANCER 48 enzymes, transporters, and molecular targets have been actively explored with regard to func-tional changes in phenotype (altered expression levels and/or activity of the encoded proteins) and their contribution to variable drug response. Recent studies also indicate that genetic varia-tions vary substantially among different ethnic groups and that the evaluation of the haplotypes (combination of polymorphisms that are inherited together) can often result in better correla-tion with phenotypes than with individual polymorphisms. The following sections describe some clinically relevant exam-ples of genetic polymorphisms to illustrate the relevance of can-cer pharmacogenomics in optimizing chemotherapy as a way to enhance efficacy and safety [Figure 6.1]. 6.3 Polymorphism in Drug-Metabolizing Enzymes 6.3.1 Thiopurine methyltransferase and 6-Mercaptopurine 6-Mercaptopurine (6-MP) is a purine antimetabolite used in the treatment of leukemia. The antitumor activity of 6-MP is via the inhibition of the formation of nucleotides necessary for DNA and RNA synthesis. Thiopurine methyltransferase (TPMT) catalyzes the S-methylation of 6-MP to form inactive metabolites. Genetic variations in the TPMT gene have profound effects on the bioavailability and toxicity of 6-MP. It has been demonstrated that about 1 in 300 individuals inherit TPMT deficiency as an autosomal recessive trait. Patients who carry TPMT polymorphisms are at risk for severe hematologic toxicities when treated with 6-MP because these polymorphisms lead to a decrease in the rate of 6-MP metabolism. Figure 6.1: Metabolism of 6-Mercaptopurine The molecular basis for polymorphic TPMT activity has been well defined. Three particular TPMTalleles, designated as TPMT*2, TMPT*3A, and TPMT*3C, have been shown to account for nearly 95% of the observed cases of TPMT defi-ciency. Each of these mutant alleles encodes TPMT pro-teins that undergo rapid degradation, leading to enzyme deficiency. The types and frequencies of TPMTalleles have been reported to differ among ethnic groups. A recent trial estimated that 71% of patients with bone marrow intolerance to 6-MP were phenotypically TPMT deficient and that these patients were more likely to be hospitalized, receive platelet transfusions, and miss scheduled doses of chemotherapy.
  • 50. CHAPTER 6. PHARMACOGENETICS OF CANCER 49 Figure 6.2: 6-Mercaptopurine dosing according to TPMT genotype Appropriate 6-MP dose reductions for TPMT-deficient patients have allowed for similar tox-icity and survival outcomes as patients with normal TPMT levels. Genotyping methods have been established for the molecular diagnosis of TPMT deficiency and can assist with determin-ing a safe starting dose for 6-MP therapy. 6.3.2 UDP-glucuronosyltransferase 1A1 and Irinotecan [Irinotecan metabolism and Genotype based toxicities are discussed in Chapter 1: Figure 1.3 and Figure 1.4] Variations in UGT1A1 activity most commonly arise from polymorphisms in the UGT1A1 promoter region that contains several repeating TA elements. The presence of seven TA re-peats (referred to as UGT1A1*28), instead of the wild-type number of six, results in reduced UGT1A1 expression and activity. Accordingly, UGT1A1*28has been shown to be associated with reduced glucuronidation of SN-38, increased exposure to SN-38, and increased clini-cal toxicity for patients treated with irinotecan. The frequencies of UGT1A1*28alleles vary significantly among different ethnic groups: UGT1A1*28alleles are present in approximately 35% of Caucasians and African Americans while their frequency is much lower in Asians.. A recent prospective study demonstrated, with sufficient statistical power, that patients with a UGT1A1*28 allele are at higher risk of grade 4 neutropenia. While that study clearly illustrates the importance of UGT1A1 pharmacogenetics as a molecular predictor of irinotecan toxicity, clinical guidelines have yet to be developed with regard to dosage adjustment or selection of
  • 51. CHAPTER 6. PHARMACOGENETICS OF CANCER 50 alternative non irinotecan-containing regimens. 6.3.3 Dihydropyrimidine dehydrogenase and 5-FU Based on its activity against a variety of tumors and synergistic interactions with other chemother-apy agents, 5-fluorouracil (5-FU) and its derivatives remain some of the most commonly pre-scribed chemotherapy agents. Approximately 5% of administered 5-FU undergoes anabolism into cytotoxic nucleotides responsible for its antitumor activity, whereas the other 80%-95% under-goes catabolism into biologically inactive metabolites that are excreted in the urine and bile. Dihydropyrimidine dehydrogenase (DPD) catalyzes the rate-limiting step in 5-FU catabolism; therefore, variability in this enzyme activity is one of the major factors that influences systemic exposure to fluorodeoxyuridine monophosphate (FdUMP) and the incidence of adverse effects to 5-FU. DPD activity is completely or partially deficient in 0.1% and 3%5% of individuals in the general population, respectively, and DPD deficiency has been associated with severe toxicity and fatal outcomes after 5-FU treatment. Figure 6.3: 5-FU metabolism and Rate limiting role of DPYD enzyme DPD deficiency appears to be a genetic disorder arising from multiple polymorphisms in the DPYDgene resulting in decreased enzyme activity. Analyses of the preva-lence of the various mutations in the DPYD allele have shown that a guanidine to adenine point mutation in the invariant splice donor site (DPYD*2A) is by far the most common . 6.3.4 MTHFR and Methotrexate Methotrexate inhibits dihydrofolate reductase (DHFR), which in turn causes accumulation of dihydrofolate and depletion of cellular folates needed for DNA synthesis. Methotrexate is asso-ciated with significant side effects and carries multiple black box warnings regarding potential toxicities, including acute renal failure and bone marrow suppression. Side effects can range
  • 52. CHAPTER 6. PHARMACOGENETICS OF CANCER 51 from mild and self limiting to more severe and sometimes fatal. There is significant interindi-vidual variation in response to methotrexate therapy. As many as one third of patients fail to respond to treatment either due to lack of efficacy or adverse events. The cytotoxic effect of methotrexate is dependent on inhibition of folate metabolism, so the folate pathway has been examined for enzyme variants that could be associated with efficacy and/or toxicity. One of these enzymes, 5,10-methylenetetrahydrofolate reductase (MTHFR), plays a key role, modulating DNA synthesis and intracellular folate pools. Two tightly linked, common variants in the MTHFR gene, C677T and A1298C, have been shown to cause reduced levels of MTHFR enzyme activity and corresponding reductions in available folate. Therefore, these variants may act as a useful marker for predicting individual differences in efficacy or toxicity of methotrexate. Table 6.2: Frequency of MTHFR allelic variation among different ethnic groups To date, numerous studies have examined the effect of the C677T and A1298C variants on toxicity and efficacy of methotrexate across disease states, methods of dosing, and ethnicities-and have shown conflicting results. 1. Two recent meta-analyses of rheumatoid arthritis patients (RA) concluded that there is no significant relationship between MTHFR variant alleles and toxicity or efficacy of methotrexate. The authors acknowledge that the differences in their analyses compared to the previous meta-analysis cited could be due to the data sets used, heterogeneity of patients, and differences in methods of analysis. 2. In recent studies of psoriatic arthritis and juvenile ALL, homozygosity of the C677 variant has been associated with toxicity. 3. A small study of juvenile ALL patients showed that patients with normal MTHFR alleles tolerated a higher dosage of methotrexate with lower risk of toxicity than patient with variants treated with a standard dosage, suggesting that the MTHFR genotype can help guide dosage in this population.