WO1999057322A2 - Genotyping the human udp-glucuronosyltransferase 1 (ugt1) gene - Google Patents

Genotyping the human udp-glucuronosyltransferase 1 (ugt1) gene Download PDF

Info

Publication number
WO1999057322A2
WO1999057322A2 PCT/US1999/009702 US9909702W WO9957322A2 WO 1999057322 A2 WO1999057322 A2 WO 1999057322A2 US 9909702 W US9909702 W US 9909702W WO 9957322 A2 WO9957322 A2 WO 9957322A2
Authority
WO
WIPO (PCT)
Prior art keywords
ugt1
seq
expression
polymorphisms
polymorphism
Prior art date
Application number
PCT/US1999/009702
Other languages
French (fr)
Other versions
WO1999057322A3 (en
Inventor
Laura Penny
Margaret Galvin
Original Assignee
Axys Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Axys Pharmaceuticals, Inc. filed Critical Axys Pharmaceuticals, Inc.
Priority to EP99921638A priority Critical patent/EP1084271A2/en
Priority to AU38792/99A priority patent/AU3879299A/en
Publication of WO1999057322A2 publication Critical patent/WO1999057322A2/en
Publication of WO1999057322A3 publication Critical patent/WO1999057322A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6881Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for tissue or cell typing, e.g. human leukocyte antigen [HLA] probes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6827Hybridisation assays for detection of mutation or polymorphism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • Table 1 provides a summary of the major endobiotic and xenobiotic substrates, as well as exemplary non- substrates, of four UGT1 isoenzymes (UGT1*1 (same as UGT1 A), UGT1*4 (same as UGT1D), UGT1 * 6 (same as UGT1F), and UGT1*02 (same as UGT1G) (see Burchell et al. 91995) Life Sci. 57:1819-31).
  • the basal expression level in different tissue may be determined by analysis of tissue samples from individuals typed for the presence or absence of a specific polymorphism. Any convenient method may be use, e.g. ELISA, RIA, etc. for protein quantitation, northern blot or other hybridization analysis, quantitative RT-PCR, etc. for mRNA quantitation.
  • the tissue specific expression is correlated with the genotype.
  • the alteration of UGT1 expression in response to a modifier is determined by administering or combining the candidate modifier with an expression system, e.g. animal, cell, in vitro transcription assay, etc.
  • the genotype of an individual is determined with respect to the provided UGT1 gene polymorphisms.
  • the genotype is useful for determining the presence of a phenotypically evident polymorphism, and for determining the linkage of a polymorphism to phenotypic change.
  • TTTGTCTTCCAATTACATGC SEQ ID AGTAGATATGGAAGCACTTGTAAG (SEQ NO:23) ID N0:24
  • each primary PCR reaction was re- amplified using the secondary PCR primers, also listed in Table 2.
  • the thermal cycling profile that was used for the primary PCR for an exon was used for the secondary PCR.
  • IH Touch-Down PCR step 10 cycles same as Primary PCR 66 C (minus 1 C per cycle), for 15 sec Total Number of cycles: 35
  • PCR products from 48 individuals were prepared for sequencing by treating 8 ⁇ L of each PCR product with 0.15 ⁇ L of exonuclease I (1.5 U/reaction), 0.3 ⁇ L of Shrimp Alkaline Phosphatase (0.3 U/reaction), q.s. to 10 ⁇ L with MilliQ water, and incubated at 37°C for 15 min, followed by 72°C for 15 min.
  • Cycle sequencing was performed on the GeneAmp PCR System 9600 PCR machine (Perkin Elmer) using the ABI Prism dRhodamine Terminator Cycle Sequencing Ready Reaction Kit according to the manufacturer's directions, with the following changes: (1) 2 ⁇ L of dRhodamine terminator premix, instead of 8 ⁇ L; and (2) 10% (v/v) Dimethylsulfoxide was added to each individual nucleotide.
  • the oligonucleotide primers (unlabeled), at 3 picomoles per reaction, used for the sequencing reactions are listed in Table 4.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Immunology (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

Genetic polymorphisms are identified in the human UGT1 gene that alter UGT1-dependent drug metabolism. Nucleic acids comprising the polymorphic sequences are used to screen patients for altered metabolism for UGT1 substrates, potential drug-drug interactions, and adverse/side effects, as well as diseases that result from environmental or occupational exposure to toxins. The nucleic acids are used to establish animal, cell and in vitro models for drug metabolism.

Description

GENOTYPING THE HUMAN UDP-GLUCURONOSYLTRANSFERASE 1 (UGT1) GENE
INTRODUCTION The metabolic processes commonly involved in the biotransformation of xenobiotics have been classified into functionalization reactions (phase I reactions), in which lipophilic compounds are modified via monooxygenation, dealkylation, reduction, aromatization, or hydrolysis. These modified molecules can then be substrates for the phase II reactions, often called conjugation reactions, as they conjugate a functional group with a polar, endogenous compound. Drug glucuronidation, a major phase II conjugation reaction in the mammalian detoxification system, is catalyzed by the UDP-glucuronosyltransferases (UGTs) (Batt AM, et ai. (1994) Clin Chim Acta 226:171-190: Burchell et al. (1995 Life Sci. 57:1819-31).
The UGTs are a family of enzymes that catalyze the glucuronic acid conjugation of a wide range of endogenous and exogenous substrates including phenols, alcohols, amines and fatty acids. The reactions catalyzed by UGTs permit the conversion of a large range of toxic endogenous/xenobiotic compounds to more water-soluble forms for subsequent excretion (Parkinson A (1996) Toxicol Pathol 24:48-57).
The UGT isoenzymes are located primarily in hepatic endoplasmic reticulum and nuclear envelope (Parkinson A (1996) Toxicol Pathol 24:48-57), though they are also expressed in other tissues such as kidney and skin. UGTs are encoded by a large multigene superfamily that has evolved to produce catalysts with differing but overlapping substrate specificities. Three families, UGT1 , UGT2, and UGT8, have been identified within the superfamily. UGTs are assigned to one of the subfamilies based on amino acid sequence identity, e.g., UGT1 family members have greater than 45% amino acid sequence identity (Mackenzie et al. 1997) Pharmacogenetics 7:255-69).
The UGT1 locus is located on chromosome 2q37, and contains at least 12 promoters/first exons, which are apparently able to splice with common exons 2 through 5, producing gene products having strikingly different N-terminal halves (amino acid sequence identities ranging from 24% to 49%), but identical C-terminal halves (Fig. 1). At least eight different isoenzymes are encoded by the UGT1 locus; at least one or more first exons encode pseudogenes. The different N-terminal halves encoded by the first exons confer different substrate binding specificities upon the UGT1 isoenzymes, while exons 2-5, which are present in all UGT1 isoenzyme mRNAs, encode the UDP-glucuronic acid binding domain, membrane anchorage site, and ER retention signal that are common to all UGT proteins (Ritter et al. (1992) J Biol Chem 267:3257-3261). UGT1 locus isoenzymes are best known for their role in glucoronidation and metabolism of many substrates, including bilirubin (1A1 , 1D1), planar and non-planar phenols, naphthols (1F1) (Ouzzine M, et al. (1994) Arch Biochem Biophys 310:196-204), anthraquinones, flavones, aliphatic alcohols, aromatic carboxylic acids, and steroids (Ebner T, et al. (1993) Drug Metab Dispos 21:50- 55).
In addition to UGT1 exon usage, metabolism of endogenous and exogenous substrates can also be affected by competitive binding phenomena. For example, in some cases exogenous substrates for the UGT1 enzymes have a higher binding affinity or avidity for the enzyme than the endogenous UGT1 substrates. For example, UGT1*1 , the major bilirubin-metabolizing form of UGT1 , more readily binds both octyl-gallate and emodin than it binds bilirubin, thus indicating the potential of these xenobiotics to cause jaundice by inhibition of bilirubin binding to UGT1*1 (where 1*1 indicates that the first exon is used in the spliced gene product). UGT1*1 is also responsible for glucuronidation of the oral contraceptive ethinylestradiol (Ebner et al. (1993) Mol. Pharmacol. 43:649-54), and can also glucuronidate phenols, anthroquinones, flavones, and certain endogenous steroids. As noted above, the first exon present in UGT1 can affect substrate binding specificity of the UGT1 gene product (for a review, see Burchell (1995) Life Sci. 57:1819- 31). For example, UGT1*2 accepts a wide range of compounds as substrates including non-planar phenols, anthraquinones, flavones, aliphatic alcohols, aromatic carboxylic acids, steroids (4-hydroxyestrone, estrone) and many drugs of varied structure (Ebner et al. (1993) Drug. Metab. Disp. 21:50-5; Burchell (1995) Life Sci. 57:1819-31). In contrast, UGT1*6 exhibits only limited substrate specificity for planar phenolic compounds relative to other human UGTs.
Polymorphisms can markedly affect binding of the endogenous substrate, which can be manifested as clinical syndromes. At least two conditions, Crigler-Najjar syndrome and Gilbert syndrome, are associated with UGT1 polymorphisms. Both of these syndromes are hereditary and are associated with predominantly unconjugated hyperbilirubinemia. Crigler- Najjar syndrome is associated with intense, persistent jaundice which begins at birth. Some affected infants die in the first weeks or months of life with kernicterus; others survive with little or no neurologic defect. Crigler-Najjar syndrome is caused by a defect in the ability of UGT1 to catalyze UDP-glucuronidation of bilirubin, resulting in accumulation of bilirubin in the blood (Erps et al. (1994) J. Clin. Invest. 93:564-70). Gilbert syndrome is a benign mild form of unconjugated hyperbilirubinemia that is characterized by normal liver function tests, normal liver histology, delayed clearance of bilirubin from the blood, and mild jaundice that tends to fluctuate in severity. As with Crigler-Najjar syndrome, Gilbert syndrome is associated with a defect in UGT1. Specific UGT polymorphisms that are known to be associated with disease are indicated in Fig. 1.
Alteration of the expression or function of UGTs may also affect drug metabolism. For example, there may be common polymorphisms in the human UGT1 gene that alter expression or function of the protein product and cause drug exposure-related phenotypes. Thus, there is a need in the field to identify UGT1 polymorphisms in order to provide a better understanding of drug metabolism and the diagnosis of drug exposure-related phenotypes.
RELEVANT LITERATURE
Genbank accession number M84122 provides UGT1 exon 2, M84123 provides exons 3 and 4, M84124 provides 5, M84125 provides exon 1A, M84127 provides exon 1C,
M84128 provides exon 1 D, M84129 provides exon 1E, M84130 provides exon 1 F, U39570 provides exon 1G, U42604 provides exon 1 H, U39550 provides exon U. The UGT gene superfamily and recommended nomenclature for describing UGT genes and alleles are reviewed in Mackenzie et al. (1997) Pharmacogenet. 7:255-69.
The two UGT1A6 genetic polymorphisms are described in Ciotti et al. (1997) Am. J.
Hum. Genet. 61(Supp):A249. The identification of Asp446 as a critical residue in UGT1 is described in Iwano et al. (1997) Biochem. J. 325:587-91. A review of the substrate specificity of human UDP-glucuronosyltransferases is provided by Burchell et al. (1995) Life Sci. 57:1819-31. For a review of drug glucoronidation in humans, see Miners et al. (1991) Pharmacol. Ther. 51:347-69.
At least twelve UGT1A1 polymorphisms have been identified and linked to disease.
These UGT1A1 alleles, each of described in OMIM Entry 191740 (at http://www.ncbi.nlm.nih.gov/htbin-post/Omim/dispmim7191740) and in OMIM Entry 143500
(at http://www.ncbi.nlm.nih.gov/htbin-post/Omim/dispmim7143500), include:
1) UGT1*FB (UGT1A1 , 13-BP DEL, EX2; 191740.0001), which contains a 13 bp deletion in exon 2 and is associated with Crigler-Najjar syndrome type I (CN-I);
2) UGT1A1 , EXON4, C-T, SER-PHE (191740.0002), which contains a C-to-T transition in exon 4 (resulting in an amino acid change from serine to phenylalanine) is associated with CN-I and deficiency of both bilirubin-UGT and phenol-UGT activities in the liver;
3) UGT1A1 , GLN331TER (191740.0003), which contains a C-to-T transition 6 bp upstream from the 3-prime end of exon 2 of the common region (replacement of a glutamine codon with a stop codon), is associated with CN-I; 4) UGT1A1 , ARG341TER (191740.0004), which contains a nonsense mutation (CGA-to-TGA) in exon 3 and is associated with CN-I and a total absence of all phenol/bilirubin UGT proteins and their activities in liver homogenate by enzymologic and immunochemical analysis; 5) UGT1A1 , GLN331ART (191740.0005), which contains an A-to-G transition 5 bp upstream of the exon 2/intron 2 boundary (resulting in a glutamine-to-arginine substitution), is associated with Crigler-Najjar Syndrome, type II (CN-II);
6) UGT1A1 , PHE170DEL (191740.0006), which contains a deletion of the phenylalanine codon at position 170 in exon 1 , and is associated with CN-I; 7) UGT1A1 , SER376PHE (191740.0007), which contains a C-to-T transition in codon 376 (resulting in a change of serine to phenylalanine) and is associated with CN-I;
8) UGT1A1 , GLY309GLU (191740.0008), which contains a G-to-A transition in codon 309 (resulting in a glycine to glutamic acid change) and is associated with CN-I;
9) UGT1A1 , NT840, C-A, CYS-TER (191740.0009), which contains a C-to-A transversion at base position 840 in exon 1 (resulting in replacing a cysteine with a stop codon), is associated with CN-I;
10) UGT1A1 , PRO229GLN (191740.00010), which contains C-to-A transversion at nucleotide 686 (changing proline-229 to glutamine), is associated with Gilbert syndrome;
11) UGT1A1 , 2-BP INS, TA INS, TATAA ELEMENT (191740.00011) contains 2 extra bases (TA) in the TATAA element of the 5-prime promoter region of the gene (where normally an A(TA)6TAA element is present between nucleotides -23 and -3) and is associated with Gilbert syndrome; and
12) UGT1A1 , 1-BP INS, 470T INS (191740.00012), which contains 470insT mutation in exon 1 and is associated with CN-I.
SUMMARY OF THE INVENTION Genetic sequence polymorphisms are identified in the UGT1 gene. Nucleic acids comprising the polymorphic sequences are used in screening assays, and for genotyping individuals. The genotyping information is used to predict an individuals' rate of metabolism for UGT1 substrates, potential drug-drug interactions, and adverse/side effects.
Accordingly, in one aspect the invention features an isolated nucleic acid molecule comprising a UGT1 sequence polymorphism of SEQ ID NOS:87-124, as part of other than a naturally occurring chromosome. In related aspects, the invention features nucleic acid probes for detection of UGT1 locus polymorphisms, where the probe comprises a polymorphic sequence of SEQ ID NOS:87-124. In another aspect the invention features an array of oligonucleotides comprising two or more probes for detection of UGT1 locus polymorphisms, where the probes comprise at least one form of a polymorphic sequences of SEQ ID NOS:87-124.
In still another aspect, the invention features a method for detecting in an individual a polymorphism in UGT1 metabolism of a substrate, where the method comprises analyzing the genome of the individual for the presence of at least one UGT1 polymorphism of SEQ ID NOS:87-124; wherein the presence of the predisposing polymorphism is indicative of an alteration in UGT1 expression or activity.
In one embodiment, the analyzing step of the method is accomplished by detection of specific binding between the individual's genomic DNA with an array of oligonucleotides comprising two or more probes for detection of UGT1 locus polymorphisms, where the probes comprise at least one form of a polymorphic sequence of SEQ ID NOS:87-124.
In other embodiments of the method, the alteration is UGT1 expression or activity is tissue specific, or is in response to a UGT1 modifier. The UGT1 modifier may either induce or inhibit UGT1 expression.
BRIEF DESCRIPTION OF THE DRAWINGS Fig. 1 is a schematic showing the UGT1 locus. Each of the first exons is denoted by both its alphabetic and numerical nomenclatures (e.g., 1A and 1.1). Fig. 2 is a schematic showing exons 1A-1J of the UGT1 locus and the polymorphisms described in the present application.
Fig. 3 is a schematic showing the exons 1A-1 F, and 2-5 of the UGT1 locus and the polymorphisms that have been publicly disclosed.
BRIEF DESCRIPTION OF THE SEQUENCE LISTING
UGT1 Reference Sequences. SEQ ID NOS: 1 , 3, 5, 7, 9, 11 , 13, and 15 are the UGT1 reference polynucleotide sequences for UGT1 exons 1A, 1C, 1D, 1 E, 1 F, 1G, 1 H, and 1 J. The polypeptide sequences are encoded by these reference exon sequences are SEQ ID NOS:2, 4, 6, 8, 12, 14, and 16. SEQ ID NOS: 17 and 18 are the reference polynucleotide and amino acid sequences for UGT1 exons 2-5.
PCR Primers. The primary and secondary PCR primers for amplification of polymorphic sequences are presented as SEQ ID NOS:19-50.
Sequencing Primers. The primers used in sequencing isolated polymorphic sequences are presented as SEQ ID NOS:51-86. Polymorphisms. Polymorphic sequences of the invention are presented as SEQ ID
NOS:88-124. DESCRIPTION OF THE SPECIFIC EMBODIMENTS Pharmacogenetics is the linkage between an individual's genotype and that individual's ability to metabolize or react to a therapeutic agent. Differences in metabolism or target sensitivity can lead to severe toxicity or therapeutic failure by altering the relation between bioactive dose and blood concentration of the drug. Relationships between polymorphisms in metabolic enzymes or drug targets and both response and toxicity can be used to optimize therapeutic dose administration.
Genetic polymorphisms are identified in the UGT1 gene. Nucleic acids comprising the polymorphic sequences are used to screen patients for altered metabolism for UGT1 substrates, potential drug-drug interactions, and adverse/side effects, as well as diseases that result from environmental or occupational exposure to toxins. The nucleic acids are used to establish animal, cell culture and in vitro cell-free models for drug metabolism.
Definitions It is to be understood that this invention is not limited to the particular methodology, protocols, cell lines, animal species or genera, constructs, and reagents described, as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. As used herein the singular forms "a", "and", and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a construct" includes a plurality of such constructs and reference to "the UGT1 nucleic acid" includes reference to one or more nucleic acids and equivalents thereof known to those skilled in the art, and so forth. All technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs unless clearly indicated otherwise.
UGT1 polymorphic sequences. The sequence of the UGT1 gene is known in the art, and accessible in public databases, as cited above. This sequence is useful as a reference for the genomic location of the human gene, and for specific coding region sequences. As used herein, the term "UGT1 gene" is intended to refer to both the wild-type and variant sequences, unless specifically denoted otherwise. Nucleic acids of particular interest comprise the provided variant nucleotide sequence(s). For screening purposes, hybridization probes may be used where both polymorphic forms are present, either in separate reactions, or labeled such that they can be distinguished from each other. Assays may utilize nucleic acids that hybridize to one or more of the described polymorphisms. The genomic UGT1 sequence is of particular interest. A polymorphic UGT1 gene sequence, i.e. including one or more of the provided polymorphisms, is useful for expression studies to determine the effect of the polymorphisms on enzymatic activity. The polymorphisms are also used as single nucleotide polymorphisms to detect genetic association with phenotypic variation in UGT1 activity and expression.
The UGT1 exon structure is illustrated in Fig. 1. The UGT1 locus contains at least 12 promoters/first exons, which are apparently able to splice with common exons 2 through 5, producing gene products having different N-terminal halves but identical C-terminal halves. The first exon utilized at least in part determines the substrate specificity of the resulting UGT1 gene product. Each of the first exons in Fig. 1 is denoted by both its alphabetic and numerical nomenclatures (e.g., 1A and 1.1). Polymorphisms in the UGT1 first exon can be associated with alteration of substrate binding specificity and/or disease. Fig. 2 shows UGT1 exons 1A-1 J and the polymorphisms described in the present application. Fig. 3 shows UGT1 exons 1A-1F and 2-5 and the polymorphisms in these exons that have been publicly disclosed. Polymorphisms denoted by an asterisk (*) have been assigned the indicated "allele name" (e.g., *12). The specific associated disease is indicated below in parentheses for several of these disease-associated polymorphisms. Except for the "mutation" that is associated with Gilbert's (*28, which is not universally agreed upon in the literature), all mutations in exons 1D, 1A, and 2-5 were isolated from individuals with disease.
Fragments of the DNA sequence are obtained by chemically synthesizing oligonucleotides in accordance with conventional methods, by restriction enzyme digestion, by PCR amplification, etc. For the most part, DNA fragments will be of at least 15 nt, usually at least 20 nt, often at least 50 nt. Such small DNA fragments are useful as primers for PCR, hybridization screening, etc. Larger DNA fragments, i.e. greater than 100 nt are useful for production of the encoded polypeptide, promoter motifs, etc. For use in amplification reactions, such as PCR, a pair of primers will be used. The exact composition of primer sequences is not critical to the invention, but for most applications the primers will hybridize to the subject sequence under stringent conditions, as known in the art. The UGT1 nucleic acid sequences are isolated and obtained in substantial purity, generally as other than an intact mammalian chromosome. Usually, the DNA will be obtained substantially free of other nucleic acid sequences that do not include a UGT1 sequence or fragment thereof, generally being at least about 50%, usually at least about 90% pure and are typically "recombinant", i.e. flanked by one or more nucleotides with which it is not normally associated on a naturally occurring chromosome. UGT1 polypeptides. The UGT1 genetic sequence, including polymorphisms, may be employed for synthesis of a complete UGT1 protein, or polypeptide fragments thereof, particularly fragments corresponding to functional domains; binding sites; etc.; and including fusions of the subject polypeptides to other proteins or parts thereof. For expression, an expression cassette may be employed, providing for a transcriptional and translational initiation region, which may be inducible or constitutive, where the coding region is operably linked under the transcriptional control of the transcriptional initiation region, and a transcriptional and translational termination region. Various transcriptional initiation regions may be employed that are functional in the expression host. The polypeptides may be expressed in prokaryotes or eukaryotes in accordance with conventional ways, depending upon the purpose for expression. Small peptides can also be synthesized in the laboratory.
Substrate. A substrate is a chemical entity that is modified by UGT1 , usually under normal physiological conditions. Although the duration of drug action tends to be shortened by metabolic transformation, drug metabolism is not "detoxification". Frequently the metabolic product has greater biologic activity than the drug itself. In some cases the desirable pharmacologic actions are entirely attributable to metabolites, the administered drugs themselves being inert. Likewise, the toxic side effects of some drugs may be due in whole or in part to metabolic products. Substrates can be either endogenous substrates (e.g., substrates normally found within the natural environment of UGT1 , such as the bilirubin or other endobiotic compound) or exogenous (e.g., substrates that are not normally found within the natural environment of UGT1 , such as ethinyl estradiol or other xenobiotic compound). Exemplary UGT1 substrates (i.e., substrates of wild-type UGT1 and/or UGT1 polypeptides encoded by UGT1 polymorphisms) include, but are not necessarily limited to endobiotics such as bilirubin, bilirubin monoglucoronide, bile acids, steroids, thyroxine, biogenic amines, fat- soluble vitamins, UDPGA, 17β estradiol, estriol, 2-hydroxy-estriol, T4,rT3, and the like; and xenobiotics such as hydroxylated polycyclic aromatic hydrocarbons, heterocyclics, carcinogens, plant metabolites, octyl gallate, ethinylestradiol, anthraflavic acid, quercetin, 1- naphthol, naphthylamines, 4-aminobiphenyl, benzidine, imipamine, BP-3,6-quinol, 5- hydroxy-BP, acetaminophen, vanillin, naproxen, 4-methylumbelliferone, monohalogenated phenols, propofol, 4t-pentylphenol, 4-hydroxybiphenyl, carvacrol, emodin, galangin, bulky phenols, carboxylic acids, 5-hydroxy 2AAF, 8-hydroxy 2AAF, and the like. Table 1 provides a summary of the major endobiotic and xenobiotic substrates, as well as exemplary non- substrates, of four UGT1 isoenzymes (UGT1*1 (same as UGT1 A), UGT1*4 (same as UGT1D), UGT1*6 (same as UGT1F), and UGT1*02 (same as UGT1G) (see Burchell et al. 91995) Life Sci. 57:1819-31).
Table 1 Substrate Specificity of Human Liver UGT1 Isoenz mes
Figure imgf000011_0001
Modifier. A modifier is a chemical agent that modulates the action of UGT1 , either through altering its enzymatic activity (enzymatic modifier) or through modulation of expression (expression modifier, e.g., by affecting transcription or translation). In some cases the modifier may also be a substrate. For example, the UGT1 gene contains an electrophile responsive element (USPN 5,589,504); thus, compounds such as metabolites of planar aromatic compounds and phenolic antioxidants, as well as reactive oxygen species including peroxides would be expression modifiers via their effect on the electrophile responsive element. Endogenous and exogenous inducers that are capable of inducing particular UGT activities include phenobarbital, dioxin, peroxisome proliferators, rifamicin, oral contraceptive drug, carbamazepine, cigarette smoke, cabbage, brussel sprouts, polycyclic/aromatic hydrocarbons, and derivatives of indole 3-carbonil (see Burchell et al. (1995), supra, Parkinson In: "Biotransformation of Xenobiotics." Chapter 6, Casarett & Doull's Toxicology. 5th Ed., C. Klaassen, ed.)).
Pharmacokinetic parameters. Pharmacokinetic parameters provide fundamental data for designing safe and effective dosage regimens. A drug's volume of distribution, clearance, and the derived parameter, half-life, are particularly important, as they determine the degree of fluctuation between a maximum and minimum plasma concentration during a dosage interval, the magnitude of steady state concentration and the time to reach steady state plasma concentration upon chronic dosing. Parameters derived from in vivo drug administration are useful in determining the clinical effect of a particular UGT1 genotype. Expression assay. An assay to determine the effect of a sequence polymorphism on UGT1 expression. Expression assays may be performed in cell-free extracts, or by transforming cells with a suitable vector. Alterations in expression may occur in the basal level that is expressed in one or more cell types, or in the effect that an expression modifier has on the ability of the gene to be inhibited or induced. Expression levels of a variant alleles are compared by various methods known in the art. Methods for determining promoter or enhancer strength include quantitation of the expressed natural protein; insertion of the variant control element into a vector with a reporter gene such as β-galactosidase, luciferase, chloramphenicol acetyltransferase, etc. that provides for convenient quantitation; and the like.
Gel shift or electrophoretic mobility shift assay provides a simple and rapid method for detecting DNA-binding proteins (Ausubel, F.M. et al. (1989) In: Current Protocols in Molecular Biology, Vol. 2, John Wiley and Sons, New York). This method has been used widely in the study of sequence-specific DNA-binding proteins, such as transcription factors. The assay is based on the observation that complexes of protein and DNA migrate through a nondenaturing polyacrylamide gel more slowly than free DNA fragments or double-stranded oligonucleotides. The gel shift assay is performed by incubating a purified protein, or a complex mixture of proteins (such as nuclear or cell extract preparations), with an end-labeled DNA fragment containing the putative protein binding site. The reaction products are then analyzed on a nondenaturing polyacrylamide gel. The specificity of the DNA-binding protein for the putative binding site is established by competition experiments using DNA fragments or oligonucleotides containing a binding site for the protein of interest, or other unrelated DNA sequences. Expression assays can be used to detect differences in expression of polymorphisms with respect to tissue specificity, expression level, or expression in response to exposure to various substrates, and/or timing of expression during development. For example, since UGT1A and UGT1E are expressed in liver, UGT1A and UGT1 E polymorphisms could be evaluated for expression in tissues other than liver, or expression in liver tissue relative to a reference UGT1A or UGT1E polypeptide. Similarly, expression of polymorphisms in UGT1F, which is normally expressed in liver, kidney and skin, could be assayed in each of these tissues and the relative levels of expression compared to a reference UGT1 F polypeptide. Substrate screening assay. Substrate screening assays are used to determine the metabolic activity of a UGT1 protein or peptide fragment on a substrate. Many suitable assays are known in the art, including the use of primary or cultured cells, genetically modified cells (e.g., where DNA encoding the UGT1 polymorphism to be studied is introduced into the cell within an artificial construct), cell-free systems, e.g. microsomal preparations or recombinantly produced enzymes in a suitable buffer, or in animals, including human clinical trials (see, e.g., Burchell et al. (1995) Life Sci. 57:1819-1831. specifically incorporated herein by reference. Where genetically modified cells are used, since most cell lines do not express UGT1 activity (liver cells lines being the exception), introduction of artificial construct for expression of the UGT1 polymorphism into many human and non-human cell lines does not require additional modification of the host to inactivate endogenous UGT1 expression/activity. Clinical trials may monitor serum, urine, etc. levels of the substrate or its metabolite(s).
Typically a candidate substrate is input into the assay system, and the conversion to a metabolite is measured over time. The choice of detection system is determined by the substrate and the specific assay parameters. Assays are conventionally run, and will include negative and positive controls, varying concentrations of substrate and enzyme, etc.
Genotyping: UGT1 genotyping is performed by DNA or RNA sequence and/or hybridization analysis of any convenient sample from a patient, e.g. biopsy material, blood sample (serum, plasma, etc.), buccal cell sample, etc. A nucleic acid sample from an individual is analyzed for the presence of polymorphisms in UGT1 , particularly those that affect the activity or expression of UGT1. Specific sequences of interest include any polymorphism that leads to changes in basal expression in one or more tissues, to changes in the modulation of UGT1 expression by modifiers, or alterations in UGT1 substrate specificity and/or activity. Linkage Analysis: Diagnostic screening may be performed for polymorphisms that are genetically linked to a phenotypic variant in UGT1 activity or expression, particularly through the use of microsatellite markers or single nucleotide polymorphisms (SNP). The microsatellite or SNP polymorphism itself may not phenotypically expressed, but is linked to sequences that result in altered activity or expression. Two polymorphic variants may be in linkage disequilibrium, i.e. where alleles show non-random associations between genes even though individual loci are in Hardy-Weinberg equilibrium.
Linkage analysis may be performed alone, or in combination with direct detection of phenotypically evident polymorphisms. The use of microsatellite markers for genotyping is well documented. For examples, see Mansfield et al. (1994) Genomics 24:225-233; and Ziegle et al. (1992) Genomics 14:1026-1031. The use of SNPs for genotyping is illustrated in Underhill et al. (1996) Proc Natl Acad Sci U S A 93:196-200.
Transgenic animals. The subject nucleic acids can be used to generate genetically modified non-human animals or site specific gene modifications in cell lines. The term "transgenic" is intended to encompass genetically modified animals having a deletion or other knock-out of UGT1 gene activity, having an exogenous UGT1 gene that is stably transmitted in the host cells, or having an exogenous UGT1 promoter operably linked to a reporter gene. Transgenic animals may be made through homologous recombination, where the UGT1 locus is altered. Alternatively, a nucleic acid construct is randomly integrated into the genome. Vectors for stable integration include plasmids, retroviruses and other animal viruses, YACs, and the like. Of interest are transgenic mammals, e.g. cows, pigs, goats, horses, etc., and particularly rodents, e.g. rats, mice, etc.
Genetically Modified Cells. Primary or cloned cells and cell lines are modified by the introduction of vectors comprising UGT1 gene polymorphisms. The gene may comprise one or more variant sequences, preferably a haplotype of commonly occurring combinations. In one embodiment of the invention, a panel of two or more genetically modified cell lines, each cell line comprising a UGT2B4 polymorphism, are provided for substrate and/or expression assays. The panel may further comprise cells genetically modified with other genetic sequences, including polymorphisms, particularly other sequences of interest for pharmacogenetic screening, e.g. UGT1 , other UGT2 sequences, cytochrome oxidase polymorphisms, etc.
Vectors useful for introduction of the gene include plasmids and viral vectors, e.g. retroviral-based vectors, adenovirus vectors, etc. that are maintained transiently or stably in mammalian cells. A wide variety of vectors can be employed for transfection and/or integration of the gene into the genome of the cells. Alternatively, micro-injection may be employed, fusion, or the like for introduction of genes into a suitable host cell.
Genotyping Methods The effect of a polymorphism in the UGT1 gene sequence on the response to a particular substrate or modifier of UGT1 is determined by in vitro or in vivo assays. Such assays may include monitoring the metabolism of a substrate during clinical trials to determine the UGT1 enzymatic activity, specificity or expression level. Generally, in vitro assays are useful in determining the direct effect of a particular polymorphism, while clinical studies will also detect an enzyme phenotype that is genetically linked to a polymorphism. The response of an individual to the substrate or modifier can then be predicted by determining the UGT1 genotype, with respect to the polymorphism. Where there is a differential distribution of a polymorphism by racial background, guidelines for drug administration can be generally tailored to a particular ethnic group. The basal expression level in different tissue may be determined by analysis of tissue samples from individuals typed for the presence or absence of a specific polymorphism. Any convenient method may be use, e.g. ELISA, RIA, etc. for protein quantitation, northern blot or other hybridization analysis, quantitative RT-PCR, etc. for mRNA quantitation. The tissue specific expression is correlated with the genotype. The alteration of UGT1 expression in response to a modifier is determined by administering or combining the candidate modifier with an expression system, e.g. animal, cell, in vitro transcription assay, etc. The effect of the modifier on UGT1 transcription and/or steady state mRNA levels is determined. As with the basal expression levels, tissue specific interactions are of interest. Correlations are made between the ability of an expression modifier to affect UGT1 activity, and the presence of the provided polymorphisms. A panel of different modifiers, cell types, etc. may be screened in order to determine the effect under a number of different conditions.
A UGT1 polymorphism that results in altered enzyme activity or specificity is determined by a variety of assays known in the art. The enzyme may be tested for metabolism of a substrate in vitro, for example in defined buffer, or in cell or subcellular lysates, where the ability of a substrate to be metabolized by UGT1 under physiologic conditions is determined. Where there are not significant issues of toxicity from the substrate or metabolite(s), in vivo human trials may be utilized, as previously described.
The genotype of an individual is determined with respect to the provided UGT1 gene polymorphisms. The genotype is useful for determining the presence of a phenotypically evident polymorphism, and for determining the linkage of a polymorphism to phenotypic change.
A number of methods are available for analyzing nucleic acids for the presence of a specific sequence. Where large amounts of DNA are available, genomic DNA is used directly. Alternatively, the region of interest is cloned into a suitable vector and grown in sufficient quantity for analysis. The nucleic acid may be amplified by conventional techniques, such as the polymerase chain reaction (PCR), to provide sufficient amounts for analysis. The use of the polymerase chain reaction is described in Saiki et al. (1985) Science 230:1350-1354, and a review of current techniques may be found in Sambrook et al. Molecular Cloning: A Laboratory Manual, CSH Press 1989, pp.14.2-14.33.
Amplification may be used to determine whether a polymorphism is present, by using a primer that is specific for the polymorphism. Alternatively, various methods are known in the art that utilize oligonucleotide ligation as a means of detecting polymorphisms, for examples see Riley et al. (1990) Nucleic Acids Res 18:2887-2890; and Delahunty et al. (1996) Am J Hum Genet 58:1239-1246.
A detectable label may be included in an amplification reaction. Suitable labels include fluorochromes, e.g. fluorescein isothiocyanate (FITC), rhodamine, Texas Red, phycoerythrin, allophycocyanin, 6-carboxyfluorescein (6-FAM), 2',7'-dimethoxy-4',5'- dichloro-6-carboxyfluorescein (JOE), 6-carboxy-X-rhodamine (ROX), 6-carboxy-2',4',7',4,7- hexachlorofluorescein (HEX), 5-carboxyfluorescein (5-FAM) or N,N,N',N'-tetramethyl-6- carboxyrhodamine (TAMRA), radioactive labels, e.g. 32P, 35S, 3H; etc. The label may be a two stage system, where the amplified DNA is conjugated to biotin, haptens, etc. having a high affinity binding partner, e.g. avidin, specific antibodies, etc., where the binding partner is conjugated to a detectable label. The label may be conjugated to one or both of the primers. Alternatively, the pool of nucleotides used in the amplification is labeled, so as to incorporate the label into the amplification product.
The sample nucleic acid, e.g. amplified or cloned fragment, is analyzed by one of a number of methods known in the art. The nucleic acid may be sequenced by dideoxy or other methods. Hybridization with the variant sequence may also be used to determine its presence, by Southern blots, dot blots, etc. The hybridization pattern of a control and variant sequence to an array of oligonucleotide probes immobilized on a solid support, as described in U.S. 5,445,934, or in WO95/35505, may also be used as a means of detecting the presence of variant sequences. Single strand conformational polymorphism (SSCP) analysis, denaturing gradient gel electrophoresis (DGGE), mismatch cleavage detection, and heteroduplex analysis in gel matrices are used to detect conformational changes created by DNA sequence variation as alterations in electrophoretic mobility. Alternatively, where a polymorphism creates or destroys a recognition site for a restriction endonuclease (restriction fragment length polymorphism, RFLP), the sample is digested with that endonuclease, and the products size fractionated to determine whether the fragment was digested. Fractionation is performed by gel or capillary electrophoresis, particularly acrylamide or agarose gels.
In one embodiment of the invention, an array of oligonucleotides are provided, where discrete positions on the array are complementary to one or more of the provided polymorphic sequences, e.g. oligonucleotides of at least 12 nt, frequently 20 nt, or larger, and including the sequence flanking the polymorphic position. Such an array may comprise a series of oligonucleotides, each of which can specifically hybridize to a different polymorphism. For examples of arrays, see Hacia et al. (1996) Nat Genet 14:441-447 and DeRisi et al. (1996) Nat Genet 14:457-460. Arrays of interest may further comprise sequences, including polymorphisms, of other genetic sequences, particularly other sequences of interest for pharmacogenetic screening, e.g. UGT1 , other UGT2 sequences, cytochrome oxidase polymorphisms, etc.
The genotype information is used to predict the response of the individual to a particular UGT1 substrate or modifier. Where an expression modifier inhibits UGT1 expression, then drugs that are a UGT1 substrate will be metabolized more slowly if the modifier is co-administered. Where an expression modifier induces UGT1 expression, a co-administered substrate will typically be metabolized more rapidly. Similarly, changes in UGT1 activity will affect the metabolism of an administered drug. The pharmacokinetic effect of the interaction will depend on the metabolite that is produced, e.g. a prodrug is metabolized to an active form, a drug is metabolized to an inactive form, an environmental compound is metabolized to a toxin, etc. Consideration is given to the route of administration, drug-drug interactions, drug dosage, etc.
EXAMPLES The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the subject invention, and are not intended to limit the scope of what is regarded as the invention. Efforts have been made to ensure accuracy with respect to the numbers used (e.g., amounts, temperature, concentrations, etc.) but some experimental errors and deviations should be allowed for. Unless otherwise indicated, parts are parts by weight, molecular weight is average molecular weight, temperature is in degrees centigrade; and pressure is at or near atmospheric. EXAMPLE: IDENTIFICATION OF UGT1 POLYMORPHISMS MATERIALS AND METHODS
DNA Samples. Blood specimens were collected from approximately 48 individuals after obtaining informed consent. All samples were stripped of personal identifiers to maintain confidentiality. Genomic DNA was isolated from these samples using standard techniques. Genomic DNA was stored either as a concentrated solution, or in a dried form in microtiter plates.
PCR amplifications. The primers used to amplify all exons are shown in Table 2, and were designed with NBI's Oligo version 5.0 program. Table 2. PCR Primers. (Ex = Exon)
PRIMARY PCR AMPLIFICATION
EX FORWARD PRIMER REVERSE PRIMER
1A TGGTGTATCGATTGGTTTT (SEQ ID CATATATCTGGGGCTAGTTAATC (SEQ ID NO: 19) NO:20)
1C ACAAGGTAATTAAGATGAAGAAAGCA ACCTGAGATAGTGGCTTCCT (SEQ ID (SEQ ID N0.-21) N0:22)
ID TTTGTCTTCCAATTACATGC (SEQ ID AGTAGATATGGAAGCACTTGTAAG (SEQ NO:23) ID N0:24)
IE TCTCAGTGACAAGGTAATTAAGAC(SE CATTGATTGGATAAAGGCA (SEQ ID NO:26) Q ID N0:25)
IF AATTTGGGTTCTTACATATCAA(SEQ ID GAGTGAGGGAGGACAGAG (SEQ ID NO:28) N0:27)
IG ATAAGTACACGCCTTCTTTTG (SEQ ID GCTGCTTTATACAATTTGCTAC (SEQ ID N0:29) NO:30)
IH CGCCTACGTATCATAGCAGTTA(SEQ ID GGAAAGAAATTTGAAATGCAAC (SEQ ID N0:31) NO:32)
U TCTTTCCGCCTACTGTATCA (SEQ ID TTCAAGAAGGGCAGTTTTAT (SEQ ID N0:33) NO:34)
SECONDARY PCR AMPLIFICATION
EX FORWARD PRIMER REVERSE PRIMER
1 A CTCTGGCAGGAGCAAAG (SEQ ID ATACACACCTGGGATAGTGG (SEQ ID
NO:35) NO:36) 1C GGTAATTAAGATGAAGAAAGCA(SEQ CTGAGATAGTGGCTTCCTG (SEQ IDNO:38)
ID NO:37) ID GTGGCTCAATGACAAGG (SEQ ID ATATGGAAGCACTTGTAAGTAAA(SEQ ID
NO:39) NO:40)
IE TTAAGACGAAGGAAACAATTCT(SEQ ACCTGAGATAGTGGCTTCC (SEQ ID NO:42) ID NO:41)
1 F ATCA AAGGGTAA A ATTC AGA (SEQ ID GGCAGTCCAAAAGAAATA (SEQ ID NO:44) NO:43)
IG TTTTGAGGGCAGGTTCTA (SEQ ID AATGGGACAAATGTAAATGATA (SEQ ID NO:45) NO:46) IH TTCTCTCATGGCTCGCA (SEQ ID NO:47) ATGTCAAATCACAATTCAGTAAGG (SEQ ID NO:48)
1 J CCGCCTACTGTATCATAGCA (SEQ ID CAACGAAATGTCAAATCACAG (SEQ ID NO:49) NO:50) Publicly available genomic sequences were used as references. Twenty-five nanograms of genomic DNA were amplified in the primary amplifications using the Perkin Elmer GeneAmp PCR kit according to the manufacturer's instructions in 25 μl reactions with AmpliTaq Gold DNA polymerase. Reactions contained 25 mM MgCI2 and 0.2 μM of each primer. Thermal cycling was performed using a GeneAmp PCR System 9600 PCR machine (Perkin Elmer), utilizing a touch-down PCR protocol. The protocol, unless indicated otherwise in Table 3, consisted of an initial incubation of 95°C for 10 min, followed by eight cycles of 95°C for 20 sec, 66°C (minus 1 °C per cycle) for 15 sec, 72«C for 2 min, and twenty-seven cycles of 95°C for 20 sec, 54°C for 15 sec, 72°C for 2 min, and one final extension step of 72°C for 10 min.
For the secondary PCR reactions, one microliter of each primary PCR reaction was re- amplified using the secondary PCR primers, also listed in Table 2. The thermal cycling profile that was used for the primary PCR for an exon was used for the secondary PCR.
Table 3. Cycling Profile Modifications
Exon Primary PCR Secondary PCR
IE Touch-Down PCR step: 8 cycles same as Primary PCR
64 C (minus 1 C per cycle), for 15 sec
Total Number of cycles: 35
IF Touch-Down PCR step: 10 cycles same as Primary PCR 64 C (minus 1 C per cycle), for 15 sec Total Number of cycles: 35
IG Touch-Down PCR step: 7 cycles same as Primary PCR
64 C (minus 1 C per cycle), for 15 sec
Total Number of cycles: 35
IH Touch-Down PCR step: 10 cycles same as Primary PCR 66 C (minus 1 C per cycle), for 15 sec Total Number of cycles: 35
DNA sequencing. PCR products from 48 individuals, approximately one-third representing each of the 3 major racial groups (see above), were prepared for sequencing by treating 8 μL of each PCR product with 0.15 μL of exonuclease I (1.5 U/reaction), 0.3 μL of Shrimp Alkaline Phosphatase (0.3 U/reaction), q.s. to 10 μL with MilliQ water, and incubated at 37°C for 15 min, followed by 72°C for 15 min. Cycle sequencing was performed on the GeneAmp PCR System 9600 PCR machine (Perkin Elmer) using the ABI Prism dRhodamine Terminator Cycle Sequencing Ready Reaction Kit according to the manufacturer's directions, with the following changes: (1) 2 μL of dRhodamine terminator premix, instead of 8 μL; and (2) 10% (v/v) Dimethylsulfoxide was added to each individual nucleotide. The oligonucleotide primers (unlabeled), at 3 picomoles per reaction, used for the sequencing reactions are listed in Table 4. Sequencing reactions, with a final volume of 5 μL, were subjected to 30 cycles at 96°C for 20 sec, 50°C for 5 sec, and 60°C for 4 min, followed by ethanol precipitation. After decanting the ethanol, samples were evaporated to dryness using a SpeedVac for roughly 15 min and were resuspended in 2 μl of loading buffer (5:1 deionized formamide:50 mM EDTA pH 8.0). The samples were then, heated to 94°C for 2 min, and electrophoresed through 5.25% polyacrylamide/6M urea gels in an ABI Prism 377 DNA Sequencer according to the manufacturer's instructions for sequence determination. All sequences were determined from both the 5' and 3' (sense and antisense) direction.
Of the forty-eight samples, 38 polymorphisms were identified. The polymorphisms are described in Table 5 below.
Table 4. Sequencing Primers (No. = Polymorphism No.) No. FORWARD PRIMER REVERSE PRIMER
1 CTCTGGCAGGAGCAAAG (SEQ IDN0:51) ACAGTGGGCAGAGACAG (SEQ ID
NO:52)
2 GTGGTTTATTCCCCGTAT (SEQ ID N0:53) ATACACACCTGGGATAGTGG (SEQ ID
N0:54) 3-5 GGTAATTAAGATGAAGAAAGCA (SEQ GAAATGGCATAGGTTGTC (SEQ ID
ID NO:55) N0:56)
6 GGCCACACTCAACTGTA (SEQ ID NO:57) CTCAAAAAAAACACAGTAGG (SEQ ID
N0:58) 7,8 ACTTTTTCTGCCCCTTAT (SEQ ID N0:59) ATATGGAAGCACTTGTAAGTAAA (SEQ
ID NO:60) 9-12 TTAAGACGAAGGAAACAATTCT (SEQ ID AATGGCATACGTTGTCA (SEQ ID N0:62) N0:61)
13,14 AGAATGGCAATTATGAACA (SEQ ID TGTGTGCCCTTAAAGTCT (SEQ ID NO:63) NO:64) 15-17 AGAATGGCAATTATGAACA (SEQ ID ACCTGAGATAGTGGCTTCC (SEQ ID
NO:65) NO:66)
18-24 CTCTGGC T CTGTCCTAC* (SEQ ID ACCTGAGATAGTGGCTTCC (SEQ ID NO:67) NO:68)
25 ATCAAAGGGTAAAATTCAGA (SEQ ID CAGCAGCTTGTCACCTAC (SEQ ID NO:69) NO:70)
26 AATTTGCTTTTGAAAGAATC (SEQ ID GGTAGGCCCAAATACTCA (SEQ ID NO:71) NO:72)
27,28 AATTTGCTTTTGAAAGAATC (SEQ ID GGCAGTCCAAAAGAAATA (SEQ ID NO:73) NO:74) 29,30 TTTTGAGGGCAGGTTCTA (SEQ ID CACCTCTGGCATGACTAC (SEQ ID
NO:75) NO:76)
31,32 TTGCAGGAGTTTGTTTAAT (SEQ ID AATGGGACAAATGTAAATGATA (SEQ NO:77) ID NO:78)
33 CATTGCAGGAGTTTGTTTA (SEQ ID CATCTGAGAACCCTAAGAGA (SEQ ID NO:79) NO:80)
34 AGAAATAGCCTCTGAAATTC (SEQ ID ATGTCAAATCACAATTCAGTAAGG(SEQ NO:81) ID NO:82)
35 CCGCCTACTGTATCATAGCA (SEQ ID GAGTGTACGAGGTTGAGTAAG (SEQ ID NO:83) NO:84) 36-38 ATTTTGCCAGTATCTTTTTAG (SEQ ID CAACGAAATGTCAAATCACAG (SEQ ID
NO:85) NO:86) * Note polymorphism in primer. The reference sequence has a "C" at the highlighted position.
Table 5. UGT1 polymorphisms. Amino acid changes numbered from first methionine for that exon (Ex).
No Ex Ntd AA SEQUENCE (SEQ ID NO:)
1 1A G227A Gly71 Arg CATCAGAGAC A GAGCATTTTACACCTT(SEQ ID NO:87)
2 1A T765C Ser251 Pro GGACCTATTGAGC C.CTGCATCTGTCT (SEQ ID NO:88)
3 1C T75C Trp 11 Arg GGTTCCCCTGCCG C GGCTGGCCACA (SEQ ID NO:89)
4 1C G 125 A GCCCTGGGCTGA A AGTGGAAAG (SEQ ID NO:90)
5 1C T184C Val 47 Ala ATGCGGGAGG C CTTGCGGGAGCT (SEQ ID NO:91)
6 1C A521G CTCTGCGCGGC G GTGCTGGCTAAG (SEQ ID NO:92) 7 7 I IDD G G884488AA TACCCCAGGCC A ATCATGCCCAACA (SEQ ID NO:93)
8 ID C43T Intronic TCCAGGCAAAA T ACTTTTTAAAAAATG(SEQ ID NO:94)
9 IE T187C Leu 48 Ser AGCATGCGGGAGGCCT C GCGGGA (SEQ ID NO:95)
10 IE C 194 G Asp 58 Glu GCGGGA G_CTCCATGCGAGAGG (SEQ ID NO:96)
11 IE T232C Leu 63 Pro TGGTGGTCCTCACCC C GGAGGTGAA (SEQ ID NO:97) 1 122 I IEE A A225577GG TACATCAAAGA G GAGAACTTTTTCAC (SEQ ID
NO:98)
13 IE C468A His 142 Asn TGATCAGGCACCTG A ATGCTACTTCC (SEQ ID NO:99)
14 IE C517G Ala 158 Gly ACCTCTGCG G GGCGGTGCTGG (SEQ ID NO: 100)
15 IE C689T AAGAACATGCT T TACCCTCTGGC (SEQ ID NO:101)
16 IE C701 T CTCTGGC T CTGTCCTACC (SEQ ID NO: 102) 1 177 I IEE C C771177TT TCCTACCTTTGCTATGCTGTTTCT(SEQIDNO:103)
18 IE C786A Leu 248 He TGTCAGTGGTGGAT A TT (SEQ ID NO: 104)
19 IE G789C Val 249 Leu GGTGGAT A TT Cl TCAGC (SEQ ID NO: 105)
20 IE C795T His 251 Tyr TCAGC T ATGCATC (SEQ ID NO: 106)
21 IE T803C Ser253Phe GCATC C GTGTGGCTGTTCCGA (SEQ ID NO: 107) 2 222 I IEE G G881199CC G Gllyy 225599 AArrgg TGGCTGTTCCGA CGGGACTT(SEQ ID NO: 108)
23 IE T827C GGGACTT C GTGATGGA (SEQ ID NO: 109)
24 IE T836C GTGATGGA C TACCCCAGGCCGAT (SEQ ID NO:l 10)
25 IF T161G Ser 7 Ala CCTGCCTCCTTCGC G.CATTTCAGAG (SEQ ID NO: 111)
26 IF A457G GCGATCATTCCT G ACTGCTCCTCAG (SEQ ID NO:l 12) 2 277 I IFF A A668833GG T Thhrr 118811 AAllaa CCCTGGAGCATG CATTCAGCAG (SEQ ID NO: 113)
28 IF A694C Arg 184 Ser CATTCAGCAG C.AGCCCAGACCCT (SEQ ID NO:l 14)
29 IG T35G TACTTCTTCCAC G TACTATATTA (SEQ ID NO: 115)
30 IG C124A GGCCTCCTTCC A CTATATGTGTGT (SEQ ID NO:l 16)
31 IG T712C Trp 208 Arg GGAGAGAGTA C GGAACCACAT (SEQ ID NO:l 17) 3 322 I IGG G G884466AA TCAATTTGGTT A TTGCGAACTGA (SEQ ID NO: 118)
33 IH G518C Gly 173 Ala CAGGGGAATAG C TTGCCACTAT (SEQ ID NO: 119)
34 IH A765G TGTTGCGAAC G GACTTTGTTTTGG (SEQ ID NO: 120)
35 U G127A TTCACCAGCA A TCGGTGGTGG (SEQ ID NO: 121)
36 U C694T CTAGAAATAGC T TCTGAAATTCTCC (SEQ ID NO: 122) 3 377 1 U J C C 773311 AA L Leeuu 224444 HHee CGGCATATGAT A TCTACAGTCACA (SEQ ID NO: 123)
38 U T761 C Arg 254 Stop TCAATTTGGTTG C TGCGAACAGGAC (SEQ ID NO: 124) The asterisk associated with the second nucleotide residue in polymorphism no.19 is in the sequence surrounding the newly discovered polymorphism at residue 789
(nucleotide change from C at residue 786 to A). All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention.
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

Claims

What is Claimed is:
1. An isolated nucleic acid molecule comprising a UGT1 sequence polymorphism of SEQ ID NOS: 87-124, as part of other than a naturally occurring chromosome.
2. A nucleic acid probe for detection of UGT1 locus polymorphisms, comprising a polymorphic sequence of SEQ ID NOS:87-124.
3. A nucleic acid probe according to Claim 2, wherein said probe is conjugated to a detectable marker.
4. An array of oligonucleotides comprising: two or more probes for detection of UGT1 locus polymorphisms, said probes comprising at least one form of a polymorphic sequences of SEQ ID NOS:87-124.
5. A method for detecting in an individual a polymorphism in UGT1 metabolism of a substrate, the method comprising: analyzing the genome of said individual for the presence of at least one UGT1 polymorphism of SEQ ID NOS:87-124; wherein the presence of said predisposing polymorphism is indicative of an alteration in UGT1 expression or activity.
6. A method according to Claim 5, wherein said analyzing step comprises detection of specific binding between the genomic DNA of said individual with an array of oligonucleotides comprising: two or more probes for detection of UGT1 locus polymorphisms, said probes comprising at least one form of a polymorphic sequence of SEQ ID NOS:87-124.
7. A method according to Claim 5, wherein said alteration in UGT1 expression is tissue specific.
8. A method according to Claim 5, wherein said alteration in UGT1 expression is in response to a UGT1 modifier.
9. A method according to Claim 8, wherein said modifier induces UGT1 expression.
10. A method according to Claim 8, wherein said modifier inhibits UGT1 expression.
PCT/US1999/009702 1998-05-07 1999-05-04 Genotyping the human udp-glucuronosyltransferase 1 (ugt1) gene WO1999057322A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP99921638A EP1084271A2 (en) 1998-05-07 1999-05-04 Genotyping the human udp-glucuronosyltransferase 1 (ugt1) gene
AU38792/99A AU3879299A (en) 1998-05-07 1999-05-04 Genotyping the human udp-glucuronosyltransferase 1 (ugt1) gene

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US8480798P 1998-05-07 1998-05-07
US60/084,807 1998-05-07

Publications (2)

Publication Number Publication Date
WO1999057322A2 true WO1999057322A2 (en) 1999-11-11
WO1999057322A3 WO1999057322A3 (en) 2000-01-13

Family

ID=22187334

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/009702 WO1999057322A2 (en) 1998-05-07 1999-05-04 Genotyping the human udp-glucuronosyltransferase 1 (ugt1) gene

Country Status (4)

Country Link
US (2) US6479236B2 (en)
EP (1) EP1084271A2 (en)
AU (1) AU3879299A (en)
WO (1) WO1999057322A2 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001079230A2 (en) * 2000-04-18 2001-10-25 Genaissance Pharmaceuticals, Inc. Haplotypes of the ugt1a1 gene
WO2002053770A2 (en) 2001-01-05 2002-07-11 Medizinische Hochschule Hannover Method for the prediction of the risk potential for cancerous diseases and inflammatory intestinal diseases and corresponding tests
WO2002077214A1 (en) * 2001-03-22 2002-10-03 Pe Corporation (Ny) Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
WO2002006523A3 (en) * 2000-07-14 2003-04-17 Hoffmann La Roche Method for detecting pre-disposition to hepatotoxicity
EP1352970A1 (en) * 2000-12-12 2003-10-15 Nagoya Industrial Science Research Institute Method of estimating the risk of expression of adverse drug reaction caused by the administration of a compound, which is either metabloized per se by ugt1a1 or whose intermediate is metabolized by the enzyme
US6713257B2 (en) 2000-08-25 2004-03-30 Rosetta Inpharmatics Llc Gene discovery using microarrays
WO2006027182A1 (en) * 2004-09-06 2006-03-16 Medizinische Hochschule Hannover Methods and its kits based on ugt1a7 promoter polymorphism
EP2071025A1 (en) * 2006-11-30 2009-06-17 Arkray, Inc. Primer set for amplification of ugt1a1 gene, reagent for amplification of ugt1a1 gene comprising the same, and use of the same
WO2010025952A1 (en) * 2008-09-05 2010-03-11 Medizinische Hochschule Hannover Method for genotyping ugt1a1*28 promoter region variant and systems useful therefor
US7807350B2 (en) 2003-05-30 2010-10-05 The University Of Chicago Methods for predicting irinotecan toxicity
US7807447B1 (en) 2000-08-25 2010-10-05 Merck Sharp & Dohme Corp. Compositions and methods for exon profiling

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6395481B1 (en) * 1999-02-16 2002-05-28 Arch Development Corp. Methods for detection of promoter polymorphism in a UGT gene promoter
AU2002240066A1 (en) * 2001-01-26 2002-08-06 University Of Chicago Determination of ugt2b7 gene polymorphisms for predicting ugt2b7 substrate toxicity and for optimising drug dosage
AU2003257344A1 (en) * 2002-09-20 2004-04-08 Universite Laval Method for determining the predisposition of patients to toxicity or lack of efficacy of a drug
US20040203034A1 (en) * 2003-01-03 2004-10-14 The University Of Chicago Optimization of cancer treatment with irinotecan
US20090247475A1 (en) * 2004-03-05 2009-10-01 The Regents Of The University Of California Methods and compositions relating to pharmacogenetics of different gene variants in the context of irinotecan-based therapies
US20080313748A1 (en) * 2004-09-03 2008-12-18 Regents Of The University Of California Transgenic Non-Human Animals Comprising the Human Udp-Glucuronosyltransferase 1A (Ugt1a) Gene Locus and Methods of Using Them
KR100849166B1 (en) 2005-02-07 2008-07-30 인제대학교 산학협력단 - 14 Single nucleotide polymorphic of UDP-glucuronosyltransferase 1A4 and use thereof
EP1790343A1 (en) * 2005-11-11 2007-05-30 Emotional Brain B.V. Pharmaceuticals formulations and uses thereof in the treatment of female sexual dysfunction
US20100159454A1 (en) * 2006-09-11 2010-06-24 Inje University Industry Academic Cooperation Foundation HtSNPs FOR DETERMINING A GENOTYPE OF CYTOCHROME P450 1A2, 2A6 AND 2D6, PXR AND UDP-GLUCURONOSYLTRANSFERASE 1A GENE AND MULTIPLEX GENOTYPING METHODS USING THEREOF
WO2008032921A1 (en) * 2006-09-11 2008-03-20 Inje University Industry-Academic Cooperation Foundation Htsnps for determining a genotype of cytochrome p450 1a2, 2a6 and 2d6, pxr and udp-glucuronosyltransferase 1a gene and multiplex genotyping methods using thereof
DE102009015978A1 (en) * 2009-03-26 2010-09-30 Robert Bosch Gesellschaft Für Medizinische Forschung Mbh A method for determining the predisposition of a patient to altered biotransformation and the development of adverse drug reactions in a treatment of the patient with atorvastatin

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992012987A1 (en) * 1991-01-10 1992-08-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services The genetic locus ugt1 and a mutation therein
WO1997032042A2 (en) * 1996-03-01 1997-09-04 The University Court Of The University Of Dundee Drug trial assay system

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU7097494A (en) * 1993-06-01 1994-12-20 Targeted Genetics Corporation Envelope fusion vectors for use in gene delivery

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992012987A1 (en) * 1991-01-10 1992-08-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services The genetic locus ugt1 and a mutation therein
WO1997032042A2 (en) * 1996-03-01 1997-09-04 The University Court Of The University Of Dundee Drug trial assay system

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BOSMA P J ET AL: "THE GENETIC BASIS OF THE REDUCED EXPRESSION OF BILIRUBIN UDP -GLUCURONOSYLTRANSFERASE 1 IN GILBERT'S SYNDROME" THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 333, no. 18, 2 November 1995, pages 1171-1175, XP002040437 *
CIOTTI ET AL.: "GENETIC POLYMORPHISM IN THE HUMAN UGT1A6 (PLANAR PHENOL) UDP-GLUCURONOSYLTRANSFERASE: PHARMACOLOGICAL IMPLICATIONS" PHARMACOGENETICS, vol. 7, 1997, pages 485-495, XP002113205 *
MACKENZIE ET AL.: "THE UDP GLYCOSYLTRANSFERASE GENE SUPERFAMILY: RECOMMENDED NOMENCLATURE UPDATE BASED ON EVOLUTIONARY DIVERGENCE" PHARMACOGENETICS, vol. 7, 1997, pages 255-269, XP002113206 cited in the application *
MONAGHAN ET AL.: "GENETIC VARIATION IN BILIRUBIN UDP-GLUCURONOSYLTRANSFERASE GENE PROMOTER AND GILBERT'S SYNDROME" LANCET, vol. 347, 1996, pages 578-581, XP002040439 *
OWENS I S ET AL: "THE NOVEL BILIRUBIN/PHENOL UDP-GLUCURONOSYLTRANSFERASE UGT1 GENE LOCUS: IMPLICATIONS FOR MULTIPLE NONHEMOLYTIC FAMILIAL HYPERBILIRUBINEMIA PHENOTYPES" PHARMACOGENETICS, vol. 2, no. 3, 1 June 1992, pages 93-108, XP002040438 *
RITTER ET AL.: "A NOVEL COMPLEX LOCUS UGT1 ENCODES HUMAN BILIRUBIN, PHENOL, AND OTHER UDP-GLUCURONOSYLTRANSFERASE ISOZYMES WITH IDENTICAL CARBOXYL TERMINI" J.BIOL.CHEM., vol. 267, no. 5, 1992, pages 3257-3261, XP002113204 *

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001079230A2 (en) * 2000-04-18 2001-10-25 Genaissance Pharmaceuticals, Inc. Haplotypes of the ugt1a1 gene
WO2001079230A3 (en) * 2000-04-18 2002-03-21 Genaissance Pharmaceuticals Haplotypes of the ugt1a1 gene
WO2002006523A3 (en) * 2000-07-14 2003-04-17 Hoffmann La Roche Method for detecting pre-disposition to hepatotoxicity
US6713257B2 (en) 2000-08-25 2004-03-30 Rosetta Inpharmatics Llc Gene discovery using microarrays
US7807447B1 (en) 2000-08-25 2010-10-05 Merck Sharp & Dohme Corp. Compositions and methods for exon profiling
EP1352970A1 (en) * 2000-12-12 2003-10-15 Nagoya Industrial Science Research Institute Method of estimating the risk of expression of adverse drug reaction caused by the administration of a compound, which is either metabloized per se by ugt1a1 or whose intermediate is metabolized by the enzyme
EP1352970A4 (en) * 2000-12-12 2004-08-25 Inst Nagoya Ind Science Res Method of estimating the risk of expression of adverse drug reaction caused by the administration of a compound, which is either metabloized per se by ugt1a1 or whose intermediate is metabolized by the enzyme
WO2002053770A2 (en) 2001-01-05 2002-07-11 Medizinische Hochschule Hannover Method for the prediction of the risk potential for cancerous diseases and inflammatory intestinal diseases and corresponding tests
WO2002053770A3 (en) * 2001-01-05 2003-07-24 Hannover Med Hochschule Method for the prediction of the risk potential for cancerous diseases and inflammatory intestinal diseases and corresponding tests
US7264928B2 (en) 2001-01-05 2007-09-04 Medizinische Hochschule Hannover Method for the prediction of the risk potential for cancerous diseases and inflammatory intestinal diseases and corresponding tests
WO2002077214A1 (en) * 2001-03-22 2002-10-03 Pe Corporation (Ny) Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
US6713295B2 (en) 2001-03-22 2004-03-30 Applera Corporation Isolated human UDP-glycosyltransferase proteins
US7807350B2 (en) 2003-05-30 2010-10-05 The University Of Chicago Methods for predicting irinotecan toxicity
WO2006027182A1 (en) * 2004-09-06 2006-03-16 Medizinische Hochschule Hannover Methods and its kits based on ugt1a7 promoter polymorphism
EP2071025A4 (en) * 2006-11-30 2009-12-09 Arkray Inc Primer set for amplification of ugt1a1 gene, reagent for amplification of ugt1a1 gene comprising the same, and use of the same
EP2071025A1 (en) * 2006-11-30 2009-06-17 Arkray, Inc. Primer set for amplification of ugt1a1 gene, reagent for amplification of ugt1a1 gene comprising the same, and use of the same
US20110281265A1 (en) * 2006-11-30 2011-11-17 Mitsuharu Hirai Primer set for amplifying UGT1A1 gene, reagent for amplifying UGT1A1 gene containing the same, and the uses thereof
US8357516B2 (en) 2006-11-30 2013-01-22 Arkray, Inc. Primer set for amplification of UGT1A1 gene, reagent for amplification of UGT1A1 gene containing the same, and the uses thereof
US9175272B2 (en) 2006-11-30 2015-11-03 Arkray, Inc. Probes for detection of UGT1A1 gene, reagent containing the same, and the uses thereof
WO2010025952A1 (en) * 2008-09-05 2010-03-11 Medizinische Hochschule Hannover Method for genotyping ugt1a1*28 promoter region variant and systems useful therefor

Also Published As

Publication number Publication date
EP1084271A2 (en) 2001-03-21
WO1999057322A3 (en) 2000-01-13
US20030157517A1 (en) 2003-08-21
US20020061518A1 (en) 2002-05-23
US6479236B2 (en) 2002-11-12
AU3879299A (en) 1999-11-23

Similar Documents

Publication Publication Date Title
US6479236B2 (en) Genotyping the human UDP-glucuronosyltransferase 1 (UGT1) gene
US6432639B1 (en) Isolated CYP3A4 nucleic acid molecules and detection methods
Shield et al. Human catechol O-methyltransferase genetic variation: gene resequencing and functional characterization of variant allozymes
Hassett et al. Human microsomal epoxide hydrolase: genetic poloymorphism and functional expression in vitro of amino acid variants
Thomae et al. Human sulfotransferase SULT2A1 pharmacogenetics: genotype-to-phenotype studies
Hildebrandt et al. Human SULT1A3 pharmacogenetics: gene duplication and functional genomic studies
US6448003B1 (en) Genotyping the human phenol sulfotransferbase 2 gene STP2
Sun et al. A signature of balancing selection in the region upstream to the human UGT2B4 gene and implications for breast cancer risk
US6586175B1 (en) Genotyping the human UDP-glucuronosyltransferase 2B7 (UGT2B7) gene
US6551812B1 (en) Compositions and methods relating to the peroxisomal proliferator activated receptor-α mediated pathway
Egoshi et al. Identification of five novel SLC3A1 (rBAT) gene mutations in Japanese cystinuria
WO2000079003A1 (en) Polymorphisms in the human hmg-coa reductase gene
AU754923B2 (en) Method to determine predisposition to hypertension
US6444427B1 (en) Polymorphisms in a diacylglycerol acyltransferase gene, and methods of use thereof
AU2005264056A1 (en) Novel allelic variant of CYP2C19 associated with drug metabolism
EP1100961A1 (en) Genetic polymorphisms in the human neurokinin 2 receptor gene and their use in diagnosis and treatment of diseases
WO2000039331A1 (en) Single nucleotide polymorphism in a pyruvate dehydrogenase kinase isoenzyme 2 (pdk2) in humans
CA2378665A1 (en) Dna polymorphisms in sterol regulator element binding proteins
WO2002100894A2 (en) Dna encoding a mutant peroxisome proliferator-activated receptor gamma coactivator-1 (pgc-1), detection methods and test kits therefor
WO2004070059A2 (en) Polymorphisms in the esr1 gene and diagnosis of an estrogen-sensitive disorder

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 1999921638

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: KR

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1999921638

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1999921638

Country of ref document: EP