throbber
(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT)
`
`(19) World Intellectual Property Organization
`International Bureau
`
`11111111111111111111111111111111111111111111111111111111111111111111111111111111
`
`(43) International Publication Date
`25 October 2001 (25.10.2001)
`
`PCT
`
`(10) International Publication Number
`WO 01/79554 Al
`
`(51) International Patent Classification7:
`C07H 21/04
`
`C12Q 1/68,
`
`(21) International Application Number: PCT/USOl/12087
`
`(72) Inventor; and
`(75) Inventor/Applicant (jor US only): WOOSLEY, Ray(cid:173)
`mond, L. [US/US]; 4106 18th Road, North Arlington, VA
`22207 (US).
`
`(22) International Filing Date:
`
`13 April2001 (13.04.2001)
`
`(25) Filing Language:
`
`English
`
`(74) Agent: TESKIN, Robin, L.; Pillsbury Winthrop LLP,
`1100 New York Avenue, NW, Washington, DC 20005
`(US).
`
`(26) Publication Language:
`
`English
`
`(81) Designated States (national): CA, JP, US.
`
`(84) Designated States (regional): European patent (AT, BE,
`CH, CY, DE, DK, ES, Fl, FR, GB, GR, IE, IT, LU, MC,
`NL, PT, SE, TR).
`
`Published:
`with international search report
`
`For two-letter codes and other abbreviations .. refer to the "Guid(cid:173)
`ance Notes on Codes and Abbreviations" appearing at the begin(cid:173)
`ning of each regular issue of the PCT Gazette.
`
`(30) Priority Data:
`60/196,916
`
`13 April :woo (13.04.2000) US
`
`(63) Related by continuation (CON) or continuation-in-part
`(CIP) to earlier application:
`us
`Filed on
`
`60/196,916 (CON)
`13 April2000 (13.04.2000)
`
`(71) Applicant
`(for all designated States except US):
`GEORGETOWN UNIVERSITY
`[US/US];
`3900
`Reservior Road, N.W., Washington, DC 20007 (US).
`
`iiiiiiiiiiii
`
`iiiiiiiiiiii
`
`--iiiiiiiiiiii
`iiiiiiiiiiii =
`iiiiiiiiiiii -iiiiiiiiiiii
`!!!!!!! --
`--==
`iiiiiiiiiiii ---==
`
`~
`
`<-------------------------------------------------------
`~ (54) Title: GENETIC DIAGNOSIS FOR QT PROLONGATION RELATED ADVERSE DRUG REACTIONS
`ln
`ln (57) Abstract: The specification is directed to a method of diagnosing whether a subject is predisposed to an adverse reaction to
`~ one or more pharmaceutical agents which may induce a prolonged QT interval or acquired LQTS in that individual. The diagnosis is
`S risk for prolonged QT intervals or Torsades de Pointes (TdP). Genetic screening for determining the predisposition of prolonged
`-... genetic analysis of at least two polymorphisms or mutations which the individual may have, which are associated with an increased
`0 two classes or genes, wherein the genes are (1) LQT genes, (2) altered sensitivity genes (e.g., MiRP 1) or (3) increased exposure
`> genes (e.g.. MDR genes orP450 cytochrome genes). The specification is also directed to compositions and kits for determining such
`~ predispositions to adverse drug reactions.
`
`QT intervals induced by a pharmaceutical agent is performed by identifying genetic polymorphisms or mutations located in at least
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 001
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-1-
`
`GENETIC DIAGNOSIS FOR QT PROLONGATION RELATED
`
`ADVERSE DRUG REACTIONS
`
`CROSS REFERENCE TO RELATED APPLICATION
`
`5
`
`This application claims benefit of priority from U.S. Provisional Patent
`
`Application Serial No. 60/196,916, filed April13, 2000, the entirety of which is
`
`incorporated herein by reference.
`
`FIELD OF THE INVENTION
`
`1 0
`
`The invention relates to methods of determining a predisposition for QT
`
`interval prolongation in a subject after the administration of a pharmaceutical agent or
`
`agents. Compositions and kits for determining said predispositions to the QT interval
`
`prolongation are also described.
`
`15
`
`BACKGROUND OF THE INVENTION
`
`The invention relates to a method of screening a subject for a predisposition to
`
`an adverse drug reaction involving prolonged QT intervals. The genetic screening of
`
`patients for said predisposition focuses on genes associated with QT interval
`
`prolongation, including LQT genes, P-glycoprotein membrane pump proteins (P-gp ),
`
`20 multidrug resistance genes and cytochrome P450-mediated drug metabolism genes.
`
`I.
`
`1.
`
`LOT and Cytochrome P450 Genes and Polymorphisms
`
`LQT Genes
`
`Genes associated with long QT (LQT) syndrome (LQTS) include KVLQTJ
`
`25
`
`(LQTJ), HERG (LQT2), SCN5A (LQT3) and MinK (LQT5). A fifth gene locus exists
`
`on human chromosome 4 (e.g., LQT4). Recently, a sixth gene (LQT6) has been
`
`identified (Wang et al., Ann. Med. 30: 58-65 (1998)). All but LQT3 encode cardiac
`potassium ion (Kl channel proteins; LQT3 encodes a cardiac sodium ion (Na l
`channel protein (Vincent, Annu. Rev. Med. 49: 263-74 (1998)). At least 180
`
`30 mutations have been identified among these genes (Abbott et al., Cell97: 175-87
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 002
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-2-
`
`(1999); Vincent, Annu. Rev. Med. 49: 263-74 (1998); Curran et al., Cell80: 795-803
`
`(1995); Berthet et al., Circulation 99: 1464-70 (1999); Dausse et al., J. Mol. Cell
`
`Cardiol. 28: 1609-15 (1996); Chen et al., J. Biol. Chern. 274: 10113-8 (1999); and
`
`Sanguinetti et al., Proc. Nat!. Acad. Sci. US.A. 93: 2208-12 (1996)). Some ofthese
`
`5 mutations cause altered ion channel function resulting in non-drug induced prolonged
`
`QT intervals and a pro:pensity for Torsades de Paiutes (TdP) (See, e.g., Berthet et al.,
`
`Circulation 99: 1464-70 (1999)). Accordingly, genetic screening can be performed on
`
`subjects suspected of having long QT syndrome, as well as other patients (see, e.g.,
`
`Satler et al., Hum. Genet. 102: 265-72 (1998)). Larson et al., Hum. Mutat. 13: 318-27
`
`10
`
`(1999) reported a high-throughout single strand polymorphism (SSCP) analysis for
`
`detecting point mutations associated with LQTS.
`
`U.S. Patent No. 5,599,673 claims two (e.g., HERG and SCN5A) of the six LQT
`
`genes. Two BERG-related genes have also been claimed (U.S. Patent No. 5,986,081).
`
`futemational PCT Application WO 97/23598 describes a method of assessing a
`
`15
`
`patient's risk for long QT syndrome (LQTS) by screening for genetic mutations in the
`
`MinK gene. However, these patents do not disclose methods of diagnosing a patient's
`
`predisposition to an adverse drug reaction involving elongation of the QT interval due
`
`to mutations in any of the LQT genes.
`
`Drugs have been identified that cause QT interval prolongation, an:d thereby
`
`20
`
`adverse drug reactions. Certain antihistamines, such as terfenadine (e.g., Seldane®)
`
`and astemizole (e.g., Hismanal®), reportedly block potassium channels (Woosley,
`
`Annu. Rev. Pharmacal. Toxicol. 36: 233-52 (1996)) and inhibit the HERG protein,
`
`and thereby were postulated to induce Torsades de Pointes (Wang et al., 1998). All
`
`antiarrhythmic drugs that lengthen repolarization reportedly can cause Torsades de
`
`25
`
`Pointes (Drici et al., Circulation 94: 1471-4 (1996)). Additional non-cardiac and
`
`cardiac drugs capable of inducing QT prolongation including many that were
`
`identified by the inventor were released on March 27, 1998 at the following web site:
`
`www.qtdrugs.org. However, Wei et al., Circulation 92: I-125 (1995) could not
`
`identify HERG or SCN5A gene mutations that were linked to acquired LQTS in
`
`30
`
`patients treated with an anti-arrhythmic agent. To the best knowledge of the inventor,
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 003
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-3-
`
`no one has described diagnosing a predisposition towards an adverse drug or drug(cid:173)
`
`drug reaction whic~ causes QT interval elongation by screening patients for one or
`
`more polymorphisms in one or more LQT genes.
`
`5
`
`1.
`
`Cytochrome P450 Genes
`
`The cytochrome P450 enzymes have also been linked to adverse drug
`
`reactions. CYP2D6 was the first cytochrome P450 isoform found to be genetically
`
`polymorphic in its distribution (Eichelbaum et al., Eur. J. Clin. Pharmacal. 16: 183-7
`
`I
`
`(1979); and Mahgoub et al., Lancet 2: 584-6 (1977)), and it is now clear that-this
`
`10
`
`enzyme metabolizes a large number of drugs (fuaba et al., Can. J. Physiol.
`
`Pharmacal. 73: 331-8 (1995); and Buchert et al., Pharmacogenetics 2: 2-11 (1992)).
`
`At least 30 mutations exist which alter the activity or specificity ofCYP2D6 (Jordan
`
`et al., Endocr. Rev. 20: 253-78 (1999)). These include alleles that contain single point
`
`mutations resulting in no activity (e.g., CYP2D6*4), alleles in which the CYP2D6
`
`15
`
`gene has been deleted (e.g., CYP2D6*5) and alleles in which it has been duplicated
`
`(e.g., CYP2D6*2_n) (Aklillu et al., J. Pharmacal. Exp. Ther. 278: 441-6 (1996)).
`
`There are numerous cytochrome P450 genes which are involved in the
`
`metabolism of drugs and drug metabolites. Several of them include CYP IA2,
`
`CYP2C19, CYP2C9, CYP2D6, CYP2El, CYP3A4, CYP3A5 and CYP3A7. Allelic
`
`20
`
`variations exist amongst these genes. Certain of these allelic variations combine to
`
`produce a poor metabolizer phenotype in 7% of Caucasians, but smaller percentages
`
`of Africans and Asians and the "ultrarapid" phenotype in ~5% of Caucasian and up to
`
`30% Africans. As ethnic-specific alleles for both Asians (Y okoi et al., Pharm. Res.
`
`15: 517-24 (1998)) and Africans (Aklillu et al., J. Pharmacal. Exp. Ther. 278: 441-6
`
`25
`
`(1996); and Oscarson et al., Mol. Pharmacal. 52: 1034-40 (1997)) have been
`
`identified, that may alter the mean activity of the enzymes in these populations (see
`
`Table 1 below), it is also important to test for these alleles in studies of the
`
`relationship between genotype and phenotype.
`
`30
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 004
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-4-
`
`Table 1
`
`Chromosome Distribution of Cytochrome P450 Gene
`
`Chr.
`15
`
`Chr. 10
`Polymorphic
`
`Chr. 10
`Polymorphic
`
`Chr. 22
`
`Chr.lO Chr. 7
`
`3-5%
`Caucasian
`PMs
`
`1-3%
`Caucasian
`PMs
`
`15-20% Asian
`PMs
`
`Polymorphic
`
`5-10%
`Caucasian
`PMs
`
`In fact, due to the metabolic differences, methods have been reported which identify a
`
`5
`
`drug which interacts with the CYP2C19 gene product, S-mephenytoin 4'-hydroxylase
`
`(U.S. Patent No. 5,786,191).
`
`Methods for detecting the presence or absence ofmutations in certain ofthe
`
`cytochrome P450 genes have been described. For example, U.S. Patent No.
`
`5,891,633 relates to a method of identifying mutations in the cytochrome P450 genes
`
`10
`
`CYP2C9 and CYP2A6.
`
`futemational PCT Application WO 95/30772 reportedly describes a CYP2D6
`
`gene polymorphisms involving a 9 bp insertion in exon 9, which is associated with a
`
`slower than normal rate of drug metabolism in individuals bearing it and may be
`
`therefore useful diagnostically. PCR primers have been described for detecting
`
`15 mutations in drug metabolism enzymes, including detection of the debrisoquine
`
`polymorphism, mephenytoin polymorphism and the acetylation polymorphism (U.S.
`
`Patent Nos. 5,648,484 and 5,844,108). Additional mutations have been identified in
`
`CYP2D6 bufuralol-1 '-hydroxylase, including mutations at positions 271, 281, 294,
`
`and 506 whlch result in metabolizer/poor metabolizer phenotypes as described in
`
`20
`
`futemational PCT Application WO 91/10745 and U.S. Patent No. 5,981,174.
`
`Japanese Patent No. 8168400 provides a method of determining mutations in
`
`exons 6 and 7 ofthe CYP2C19 gene. Japanese Patent No. 10014585 describes
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 005
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-5-
`
`primers and methods of detecting a mutation in exon 5 of CYP2C19, which is related
`
`to the abnormal metabolism of diazepam, imipramine, omeprazole and propranolol.
`
`U.S. Patent No. 5,912,120 claims a method of diagnosing a patient having a
`
`deficiency in S-mephenytoin 4'-hydroxylase activity by detecting polymorphisms at
`
`5
`
`nucleotides 681 or 636.
`
`U.S. Patent 5,719,026 provides methods and primers for detecting a
`
`polymorphisms in CYP JA2 and assessing the changes in the drug activity of
`
`theophylline associated with those polymorphisms.
`
`Japanese Patent No. 10286090 reportedly describes methods and primers to
`
`10
`
`detect mutations in CYP2El. These mutations are reported as being useful for
`
`determining the safety margin for drug administration for the treatment or related
`
`diseases.
`
`Despite these teachings and to the best of the inventor's knowledge, no one has
`
`described or suggested that a combination of polymorphisms in LQT and cytochrome
`
`15
`
`P450 genes can induce acquired LQTS in a subject in response to the administration
`
`of a drug or drugs.
`
`C.
`
`P-glycoprotein Pump Genes
`
`P-Glycoprotein Pump (P-gp) in the development of drug-resistant tumor cells
`
`20
`
`has been extensively studied (Lo et al., J. Clin. Pharmacal. 39: 995-1005 (1999)). P(cid:173)
`
`gp is an ATP-dependent drug pump that extrudes a broad range of cytotoxic agents
`
`from the cells end is encoded by a gene called MDR -1, for multi drug resistance (Loo
`
`et al., Biochem. Cell. Bioi. 77: 11-23 (1999); and Robert, Eur. J. Clin. Invest. 29: 536-
`
`45 (1999)). The human P-gp sequence has been described by Chen et al., Cell47:
`
`25
`
`381-9 (1986) and has the GenBank Accession No. M14758.
`
`Its physiological role may be to protect the body from endogenous and
`
`exogenous cytotoxic agents. The protein has clinical importance because it
`
`contributes to the phenomenon of multi drug resistance during chemotherapy (Loo et
`
`al., 1999) and the development of simultaneous resistance to multiple cytotoxic drugs
`
`30
`
`in cancer cells (Ambudkar et al., Annu. Rev. Pharmacal. Toxicol. 39: 361-98 (1999)).
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 006
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-6-
`
`Specifically, the over expression ofthis membrane pump appears to extrude many
`
`xenobiotics out of the tumor cells (Robert, 1999). However, considerable controversy
`
`remains about the mechanism of action of this efflux pump and its function in normal
`
`cells (Ambudkar eta!., 1999).
`
`5
`
`Multidrug resistance (MDR) can be diagnosed in tumors using molecular
`
`biology techniques (e.g., gene expression at the mRNA level), by immunological
`
`techniques (e.g., quantification of the P-glycoprotein itself) or by functional
`
`approaches (e.g., measuring dye exclusion) (Robert, 1999).
`
`Drugs have been developed which reverse or modulate MDR. For example,
`
`10
`
`PSC-833 is a non-immunosuppressive cyclosporin derivative that potently and
`
`specifically inhibits P-gp (Atadja eta!., Cancer Metastasis Rev. 17: 163-8 (1998)).
`
`Also, compounds have been identified which increase or modulate the bioavailability
`
`of pharmaceutical compounds. See, e.g., U.S. Patent Nos. 6,004,927; 5,962,522;
`
`5,916,566; 5,716,928; 5,665,386; and 5,567,592. P-gp activity has been altered by
`
`15
`
`expression of antisense nucleotides specific to MDR-1 (U.S. Patent No. 6,001,991).
`
`Methods and assays have also been developed which assess whether multidrug
`
`resistance has been reversed (U.S. Patent No. 5,403,574).
`
`Mutations have also been identified which alter an agents interaction with P(cid:173)
`
`gp. For instance U.S. Patent No. 5,830,697 discloses single and double mutations
`
`20
`
`(Phe335 and/or 336) which alters the spectrum of cross-reactivity to cytotoxins and
`
`resistance to modulation by cyclosporins. Another mutation at V185G in P-gp
`
`confers increased resistence to colchicine (U.S. Patent No. 5,830,697). P-gp
`
`sensitivity to vinblastine, colchicine, VP16 and adriamycin, common
`chemotherapeutic agents, was up- and down-regulated by altering 61His to another
`
`25
`
`amino acid residue (Taguchi eta!., Biochemist1y 36: 8883-9 (1997)). Moreover,
`
`different drugs interact differently with P-gp and mutated forms ofP-gp, such that one
`
`mutation may influence the activity of one drug and not another (See, e.g., Chen et a!.,
`
`J. Biol. Chem. 272: 5974-82 (1997); Bakos eta!., Biochem. J. 323: 777-83 (1997);
`
`and Gros eta!., Proc. Nat!. Acad. Sci. USA 88: 7289-93 (1991)). However, despite the
`
`30
`
`information regarding the influence such mutations may have on drug activity, no
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 007
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-7-
`
`association has been made linking P-gp by itself, or in combination with another
`
`protein in influencing QT intervals or inducing Torsades de Pointes.
`
`II.
`
`Nucleic Acid Hybridization
`
`5
`
`The capacity of a nucleic acid "probe" molecule to hybridize (i.e., base pair) to
`
`a complementary nucleic acid "target" molecule forms the cornerstone for a wide
`
`array of diagnostic and therapeutic procedures. Hybridization assays are extensively
`
`used in molecular biology and medicine. Methods of performing such hybridization
`
`reactions are disclosed by, for example, SAMBROOK ET AL., MOLECULAR CLONING: A
`
`10
`
`LABORATORY MANUAL (Cold Spring Harbor Laboratory Press, Cold Spring Harbor,
`
`N.Y. (1989)), HAYMES ET AL., NUCLEIC ACID HYBRIDIZATION: A PRACTICAL
`
`APPROACH (IRL Press, Washington, D.C. (1985)) and KELLERET AL., DNA PROBES
`
`(2nd Ed., Stockton Press, New York (1993)).
`
`Many hybridization assays require the immobilization of one component to a
`
`15
`
`solid support. Nagata et al., FEBS Letters 183: 379-82 (1985) described a method for
`
`quantifying DNA which involved binding unknown amounts of cloned DNA to
`
`microtiter wells in the presence of 0.1 M MgCh. A complementary biotinylated probe
`
`was then hybridized to the DNA in each well and the bound probe measured
`
`colorimetrically. Dahlen et al., Mol .. Cell. Probes 1: 159-168 (1987) have discussed
`
`20
`
`sandwich hybridization in microtiter wells using cloned capture DNA adsorbed to the
`
`wells. An assay for detecting HN -1 DNA using PCR amplification and capture
`
`hybridization in microtiter wells also has been reported (Keller et al., J. Clin.
`
`Microbial. 29: 638-41 (1991)). The NaCl-mediated binding of oligomers to
`
`polystyrene wells has been discussed by Cros et al. (French Patent No. 2,663,040) and
`
`25
`
`by Nikiforov et al., PCR Methods Applic. 3: 285-291 (1994). A cationic
`
`detergent-mediated binding of oligomers to polystyrene wells has been described by
`
`Nikiforov et al., Nucleic Acids Res. 22: 4167-75 (1994).
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 008
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-8-
`
`III. Analysis Of Single Nucleotide DNA Polymorphisms
`
`Many genetic diseases and traits (i.e. hemophilia, sickle-cell anemia, cystic
`
`fibrosis, etc.) reflect the consequences of mutations that have arisen in the genomes of
`
`some members of a species through mutation or evolution (Gusella, Ann. Rev.
`
`5
`
`Biochem. 55: 831-54 (1986)). fu. some cases, suchpolymorphisms are linked to a
`
`genetic locus responsible for the disease or trait; in other cases, the polymorphisms are
`
`the determinative characteristic of the condition .
`
`.Single nucleotide polymorphisms (SNPs) differ significantly from the variable
`
`nucleotide type polymorphisms (VNTRs ), that arise from spontaneous tandem
`
`10
`
`duplications of di- or tri-nucleotide repeated motifs of nucleotides (Weber, U.S. Pat.
`
`No. 5,075,217; Armour et al., FEBS Lett. 307: 113-5 (1992); Hom et·al., PCT
`
`Application No. WO 91/14003; Moore et al., Genomics 10: 654-60 (1991); Hillel et
`
`al., Genet. 124: 783-9 (1990)), and from the restriction fragment length
`
`polymorphisms ("RFLPs") that comprise variations which alter the lengths of the
`
`15
`
`fragments that are generated by restriction endonuclease cleavage (e.g., Fischer et al.,
`
`(PCT Application No. WO 90/13668); and Uhlen (PCT Application No. WO
`
`90/11369)).
`
`Because SNPs constitute sites of variation flanked by regions of invariant
`
`sequence, their analysis requires no more than the determination of the identity of the
`
`20
`
`single nucleotide present at the site of variation; it is unnecessary to determine a
`
`complete gene sequence for each patient. Several methods have been developed to
`
`facilitate the analysis of such single nucleotide polymorphisms.
`
`Mundy, C. R. (U.S. Pat. No. 4,656, 127), for example, discusses a method for
`
`determining the identity of the nucleotide present at a particular polymorphic site that
`
`25
`
`employs a specialized exonuclease-resistant nucleotide derivative.
`
`Cohen et al. (French Patent 2,650,840; and PCT Application No. WO
`
`91/02087) discuss a solution-based method for determining the identity of the
`
`nucleotide of a polymorphic site. As in the Mundy method ofU.S. Patent. No.
`
`4,656,127, a primer is employed that is complementary to allelic sequences
`
`30
`
`immediately 3' to a polymorphic site.
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 009
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-9-
`
`Additional SNP detection methods include the Genetic Bit Analysis method
`
`described by Goelet et al. (PCT Application No. 92115712). The method ofGoelet et
`
`al. uses mixtures of labeled terminators and a primer that is complementary to the
`
`sequence 3' to a polymorphic site.
`
`5
`
`Cheesman (U.S. Pat. No. 5,302,509) describes a method for sequencing a
`
`single stranded DNA molecule using fluorescently labeled 3 '-blocked nucleotide
`
`triphosphates. An apparatus for the separation, concentration and detection of a DNA
`
`molecule in a liquid sample has been described by Ritterband et al. (PCT Patent
`
`Application No. WO 95/17676).
`
`10
`
`Several primer-guided nucleotide incorporation procedures for assaying
`
`polymorphic sites in DNA have been described (Prezant et al., Hum. Mutat. 1: 159-64
`
`(1992); Ugozzoli et al., GATA 9: 107-12 (1992); and Nyren et al., Anal. Biochem.
`
`208: 171-5 (1993)).
`
`15
`
`IV. Methods of Immobilizing Nucleic Acids to a Solid-Phase
`
`Several ofthe above-described methods involve procedures in which one or
`
`more of the nucleic acid reactants are immobilized to a solid support. Currently,
`
`96-well polystyrene plates are widely used in solid-phase immunoassays, PCR
`
`product detection methods that use plates as a solid support and DNA chips have been
`
`20
`
`described. The microtiter plate method requires the immobilization of a suitable
`
`oligonucleotide probe into the microtiter wells, followed by the capture ofthe PCR
`
`product by hybridization and colorimetric detection of a suitable hapten.
`
`Covalent disulfide bonds have been previously used to immobilize both
`
`proteins and oligonucleotides. Chu et al. (Nucl. Acids Res. 16: 3671-91 (1988))
`
`25
`
`discloses a method for coupling oligonucleotides to nucleic acids or proteins via
`
`cleavable disulfide bonds.
`
`Gentalen et al., Nucl. Acids Res. 27: 1485-91 (1999) describe a cooperative
`
`hybridization method to establish physical linkage between two loci on a DNA strand
`
`by using hybridization to a new type of high-density oligonucleotide array. This same
`
`30 method can be used to determine SNP haplotypes.
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 010
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-10-
`
`Yershov et al., Proc. Nat!. Acad. Sci. USA 93: 4913-8 (1996) describe an
`
`oligonucleotide microchip which has been used to detect beta-thalassemia mutations
`
`in patients by hybridizing PCR-amplified DNA with the microchips. This technology
`
`was suggested for large scale diagnostics in gene polymorphism studies.
`
`5
`
`Guo et al., Nucl. Acids Res. 22: 5456-65 (1994) describe a simple method for
`
`the analysis of genetic polymorphisms allele-specific oligonucleotide raised bound to
`
`glass supports. This method was demonstrated in parallel analysis of 5 point
`
`mutations from exon 4 of the human tyrosinase gene.
`
`More recently, Gilles et al., Nat. Biotechnol. 17: 365-70 (1999) have described
`
`10
`
`a rapid assay for SNP detection utilizing electronics circuitry on silicon microchips.
`
`Holloway et al., Hum. Mutat. 14: 340-7 (1999) also compares methods for high(cid:173)
`
`throughput SNP typing using TaqMan® liquid phase hybridization, PCR-SSOP or
`
`ARMS-microplate array diagonal gel electrophoresis (MADGE).
`
`As the world population ages and new drugs are identified, more and more
`
`15
`
`patients will administer one or more phannaceutical compositions, such that an
`
`individual drug or drug combination can cause adverse drug reactions. Therefore, not
`
`withstanding what has been previously reported in the literature, the inventor herein
`
`describes methods and compositions for diagnosing drug interactions which involve at
`
`least one mutation in a LQT gene. Additional mutations may exist in certain
`
`20
`
`cytochrome 450 genes and P-glycoprotein pumps, which work in concert with a LQT
`
`gene mutation or other ion chrumel (e.g., K+ or Nal gene polymorphisms to produce
`
`an adverse drug or drug-drug reaction. The specification also discloses kits and
`
`compositions for diagnosing a subject's predisposition to QT interval elongation in
`
`response to the administration of one or more pharmaceutical agents.
`
`25
`
`SUMMARY OF THE INVENTION
`
`It is an object of the invention to provide novel and improved methods for
`
`determining whether a subject has a predisposition for QT interval elongation or
`
`Torsades de Pointes due to one or more pharmaceutical agents. The methods
`
`30
`
`comprise the step of screening a biological sample from the subject through a nucleic
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 011
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-11-
`
`acid array, wherein said nucleic acid array contains probes for at least two genetic
`
`mutations or polymorphisms. These two genetic mutations or polymorphisms are
`
`located in two or more of the group of genes consisting of (1) LQT genes, (2) altered
`
`sensitivity genes, and (3) increased exposure genes. Preferred genes include LQT
`
`5
`
`genes andMDR genes (e.g., MDR-1). The nucleic acid array can be in the form of a
`
`chip, a microchip, a bead or a microsphere. The LQT gene which may contain a
`
`polymorphism which induces QT interval elongation include LQTI, LQT2, LQT3,
`
`LQT4, LQT5 and LQT6.
`
`The method may further comprise screening both LQT and increased exposure
`
`10
`
`gene (e.g., cytochrome P450 genes) mutations and polymorphisms. The P450
`
`cytochrome isoforms which may contain a mutation which can result in excessive
`
`accumulation of drugs and thereby induce QT interval elongation include: CYP IA2,
`
`CYP2C19, CYP2C9, CYP2D6, CYP2El, CYP3A, CYP3A5 and CYP3A7.
`
`A further object of the invention is to provide a method for determining
`
`15 whether a subject has a predisposition for QT interval elongation (e.g., acquired
`
`LQTS) when treated with one or more pharmaceutical agents comprising the step of
`
`screening a biological sample from the subject through a nucleic acid array, such as a
`
`DNA array. The DNA array contains probes for two or more genetic mutations or
`
`polymorphisms in at least two or more groups of genes wherein the genes are selected
`
`20
`
`from the group consisting of(1) LQT genes, (2) altered sensitivity genes (e.g., MiRP-
`
`1 genes and its related genes), and (3) increased exposure genes (e.g., multidrug
`
`resistant genes and cytochrome P450 genes). The two or more genetic mutations or
`
`polymorphisms are found in these genes as at least one or more genetic mutations or
`
`polymorphisms in each of the two or more groups of genes. The genes can be
`
`25
`
`selected from those described above.
`
`Another object of the invention is to provide a nucleic acid array comprising
`
`nucleic acids which recognize and bind to mutations of the QT syndrome genes (e.g.,
`
`LQT genes), the altered sensitivity genes (MirR-1 genes) and/or the increased
`
`exposure genes.
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 012
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-12-
`
`Another object of the invention is to provide a method of screening one or
`
`more pharmaceutical agents in vitro for its or their ability to induce prolonged cardiac
`
`repolarization of a cell comprising the steps of A) measuring IKr and IKs currents of the
`
`cell using a voltage clamp before superfusing the cell with a candidate agent or
`
`5
`
`agents; B) superfusing and incubating the cell with the candidate agent or agents; C)
`
`measuring the Ir<r and IKs currents after superfusion and incubation of the cell with the
`
`candidate. agent or agents using a voltage clamp; and D) determining whether the IKr
`
`and IKs currents are inhibited or abolished thereby indicating that the drug prolongs
`
`repolarization.
`
`10
`
`It is another object of the invention to provide a method for identifying genetic
`
`polymorphisms and mutations, which can cause QT interval prolongation in a subject
`
`comprising the steps of inserting at least two nucleic acids each encoding a
`
`polymorphism or mutation of at least two of the following genes: a LQT gene, an
`
`altered sensitivity gene, and/or an increased drug exposure gene into a cell; B)
`
`15 measuring IKr and IKs currents of the cell before administering a drug lmown to cause a
`
`change in IKr and/or IKs; C) measuring IKr and IKs currents of the cell after superfusion
`
`of the cell with the drug; D) comparing the IKr and IKs values of the cell expressing the
`
`polymorphisms and/or mutations to the IKr and IKs values of a cell expressing a wild(cid:173)
`
`type genes; and E) determining if the presence ofthe polymorphisms and/or mutations
`
`20
`
`leads to greater inhibition or blockage ofiKr and IKs currents in the cell expressing said
`
`polymorphism or polymorphisms.
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 013
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-13-
`
`BRIEF DESCRIPTION OF THE DRAWINGS
`
`Figure 1. Recordings oflKn Ito (A and B) and IK1 (C) in the same cell
`
`before and after 5 minutes superfusion with 5 flmol/L E-4031. Panel A) IKr and Ito
`
`currents before and after superfusion with E-4031. E-4031 abolished the IKr tail
`
`5
`
`current and also reduced the time-dependent IKr current without affecting the transient
`
`outward current Cito) or the holding current; Panel B) E-4031 sensitive currents
`
`obtained by digital subtraction of currents after E-4031 exposure from currents before
`
`E-4031 exposure. Note the inward rectification of the time-dependent IKr currents at
`
`10
`
`very positive potentials compared with the tail currents; Panel C) IK1 current before
`and after superfusion with E-4031. E-4031 showed little effect on the inward IK1
`current recorded at -120 mV. The outward holding currents that represent the
`
`amplitude ofk1 at -40 mV are superimposed.
`
`Figure 2. Recordings of IKr, Ito (A and B) and IK1 (C) in the same cell
`
`before and after 5 minutes superfusion with 10 flmol/L tamoxifen. Panel A) IKr
`
`15
`
`and Ito currents recorded before and after superfusion with tamoxifen. Tamoxifen
`
`abolished the IKr tail current and also reduced the time-dependent IKr current, without
`
`affecting the transient outward current Oto); Panel B) Tamoxifen-sensitive currents
`
`obtained by digital subtraction of currents before and after tamoxifen superfusion.
`
`Note the inward rectification of the time-dependent IKr currents at very positive
`
`20
`
`potentials compared with the tail currents and their similarity to the E-4031 sensitive
`
`currents; Panel C) IK1 current recorded before and after superfusion with tamoxifen.
`
`Tamoxifen showed no block ofiK1 inward current. The outward holding currents,
`which represent the an1plitude ofiK1 at -40 mV, were superimposed together.
`Figure 3. Time-dependent block of IKr by tamoxifen. IKr currents were
`
`25
`
`recorded in the same cell before drug administration, 3, 5 and 9 min. after superfusion
`
`with 1 J.Lmol/1 tamoxifen.
`
`Figure 4. Voltage- and concentration-dependent block of IKr by
`tamoxifen. IKr tail currents ~ere measured 5-7 min. after superfusion with tamoxifen.
`
`Panel A) Effect of 1 Jlmol/L I-V relationship; Panel B) Effect of3.3 J.Lmol/1
`
`Roxane Labs., Inc.
`Exhibit 1014
`Page 014
`
`

`
`wo 01/79554
`
`PCT/USOl/12087
`
`-14-
`
`tamoxifen on I-V (current-voltage) relationship. Data are expressed as mean± SD,
`
`n=4, *p<0.05.
`
`Figure 5. Comparison of IKr block by tamoxifen and quinidine. Panel A)
`
`IKr currents recorded from the same cell before drug administration, 5 minutes after
`
`5
`
`super:fusion with 10 )lmol/L tamoxifen and 5 minutes after washout. IKr tail currents
`
`were abolished by tamoxifen without recovery. Panel B) IKr currents recorded from
`
`another cell before dmg administration, 5 minutes after superfusion with 10 )lmol/L
`
`quinidine and 3 minutes after washout demonstrate that IKr tail currents were reduced
`
`but not abolished by quinidine, with partial recovery after 3 minutes washout. Panel
`
`10
`
`C) Inhibition ofiKr by 3.3 ~-tmol/L tamoxifen and 3.3 )lmol/L quinidine. Data are
`
`expressed as mean± SD, n=4, **p<O.Ol.
`
`Figure 6. Effect of tamoxifen on the action potential duration (APD).
`
`Action potentials were elicited by injecting 100 pA depolarizing currents of 2 ms, at a
`
`frequency of 0.45 HZ. Shown in the figure are two superimposed action potential
`
`15
`
`tracings recorded in a single ventricular myocyte before and 4 minutes after exposure
`
`to 3.3 )lmol/L tamoxifen. Th

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket