technologies for detecting genetic polymorphisms in pharmacogenomics

9
Molecular Diagnosis Vol. 4 No. 4 1999 Technologies for Detecting Genetic Polymorphisms in Pharmacogenomics MICHAEL M. SHI, MD, PhD, MICHAEL R. BLEAVINS, PhD, FELIX A. de la IGLESIA, MD Ann Arbor, Michigan Background: Pharmacogenomics is an emerging scientific discipline examining the genetic basis for individual variations in response to therapeutics. Methods and Results: Genetic polymorphisms are a major cause of individual dif- ferences in drug response. Metabolic phenotyping can be accomplished by admin- istrating a probe drug or substrate and measuring the metabolites and clinical outcomes. However, this approach tends to be labor intensive and requires repeated sample collection from the individual being tested. Alternatively, geno- typing allows determination of individual DNA sequence differences for a partic- ular trait. Commonly used genotyping methods include gel electrophoresis-based techniques, such as polymerase chain reaction (PCR) coupled with restriction fragment length polymorphism analysis, multiplex PCR, and allele-specific ampli- fication. Fluorescent dye-based high-throughput genotyping procedures are increas- ing in popularity, including oligonucleotide ligation assay, direct heterozygote sequencing, and TaqMan (Perkin Elmer, Foster City, CA) allelic discrimination. High-density chip array and mass spectrometry technologies are the newest advances in the genotyping field, but their wide application is yet to be developed. Novel mutations/polymorphisms also can be identified by conformation-based mutation screening and direct high-throughput heterozygote sequencing. Conclusions: Rapid and accurate detection of genetic polymorphisms has great potential for application to drug development, animal toxicity studies, improve- ment of human clinical trials, and postmarket monitoring surveillance for drug efficacy and toxicity. Key words: pharmacogenomics, genotyping, phenotyping, polymorphisms. Pharmacogenomics is an emerging scientific disci- pline that examines the genetic basis for individual vari- ations in response to therapeutic agents or biotechno- From the Genomic Pathology Laboratory, Pathology and Experimental Toxicology, Parke-Davis Pharmaceutical Research, Warner-Lambert Company; and the Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan. Reprint requests: Michael M. Shi, MD, PhD, Pathology and Experimental Toxicology, Parke-Davis Pharmaceutical Re- search, Warner-Lambert Company, 2800 Plymouth Road, Ann Arbor, MI 48105. Email: [email protected] Copyright © 1999 by Churchill Livingstone ® 1084-8592/99/0404-0011 $10.00/0 logical interventions [1].Variations can result from genetic or environmental factors, such as age, sex, body size, diet, alcohol or tobacco consumption, pregnancy, kidney or liver dysfunction, concurrent disease states, and drug interactions [2]. Interindividual variations also result from genetically determined differences in absorption, distribution, metabolism, and excretion of therapeutic agents. These genetic factors can alter the effects of a given dose, leading to a spectrum of responses ranging from clinical benefits to adverse effects and therapeutic failures. Recent research shows that certain genetic polymorphisms cause significantly different pharmaco- logical and toxicological effects among individuals on exposure to a particular drug [3]. Most genetic varia- 343

Upload: umich

Post on 29-Nov-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

Molecular Diagnosis Vol. 4 No. 4 1999

Technologies for Detecting Genetic Polymorphisms in Pharmacogenomics

M I C H A E L M. SHI, MD, PhD, M I C H A E L R. B L E A V I N S , PhD,

F E L I X A. de la I G L E S I A , M D

A n n Arbor, Michigan

Background: Pharmacogenomics is an emerging scientific discipline examining the genetic basis for individual variations in response to therapeutics. Methods and Results: Genetic polymorphisms are a major cause of individual dif- ferences in drug response. Metabolic phenotyping can be accomplished by admin- istrating a probe drug or substrate and measuring the metabolites and clinical outcomes. However, this approach tends to be labor intensive and requires repeated sample collection from the individual being tested. Alternatively, geno- typing allows determination of individual DNA sequence differences for a partic- ular trait. Commonly used genotyping methods include gel electrophoresis-based techniques, such as polymerase chain reaction (PCR) coupled with restriction fragment length polymorphism analysis, multiplex PCR, and allele-specific ampli- fication. Fluorescent dye-based high-throughput genotyping procedures are increas- ing in popularity, including oligonucleotide ligation assay, direct heterozygote sequencing, and TaqMan (Perkin Elmer, Foster City, CA) allelic discrimination. High-density chip array and mass spectrometry technologies are the newest advances in the genotyping field, but their wide application is yet to be developed. Novel mutations/polymorphisms also can be identified by conformation-based mutation screening and direct high-throughput heterozygote sequencing. Conclusions: Rapid and accurate detection of genetic polymorphisms has great potential for application to drug development, animal toxicity studies, improve- ment of human clinical trials, and postmarket monitoring surveillance for drug efficacy and toxicity. Key words: pharmacogenomics, genotyping, phenotyping, polymorphisms.

Pharmacogenomics is an emerging scientific disci- pline that examines the genetic basis for individual vari- ations in response to therapeutic agents or biotechno-

From the Genomic Pathology Laboratory, Pathology and Experimental Toxicology, Parke-Davis Pharmaceutical Research, Warner-Lambert Company; and the Department of Pathology, The University of Michigan Medical School, Ann Arbor, Michigan.

Reprint requests: Michael M. Shi, MD, PhD, Pathology and Experimental Toxicology, Parke-Davis Pharmaceutical Re- search, Warner-Lambert Company, 2800 Plymouth Road, Ann Arbor, MI 48105. Email: [email protected]

Copyright © 1999 by Churchill Livingstone ® 1084-8592/99/0404-0011 $10.00/0

logical interventions [1].Variations can result from genetic or environmental factors, such as age, sex, body size, diet, alcohol or tobacco consumption, pregnancy, kidney or liver dysfunction, concurrent disease states, and drug interactions [2]. Interindividual variations also result from genetically determined differences in absorption, distribution, metabolism, and excretion of therapeutic agents. These genetic factors can alter the effects of a given dose, leading to a spectrum of responses ranging from clinical benefits to adverse effects and therapeutic failures. Recent research shows that certain genetic polymorphisms cause significantly different pharmaco- logical and toxicological effects among individuals on exposure to a particular drug [3]. Most genetic varia-

343

344 Molecular Diagnosis Vol. 4 No. 4 December 1999

tions do not affect protein function and therefore have no phenotypic expression. However, other polymor- phic genes encode proteins with dysfunctional or non- functional activities in comparison with normal func- tion (wild-type). Thus, determination of these genetic polymorphisms may be of significant value in predict- ing drug efficacy or toxicity in a variety of therapeutic scenarios.

Genetic polymorphisms are usually determined by phenotyping or genotyping. Phenotypes constitute ob- servable characteristics of a cell or organism, usually monitored by direct observations or biochemical or functional analyses. In pharmacogenomics, phenotypes can be monitored at additional levels, such as low or ex- aggerated pharmacological effects, frequency of side ef- fects, and metabolic rate for certain drugs. In contrast, genotyping determines individual DNA sequence dif- ferences for particular traits independently of their func- tional effects. This approach is increasingly being used in biomedical research and clinical molecular diagnostics to replace traditional serological testing to some extent.

Establishing the genotypic and phenotypic correla- tion through clinical pharmacogenomics will allow strat- ification of patient subpopulations according to differ- ent drug responses. These data offer the potential for wider and safer drug use based on individual genetic makeup. When applied to drug discovery and develop- ment, pharmacogenomics also may help the deve- lopment of therapeutic interventions targeting the spe- cific responder patient groups. Advanced technologies to identify genetic polymorphisms rapidly, accurately, and economically are becoming a priority in the imple- mentation of pharmacogenomics to drug development, clinical trials, and clinical monitoring for drug efficacy and toxicity.

Phenotyping in Pharmacogenomics

Genetic variations contribute to interpersonal differ- ences in drug response at both the pharmacokinetic and pharmacodynamic levels. Over the past 2 decades, many genetic polymorphisms in drug-metabolizing enzymes have been described [3]. When these genetic variations significantly affect the enzyme's function, different clin- ical outcomes can occur among people exposed to the same drug. Phenotyping for these drug-metabolizing en- zymes can be divided into functional phenotyping or metabolic phenotyping. Functional phenotyping is the direct measurement of the properties of a biomarker specific to a system, organ, or tissue, such as enzyme ac- tivities and drug-receptor interactions. Although di- rectly reflecting a protein's properties, functional pheno- typing is usually hampered by tissue availability because

of the necessary invasive procedures for tissue sampling, such as liver biopsy.

In some cases, it may be possible to use surrogate tis- sues or peripheral blood for functional analysis. Sulfa- tion and glucuronidation are two major pathways for the metabolism of many drugs and are catalyzed by the liver phase II enzymes, sulfotransferases (SULTs), and uri- dine diphosphate glucuronosyltransferases (UGTs), re- spectively. Significant interindividual variations have been reported for both enzymes [4]. Deficiencies in UGT activity are responsible for the bilirubin level elevations seen in Crigler-Najjar type I and II [5] and Gilbert syn- dromes, which affect approximately 2% to 9% of the population [6]. Gilbert syndrome is a benign form of unconj ugated hyperbilirubinemia in the absence of liver disease or overt hemolysis. Patients with Gilbert syn- drome have inherited abnormalities of UGT1A1 activ- ity and, as a result, a defect in bilirubin glucuronide for- mation. This genetic impairment results in enhanced drug toxicities. For example, patients with Gilbert syn- drome are predisposed to severe diarrhea and hemato- logic toxicities caused by the chemotherapeutic agent irinotecan because of defective UGTIA1 activity [7]. Irino- tecan is metabolized to its active metabolite, SN38, conju- gated by UGT1A1 to form SN38-glucuronide, and then se- creted into the bile. Gilbert syndrome often is undiagnosed because the enzymatic assays usually require liver biopsy specimens to measure the enzyme activity. Recent reports have shown that the enzyme activities of both SULT1A1 and UGT1A1 can be analyzed using circulating platelets [8,9]. Platelets are a readily accessible source of both en- zymes, and the enzymatic activities of platelet UGTs have been reported to correlate with the activity found in the liver [9,10]. Functional phenotyping also has some limita- tion when enzymatic activities overlap substrate specificity. Although UGTIA1 is the primary isoform responsible for the bilirubin conjugation, other isoforms, such as UGT1A4, also are capable of catalyzing this reaction [11]. It therefore is difficult to specifically identify the protein subtype responsible for a particular phenotype or pathway using only functional phenotyping.

In general, a significant challenge resides in the iden- tification of surrogate markers for specific phenotyping, such as drug-metabolizing enzymes. More commonly, phenotyping is accomplished by administrating a probe drug and measuring metabolites in plasma or urine (metabolic phenotyping [12,13]). To date, cytochrome P-4502D6 (CYP2D6) is an extensively characterized drug-metabolizing enzyme. This microsomal isozyme is responsible for the oxidative metabolism of approxi- mately 50 clinically relevant drugs from multiple thera- peutic classes. These substrates include antiarrhythmics, tricyclic antidepressants, beta-adrenergic blockers, neu- roleptics, and other classes of drugs frequently having narrow therapeutic windows. For these drugs, slightly

less than targeted plasma concentrations cannot have the desired therapeutic effect, whereas slightly greater con- centrations can cause toxicity. CYP2D6 has three clini- cally distinct phenotypes: the normal (or extensive) me- tabolizer, slow (or poor) metabolizer, and fast (or ultraextensive) metabolizer. The same dose of a drug metabolized through CYP2D6 will result in plasma con- centrations that vary greatly among individuals with these distinct phenotypes. In normal metabolizers, steady-state plasma concentrations are within the ex- pected therapeutic range, and toxic effects are minimal or nonexistent. In fast metabolizers, steady-state drug levels are less than the therapeutic concentration, and ef- ficacy is subtherapeutic. Poor metabolizers have greater- than-expected drug levels and are therefore susceptible to undesired toxicity. Debrisoquine hydroxylation is commonly used for CYP2D6 metabolic phenotyping [14]. For this assay, a dose of debrisoquine is adminis- tered to the subject, and the ratio between the parent drug and its metabolites is monitored in urine. The met- abolic ratio is expressed as debrisoquine divided by 4- hydroxydebrisoquine, the major metabolite of the parent drug. If the metabolic ratio is greater than the normal range of 0.5 to 12.6, the individual is categorized as a slow or poor metabolizer. A metabolic ratio less than the ref- erence range indicates an ultraextensive metabolizer. Si- milar approaches are used for metabolic phenotyping of other cytochrome P-450 isoforms. The methods used for CYP1A2, 2C19, 2D6, 2El, and 3A4 are listed in Table 1.

Phenotyping methods were used long before molecu- lar biological tools became available and therefore have been fully validated. However, these metabolic pheno- typing procedures involve sophisticated analytic tech- niques and require significant time for obtaining results. Metabolic results also can be complicated by sample sta- bility, as well as the external factors of age, nutrition, dis- ease status, and concurrent medications. In addition, dif- ferent substrates of a polymorphic enzyme typically have different specificities, and the phenotype may not accu- rately represent the disposition of a particular drug sub- strate. Also, poor metabolizers may experience unpleas- ant side effects from the probe drugs, thus further limiting the clinical application of metabolic phenotyping.

Technologies for Pharmacogenomics • Shi et al. 345

Genotyping in Pharmacogenomics

A variety of genotyping techniques have been de- veloped for identified polymorphisms. Commonly used methods include gel electrophoresis-based tech- niques, such as polymerase chain reaction (PCR) cou- pled with restriction fragment length polymorphism (RFLP) analysis, multiplex PCR, and allele-specific amplification. Fluorescent dye-based high-throughput genotyping procedures have gained increased popularity, including the oligonucleotide ligation assay (OLA), het- erozygote sequencing, TaqMan (Perkin Elmer, Foster City, CA) allelic discrimination, and high-density chip array technology. Novel polymorphisms also can be iden- tified by single-strand conformation polymorphism (SSCP) and direct heterozygote sequencing. Mass spec- trometry (MS) genotyping similarly has been used for both known and unknown polymorphisms.

One significant advantage of genotyping over pheno- typing is the less invasive nature of DNA sampling. Buc- cal swabs, mouthwashes, hair roots, and blood spot collec- tions make DNA sampling easier and more acceptable to patients. In contrast to phenotyping, genotyping results are not modulated by drug-drug or drug-food interac- tions and reliably identify heterozygotes with intermedi- ate phenotypes. This approach is not without limitations because many functionally important allelic variants are not yet identified or linked to phenotypic function. Never- theless, with increasing sequence information becoming available, more and more genetic polymorphisms are being characterized by genotype/phenotype association studies. Simple, reliable, high-throughput, and cost- effective DNA technologies are highly desirable for large-scale genotyping projects and association studies in pharmacogenomics.

Gel-based Genotyping. Gel-based genotyping meth- ods include PCR-RFLP, allele-specific PCR, and multi- plex PCR. PCR amplifies specific regions of DNA se- quences between two custom-designed oligonucleotide primers, allowing small quantities of DNA containing the polymorphic site to be increased dramatically. These detectable levels of specific DNA then are di-

Table 1. Metabolic Phenotyping of Drug-Metabolizing Enzymes

Enzyme Probe Drug React ion References

CYPIA2 Caffeine 3N-Demethylation 15 CYP2D6 Debrisoquine Hydroxylation 14

Dextromethorphan N-Demethylation 16 CYP2E1 Chlorzoxazone 6-Hydroxylation 17 CYP3A4 Erythromycin 3N-Demethylation 18

Nifedipine Oxidation 19

346 Molecular Diagnosis Vol. 4 No. 4 December 1999

gested with appropriate restriction enzymes, with vis- ualization by gel staining after electrophoresis. The re- striction digestion pat tern differs if the genetic polymorphism results in a gain or loss of the restric- tion site. Many restriction enzymes exist that cleave at a wide variety of DNA sequences. A PCR-RFLP method to detect the CYP2D6*4 allele (B mutation) is de- scribed in Fig. 1. A guanosine (G) to adenosine (A) transition at the junction of the third intron and fourth exon (nucleotide 1934) causes a messenger RNA splic- ing site mutation. This results in no functional CYP2D6 enzyme in homozygous mutant individuals and the slow-metabolizer phenotype [20]. DNA is purified from human peripheral blood samples and subjected to PCR amplification of a 355-bp fragment of the gene spanning the polymorphic site. The wild-type allele PCR product is recognized by the restriction enzyme BstNI, which digests the D N A into 250-bp and 105-bp fragments. The G to A change in the mutant allele ren- ders the restriction site unrecognizable to BstNI, giv-

E < z a £1.

O O

= E

-355 bp -250 bp

-105 bp

Fig. 1. PCR-RFLP for genotyping CYP2D6*4, Forward primer: 5'-CCT TCG CCA ACC ACT CCG-3'; reverse primer: 5'-AAA TCC TGC TCT TCC GAG GC-3'. After PCR and restriction enzyme B.stNI digestion, the PCR prod- ucts can be resolved by agarose gel electrophoresis and visual- ized under ultraviolet (UV) light after ethidium bromide staining. The presence of DNA bands of 250 and 105 bp indi- cates a CYP2D6 homozygous wild-type genotype (wt/wt). The presence of a single DNA 355-bp band indicates the absence of this restriction endonuclease site and a CYP2D6*4 homozy- gous mutant genotype (rout/rout). Subjects with 3 bands of 355, 250, and 105 bp indicate a heterozygous genotype (wt/mut).

ing rise to a single undigested band of 355 bp. Three genotypes (homozygous wild-type, heterozygous, and homozygous mutant) can be resolved by analyzing the digestion patterns after gel electrophoresis.

A significant limitation of the PCR-RFLP method is the requirement that the polymorphisms alter a restric- tion enzyme cutting site. To detect mutations that do not create or eliminate restriction sites, allele-specific PCR can be performed using mutation-specific amplifica- tion.This technique has been applied for identifying major CYP2D6 poor-metabolizer genotypes [21]. In contrast to poor metabolizers, approximately 7% of whites are ul- traextensive metabolizers of CYP2D6. These individuals have alleles with duplication of CYP2D6 genes and ex- press excessive amounts of CYP2D6 enzymes. As a result, ultraextensive metabolizers require greater-than- average doses of drugs metabolized by CYP2D6 to reach therapeutic plasma concentrations. This C Y P 2 D 6

variation also can be identified by PCR amplification [22]. Allele-specific amplification methods depend on the nature of the mutation and the surrounding se- quences, with guanine cytosine (GC)-rich regions making PCR assays difficult to optimize and multiplex.

Overall, gel-based genotyping assays are relatively straightforward and are useful when dealing with a small number of samples of no more than a few hundred. The methods are labor intensive and require experienced and skilled technical staff for final analysis. These re- quirements make gel-based techniques difficult to apply for large-scale genotyping in clinical trials and routine testing in diagnostic laboratories.

OLA Genotyping. The PCR-OLA takes advantage of fluorescent detection systems. The three oligonucle- otides used in this assay are two allele-specific oligo- nucleotide probes (one specific for the wild-type allele and the other specific for the mutant allele) plus a fluo- rescent common probe. The 3'-ends of the allele-specific probes are immediately adjacent to the 5'-end of the common probe. To perform OLA, the gene fragment containing the polymorphic site is amplified by PCR and incubated with the probes. In the presence of thermally stable DNA ligase, ligation of the fluorescent-labeled probe to the allele-specific probe(s) only occurs when there is a perfect match between the mutant or the nor- mal probe and the PCR product template. These ligation products are then separated by electrophoresis on a fluorescent-based DNA sequencer, which permits the recognition of the normal genotypes, mutants, heterozy- gotes, and unligated probes. By varying the combinations of color dyes and probe lengths, multiple mutations can be detected in a single reaction. This technology is par- ticularly useful for disorders in which there are many mutations causing a single disease, such as cystic fibrosis and familial hypercholesterolemia [23]. An OLA for genotyping CYP2D6 poor metabolizers is reported that

significantly reduces the time and cost associated with high-throughput testing [24].

Similar to allele-specific amplification, OLA relies on hybridization with specific oligonucleotide probes that can effectively discriminate between the wild-type and variant sequences. This method can be used to genotype a large panel of informative biallelic markers. The hy- bridization of allele-specific oligonucleotides is depen- dent on both the variant and surrounding sequences. Highly GC-rich DNA regions make the allele-specific li- gation step in OLA difficult to optimize and multiplex.

Fluorescence Resonance Energy Transfer Genotyp- ing. Fluorescence resonance energy transfer (FRET) oc- curs when two fluorescent dyes are in close proximity to each other and one fluorophore's emission spectrum overlaps the other's excitation spectrum [25]. One appli- cation using this approach is the TaqMan Allelic Dis- crimination assay, which uses the 5'-nuclease activity of Taq polymerase to detect a fluorescent reporter signal generated during or after PCRs [26]. For genotyping single-nucleotide polymorphisms (SNPs), one pair of TaqMan probes and one pair of PCR primers are used. TaqMan probes typically consist of 20- to 40-bp oligonu- cleotides complementary to the polymorphic region, with the polymorphic nucleotide located at approximately the middle of the sequences. The assay uses two TaqMan probes that differ at the polymorphic site, with one probe

Technologies for Pharmacogenomics • Shi et al. 347

complementary to the wild-type allele and the other to the variant allele. A 5'-reporter dye (6-carboxy-4,7,2',7'- tetrachlorofluorescenin [TET]) and a 3'-quencher dye (6-carboxy-N,N,N',N'-tetrachlorofluorescein [TAMRA]) are covalently linked to the wild-type allele probe. Sim- ilarly, the variant allele probe is labeled with a 5'- reporter dye (6-carboxyfluorescein [FAM]) and the same 3'-quencher dye, TAMRA. When the probes are intact, fluorescence is quenched because of the physical proximity of the reporter and quencher dyes.

During the PCR annealing step, the TaqMan probes hybridize to the targeted polymorphic site. During the PCR extension phase, the 5'-reporter dye is cleaved by the 5'-nuclease activity of the Taq polymerase, leading to an increase in the reporter's characteristic fluores- cence. Specific genotyping is determined by measuring the signal intensity of TET and FAM immediately after the PCR. A high-throughput CYP2D6*4 TaqMan geno- typing assay also has been developed (Fig. 2). In addi- tion to detecting SNPs, small gene deletions and inser- tions can be identified by this method.

FRET genotyping offers high sample throughput and accurate detection of SNPs. This fluorescence-based genotyping approach significantly reduces assay com- plexity by eliminating restriction enzyme digestion, gel electrophoresis, resolution of PCR products, and visual assessment of bands. In addition, the assay can use 96-well

Fig. 2. TaqMan genotyping for CYP2D6*4. Forward primer 5'-CGC CTI" CGC CAA CCA CT-3' and reverse primer 5'-CTT TGT CCA AGA GAC CGT TGG-3'. Allele-1 (mutant) TaqMan probe TET-CAC CCC CAA GAC GCC CCT TF-TAMRA, allele 2 (wild-type) probe 6FAM-CAC CCC CAG GAC GCC CCT-TAMRA. The CYP2D6 genotypes were determined according to the ratio of FAM and TET signal. Allele-i, A/A (mut/mut, FAM~TET); allele-2, G/G (wt/wt, FAM,,TET); allele-1 and 2, G/A (wt/mut, FAM --~ TET).

348 Molecular Diagnosis Vol. 4 No. 4 December 1999

plate formats and closed-tube PCRs, minimizing the po- tential for contamination. The ability to automate data handling further enhances accuracy by eliminating oper- ator bias. However, because FRET (TaqMan) genotyp- ing involves at least several steps to optimize conditions and is difficult to multiplex, it will not be the method of choice for determining multiple markers in a small num- ber of samples. This assay, however, may be very useful for large-scale characterization of the same genetic polymorphism.

Conformation-based Mutation Screening. Mutation detection is a key step for the identification of new poly- morphisms. A commonly used strategy for detecting SNPs is to PCR-amplify genes of interest and scan the PCR products for the presence of mutations by confir- mation-based mutation scanning methods, followed by sequencing-positive PCR products. One of the most widely used techniques for identifying unknown muta- tions is SSCP analysis. SSCP is based on the principle that the 3-dimensional conformation of a single- stranded DNA molecule has a specific sequence-based secondary structure in a nondenaturing gel matrix. SSCP involves three major steps: (1) PCR amplification of the region with potential polymorphisms, (2) forma- tion of single-stranded DNA by denaturing the double- stranded PCR products with formamide and heat, and (3) separation of the single-stranded DNA using a nondena- turing polyacrylamide gel. A fragment with a single-base modification generally forms a different conformer and migrates differently than wild-type DNA, and thus mu- tants can be easily identified by these mobility difference~ Whereas the original SSCP protocols incorporated radio- active labels for detection [27], fluorescence labeling now has become widely used [28].

SSCP can be used as a first-pass discrimination test for screening large numbers of samples and offers an economic way to detect new polymorphisms. The sensi- tivity of this method, however, varies for different DNA sequences and is a concern. The sensitivity of SSCP is typically 60% to 95% for fragments less than 250 bp [29]. This sensitivity can be increased to nearly 100% by coupling with either restriction enzyme fingerprinting [30] or dideoxy-sequencing fingerprinting [31]. Al- though these additional components improve sensitiv- ity, they also increase assay complexity and decrease throughput.

Other popular mutation scanning methods include conformation-sensitive gel electrophoresis [32], chemi- cal or enzymatic mismatch cleavage detection [33-35], denaturing gradient gel electrophoresis [36], and dena- turing high-performance liquid chromatography [37]. The underlying principle of these methods is that the melting characteristics of double-stranded DNA is largely defined by its sequence. Therefore, a single-base mis- match can produce conformation changes in the double

helix that causes the differential migration of homo- duplexes and heteroduplexes containing base mismatches during gel electr0ph0resis. Conformation-sensitive gel electrophoresis has been developed to distinguish homo- duplexes from heteroduplexes containing a single mis- matched base pair by polyacrylamide gel electrophoresis in a mildly denaturing solvent system. This method has been shown to be highly sensitive to pick up mutations in areas of highly repetitive and GC-rich sequences [38].

Mismatch cleavage detection takes advantage of the sensitivity of mismatched bases to cleavage by enzymes or chemicals. After PCR amplification, the wild-type and mutant products are subjected to denaturation/ renaturation to create heteroduplex molecules. After in- cubation with the resolvases or chemicals, the products are resolved electrophoretically side by side to score the presence of mismatch cleaved molecules [33-35]. The DNA fragment could be up to 1,000 bp, with a sen- sitivity greater than 90%.

In denaturing gradient gel electrophoresis, genomic DNA are PCR amplified, and the products are then re- solved in a polyacrylamide gel with an increasing dena- turing gradient of formamide and urea under careful temperature control. Heteroduplex DNA fragments with a single mismatched base pair are shown as migra- tional differences from homoduplexes. The advantages of this method are its accuracy and relatively inexpen- sive cost. The disadvantages are its low throughput and difficulty to optimize [36].

Denaturing high-performance liquid chromatography is another form of heteroduplex analysis for detecting DNA mobility differences by chromatographic analysis in slightly denaturing conditions. Temperature-modulated heteroduplex analysis is a newly commercialized method from Transgenomic, Inc. (San Jose, CA) [39]. The mu- tant sample is first hybridized with wild-type DNA to form a mixture of homoduplexes and heteroduplexes. The heteroduplexes can be separated from the homodu- plexes by column chromatography at a temperature to partially denature the mismatched DNA. The denatur- ing temperature can be selected for optimal resolution. The procedure is fully automated and could be applied for high-throughput mutation detection.

High-throughput DNA Sequencing. DNA sequenc- ing is the most direct diagnostic method to determine a target gene and remains the gold standard for detecting mutations. In this mode, an individual's sequence can be compared with the wild-type sequence to identify poly- morphisms. The major platform used by research and diagnostic laboratories is now fluorescent-based se- quencing using multiple fluorochromes. With the im- provement of computer software and detection systems, fluorescent DNA sequencing has become fully auto- mated in all phases of the sequencing process.

The fluorescent DNA sequencer also offers precise

Technologies for Pharmacogenomics • Shi et al. 349

allele sizing and quantitation of DNA fragments. These advances have set the stage for other molecular biologi- cal applications based on multicolor fluorescence, in- cluding microsatellite polymorphism sizing and tandem repeat genotyping [40], gene mapping [41], detection of loss of heterozygosity [42], and microsatellite instability [43]. The sensitivity and specificity of these fluorescent genotyping methods for detecting heterozygous geno- types are superior to conventional RFLP or allele-specific PCR. These approaches are emerging as vital tech- niques for disease gene mapping in research and in di- agnostic laboratories.

DNA Microarray Chip. The DNA microarray chip is a hybridization-based genotyping technique that offers si- multaneous analysis of many polymorphisms. High- density microarrays are created by attaching hundreds of thousands of oligonucleotides to a solid silicon surface in an ordered array.The DNA sample of interest is PCR am- plified to incorporate fluorescent-labeled nucleotides and then hybridized to the chip array. Each oligonucleotide in the high-density array acts as an allele-specific probe. Per- fectly matched sequences hybridize more efficiently to their corresponding oligomers on the array and therefore give stronger fluorescent signals over mismatched probe- target combinations. The hybridization signals are quanti- tated by high-resolution fluorescent scanning and ana- lyzed by computer software. DNA alterations, such as heterozygous base-pair polymorphisms or mutations, insertions, and deletions, can be identified [44]. Cur- rently, the initial start-up and chip costs of the micro- array procedures are high relative to other methods. As greater numbers of clinically important polymorphisms are identified, application of microarray chip technology to simultaneously monitor tens of thousands of genetic variations will provide significant advantages for genome-wide screening. The clinical usefulness of these chip-based genotyping technologies is currently under development and evaluation in several companies.

Although simple tandem repeats or microsatellite poly- morphisms have been used in linkage analysis, it is be- coming increasingly popular to use SNP analysis because SNPs are the most frequent DNA sequence variations found in the human genome. By creating high-density SNP maps, researchers will expand genomic capabilities for identification of critical diseases and drug response genes in nonfamilial studies. Creation of these maps now is feasible using high-throughput DNA sequencing [45] and chip hybridization [46].

MS. MS is an established method in the analytic arena and a new technology for high-throughput genotyping [47-50]. MS yields precise information on the molecular mass of the DNA fragments, the procedure can be fully automated, and both DNA strands can be analyzed in parallel. Large molecules now can be identified by MS through electrospray or matrix-assisted laser desorption

ionization (MALDI) and ion trap or time of flight (TOF) detectors. MALDI-TOF MS has been successfully ap- plied to DNA analysis [49]. The mass determination pro- cess usually involves incorporation of internal standards into the MALDI preparation, and the internal standard signals are used to calibrate the mass spectrometer. These methods are more precise and quicker than traditional gel-based analyses [51]. One previous limiting factor for SNP analysis was the mass resolution required for discrim- inating the small-molecular-weight differences between adenosine and thymidine (9Da) in PCR-generated frag- ments. By generating very short defined fragments of PCR products using restriction endonuclease digestion and direct analysis by electrospray ionization MS, the method has been dramatically improved [51]. By incor- porating the restriction site into the PCR primers, no re- striction recognition sites in the targeted DNA sequences are required. The size of the final endonuclease products can be as small as 7 bp and will allow accurate identifica- tion of all common DNA variations.

Unlike the fluorescent genotyping methods previ- ously described, MS offers specificity and accuracy with- out requiring specially labeled probes or primers. This feature reduces start-up reagent costs. The current limi- tation of MS is the difficulty applying it to ultra-high- throughput genotyping because the current MS instru- ments only handle one sample at a time. Even with multi- plex PCR, each MS instrument is capable of analyzing no more than a few thousand markers per day. In addi- tion, MS genotyping requires purified samples free of ions and other impurities, thus increasing the technical time and sample-processing costs.

Conclusions

Pharmacogenomics has the potential to improve our understanding of interindividual variations in drug re- sponses resulting from genetic polymorphisms. A major challenge we are facing today is to compare hundreds of thousands of polymorphisms among numerous individ- uals.Thus, the success of pharmacogenomics depends on the technologies that can detect polymorphisms rapidly and accurately in a cost-effective and user-friendly way on a large scale. Each of the methods previously dis- cussed has been proven to work in a variety of settings, but each method has its own limitations. Of all the op- tions available, non-gel-based and chip-based genotyping technologies will most likely evolve as the ultra-high- throughput detection systems to meet the requirements of pharmacogenomics. Once validated and proven to be cost effective, pharmacogenomics will revolutionize our concepts of modern medicine by allowing physicians to prescribe the most effective and safest drug based on the patient's genetic blueprint.

350 Molecular Diagnosis Vol. 4 No. 4 December 1999

Acknowledgment

The authors thank Scott Myrand for technical assis- tance and efforts in developing multiple new genotyping techniques.

Received April 27, 1999. Received in revised form June 18, 1999.

Accepted August 10, 1999.

References

1. Kleyn PW, Vesell ES: Genetic variation as a guide to drug development. Science 1998;281:1820-1821

2. Benet LZ, Kroetz DL, Sheiner LB: General princi- ples, In Hardman JG, Limbird LE (eds.): Goodman & Gilman's the pharmacological basis of therapeu- tics, 9th ed. McGraw-Hill, New York, 1996, 3-27

3. Kalow W (ed.): Pharmacogenetics of drug metabo- lism. International encyclopedia of pharmacology and therapeutics. Oxford, Pergamon Press, 1992, 1- 888

4. De Morals SM, Uetrecht JR Wells PG: Decreased glucuronidation and increased bioactivation of acet- aminophen in Gilbert's syndrome. Gastroenterol- ogy 1992;102:577-586

5. Mackenzie PI, Owens IS, Burchell B, et al.: The UDP glycosyltransferase gene superfamily: Recom- mended nomenclature update based on evolution- ary divergence. Pharmacogenetics 1997;7:255-269

6. Owens D, Evans J: Population studies on Gilbert's syndrome. J Med Genet 1975;12:152-156

7. Iyer L, King C, Whitington PF, et al.: Genetic predis- position to the metabolism of irinotecan (CPT-11): Role of uridine diphosphate glucuronosyltransferase 1A1 in the glucuronidation of its active metabolite (SN-38) in human liver microsomes. J Clin Invest 1998;101:847-854

8. Raftogianis RB, Wood TC, Otterness DM, van Loon JA, Weinshilboum RM: Phenol sulfotransferase phar- macogenomics in humans: Association of common SULTIA1 alleles with TS PST phenotype. Biochem Biophys Res Commun 1997;239:298-304

9. Nowell SA, Leakey JE, Warren JF, Lang NR Frame LT: Identification of enzymes responsible for the metabolism of heine in human platelets. J Biol Chem 1998;273:33342-33346

10. Jones AL, Hagen M, Coughtrie MW, Roberts RC, Glatt H: Human platelet phenolsulfotransferases: cDNA cloning, stable expression in V79 cells and identification of a novel allelic variant of the phenol- sulfating form. Biochem Biophys Res Commun 1995;208:855-862

11. Ritter JK, Crawford JM, Owens IS: Cloning of two

human liver bilirubin UDP-glucuronosyltransferase cDNAs with expression in COS-1 cells. J Biol Chem 1991 ;266: i 04321047

12. Cohen LJ, de Vane CL: Clinical implications of antidepressant pharmacokinetics and pharmacoge- netics. Ann Pharmacother 1996;30:1471-1480

13. Coutts RT: Polymorphism in the metabolism of drugs, including antidepressant drugs: Comments on phe- notyping. J Psychiatry Neurosci 1994;19:30-44

14. Kalow W, Otton SV, Kadar D, Endrenyi L, Inaba T: Ethnic difference in drug metabolism: Debriso- quine 4-hydroxylation in Caucasians and Orientals. Can J Physiol Pharmacol 1980;58:1142-1144

15. Wietholtz H, Voegelin M, Arnaud MJ, Bircher J, Preisig R: Assessment of the cytochrome P-448 dependent liver enzyme system by a caffeine breath test. Eur J Clin Pharmacol 1981;21:53-59

16. Park YH, Kullberg MR Hinsvark ON: Quantitative determination of dextromethorphan and three metab- olites in urine by reverse-phase high-performance liquid chromatography. J Pharm Sci 1984;73:24-29

17. Kharasch ED, Thummel KE, Mhyre J, Lillibridge JH: Single-dose disulfiram inhibition of chlorzox- azone metabolism: A clinical probe for P450 2El, Clin Pharmacol Ther 1993;53:643-650

18. Lown K, Kolars J, Turgeon K, Merion R, Wrighton SA, Watkins PB: The erythromycin breath test selec- tively measures P450IIIA in patients with severe liver disease. Clin Pharmacol Ther 1992;51:229-238

19. Kleinbloesem CH, van Brummelen R Faber H, Dan- hof M, Vermeulen NR Breimer DD: Variability in nifedipine pharmacokinetics and dynamics: A new oxidation polymorphism in man. Biochem Pharma- col 1984;33:3721-3724

20. Gough AC, Miles JS, Spurr NK, et al.: Identifica- tion of the primary gene defect at the cytochrome P450 CYP2D locus. Nature 1990;347:773-776

21. Heim M, Meyer UA: Genotyping of poor metabo- lizers of debrisoquine by allele-specific PCR ampli- fication. Lancet 1990;336:529-532

22. Sachse C, Brockmoller J, Bauer S, Roots I: Cyto- chrome P450 2D6 variants in a Caucasian popula- tion: Allele frequencies and phenotypic conse- quences. Am J Hum Genet 1997;60:284-295

23. Baron H, Fung S, Aydin A, Bahring S, Luft FC, Schuster H: Oligonucleotide ligation assay (OLA) for the diagnosis of familial hypercholesterolemia. Nat Biotechnol 1996;14:1279-1282

24. Hansen TS, Petersen NE, Iitia A, Blaabjerg O, Hyltoft-Petersen R Horder M: Robust nonradioac- tive oligonucleotide ligation assay to detect a com- mon point mutation in the CYP2D6 gene causing abnormal drug metabolism. Clin Chem 1995;41: 413-418

25. Clegg RM: Fluorescence resonance energy transfer and nucleic acids. Methods Enzymo11992;211:353-388

Technologies for Pharmacogenomics • Shiet al. 351

26. Livak KJ, Flood SJA, Marmaro J, Giusti W, Deetz K: Oligonucleotides with fluorescent dyes at oppo- site ends provide a quenched probe system useful for detecting PCR product and nucleic acid hybrid- ization. PCR Methods Appl 1995;4:357-362

27. Orita M, Suzuki Y, Sekiya I, Hayashi K: Rapid and sensitive detection of point mutations and DNA polymorphisms using the polymerase chain reac- tion. Genomics 1989;5:874-879

28. Makino R, Yazyu H, Kishimoto Y, Sekiya T, Hayashi K: F-SSCP: Fluorescence-based polymerase chain reaction-single-strand conformation polymorphism (PCR-SSCP) analysis. PCR Methods Appl 1992;2: 10-13

29. Eng C, Vijg J: Genetic testing: The problems and the promise. Nat Biotechnol 1997;15:422-426

30. Liu Q, Sommer SS: Restriction endonuclease finger- printing (REF): A sensitive method for screening mutations in long, contiguous segments of DNA. Biotechniques 1995;18:470-477

31. Sarkar G, Yoon HS, Sommer SS: Dideoxy finger- printing (ddE): A rapid and efficient screen for the presence of mutations. Genomics 1992;13:441-443

32. Ganguly A, Prockop D J: Detection of mismatched bases in double-stranded DNA by gel electro- phoresis. Electrophoresis 1995;16:1830-1835

33. Cotton RGH, Rodrigues NR, Campbell RD: Reac- tivity of cytosine and thymine in single-base pair mismatches with hydroxylamine and osmium tetrox- ide and its application to the study of mutations. Proc Natl Acad Sci U S A 1988;85:4397-4401

34. Mashal RD, Koontz J, Sklar J: Detection of muta- tions by cleavage of DNA heteroduplexes with bac- teriophage resolvases. Nat Genet 1995;9:177-183

35. Youil R, Kemper BW, Cotton RG: Screening for mutations by enzyme mismatch cleavage with T4 endonuclease VII. Proc Natl Acad Sci U S A 1995; 92:87-91

36. Myers RM, Maniatis T, Lerman LS: Detection and localization of single base changes by denaturing gradient gel electrophoresis. Methods Enzymol 1987; 155:501-527

37. O'Donovan MC, Oefner PJ, Roberts SC, et al.: Blind analysis of denaturing high-performance liquid chromatography as a tool for mutation detection. Genomics 1998;52:44-49

38. Korkko J, Annunen S, Pihlajamaa T, Prockop D J,

Ala-Kokko L: Conformation sensitive gel electro- phoresis for simple and accurate detection of muta- tions: Comparison with denaturing gradient gel electrophoresis and nucleotide sequencing. Proc Natl Acad Sci U S A 1998;95:1681-1685

39. Kuklin A, Munson K, Gjerde D, Haefele R, Taylor P: Detection of singlemucleotide polymorphisms with the WAVE T M DNA fragment analysis system. Genet Testing 1997/98;1:201-206

40. Perlin MW, Lancia G, Ng SK: Toward fully automated genotyping: Genotyping microsatellite markers by deconvolution. Am J Hum Genet 1995;57:1199-1210

41. Davis JL, Kawaguchi Y, Bennett ST, et al.: A genome-wide search for human type 1 diabetes sus- ceptibility genes. Nature 1994;371:130-136

42. Canzian F, Salovaara R, Hemminki A, et al.: Semi- automated assessment of loss of heterozygosity and replication error in tumors. Cancer Res 1996;56: 3331-3337

43. Thibodeau SN, Bren G, Schaid D: Microsatellite instability in cancer of the proximal colon. Science 1.993;260:816-819

44. Chee M, Yang R, Hubbell E, et al.: Accessing genetic information with high-density DNA arrays. Science 1996;274:610-614

45. Lai E, Riley J, Purvis I, Roses A: 4-Mb high-density single nucleotide polymorphism-based map around human APOE. Genomics 1998;54:31-38

46. Wang DG, Fan JB, Siao CJ, et al.: Large-scale identifi- cation, mapping, and genotyping of single-nucleotide polymorphisms in the human genome. Science 1998; 280:1077-1082

47. Beattie KL, Beattie WG, Meng L, et al.: Advances in genosensor research. Clin Chem 1995;41:700-706

48. Schmalzing D, Koutny L, Adourian A, Belgrader R Matsudaira R Ehrlich D: DNA typing in thirty sec- onds with a microfabricated device. Proc Natl Acad Sci U S A 1997;94:10273-10278

49. Ross R Hall L, Smirnov I, Hall L: High-level multi- plex genotyping by MALDI-TOF mass spectrome- try. Nat Biotechnol 1998;16:1347-1351

50. Laken SJ, Jackson PE, Kinzler KW, et al.: Genotyp- ing by mass spectrometric analysis of short DNA fragments. Nat Biotechnol 1998;16:1352-1356

51. Lewis JK, Krone JR, Nelson RW: Mass spectromet- ric methods for evaluating point mutations. Bio- techniques 1998;24:102-110